Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Viruses ; 16(4)2024 04 05.
Article in English | MEDLINE | ID: mdl-38675909

ABSTRACT

Adjuvant systemic therapies effectively reduce the risk of breast cancer recurrence and metastasis, but therapy resistance can develop in some patients due to breast cancer stem cells (BCSCs). Oncolytic adenovirus (OAd) represents a promising therapeutic approach as it can specifically target cancer cells. However, its potential to target BCSCs remains unclear. Here, we evaluated a Cox-2 promoter-controlled, Ad5/3 fiber-modified OAd designed to encode the human sodium iodide symporter (hNIS) in breast cancer models. To confirm the potential of OAds to target BCSCs, we employed BCSC-enriched estrogen receptor-positive (ER+) paclitaxel-resistant (TaxR) cells and tumorsphere assays. OAd-hNIS demonstrated significantly enhanced binding and superior oncolysis in breast cancer cells, including ER+ cells, while exhibiting no activity in normal mammary epithelial cells. We observed improved NIS expression as the result of adenovirus death protein deletion. OAd-hNIS demonstrated efficacy in targeting TaxR BCSCs, exhibiting superior killing and hNIS expression compared to the parental cells. Our vector was capable of inhibiting tumorsphere formation upon early infection and reversing paclitaxel resistance in TaxR cells. Importantly, OAd-hNIS also destroyed already formed tumorspheres seven days after their initiation. Overall, our findings highlight the promise of OAd-hNIS as a potential tool for studying and targeting ER+ breast cancer recurrence and metastasis.


Subject(s)
Adenoviridae , Breast Neoplasms , Drug Resistance, Neoplasm , Neoplastic Stem Cells , Oncolytic Virotherapy , Oncolytic Viruses , Paclitaxel , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Breast Neoplasms/therapy , Breast Neoplasms/drug therapy , Paclitaxel/pharmacology , Adenoviridae/genetics , Adenoviridae/physiology , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Oncolytic Virotherapy/methods , Female , Cell Line, Tumor , Animals , Mice , Symporters/metabolism , Symporters/genetics , Genetic Vectors/genetics
3.
Essays Biochem ; 65(6): 971-983, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34132331

ABSTRACT

The classification and treatment of breast cancer is largely defined by the expression of steroid hormone receptors (HRs), namely estrogen receptor (ER) and progesterone receptor (PR), and gene amplification/overexpression of human epidermal growth factor receptor 2 (HER2). More recently, studies of androgen receptor (AR), glucocorticoid receptor (GR), and mineralocorticoid receptor (MR) have revealed that targeting these related HRs may be a promising strategy for a more personalized approach to the treatment of specific subtypes of HR+ breast cancer. For example, GR expression is associated with a good prognosis in ER+ breast cancer, but predicts poor prognosis in triple-negative breast cancer (TNBC). GR, like ER, PRs, and AR, is a ligand-activated transcription factor, but also has significant ligand-independent signaling activities. GR transcriptional activity is classically regulated by circulating glucocorticoids (GCs; ligand-dependent). Recent studies demonstrate that GR transcriptional activity is also regulated by a variety of cellular stress stimuli that input to GR Ser134 phosphorylation via rapid activation of the p38 mitogen activated protein kinase (MAPK) signaling pathway (ligand-independent). Furthermore, ligand-independent GR activation promotes feedforward signaling loops that mediate sustained activation of stress signaling pathways to drive advanced cancer biology (i.e. migration, invasion, chemoresistance, survival, and cellular growth). In this review, we will focus on the role of GR as a key sensor and mediator of physiologic and tumor microenvironment (TME)-derived cellular stress signaling in TNBC and discuss how targeting GR and/or associated signaling pathways may provide a strategy to inhibit deadly TNBC progression.


Subject(s)
Receptors, Glucocorticoid , Triple Negative Breast Neoplasms , Humans , Phosphorylation , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
4.
Oncogene ; 40(25): 4384-4397, 2021 06.
Article in English | MEDLINE | ID: mdl-34103681

ABSTRACT

Recurrence of metastatic breast cancer stemming from acquired endocrine and chemotherapy resistance remains a health burden for women with luminal (ER+) breast cancer. Disseminated ER+ tumor cells can remain viable but quiescent for years to decades. Contributing factors to metastatic spread include the maintenance and expansion of breast cancer stem cells (CSCs). Breast CSCs frequently exist as a minority population in therapy resistant tumors. In this study, we show that cytoplasmic complexes composed of steroid receptor (SR) co-activators, PELP1 and SRC-3, modulate breast CSC expansion through upregulation of the HIF-activated metabolic target genes PFKFB3 and PFKFB4. Seahorse metabolic assays demonstrated that cytoplasmic PELP1 influences cellular metabolism by increasing both glycolysis and mitochondrial respiration. PELP1 interacts with PFKFB3 and PFKFB4 proteins, and inhibition of PFKFB3 and PFKFB4 kinase activity blocks PELP1-induced tumorspheres and protein-protein interactions with SRC-3. PFKFB4 knockdown inhibited in vivo emergence of circulating tumor cell (CTC) populations in mammary intraductal (MIND) models. Application of PFKFB inhibitors in combination with ER targeted therapies blocked tumorsphere formation in multiple models of advanced breast cancer including tamoxifen (TamR) and paclitaxel (TaxR) resistant models, murine tumor cells, and ER+ patient-derived organoids (PDxO). Together, our data suggest that PELP1, SRC-3, and PFKFBs cooperate to drive ER+ tumor cell populations that include CSCs and CTCs. Identifying non-ER pharmacological targets offers a useful approach to blocking metastatic escape from standard of care ER/estrogen (E2)-targeted strategies to overcome endocrine and chemotherapy resistance.


Subject(s)
Breast Neoplasms/genetics , Co-Repressor Proteins/genetics , Drug Resistance, Neoplasm/genetics , Nuclear Receptor Coactivator 3/genetics , Phosphofructokinase-2/genetics , Receptors, Estrogen/genetics , Transcription Factors/genetics , Animals , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Estrogens/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Paclitaxel/pharmacology , Phosphorylation/genetics , Tamoxifen/pharmacology , Up-Regulation/genetics
5.
J Mol Endocrinol ; 65(1): T35-T48, 2020 07.
Article in English | MEDLINE | ID: mdl-32209723

ABSTRACT

Steroid hormone receptors (SRs) are classically defined as ligand-activated transcription factors that function as master regulators of gene programs important for a wide range of processes governing adult physiology, development, and cell or tissue homeostasis. A second function of SRs includes the ability to activate cytoplasmic signaling pathways. Estrogen (ER), androgen (AR), and progesterone (PR) receptors bind directly to membrane-associated signaling molecules including mitogenic protein kinases (i.e. c-SRC and AKT), G-proteins, and ion channels to mediate context-dependent actions via rapid activation of downstream signaling pathways. In addition to making direct contact with diverse signaling molecules, SRs are further fully integrated with signaling pathways by virtue of their N-terminal phosphorylation sites that act as regulatory hot-spots capable of sensing the signaling milieu. In particular, ER, AR, PR, and closely related glucocorticoid receptors (GR) share the property of accepting (i.e. sensing) ligand-independent phosphorylation events by proline-directed kinases in the MAPK and CDK families. These signaling inputs act as a 'second ligand' that dramatically impacts cell fate. In the face of drugs that reliably target SR ligand-binding domains to block uncontrolled cancer growth, ligand-independent post-translational modifications guide changes in cell fate that confer increased survival, EMT, migration/invasion, stemness properties, and therapy resistance of non-proliferating SR+ cancer cell subpopulations. The focus of this review is on MAPK pathways in the regulation of SR+ cancer cell fate. MAPK-dependent phosphorylation of PR (Ser294) and GR (Ser134) will primarily be discussed in light of the need to target changes in breast cancer cell fate as part of modernized combination therapies.


Subject(s)
Breast Neoplasms/enzymology , MAP Kinase Signaling System , Progesterone/metabolism , Receptors, Steroid/metabolism , Female , Humans , Phosphorylation , Protein Processing, Post-Translational , Receptors, Steroid/chemistry
6.
Mol Cancer Res ; 16(11): 1761-1772, 2018 11.
Article in English | MEDLINE | ID: mdl-29991529

ABSTRACT

The metastatic cascade is a complex process that requires cancer cells to survive despite conditions of high physiologic stress. Previously, cooperation between the glucocorticoid receptor (GR) and hypoxia-inducible factors (HIF) was reported as a point of convergence for host and cellular stress signaling. These studies indicated p38 MAPK-dependent phosphorylation of GR on Ser134 and subsequent p-GR/HIF-dependent induction of breast tumor kinase (PTK6/Brk), as a mediator of aggressive cancer phenotypes. Herein, p-Ser134 GR was quantified in human primary breast tumors (n = 281) and the levels of p-GR were increased in triple-negative breast cancer (TNBC) relative to luminal breast cancer. Brk was robustly induced following exposure of TNBC model systems to chemotherapeutic agents (Taxol or 5-fluorouracil) and growth in suspension [ultra-low attachment (ULA)]. Notably, both Taxol and ULA resulted in upregulation of the Aryl hydrocarbon receptor (AhR), a known mediator of cancer prosurvival phenotypes. Mechanistically, AhR and GR copurified and following chemotherapy and ULA, these factors assembled at the Brk promoter and induced Brk expression in an HIF-dependent manner. Furthermore, Brk expression was upregulated in Taxol-resistant breast cancer (MCF-7) models. Ultimately, Brk was critical for TNBC cell proliferation and survival during Taxol treatment and in the context of ULA as well as for basal cancer cell migration, acquired biological phenotypes that enable cancer cells to successfully complete the metastatic cascade. These studies nominate AhR as a p-GR binding partner and reveal ways to target epigenetic events such as adaptive and stress-induced acquisition of cancer skill sets required for metastatic cancer spread.Implication: Breast cancer cells enlist intracellular stress response pathways that evade chemotherapy by increasing cancer cell survival and promoting migratory phenotypes. Mol Cancer Res; 16(11); 1761-72. ©2018 AACR.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1/metabolism , Neoplasm Proteins/metabolism , Paclitaxel/pharmacology , Protein-Tyrosine Kinases/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Glucocorticoid/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Gene Expression , Humans , MCF-7 Cells , Phenotype , Phosphorylation , Signal Transduction/drug effects , Transfection , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
7.
Mol Cancer Res ; 16(4): 707-719, 2018 04.
Article in English | MEDLINE | ID: mdl-29348189

ABSTRACT

Proline, glutamic acid, leucine-rich protein 1 (PELP1) is overexpressed in approximately 80% of invasive breast tumors. PELP1 dynamically shuttles between the nucleus and cytoplasm, but is primarily nuclear in normal breast tissue. However, altered localization of PELP1 to the cytoplasm is an oncogenic event that promotes breast cancer initiation and progression. Herein, interacting partners unique to cytoplasmic PELP1 and the mechanisms by which these interactions promote oncogenic PELP1 signaling were sought. AIB1 (amplified in breast cancer 1; also known as SRC-3 or NCOA3) was identified as a novel binding partner of cytoplasmic PELP1 in both estrogen receptor-positive (ER+) and ER-negative cell lines. Cytoplasmic PELP1 expression elevated basal phosphorylation levels (i.e., activation) of AIB1 at Thr24, enhanced ALDH+ tumorsphere formation, and upregulated specific target genes independently of hormone stimulation. Direct manipulation of AIB1 levels using shRNA abrogated cytoplasmic PELP1-induced tumorsphere formation and downregulated cytoplasmic PELP1-specific target genes. SI-2, an AIB1 inhibitor, limited the PELP1/AIB1 interaction and decreased cytoplasmic PELP1-induced tumorsphere formation. Similar results were observed in a murine-derived MMTV-AIB1 tumor cell line. Furthermore, in vivo syngeneic tumor studies revealed that PELP1 knockdown resulted in increased survival of tumor-bearing mice as compared with mice injected with control cells.Implications: These data demonstrate that cytoplasmic PELP1/AIB1-containing complexes function to promote advanced cancer phenotypes, including outgrowth of stem-like cells, associated with estrogen-independent breast cancer progression. Mol Cancer Res; 16(4); 707-19. ©2018 AACR.


Subject(s)
Breast Neoplasms/metabolism , Co-Repressor Proteins/metabolism , Cytoplasm/metabolism , Neoplastic Stem Cells/metabolism , Nuclear Receptor Coactivator 3/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cells, Cultured , Co-Repressor Proteins/genetics , Cytoplasm/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Models, Biological , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Phosphorylation , Transcription Factors/genetics
9.
J Biol Chem ; 292(1): 339-350, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27881676

ABSTRACT

Cytoplasmic localization of proline, glutamic acid, leucine-rich protein 1 (PELP1) is observed in ∼40% of women with invasive breast cancer. In mouse models, PELP1 overexpression in the mammary gland leads to premalignant lesions and eventually mammary tumors. In preliminary clinical studies, cytoplasmic localization of PELP1 was seen in 36% of women at high risk of developing breast cancer. Here, we investigated whether cytoplasmic PELP1 signaling promotes breast cancer initiation in models of immortalized human mammary epithelial cells (HMECs). Global gene expression analysis was performed on HMEC lines expressing vector control, PELP1-wt, or mutant PELP1 in which the nuclear localization sequence was altered, resulting in cytoplasmic localization of PELP1 (PELP1-cyto). Global gene expression analysis identified that PELP1-cyto expression in HMECs induced NF-κB signaling pathways. Western blotting analysis of PELP1-cyto HMECs showed up-regulation of inhibitor of κB kinase ϵ (IKKϵ) and increased phosphorylation of the NF-κB subunit RelB. To determine whether secreted factors produced by PELP1-cyto HMECs promote macrophage activation, THP-1 macrophages were treated with HMEC-conditioned medium (CM). PELP1-cyto CM induced changes in THP-1 gene expression as compared with control cell CM. Double conditioned medium (DCM) from the activated THP-1 cells was then applied to HMECs to determine whether paracrine signaling from PELP1-cyto-activated macrophages could in turn promote migration of HMECs. PELP1-cyto DCM induced robust HMEC migration, which was reduced in DCM from PELP1-cyto HMECs expressing IKKϵ shRNA. Our findings suggest that cytoplasmic localization of PELP1 up-regulates pro-tumorigenic IKKϵ and secreted inflammatory signals, which through paracrine macrophage activation regulates the migratory phenotype associated with breast cancer initiation.


Subject(s)
Breast/pathology , Cell Movement , Co-Repressor Proteins/metabolism , Cytoplasm/metabolism , Epithelial Cells/pathology , I-kappa B Kinase/metabolism , Inflammation/pathology , Macrophages/pathology , Transcription Factors/metabolism , Animals , Breast/metabolism , Co-Repressor Proteins/genetics , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , I-kappa B Kinase/genetics , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Signal Transduction , Transcription Factors/genetics , Up-Regulation
10.
J Mol Endocrinol ; 56(3): R99-R114, 2016 04.
Article in English | MEDLINE | ID: mdl-26831511

ABSTRACT

Steroid hormone receptors (SRs) are heavily posttranslationally modified by the reversible addition of a variety of molecular moieties, including phosphorylation, acetylation, methylation, SUMOylation, and ubiquitination. These rapid and dynamic modifications may be combinatorial and interact (i.e. may be sequential, complement, or oppose each other), creating a vast array of uniquely modified receptor subspecies that allow for diverse receptor behaviors that enable highly sensitive and context-dependent hormone action. For example, in response to hormone or growth factor membrane-initiated signaling events, posttranslational modifications (PTMs) to SRs alter protein-protein interactions that govern the complex process of promoter or gene-set selection coupled to transcriptional repression or activation. Unique phosphorylation events allow SRs to associate or disassociate with specific cofactors that may include pioneer factors and other tethering partners, which specify the resulting transcriptome and ultimately change cell fate. The impact of PTMs on SR action is particularly profound in the context of breast tumorigenesis, in which frequent alterations in growth factor-initiated signaling pathways occur early and act as drivers of breast cancer progression toward endocrine resistance. In this article, with primary focus on breast cancer relevance, we review the mechanisms by which PTMs, including reversible phosphorylation events, regulate the closely related SRs, glucocorticoid receptor and progesterone receptor, allowing for precise biological responses to ever-changing hormonal stimuli.


Subject(s)
Breast Neoplasms/metabolism , Protein Processing, Post-Translational , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Protein Isoforms , Receptors, Estrogen/metabolism , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , Receptors, Progesterone/chemistry , Receptors, Progesterone/genetics , Signal Transduction , Stress, Physiological , Structure-Activity Relationship
11.
PLoS One ; 10(3): e0121206, 2015.
Article in English | MEDLINE | ID: mdl-25789479

ABSTRACT

Tamoxifen (Tam) is the only FDA-approved chemoprevention agent for pre-menopausal women at high risk for developing breast cancer. While Tam reduces a woman's risk of developing estrogen receptor positive (ER+) breast cancer, the molecular mechanisms associated with risk reduction are poorly understood. Prior studies have shown that cytoplasmic proline, glutamic acid and leucine rich protein 1 (PELP1) promotes Tam resistance in breast cancer cell lines. Herein, we tested for PELP1 localization in breast epithelial cells from women at high risk for developing breast cancer and found that PELP1 was localized to the cytoplasm in 36% of samples. In vitro, immortalized HMECs expressing a nuclear localization signal (NLS) mutant of PELP1 (PELP1-cyto) were resistant to Tam-induced death. Furthermore, PELP1-cyto signaling through estrogen-related receptor gamma (ERRγ) promoted cell survival in the presence of Tam. Overexpression of ERRγ in immortalized HMECs protected cells from Tam-induced death, while knockdown of ERRγ sensitized PELP1-cyto expressing HMECs to Tam. Moreover, Tam-induced HMEC cell death was independent of apoptosis and involved accumulation of the autophagy marker LC3-II. Expression of PELP1-cyto and ERRγ reduced Tam-induced LC3-II accumulation, and knockdown of ERRγ increased LC3-II levels in response to Tam. Additionally, PELP1-cyto expression led to the upregulation of MMP-3 and MAOB, known PELP1 and ERRγ target genes, respectively. Our data indicate that cytoplasmic PELP1 induces signaling pathways that converge on ERRγ to promote cell survival in the presence of Tam. These data suggest that PELP1 localization and/or ERRγ activation could be developed as tissue biomarkers for Tam responsiveness.


Subject(s)
Co-Repressor Proteins/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Mammary Glands, Human/cytology , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Transcription Factors/metabolism , Autophagy/drug effects , Breast Neoplasms/pathology , Carcinogenesis/drug effects , Cell Line, Tumor , Co-Repressor Proteins/genetics , Female , Gene Expression Regulation/drug effects , Humans , Mammary Glands, Human/pathology , Protein Transport/drug effects , Receptors, Estrogen/genetics , Risk , Transcription Factors/genetics
12.
Mol Cell Endocrinol ; 382(1): 642-651, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-23933151

ABSTRACT

Proline, glutamic acid, and leucine rich protein 1 (PELP1) is a large multi-domain protein that has been shown to modulate an increasing number of pathways and biological processes. The first reports describing the cloning and characterization of PELP1 showed that it was an estrogen receptor coactivator. PELP1 has now been shown to be a coregulator for a growing number of transcription factors. Furthermore, recent reports have shown that PELP1 is a member of chromatin remodeling complexes. In addition to PELP1 nuclear functions, it has been shown to have cytoplasmic signaling functions as well. In the cytoplasm PELP1 acts as a scaffold molecule and mediates rapid signaling from growth factor and hormone receptors. PELP1 signaling ultimately plays a role in cancer biology by increasing proliferation and metastasis, among other cellular processes. Here we will review (1) the cloning and characterization of PELP1 expression, (2) interacting proteins, (3) PELP1 signaling, and (4) PELP1-mediated biology.


Subject(s)
Signal Transduction , Transcription Factors/metabolism , Animals , Cell Nucleus/metabolism , Humans , Models, Biological , Molecular Targeted Therapy , Protein Binding , Transcription Factors/antagonists & inhibitors
13.
PLoS One ; 7(9): e45684, 2012.
Article in English | MEDLINE | ID: mdl-23049838

ABSTRACT

Breast cancers with a basal-like gene signature are primarily triple-negative, frequently metastatic, and carry a poor prognosis. Basal-like breast cancers are enriched for markers of breast cancer stem cells as well as markers of epithelial-mesenchymal transition (EMT). While EMT is generally thought to be important in the process of metastasis, in vivo evidence of EMT in human disease remains rare. Here we report a novel model of human triple-negative breast cancer, the DKAT cell line, which was isolated from an aggressive, treatment-resistant triple-negative breast cancer that demonstrated morphological and biochemical evidence suggestive of phenotypic plasticity in the patient. The DKAT cell line displays a basal-like phenotype in vitro when cultured in serum-free media, and undergoes phenotypic changes consistent with EMT/MET in response to serum-containing media, a unique property among the breast cancer cell lines we tested. This EMT is marked by increased expression of the transcription factor Zeb1, and Zeb1 is required for the enhanced migratory ability of DKAT cells in the mesenchymal state. DKAT cells also express progenitor-cell markers, and single DKAT cells are able to generate tumorspheres containing both epithelial and mesenchymal cell types. In vivo, as few as ten DKAT cells are capable of forming xenograft tumors which display a range of epithelial and mesenchymal phenotypes. The DKAT model provides a novel model to study the molecular mechanisms regulating phenotypic plasticity and the aggressive biology of triple-negative breast cancers.


Subject(s)
Breast Neoplasms/pathology , Adult , Animals , Bone Marrow/pathology , Breast Neoplasms/metabolism , Cell Line, Tumor , Culture Media, Serum-Free/pharmacology , Cytogenetics , Epithelial-Mesenchymal Transition , Exons , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Karyotyping , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Phenotype , Prognosis
14.
J Biomed Opt ; 16(11): 116003, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22112108

ABSTRACT

This work presents simultaneous imaging and detection of three different cell receptors using three types of plasmonic nanoparticles (NPs). The size, shape, and composition-dependent scattering profiles of these NPs allow for a system of multiple distinct molecular markers using a single optical source. With this goal in mind, tags consisting of anti-epidermal growth factor receptor gold nanorods, anti-insulin-like growth factor 1-R silver nanospheres, and human epidermal growth factor receptor 2Ab gold nanospheres were developed to monitor the expression of receptors commonly overexpressed by cancer cells. These labels were chosen because they scatter strongly in distinct spectral windows. A hyperspectral darkfield microspectroscopy system was developed to record the scattering spectra of cells labeled with these molecular tags. Simultaneous monitoring of multiple tags may lead to applications such as profiling of cell line immunophenotype and investigation of receptor signaling pathways. Single, dual, and triple tag experiments were performed to analyze NP tag specificity as well as their interactions. Distinct resonance peaks were observed in these studies, showing the ability to characterize cell lines using conjugated NPs. However, interpreting shifts in these peaks due to changes in a cellular dielectric environment may be complicated by plasmon coupling between NPs bound to proximal receptors and other coupling mechanisms due to the receptors themselves.


Subject(s)
Immunophenotyping/methods , Molecular Imaging/methods , Nanoparticles/chemistry , Receptors, Cell Surface/chemistry , Cell Line , ErbB Receptors/chemistry , Humans , Receptor, ErbB-2/chemistry , Receptor, IGF Type 1/chemistry , Scattering, Radiation
15.
ACS Nano ; 5(11): 8532-40, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-21999459

ABSTRACT

The dimerization of receptors on the cell membrane is an important step in the activation of cell signaling pathways. Several methods exist for observing receptor dimerization, including coimmunoprecipitation, chemical cross-linking, and fluorescence resonance energy transfer (FRET). These techniques are limited in that only FRET is appropriate for live cells, but even that method suffers from photobleaching and bleed-through effects. In this study, we implement an alternative method for the targeting of HER-2 homodimer formation based on the plasmonic coupling of gold nanoparticles functionalized with HER-2 Ab. In the presented studies, SK-BR-3 cells, known to overexpress HER-2, are labeled with these nanoparticles and receptor colocalization is observed using plasmonic coupling. HER-2 targeted nanoparticles bound to these cells exhibit a peak resonance that is significantly red-shifted relative to those bound to similar receptors on A549 cells, which have significantly lower levels of HER-2 expression. This significant red shift indicates plasmonic coupling is occurring and points to a new avenue for assessing dimerization by monitoring their colocalization. To determine that dimerization is occurring, the refractive index of the nanoenvironment of the labels is assessed using a theoretical analysis based on the Mie coated sphere model. The results indicate scattering by single, isolated nanoparticles for the low HER-2 expressing A549 cell line, but the scattering observed for the HER-2 overexpressing SK-BR-3 cell line may only be explained by plasmonic-coupling of proximal nanoparticle pairs. To validate the conformation of nanoparticles bound to HER-2 receptors undergoing dimerization, discrete dipole approximation (DDA) models are used to assess spectra of scattering by coupled nanoparticles. Comparison of the experimental results with theoretical models indicates that NP dimers are formed for the labeling of SK-BR-3 cells, suggesting that receptor dimerization has been observed.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Protein Multimerization , Receptor, ErbB-2/chemistry , Cell Line, Tumor , Gene Expression Regulation , Gold/metabolism , Humans , Protein Structure, Quaternary , Receptor, ErbB-2/metabolism
16.
Breast Cancer Res ; 13(5): R89, 2011 Sep 17.
Article in English | MEDLINE | ID: mdl-21923922

ABSTRACT

INTRODUCTION: Protein tyrosine kinases (PTKs) are frequently overexpressed and/or activated in human malignancies, and regulate cancer cell proliferation, cellular survival, and migration. As such, they have become promising molecular targets for new therapies. The non-receptor PTK termed breast tumor kinase (Brk/PTK6) is overexpressed in approximately 86% of human breast tumors. The role of Brk in breast pathology is unclear. METHODS: We expressed a WAP-driven Brk/PTK6 transgene in FVB/n mice, and analyzed mammary glands from wild-type (wt) and transgenic mice after forced weaning. Western blotting and immunohistochemistry (IHC) studies were conducted to visualize markers of mammary gland involution, cell proliferation and apoptosis, as well as Brk, STAT3, and activated p38 mitogen-activated protein kinase (MAPK) in mammary tissues and tumors from WAP-Brk mice. Human (HMEC) or mouse (HC11) mammary epithelial cells were stably or transiently transfected with Brk cDNA to assay p38 MAPK signaling and cell survival in suspension or in response to chemotherapeutic agents. RESULTS: Brk-transgenic dams exhibited delayed mammary gland involution and aged mice developed infrequent tumors with reduced latency relative to wt mice. Consistent with delayed involution, mammary glands of transgenic animals displayed decreased STAT3 phosphorylation, a marker of early-stage involution. Notably, p38 MAPK, a pro-survival signaling mediator downstream of Brk, was activated in mammary glands of Brk transgenic relative to wt mice. Brk-dependent signaling to p38 MAPK was recapitulated by Brk overexpression in the HC11 murine mammary epithelial cell (MEC) line and human MEC, while Brk knock-down in breast cancer cells blocked EGF-stimulated p38 signaling. Additionally, human or mouse MECs expressing Brk exhibited increased anchorage-independent survival and resistance to doxorubicin. Finally, breast tumor biopsies were subjected to IHC analysis for co-expression of Brk and phospho-p38 MAPK; ductal and lobular carcinomas expressing Brk were significantly more likely to express elevated phospho-p38 MAPK. CONCLUSIONS: These studies illustrate that forced expression of Brk/PTK6 in non-transformed mammary epithelial cells mediates p38 MAPK phosphorylation and promotes increased cellular survival, delayed involution, and latent tumor formation. Brk expression in human breast tumors may contribute to progression by inducing p38-driven pro-survival signaling pathways.


Subject(s)
Mammary Glands, Animal/metabolism , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Adenosquamous/genetics , Carcinoma, Adenosquamous/metabolism , Carcinoma, Adenosquamous/pathology , Cell Line, Tumor , Cell Survival , Enzyme Activation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation , Humans , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Phosphorylation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
17.
Breast Cancer Res Treat ; 126(1): 55-62, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20390344

ABSTRACT

This study quantifies uptake of a fluorescent glucose analog, (2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose) (2-NBDG), in a large panel of breast cancer cells and demonstrates potential to monitor changes in glycolysis caused by anticancer and endocrine therapies. Expressions of glucose transporter (GLUT 1) and hexokinase (HK I), which phosphorylates 2-NBDG, were measured via western blot in two normal mammary epithelial and eight breast cancer cell lines of varying biological subtype. Fluorescence intensity of each cell line labeled with 100 lM 2-NBDG for 20 min or unlabeled control was quantified. A subset of cancer cells was treated with anticancer and endocrine therapies, and 2-NBDG fluorescence changes were measured. Expression of GLUT 1 was necessary for uptake of 2-NBDG, as demonstrated by lack of 2-NBDG uptake in normal human mammary epithelial cells (HMECs). GLUT 1 expression and 2-NBDG uptake was ubiquitous among all breast cancer lines. Reduction and stimulation of 2-NBDG uptake was demonstrated by perturbation with anticancer agents, lonidamine (LND), and a-cyano-hydroxycinnamate (a-Cinn), respectively. LND directly inhibits HK and significantly reduced 2-NBDG fluorescence in a subset of two breast cancer cell lines. Conversely, when cells were treated with a-Cinn, a drug used to increase glycolysis, 2-NBDG uptake was increased. Furthermore, tamoxifen (tam), a common endocrine therapy, was administered to estrogen receptor positive and negative (ER?/-) breast cells and demonstrated a decreased 2-NBDG uptake in ER? cells, reflecting a decrease in glycolysis. Results indicate that 2-NBDG uptake can be used to measure changes in glycolysis and has potential for use in early drug development.


Subject(s)
4-Chloro-7-nitrobenzofurazan/analogs & derivatives , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast/drug effects , Deoxyglucose/analogs & derivatives , Glucose/metabolism , Tamoxifen/pharmacology , 4-Chloro-7-nitrobenzofurazan/pharmacokinetics , Antineoplastic Agents, Hormonal/pharmacology , Blotting, Western , Breast/metabolism , Cells, Cultured , Deoxyglucose/pharmacokinetics , Female , Glucose Transporter Type 1/metabolism , Hexokinase/metabolism , Humans , Phosphorylation , Receptors, Estrogen , Treatment Outcome
18.
Curr Opin Pharmacol ; 10(6): 662-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20832360

ABSTRACT

Breast tumor kinase (Brk), also termed PTK6, is known to function in cell-type and context-dependent processes governing normal differentiation. However, in tumors in which Brk is overexpressed, this unusual soluble tyrosine kinase is emerging as a mediator of cancer cell phenotypes, including increased proliferation, survival, and migration. Nuclear and cytoplasmic substrates phosphorylated by Brk include a collection of regulatory RNA-binding proteins, adaptor molecules that link Brk to signaling pathways generally associated with the activation of growth factor receptors, and Signal Transducers and Activators of Transcription (STAT) molecules that are direct regulators of gene expression. Understanding Brk-dependent regulation of these key signaling pathways and how they influence cancer cell behavior is predicted to inform the development of improved 'targeted' cancer therapies and may provide insight into ways to avoid chemo-resistance to established treatments.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Proliferation , Female , Humans , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Phosphorylation , Protein-Tyrosine Kinases/genetics , RNA-Binding Proteins/genetics
19.
Cancer Res ; 70(11): 4759-66, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20460512

ABSTRACT

Autofluorescence spectroscopy is a powerful imaging technique that exploits endogenous fluorophores. The endogenous fluorophores NADH and flavin adenine dinucleotide (FAD) are two of the principal electron donors and acceptors in cellular metabolism, respectively. The optical oxidation-reduction (redox) ratio is a measure of cellular metabolism and can be determined by the ratio of NADH/FAD. We hypothesized that there would be a significant difference in the optical redox ratio of normal mammary epithelial cells compared with breast tumor cell lines and that estrogen receptor (ER)-positive cells would have a higher redox ratio than ER-negative cells. To test our hypothesis, the optical redox ratio was determined by collecting the fluorescence emission for NADH and FAD via confocal microscopy. We observed a statistically significant increase in the optical redox ratio of cancer compared with normal cell lines (P < 0.05). Additionally, we observed a statistically significant increase in the optical redox ratio of ER(+) breast cancer cell lines. The level of ESR1 expression, determined by real-time PCR, directly correlated with the optical redox ratio (Pearson's correlation coefficient = 0.8122, P = 0.0024). Furthermore, treatment with tamoxifen and ICI 182,870 statistically decreased the optical redox ratio of only ER(+) breast cancer cell lines. The results of this study raise the important possibility that fluorescence spectroscopy can be used to identify subtypes of breast cancer based on receptor status, monitor response to therapy, or potentially predict response to therapy. This source of optical contrast could be a potentially useful tool for drug screening in preclinical models.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Flavin-Adenine Dinucleotide/metabolism , Fulvestrant , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Microscopy, Confocal/methods , NAD/metabolism , Oxidation-Reduction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tamoxifen/pharmacology
20.
J Biomed Opt ; 15(1): 018002, 2010.
Article in English | MEDLINE | ID: mdl-20210488

ABSTRACT

We describe the potential of 5-aminolevulinic acid (ALA)-induced protoporphyrin IX (PpIX) fluorescence as a source of contrast for margin detection in commonly diagnosed breast cancer subtypes. Fluorescence intensity of PpIX in untreated and ALA-treated normal mammary epithelial and breast cancer cell lines of varying estrogen receptor expression were quantitatively imaged with confocal microscopy. Percentage change in fluorescence intensity integrated over 610-700 nm (attributed to PpIX) of posttreated compared to pretreated cells showed statistically significant differences between four breast cancer and two normal mammary epithelial cell lines. However, a direct comparison of post-treatment PpIX fluorescence intensities showed no differences between breast cancer and normal mammary epithelial cell lines due to confounding effects by endogenous fluorescence from flavin adenine dinucleotide (FAD). Clinically, it is impractical to obtain pre- and post-treatment images. Thus, spectral imaging was demonstrated as a means to remove the effects of endogenous FAD fluorescence allowing for discrimination between post-treatment PpIX fluorescence of four breast cancer and two normal mammary epithelial cell lines. Fluorescence spectral imaging of ALA-treated breast cancer cells showed preferential PpIX accumulation regardless of malignant phenotype and suggests a useful contrast mechanism for discrimination of residual cancer at the surface of breast tumor margins.


Subject(s)
Aminolevulinic Acid/pharmacokinetics , Breast Neoplasms/metabolism , Contrast Media/pharmacokinetics , Protoporphyrins/pharmacokinetics , Spectrometry, Fluorescence/methods , Analysis of Variance , Breast Neoplasms/chemistry , Cell Line, Tumor , Female , Humans , Microscopy, Confocal/methods , Phenotype , Protoporphyrins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...