Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 7317, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795711

ABSTRACT

We previously showed that environmentally-induced epigenetic inheritance of cancer occurs in rodent models. For instance, we reported that paternal consumption of an obesity-inducing diet (OID) increased breast cancer susceptibility in the offspring (F1). Nevertheless, it is still unclear whether programming of breast cancer in daughters is due to systemic alterations or mammary epithelium-specific factors and whether the breast cancer predisposition in F1 progeny can be transmitted to subsequent generations. In this study, we show that mammary glands from F1 control (CO) female offspring exhibit enhanced growth when transplanted into OID females compared to CO mammary glands transplanted into CO females. Similarly, carcinogen-induced mammary tumors from F1 CO female offspring transplanted into OID females has a higher proliferation/apoptosis rate. Further, we show that granddaughters (F2) from the OID grand-paternal germline have accelerated tumor growth compared to CO granddaughters. This between-generation transmission of cancer predisposition is associated with changes in sperm tRNA fragments in OID males. Our findings indicate that systemic and mammary stromal alterations are significant contributors to programming of mammary development and likely cancer predisposition in OID daughters. Our data also show that breast cancer predisposition is transmitted to subsequent generations and may explain some familial cancers, if confirmed in humans.


Subject(s)
Epigenesis, Genetic , Fathers , Genetic Predisposition to Disease , Mammary Neoplasms, Animal/genetics , Obesity/physiopathology , Animals , Apoptosis , Area Under Curve , Body Weight , Cell Proliferation , Disease Models, Animal , Epigenome , Epigenomics , Family Health , Female , Glucose Tolerance Test , Male , Mammary Glands, Animal/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , RNA, Transfer/metabolism , RNA-Seq , Spermatozoa/metabolism
2.
Cell Death Discov ; 4: 40, 2018.
Article in English | MEDLINE | ID: mdl-30345078

ABSTRACT

The postnatal mammary gland undergoes repeated cycles of proliferation and cell death, most notably when the fully differentiated (lactating) gland dedifferentiates to a prelactation state. Accumulation of milk proteins in the secretory epithelium creates the stress signal that triggers this process (involution). How this stress is perceived, and the cellular processes that are subsequently activated, remain unclear. We now report that Unfolded Protein Response (UPR), autophagy, and apoptosis related genes cluster separately during lactation and involution in the mouse mammary gland. Time-course experiments in rodents show that autophagy and UPR signaling are tightly co-regulated at the transition from reversible to irreversible involution. Inhibition of autophagy by chloroquine or genetic deletion of one ATG7 allele enhanced progression of mammary involution into the irreversible phase, as characterized by an early/precocious induction of apoptosis. These are the first preclinical in vivo data in support of a clinical trial testing an autophagy inhibitor for prevention of intraductal breast malignancy progression to invasive breast cancer. In marked contrast, stimulation of autophagy by low dose tunicamycin treatment reduced apoptosis and extended the reversible phase of involution by sustaining the secretory epithelium. Autophagy stimulators could be used short-term to promote lactation in women experiencing difficulties or irregularities in nursing. Taken together, these data indicate that UPR and autophagy play a key role in regulating the balance between cell survival and apoptosis during normal mammary gland regression.

3.
Development ; 144(22): 4103-4113, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28947532

ABSTRACT

In the mammary gland, vimentin intermediate filaments are expressed in stromal cells and in basal epithelial cell populations, including gland-reconstituting mammary stem cells, with largely undefined functions. Here, we have studied how vimentin deficiency affects mouse mammary gland development. We find that, in adult vimentin knockout mice (Vim-/- ), mammary ductal outgrowth is delayed. The adult Vim-/- glands display dilated ducts and a reduced basal-to-luminal mouse mammary epithelial cell (MMEC) ratio indicative of altered progenitor cell activity. Accordingly, isolated Vim-/- MMECs form fewer mammospheres and basal-like organoids in vitro than their wild-type counterparts. Importantly, reduced basal MMEC number translates into defects in Vim-/- mammary gland regeneration in vivo Global gene expression profiling of basal MMECs reveals that lack of vimentin alters multiple pathways, including adhesion, cancer and Wnt signalling. Furthermore, vimentin contributes to stem-like cell properties in MDA-MB-231 breast cancer cells, wherein vimentin depletion reduces tumoursphere formation and attenuates expression of breast cancer stem cell-associated surface markers. Together, our findings identify vimentin as a positive regulator of stemness in the developing mouse mammary gland and in breast cancer cells.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Vimentin/metabolism , Animals , Cell Count , Cell Line, Tumor , Cell Proliferation , Epithelial Cells/cytology , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Silencing , Humans , Mammary Glands, Animal/cytology , Mice, Knockout , Organoids/metabolism , Regeneration , Spheroids, Cellular/pathology , Stem Cells/cytology , Stem Cells/metabolism , Stromal Cells/metabolism , Vimentin/deficiency
4.
Clin Cancer Res ; 23(3): 814-824, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-28148690

ABSTRACT

PURPOSE: Whether it is safe for estrogen receptor-positive (ER+) patients with breast cancer to consume soy isoflavone genistein remains controversial. We compared the effects of genistein intake mimicking either Asian (lifetime) or Caucasian (adulthood) intake patterns to that of starting its intake during tamoxifen therapy using a preclinical model. EXPERIMENTAL DESIGN: Female Sprague-Dawley rats were fed an AIN93G diet supplemented with 0 (control diet) or 500 ppm genistein from postnatal day 15 onward (lifetime genistein). Mammary tumors were induced with 7,12-dimethylbenz(a)anthracene (DMBA), after which a group of control diet-fed rats were switched to genistein diet (adult genistein). When the first tumor in a rat reached 1.4 cm in diameter, tamoxifen was added to the diet and a subset of previously only control diet-fed rats also started genistein intake (post-diagnosis genistein). RESULTS: Lifetime genistein intake reduced de novo resistance to tamoxifen, compared with post-diagnosis genistein groups. Risk of recurrence was lower both in the lifetime and in the adult genistein groups than in the post-diagnosis genistein group. We observed downregulation of unfolded protein response (UPR) and autophagy-related genes (GRP78, IRE1α, ATF4, and Beclin-1) and genes linked to immunosuppression (TGFß and Foxp3) and upregulation of cytotoxic T-cell marker CD8a in the tumors of the lifetime genistein group, compared with controls, post-diagnosis, and/or adult genistein groups. CONCLUSIONS: Genistein intake mimicking Asian consumption patterns improved response of mammary tumors to tamoxifen therapy, and this effect was linked to reduced activity of UPR and prosurvival autophagy signaling and increased antitumor immunity. Clin Cancer Res; 23(3); 814-24. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Estrogen Receptor Modulators/therapeutic use , Genistein/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Phytoestrogens/pharmacology , Soy Foods , Tamoxifen/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Antineoplastic Agents, Hormonal/pharmacology , Autophagy/drug effects , Autophagy/genetics , Cytokines/blood , Diet , Endoplasmic Reticulum Chaperone BiP , Estrogen Receptor Modulators/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Genistein/administration & dosage , Genistein/blood , Isoflavones/blood , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Phytoestrogens/administration & dosage , Phytoestrogens/blood , Rats , Rats, Sprague-Dawley , Recurrence , Soy Foods/adverse effects , Tamoxifen/pharmacology , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics
5.
EMBO J ; 36(2): 165-182, 2017 01 17.
Article in English | MEDLINE | ID: mdl-27974362

ABSTRACT

SHARPIN is a widely expressed multifunctional protein implicated in cancer, inflammation, linear ubiquitination and integrin activity inhibition; however, its contribution to epithelial homeostasis remains poorly understood. Here, we examined the role of SHARPIN in mammary gland development, a process strongly regulated by epithelial-stromal interactions. Mice lacking SHARPIN expression in all cells (Sharpincpdm), and mice with a stromal (S100a4-Cre) deletion of Sharpin, have reduced mammary ductal outgrowth during puberty. In contrast, Sharpincpdm mammary epithelial cells transplanted in vivo into wild-type stroma, fully repopulate the mammary gland fat pad, undergo unperturbed ductal outgrowth and terminal differentiation. Thus, SHARPIN is required in mammary gland stroma during development. Accordingly, stroma adjacent to invading mammary ducts of Sharpincpdm mice displayed reduced collagen arrangement and extracellular matrix (ECM) stiffness. Moreover, Sharpincpdm mammary gland stromal fibroblasts demonstrated defects in collagen fibre assembly, collagen contraction and degradation in vitro Together, these data imply that SHARPIN regulates the normal invasive mammary gland branching morphogenesis in an epithelial cell extrinsic manner by controlling the organisation of the stromal ECM.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Collagen/metabolism , Mammary Glands, Human/growth & development , Animals , Extracellular Matrix/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout
6.
J Natl Cancer Inst ; 109(1)2017 01.
Article in English | MEDLINE | ID: mdl-27609189

ABSTRACT

Background: Responses to endocrine therapies vary among patients with estrogen receptor (ER+) breast cancer. We studied whether in utero exposure to endocrine-disrupting compounds might explain these variations. Methods: We describe a novel ER+ breast cancer model to study de novo and acquired tamoxifen (TAM) resistance. Pregnant Sprague Dawley rats were exposed to 0 or 0.1 ppm ethinyl estradiol (EE2), and the response of 9,12-dimethylbenz[a]anthracene (DMBA)-induced mammary tumors to 15 mg/kg TAM, with (n = 17 tumors in the controls and n = 20 tumors in EE2 offspring) or without 1.2 g/kg valproic acid and 5 mg/kg hydralazine (n = 24 tumors in the controls and n = 32 tumors in EE2 offspring) in the female offspring, was assessed. One-sided Chi2 tests were used to calculate P values. Comparisons of differentially expressed genes between mammary tumors in in utero EE2-exposed and control rats, and between anti-estrogen-resistant LCC9 and -sensitive LCC1 human breast cancer cells, were also performed. Results: In our preclinical model, 54.2% of mammary tumors in the control rats exhibited a complete response to TAM, of which 23.1% acquired resistance with continued anti-estrogen treatment and recurred. Mammary tumors in the EE2 offspring were statistically significantly less likely to respond to TAM (P = .047) and recur (P = .007). In the EE2 offspring, but not in controls, adding valproic acid and hydralazine to TAM prevented recurrence (P < .001). Three downregulated and hypermethylated genes (KLF4, LGALS3, MICB) and one upregulated gene (ETV4) were identified in EE2 tumors and LCC9 breast cancer cells, and valproic acid and hydralazine normalized the altered expression of all four genes. Conclusions: Resistance to TAM may be preprogrammed by in utero exposure to high estrogen levels and mediated through reversible epigenetic alterations in genes associated with epithelial-mesenchymal transition and tumor immune responses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Ethinyl Estradiol/adverse effects , Mammary Neoplasms, Experimental/drug therapy , Neoplasm Recurrence, Local/genetics , Prenatal Exposure Delayed Effects/chemically induced , Tamoxifen/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Adenovirus E1A Proteins/genetics , Animals , Cell Line, Tumor , DNA Methylation , Drug Resistance, Neoplasm/drug effects , Female , Galectin 3/genetics , Gene Expression/drug effects , Gene Silencing , Histocompatibility Antigens Class I/genetics , Humans , Hydralazine/administration & dosage , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/chemistry , Neoplasm Recurrence, Local/prevention & control , Pregnancy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/analysis , Tamoxifen/administration & dosage , Trans-Activators/genetics , Valproic Acid/administration & dosage
7.
Cancer Res ; 76(19): 5657-5670, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27698188

ABSTRACT

The unfolded protein response is an endoplasmic reticulum stress pathway mediated by the protein chaperone glucose regulated-protein 78 (GRP78). Metabolic analysis of breast cancer cells shows that GRP78 silencing increases the intracellular concentrations of essential polyunsaturated fats, including linoleic acid. Accumulation of fatty acids is due to an inhibition of mitochondrial fatty acid transport, resulting in a reduction of fatty acid oxidation. These data suggest a novel role of GRP78-mediating cellular metabolism. We validated the effect of GRP78-regulated metabolite changes by treating tumor-bearing mice with tamoxifen and/or linoleic acid. Tumors treated with linoleic acid plus tamoxifen exhibited reduced tumor area and tumor weight. Inhibition of either GRP78 or linoleic acid treatment increased MCP-1 serum levels, decreased CD47 expression, and increased macrophage infiltration, suggesting a novel role for GRP78 in regulating innate immunity. GRP78 control of fatty acid oxidation may represent a new homeostatic function for GRP78. Cancer Res; 76(19); 5657-70. ©2016 AACR.


Subject(s)
Breast Neoplasms/metabolism , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/physiology , Lipid Metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , CD47 Antigen/physiology , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Fatty Acids/metabolism , Female , Humans , Immunity, Innate , Metabolomics , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism , Unfolded Protein Response
8.
Cancer Res ; 75(6): 1046-55, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25576084

ABSTRACT

Interferon regulatory factor-1 (IRF1) is a tumor suppressor that regulates cell fate in several cell types. Here, we report an inverse correlation in expression of nuclear IRF1 and the autophagy regulator ATG7 in human breast cancer cells that directly affects their cell fate. In mice harboring mutant Atg7, nuclear IRF1 was increased in mammary tumors, spleen, and kidney. Mechanistic investigations identified ATG7 and the cell death modulator beclin-1 (BECN1) as negative regulators of IRF1. Silencing ATG7 or BECN1 caused estrogen receptor-α to exit the nucleus at the time when IRF1 nuclear localization occurred. Conversely, silencing IRF1 promoted autophagy by increasing BECN1 and blunting IGF1 receptor and mTOR survival signaling. Loss of IRF1 promoted resistance to antiestrogens, whereas combined silencing of ATG7 and IRF1 restored sensitivity to these agents. Using a mathematical model to prompt signaling hypotheses, we developed evidence that ATG7 silencing could resensitize IRF1-attenuated cells to apoptosis through mechanisms that involve other estrogen-regulated genes. Overall, our work shows how inhibiting the autophagy proteins ATG7 and BECN1 can regulate IRF1-dependent and -independent signaling pathways in ways that engender a new therapeutic strategy to attack breast cancer.


Subject(s)
Apoptosis , Autophagy , Breast Neoplasms/pathology , Interferon Regulatory Factor-1/physiology , Signal Transduction/physiology , Animals , Apoptosis Regulatory Proteins/physiology , Autophagy-Related Protein 7 , Beclin-1 , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Lineage , Female , Humans , Membrane Proteins/physiology , Mice , Models, Theoretical , Ubiquitin-Activating Enzymes/physiology
9.
Mol Cell Biol ; 35(2): 379-90, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25368386

ABSTRACT

Antiestrogen therapy induces the unfolded protein response (UPR) in estrogen receptor-positive (ER(+)) breast cancer. X-box binding protein 1 (XBP1), which exists in the transcriptionally inactive unspliced form [XBP1(U)] and the spliced active form [XBP1(S)], is a key UPR component mediating antiestrogen resistance. We now show a direct link between the XBP1 and NF-κB survival pathways in driving the cell fate decisions in response to antiestrogens in ER(+) breast cancer cells, both in vitro and in a xenograft mouse model. Using novel spliced and nonspliceable forms of XBP1, we show that XBP1(U) functions beyond being a dominant negative of XBP1(S). Both isoforms regulate NF-κB activity via ERα; XBP1(S) is more potent because it also directly regulates p65/RelA expression. These findings provide new insights into the fundamental signaling activities of spliced and unspliced XBP1 in breast cancer, establish NF-κB to be a mediator of these activities, and identify XBP1 and its splicing to be novel therapeutic targets.


Subject(s)
Breast Neoplasms/metabolism , Cell Lineage/drug effects , DNA-Binding Proteins/drug effects , Estrogen Antagonists/pharmacology , NF-kappa B/drug effects , Tamoxifen/pharmacology , Transcription Factors/drug effects , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Female , Humans , Mice , NF-kappa B/metabolism , RNA Splicing/genetics , Receptors, Estrogen/metabolism , Regulatory Factor X Transcription Factors , Signal Transduction/drug effects , Signal Transduction/genetics , X-Box Binding Protein 1
10.
Mol Cancer ; 13: 239, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25339305

ABSTRACT

BACKGROUND: About 70% of all breast cancers are estrogen receptor alpha positive (ER+) and are treated with antiestrogens. However, 50% of ER + tumors develop resistance to these drugs (endocrine resistance). In endocrine resistant cells, an adaptive pathway called the unfolded protein response (UPR) is elevated that allows cells to tolerate stress more efficiently than in sensitive cells. While the precise mechanism remains unclear, the UPR can trigger both pro-survival and pro-death outcomes that depend on the nature and magnitude of the stress. In this study, we identified MYC, an oncoprotein that is upregulated in endocrine resistant breast cancer, as a regulator of the UPR in glucose-deprived conditions. METHODS: ER+ human breast cancer cell lines (LCC1, LCC1, LY2 and LCC9) and rat mammary tumors were used to confirm upregulation of MYC in endocrine resistance. To evaluate functional relevance of proteins, siRNA-mediated inhibition or small molecule inhibitors were used. Cell density/number was evaluated with crystal violet assay; cell cycle and apoptosis were measured by flow cytometry. Relative quantification of glutamine metabolites were determined by mass spectrometry. Signaling molecules of the UPR, apoptosis or autophagy pathways were investigated by western blotting. RESULTS: Increased MYC function in resistant cells correlated with increased dependency on glutamine and glucose for survival. Inhibition of MYC reduced cell growth and uptake of both glucose and glutamine in resistant cells. Interestingly, in glucose-deprived conditions, glutamine induced apoptosis and necrosis, arrested autophagy, and triggered the unfolded protein response (UPR) though GRP78-IRE1α with two possible outcomes: (i) inhibition of cell growth by JNK activation in most cells and, (ii) promotion of cell growth by spliced XBP1 in the minority of cells. These disparate effects are regulated, at different signaling junctions, by MYC more robustly in resistant cells. CONCLUSIONS: Endocrine resistant cells overexpress MYC and are better adapted to withstand periods of glucose deprivation and can use glutamine in the short term to maintain adequate metabolism to support cell survival. Our findings reveal a unique role for MYC in regulating cell fate through the UPR, and suggest that targeting glutamine metabolism may be a novel strategy in endocrine resistant breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogens/pharmacology , Glucose/metabolism , Glutamine/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Unfolded Protein Response/drug effects , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum Chaperone BiP , Estrogen Receptor Modulators/pharmacology , Female , Humans , Rats, Sprague-Dawley , Signal Transduction/drug effects , Up-Regulation/drug effects
11.
Clin Cancer Res ; 20(12): 3222-32, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24928945

ABSTRACT

PURPOSE: Estrogen receptor-α (ERα)-targeted therapies including tamoxifen (TAM) or Faslodex (ICI) are used to treat ER(+) breast cancers. Up to 50% of tumors will acquire resistance to these interventions. Autophagy has been implicated as a major driver of antiestrogen resistance. We have explored the ability of hydroxychloroquine (HCQ), which inhibits autophagy, to affect antiestrogen responsiveness. EXPERIMENTAL DESIGN: TAM-resistant MCF7-RR and ICI-resistant/TAM cross-resistant LCC9 ER(+) breast cancer cells were injected into mammary fat pads of female athymic mice and treated with TAM and/or ICI in combination with oral low-dose HCQ. RESULTS: We show that HCQ can increase antiestrogen responsiveness in MCF7-RR and LCC9 cells and tumors, likely through the inhibition of autophagy. However, the combination of ICI+HCQ was less effective than HCQ alone in vivo, unlike the TAM+HCQ combination. Antiestrogen treatment stimulated angiogenesis in tumors but did not prevent HCQ effectiveness. The lower efficacy of ICI+HCQ was associated with ICI effects on cell-mediated immunity within the tumor microenvironment. The mouse chemokine KC (CXCL1) and IFNγ were differentially regulated by both TAM and ICI treatments, suggesting a possible effect on macrophage development/activity. Consistent with these observations, TAM+HCQ treatment increased tumor CD68(+) cells infiltration, whereas ICI and ICI+HCQ reduced peripheral tumor macrophage content. Moreover, macrophage elimination of breast cancer target cells in vitro was reduced following exposure to ICI. CONCLUSION: HCQ restores antiestrogen sensitivity to resistant tumors. Moreover, the beneficial combination of TAM+HCQ suggests a positive outcome for ongoing neoadjuvant clinical trials using this combination for the treatment of ER(+) ductal carcinoma in situ lesions.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Estradiol/analogs & derivatives , Estrogen Receptor Modulators/pharmacology , Hydroxychloroquine/pharmacology , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Humans , Immunoenzyme Techniques , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
FASEB J ; 28(9): 3891-905, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24858277

ABSTRACT

Approximately 70% of all newly diagnosed breast cancers express estrogen receptor (ER)-α. Although inhibiting ER action using targeted therapies such as fulvestrant (ICI) is often effective, later emergence of antiestrogen resistance limits clinical use. We used antiestrogen-sensitive and -resistant cells to determine the effect of antiestrogens/ERα on regulating autophagy and unfolded protein response (UPR) signaling. Knockdown of ERα significantly increased the sensitivity of LCC1 cells (sensitive) and also resensitized LCC9 cells (resistant) to antiestrogen drugs. Interestingly, ERα knockdown, but not ICI, reduced nuclear factor (erythroid-derived 2)-like (NRF)-2 (UPR-induced antioxidant protein) and increased cytosolic kelch-like ECH-associated protein (KEAP)-1 (NRF2 inhibitor), consistent with the observed increase in ROS production. Furthermore, autophagy induction by antiestrogens was prosurvival but did not prevent ERα knockdown-mediated death. We built a novel mathematical model to elucidate the interactions among UPR, autophagy, ER signaling, and ROS regulation of breast cancer cell survival. The experimentally validated mathematical model explains the counterintuitive result that knocking down the main target of ICI (ERα) increased the effectiveness of ICI. Specifically, the model indicated that ERα is no longer present in excess and that the effect on proliferation from further reductions in its level by ICI cannot be compensated for by increased autophagy. The stimulation of signaling that can confer resistance suggests that combining autophagy or UPR inhibitors with antiestrogens would reduce the development of resistance in some breast cancers.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Unfolded Protein Response/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Cell Proliferation , Drug Resistance, Neoplasm/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Flow Cytometry , Fulvestrant , Humans , Mice , Mice, Nude , Microscopy, Confocal , Models, Theoretical , Oxidative Stress/drug effects , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Dev Biol ; 391(1): 66-80, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24726525

ABSTRACT

To understand the functional role of the peroxisomal membrane channel Pxmp2, mice with a targeted disruption of the Pxmp2 gene were generated. These mice were viable, grew and bred normally. However, Pxmp2(-/-) female mice were unable to nurse their pups. Lactating mammary gland epithelium displayed secretory lipid droplets and milk proteins, but the size of the ductal system was greatly reduced. Examination of mammary gland development revealed that retarded mammary ductal outgrowth was due to reduced proliferation of epithelial cells during puberty. Transplantation experiments established the Pxmp2(-/-) mammary stroma as a tissue responsible for suppression of epithelial growth. Morphological and biochemical examination confirmed the presence of peroxisomes in the mammary fat pad adipocytes, and functional Pxmp2 was detected in the stroma of wild-type mammary glands. Deletion of Pxmp2 led to an elevation in the expression of peroxisomal proteins in the mammary fat pad but not in liver or kidney of transgenic mice. Lipidomics of Pxmp2(-/-)mammary fat pad showed a decrease in the content of myristic acid (C14), a principal substrate for protein myristoylation and a potential peroxisomal ß-oxidation product. Analysis of complex lipids revealed a reduced concentration of a variety of diacylglycerols and phospholipids containing mostly polyunsaturated fatty acids that may be caused by activation of lipid peroxidation. However, an antioxidant-containing diet did not stimulate mammary epithelial proliferation in Pxmp2(-/-) mice. The results point to disturbances of lipid metabolism in the mammary fat pad that in turn may result in abnormal epithelial growth. The work reveals impaired mammary gland development as a new category of peroxisomal disorders.


Subject(s)
Lipid Metabolism , Mammary Glands, Animal/metabolism , Membrane Proteins/genetics , Membrane Proteins/physiology , Peroxisomes/metabolism , Adipose Tissue/metabolism , Animals , Bile Acids and Salts/chemistry , Epithelial Cells/cytology , Fatty Acids/chemistry , Female , Homeostasis , Lactation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Polyamines/chemistry , Subcellular Fractions , Time Factors
14.
J Mammary Gland Biol Neoplasia ; 18(1): 25-42, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23392570

ABSTRACT

Women are using estrogens for many purposes, such as to prevent pregnancy or miscarriage, or to treat menopausal symptoms. Estrogens also have been used to treat breast cancer which seems puzzling, since there is convincing evidence to support a link between high lifetime estrogen exposure and increased breast cancer risk. In this review, we discuss the findings that maternal exposure to the synthetic estrogen diethylstilbestrol during pregnancy increases breast cancer risk in both exposed mothers and their daughters. In addition, we review data regarding the use of estrogens in oral contraceptives and as postmenopausal hormone therapy and discuss the opposing effects on breast cancer risk based upon timing of exposure. We place particular emphasis on studies investigating how maternal estrogenic exposures during pregnancy increase breast cancer risk among daughters. New data suggest that these exposures induce epigenetic modifications in the mammary gland and germ cells, thereby causing an inheritable increase in breast cancer risk for multiple generations.


Subject(s)
Aging , Breast Neoplasms/chemically induced , Endocrine Disruptors/toxicity , Estradiol Congeners/adverse effects , Mammary Glands, Human/drug effects , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Carcinogens/toxicity , Contraceptives, Oral, Hormonal/adverse effects , Diethylstilbestrol/adverse effects , Environmental Exposure , Epigenesis, Genetic/drug effects , Estradiol Congeners/therapeutic use , Estrogen Replacement Therapy/adverse effects , Estrogens, Non-Steroidal/adverse effects , Female , Fetal Development/drug effects , Humans , Mammary Glands, Animal/drug effects , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Maternal Exposure/adverse effects , Pregnancy , Risk
15.
Mol Cancer Ther ; 12(4): 448-59, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23395885

ABSTRACT

In estrogen receptor-positive (ER+) breast cancer cells, BCL2 overexpression contributes to antiestrogen resistance. Direct targeting of the antiapoptotic BCL2 members with GX15-070 (obatoclax), a BH3-mimetic currently in clinical development, is an attractive strategy to overcome antiestrogen resistance in some breast cancers. Recently, GX15-070 has been shown to induce both apoptosis and autophagy, yet the underlying cell death mechanisms have yet to be elucidated. Here, we show that GX15-070 is more effective in reducing the cell density of antiestrogen-resistant breast cancer cells versus sensitive cells and that this increased sensitivity of resistant cells to GX15-070 correlates with an accumulation of autophagic vacuoles. Formation of autophagosomes in GX15-070-treated cells was verified by changes in expression of the lipidation of microtubule-associated protein-1 light chain-3 and both confocal and transmission electron microscopy. While GX15-070 treatment promotes autophagic vacuole and autolysosome formation, p62/SQSTM1, a marker for autophagic degradation, levels accumulate. Moreover, GX15-070 exposure leads to a reduction in cathepsin D (CTSD) and L (CTSL1) protein expression that would otherwise digest autolysosome cargo. Thus, GX15-070 has dual roles in promoting cell death: (i) directly inhibiting antiapoptotic BCL2 family members, thereby inducing apoptosis; and (ii) inhibiting downstream CTSD and CTSL1 protein expression to limit the ability of cells to use degraded material to fuel cellular metabolism and restore homeostasis. Our data highlight a new mechanism of GX15-070-induced cell death that could be used to design novel therapeutic interventions for antiestrogen resistant breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Cathepsin D/metabolism , Cathepsin L/metabolism , Estrogen Antagonists/pharmacology , Pyrroles/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Beclin-1 , Breast Neoplasms/genetics , Cathepsin D/antagonists & inhibitors , Cathepsin L/antagonists & inhibitors , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Indoles , Lysosomes/metabolism , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Phagosomes/metabolism , Pyrroles/chemistry , Pyrroles/toxicity , Sequestosome-1 Protein , Transplantation, Heterologous
16.
Nat Commun ; 3: 1053, 2012.
Article in English | MEDLINE | ID: mdl-22968699

ABSTRACT

Maternal exposures to environmental factors during pregnancy influence the risk of many chronic adult-onset diseases in the offspring. Here we investigate whether feeding pregnant rats a high-fat (HF)- or ethinyl-oestradiol (EE2)-supplemented diet affects carcinogen-induced mammary cancer risk in daughters, granddaughters and great-granddaughters. We show that mammary tumourigenesis is higher in daughters and granddaughters of HF rat dams and in daughters and great-granddaughters of EE2 rat dams. Outcross experiments suggest that the increase in mammary cancer risk is transmitted to HF granddaughters equally through the female or male germ lines, but it is only transmitted to EE2 granddaughters through the female germ line. The effects of maternal EE2 exposure on offspring's mammary cancer risk are associated with changes in the DNA methylation machinery and methylation patterns in mammary tissue of all three EE2 generations. We conclude that dietary and oestrogenic exposures in pregnancy increase breast cancer risk in multiple generations of offspring, possibly through epigenetic means.


Subject(s)
Dietary Fats/adverse effects , Ethinyl Estradiol/adverse effects , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/etiology , Maternal Exposure/adverse effects , Animals , Female , Mammary Glands, Animal/pathology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley
17.
Cancer Res ; 72(13): 3337-49, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22752300

ABSTRACT

While more than 70% of breast cancers express estrogen receptor-α (ER+), endocrine therapies targeting these receptors often fail. The molecular mechanisms that underlie treatment resistance remain unclear. We investigated the potential role of glucose-regulated protein 78 (GRP78) in mediating estrogen resistance. Human breast tumors showed increased GRP78 expression when compared with normal breast tissues. However, GRP78 expression was reduced in ER+ breast tumors compared with HER2-amplifed or triple-negative breast tumors. ER+ antiestrogen-resistant cells and ER+ tumors with an acquired resistant antiestrogen phenotype were both shown to overexpress GRP78, which was not observed in cases of de novo resistance. Knockdown of GRP78 restored antiestrogen sensitivity in resistant cells, and overexpression of GRP78 promoted resistance in sensitive cells. Mechanistically, GRP78 integrated multiple cellular signaling pathways to inhibit apoptosis and stimulate prosurvival autophagy, which was dependent on TSC2/AMPK-mediated mTOR inhibition but not on beclin-1. Inhibition of autophagy prevented GRP78-mediated endocrine resistance, whereas caspase inhibition abrogated the resensitization that resulted from GRP78 loss. Simultaneous knockdown of GRP78 and beclin-1 synergistically restored antiestrogen sensitivity in resistant cells. Together, our findings reveal a novel role for GRP78 in the integration of cellular signaling pathways including the unfolded protein response, apoptosis, and autophagy to determine cell fate in response to antiestrogen therapy.


Subject(s)
Apoptosis , Autophagy , Estrogen Receptor Modulators/pharmacology , Heat-Shock Proteins/physiology , Animals , Endoplasmic Reticulum Chaperone BiP , Female , Humans , Rats , Rats, Sprague-Dawley
18.
Mol Cancer Res ; 10(5): 597-604, 2012 May.
Article in English | MEDLINE | ID: mdl-22522458

ABSTRACT

TGF-ß regulates several steps in cancer metastasis, including the establishment of bone metastatic lesions. TGF-ß is released from bone during osteoclastic bone resorption and it stimulates breast cancer cells to produce osteolytic factors such as interleukin 11 (IL-11). We conducted a cell-based siRNA screen and identified heparan sulfate 6-O-sulfotransferase 2 (HS6ST2) as a critical gene for TGF-ß-induced IL-11 production in highly bone metastatic MDA-MB-231(SA) breast cancer cells. HS6ST2 attaches sulfate groups to glucosamine residues in heparan sulfate glycosaminoglycans. We subsequently showed how heparin and a high-molecular-weight Escherichia coli K5-derived heparin-like polysaccharide (K5-NSOS) inhibited TGF-ß-induced IL-11 production in MDA-MB-231(SA) cells. In addition, K5-NSOS inhibited bone resorption activity of human osteoclasts in vitro. We evaluated the therapeutic potential of K5-NSOS and fragmin in a mouse model of breast cancer bone metastasis. MDA-MB-231(SA) cells were inoculated into the left cardiac ventricle of athymic nude mice which were treated with fragmin, K5-NSOS, or vehicle once a day for four weeks. Both heparin-like glycosaminoglycans inhibited weight reduction, decreased osteolytic lesion area, and reduced tumor burden in bone. In conclusion, our data imply novel mechanisms involved in TGF-ß induction and support the critical role of heparan sulfate glycosaminoglycans in cancer metastasis as well as indicate that K5-NSOS is a potential antimetastatic and antiresorptive agent for cancer therapy. This study illustrates the potential to translate in vitro siRNA screening results toward in vivo therapeutic concepts.


Subject(s)
Bacterial Capsules , Heparin , Osteoclasts/drug effects , Sulfotransferases , Animals , Bacterial Capsules/chemistry , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Bone Resorption/genetics , Bone Resorption/pathology , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Heparin/chemistry , Heparin/pharmacology , Humans , Interleukin-11/antagonists & inhibitors , Interleukin-11/metabolism , Mice , Mice, Nude , RNA, Small Interfering , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/genetics , Sulfotransferases/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
19.
Cancer Res ; 72(6): 1321-31, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22422988

ABSTRACT

How breast cancer cells respond to the stress of endocrine therapies determines whether they will acquire a resistant phenotype or execute a cell-death pathway. After a survival signal is successfully executed, a cell must decide whether it should replicate. How these cell-fate decisions are regulated is unclear, but evidence suggests that the signals that determine these outcomes are highly integrated. Central to the final cell-fate decision is signaling from the unfolded protein response, which can be activated following the sensing of stress within the endoplasmic reticulum. The duration of the response to stress is partly mediated by the duration of inositol-requiring enzyme-1 activation following its release from heat shock protein A5. The resulting signals appear to use several B-cell lymphoma-2 family members to both suppress apoptosis and activate autophagy. Changes in metabolism induced by cellular stress are key components of this regulatory system, and further adaptation of the metabolome is affected in response to stress. Here we describe the unfolded protein response, autophagy, and apoptosis, and how the regulation of these processes is integrated. Central topologic features of the signaling network that integrate cell-fate regulation and decision execution are discussed.


Subject(s)
Autophagy , Breast Neoplasms/metabolism , Carcinoma/metabolism , Endoplasmic Reticulum Stress , Unfolded Protein Response , Animals , Apoptosis , Female , Humans , Mice , Signal Transduction , Tumor Microenvironment
20.
J Agric Food Chem ; 60(23): 5693-708, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22300613

ABSTRACT

Breast cancer is the most commonly diagnosed cancer among women worldwide. Many women have become more aware of the benefits of increasing fruit consumption, as part of a healthy lifestyle, for the prevention of cancer. The mechanisms by which fruits, including berries, prevent breast cancer can be partially explained by exploring their interactions with pathways known to influence cell proliferation and evasion of cell-death. Two receptor pathways, estrogen receptor (ER) and tyrosine kinase receptors, especially the epidermal growth factor receptor (EGFR) family, are drivers of cell proliferation and play a significant role in the development of both primary and recurrent breast cancer. There is strong evidence to show that several phytochemicals present in berries such as cyanidin, delphinidin, quercetin, kaempferol, ellagic acid, resveratrol, and pterostilbene interact with and alter the effects of these pathways. Furthermore, they also induce cell death (apoptosis and autophagy) via their influence on kinase signaling. This review summarizes in vitro data regarding the interaction of berry polyphenols with the specific receptors and the mechanisms by which they induce cell death. This paper also presents in vivo data of primary breast cancer prevention by individual compounds and whole berries. Finally, a possible role for berries and berry compounds in the prevention of breast cancer and a perspective on the areas that require further research are presented.


Subject(s)
Fruit/chemistry , Polyphenols/pharmacology , Signal Transduction/drug effects , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Polyphenols/blood , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...