Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Genes Dev ; 32(3-4): 244-257, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29483153

ABSTRACT

The discoidin domain receptor 1 (DDR1) is overexpressed in breast carcinoma cells. Low DDR1 expression is associated with worse relapse-free survival, reflecting its controversial role in cancer progression. We detected DDR1 on luminal cells but not on myoepithelial cells of DDR1+/+ mice. We found that DDR1 loss compromises cell adhesion, consistent with data that older DDR1-/- mammary glands had more basal/myoepithelial cells. Basal cells isolated from older mice exerted higher traction forces than the luminal cells, in agreement with increased mammary branches observed in older DDR1-/- mice and higher branching by their isolated organoids. When we crossed DDR1-/- mice with MMTV-PyMT mice, the PyMT/DDR1-/- mammary tumors grew faster and had increased epithelial tension and matricellular fibrosis with a more basal phenotype and increased lung metastases. DDR1 deletion induced basal differentiation of CD90+CD24+ cancer cells, and the increase in basal cells correlated with tumor cell mitoses. K14+ basal cells, including K8+K14+ cells, were increased adjacent to necrotic fields. These data suggest that the absence of DDR1 provides a growth and adhesion advantage that favors the expansion of basal cells, potentiates fibrosis, and enhances necrosis/hypoxia and basal differentiation of transformed cells to increase their aggression and metastatic potential.


Subject(s)
Discoidin Domain Receptor 1/genetics , Mammary Neoplasms, Experimental/pathology , Animals , Breast Neoplasms/metabolism , Cell Hypoxia , Discoidin Domain Receptor 1/metabolism , Disease-Free Survival , Epithelial Cells/metabolism , Female , Fibrosis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mice
2.
Angew Chem Int Ed Engl ; 59(9): 3439-3443, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31765069

ABSTRACT

Metabolomics is a powerful systems biology approach that monitors changes in biomolecule concentrations to diagnose and monitor health and disease. However, leading metabolomics technologies, such as NMR and mass spectrometry (MS), access only a small portion of the metabolome. Now an approach is presented that uses the high sensitivity and chemical specificity of surface-enhanced Raman scattering (SERS) for online detection of metabolites from tumor lysates following liquid chromatography (LC). The results demonstrate that this LC-SERS approach has metabolite detection capabilities comparable to the state-of-art LC-MS but suggest a selectivity for the detection of a different subset of metabolites. Analysis of replicate LC-SERS experiments exhibit reproducible metabolite patterns that can be converted into barcodes, which can differentiate different tumor models. Our work demonstrates the potential of LC-SERS technology for metabolomics-based diagnosis and treatment of cancer.


Subject(s)
Metabolome , Metabolomics/methods , Neoplasms/diagnosis , Animals , Chromatography, High Pressure Liquid , Disease Models, Animal , Mice , Neoplasms/metabolism , Spectrum Analysis, Raman , Wnt1 Protein/metabolism
3.
BMC Cancer ; 16: 204, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26964534

ABSTRACT

BACKGROUND: Breast cancer incidence and mortality vary significantly among different nations and racial groups. African nations have the highest breast cancer mortality rates in the world, even though the incidence rates are below those of many nations. Differences in disease progression suggest that aggressive breast tumors may harbor a unique molecular signature to promote disease progression. However, few studies have investigated the pathology and clinical markers expressed in breast tissue from regional African patient populations. METHODS: We collected 68 malignant and 89 non-cancerous samples from Kenyan breast tissue. To characterize the tumors from these patients, we constructed tissue microarrays (TMAs) from these tissues. Sections from these TMAs were stained and analyzed using immunohistochemistry to detect clinical breast cancer markers, including estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor 2 receptor (HER2) status, Ki67, and immune cell markers. RESULTS: Thirty-three percent of the tumors were triple negative (ER-, PR-, HER2-), 59% were ER+, and almost all tumors analyzed were HER2-. Seven percent of the breast cancer patients were male, and 30% were <40 years old at diagnosis. Cancer tissue had increased immune cell infiltration with recruitment of CD163+ (M2 macrophage), CD25+ (regulatory T lymphocyte), and CD4+ (T helper) cells compared to non-cancer tissue. CONCLUSIONS: We identified clinical biomarkers that may assist in identifying therapy strategies for breast cancer patients in western Kenya. Estrogen receptor status in particular should lead initial treatment strategies in these breast cancer patients. Increased CD25 expression suggests a need for additional treatment strategies designed to overcome immune suppression by CD25+ cells in order to promote the antitumor activity of CD8+ cytotoxic T cells.


Subject(s)
Biomarkers, Tumor/genetics , Cell Proliferation/genetics , Triple Negative Breast Neoplasms/epidemiology , Triple Negative Breast Neoplasms/pathology , Aged , Black People/genetics , Female , Humans , Immunohistochemistry , Kenya , Middle Aged , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , T-Lymphocytes, Cytotoxic/immunology , Tissue Array Analysis , Triple Negative Breast Neoplasms/genetics
4.
Cancer Cell ; 13(2): 141-52, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18242514

ABSTRACT

How breast cancers are able to disseminate and metastasize is poorly understood. Using a hyperplasia transplant system, we show that tumor dissemination and metastasis occur in discrete steps during tumor progression. Bioinformatic analysis revealed that loss of the transcription factor GATA-3 marked progression from adenoma to early carcinoma and onset of tumor dissemination. Restoration of GATA-3 in late carcinomas induced tumor differentiation and suppressed tumor dissemination. Targeted deletion of GATA-3 in early tumors led to apoptosis of differentiated cells, indicating that its loss is not sufficient for malignant conversion. Rather, malignant progression occurred with an expanding GATA-3-negative tumor cell population. These data indicate that GATA-3 regulates tumor differentiation and suppresses tumor dissemination in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cell Differentiation , GATA3 Transcription Factor/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , Epithelial Cells/pathology , Female , GATA3 Transcription Factor/deficiency , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hyperplasia , Immunohistochemistry , Mammary Glands, Animal , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Neoplastic Stem Cells/pathology
5.
BMC Genomics ; 15: 520, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24962896

ABSTRACT

BACKGROUND: The ZNF217 gene, encoding a C2H2 zinc finger protein, is located at 20q13 and found amplified and overexpressed in greater than 20% of breast tumors. Current studies indicate ZNF217 drives tumorigenesis, yet the regulatory mechanisms of ZNF217 are largely unknown. Because ZNF217 associates with chromatin modifying enzymes, we postulate that ZNF217 functions to regulate specific gene signaling networks. Here, we present a large-scale functional genomic analysis of ZNF217, which provides insights into the regulatory role of ZNF217 in MCF7 breast cancer cells. RESULTS: ChIP-seq analysis reveals that the majority of ZNF217 binding sites are located at distal regulatory regions associated with the chromatin marks H3K27ac and H3K4me1. Analysis of ChIP-seq transcription factor binding sites shows clustering of ZNF217 with FOXA1, GATA3 and ERalpha binding sites, supported by the enrichment of corresponding motifs for the ERalpha-associated cis-regulatory sequences. ERalpha expression highly correlates with ZNF217 in lysates from breast tumors (n = 15), and ERalpha co-precipitates ZNF217 and its binding partner CtBP2 from nuclear extracts. Transcriptome profiling following ZNF217 depletion identifies differentially expressed genes co-bound by ZNF217 and ERalpha; gene ontology suggests a role for ZNF217-ERalpha in expression programs associated with ER+ breast cancer studies found in the Molecular Signature Database. Data-mining of expression data from breast cancer patients correlates ZNF217 with reduced overall survival. CONCLUSIONS: Our genome-wide ZNF217 data suggests a functional role for ZNF217 at ERalpha target genes. Future studies will investigate whether ZNF217 expression contributes to aberrant ERalpha regulatory events in ER+ breast cancer and hormone resistance.


Subject(s)
Breast Neoplasms/metabolism , Chromatin/metabolism , Estrogen Receptor alpha/genetics , Trans-Activators/physiology , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cluster Analysis , Female , GATA3 Transcription Factor/physiology , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Genome, Human , Hepatocyte Nuclear Factor 3-alpha/physiology , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Protein Binding , Transcriptome
6.
Bone ; 181: 117028, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38309412

ABSTRACT

INTRODUCTION: Osteocytes modulate bone adaptation in response to mechanical stimuli imparted by the deforming bone tissue in which they are encased by communicating with osteoclasts and osteoblasts as well as other osteocytes in the lacuna-canalicular network through secreted cytokines and chemokines. Understanding the transcriptional response of osteocytes to mechanical stimulation in situ could identify new targets to inhibit bone loss or enhance bone formation in the presence of diseases like osteoporosis or metastatic cancer. We compared the mechanically regulated transcriptional response of osteocytes in trabecular bone following one or three days of controlled mechanical loading. METHODS: Porcine trabecular bone explants were cultured in a bioreactor for 48 h and subsequently loaded twice a day for one day or 3 days. RNA was isolated and sequenced, and the Tuxedo suite was used to identify differentially expressed genes and pathway analysis was conducted using Ingenuity Pathway Analysis (IPA). RESULTS: There were about 4000 differentially expressed genes following in situ culture relative to fresh bone. One hundred six genes were differentially expressed between the loaded and non-loaded groups following one day of loading compared to 913 genes after 3 d of loading. Only 45 of these were coincident between the two time points, indicating an evolving transcriptome. Clustering and principal component analysis indicated differences between the loaded and non-loaded groups after 3 d of loading. DISCUSSION: With sustained loading, there was a nine-fold increase in the number of differentially expressed genes, suggesting that osteocytes respond to loading through sequential activation of downstream genes in the same pathways. The differentially expressed genes were related to osteoarthritis, osteocyte, and chondrocyte signaling pathways. We noted that NFkB and TNF signaling are affected by early loading and this may drive downstream effects on the mechanobiological response. Moreover, these genes may regulate catabolic effects of mechanical disuse through their actions on pre-osteoclasts in the bone marrow niche.


Subject(s)
Cancellous Bone , Osteocytes , Animals , Swine , Osteocytes/metabolism , Transcriptome/genetics , Bone and Bones , Osteoblasts , Stress, Mechanical
7.
Cancer Cell ; 7(6): 499-500, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950897

ABSTRACT

It is now becoming apparent that multiple types of stromal cells, including macrophages, mast cells, adipocytes, and fibroblasts make pivotal contributions to carcinogenesis. In the May 6 issue of Cell, Orimo and colleagues (Orimo et al., 2005) show that carcinoma-associated fibroblasts can promote epithelial tumorigenesis by secreting the chemokine SDF-1alpha (CXCL12). SDF-1alpha stimulates carcinoma cell proliferation and recruitment of endothelial precursor cells.


Subject(s)
Neoplasms/metabolism , Stromal Cells/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Movement , Chemokine CXCL12 , Chemokines, CXC/metabolism , Endothelial Cells/cytology , Fibroblasts/metabolism , Humans , Leukocytes/metabolism , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cells/cytology , Models, Biological , Neoplasms/etiology , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology
8.
Biomaterials ; 292: 121913, 2023 01.
Article in English | MEDLINE | ID: mdl-36442437

ABSTRACT

Here, we report a CD138 receptor targeting liposomal formulation (TNP[Prodrug-4]) that achieved efficacious tumor growth inhibition in treating multiple myeloma by overcoming the dose limiting severe toxicity issues of a highly potent drug, Mertansine (DM1). Despite the promising potential to treat various cancers, due to poor solubility and pharmacokinetic profile, DM1's translation to the clinic has been unsatisfactory. We hypothesized that the optimal prodrug chemistry would promote efficient loading of the prodrug into targeted nanoparticles and achieve controlled release following endocytosis by the cancer cells, consequently, accomplish the most potent tumor growth inhibition. We evaluated four functional linker chemistries for synthesizing DM1-Prodrug molecules and evaluated their stability and cancer cell toxicity in vitro. It was determined that the phosphodiester moiety, as part of nanoparticle formulations, demonstrated most favorable characteristics with an IC50 of ∼16 nM. Nanoparticle formulations of Prodrug-4 enabled its administration at 8-fold higher dosage of equivalent free drug while remaining below maximum tolerated dose. Importantly, TNP[Prodrug-4] achieved near complete inhibition of tumor growth (∼99% by day 10) compared to control, without displaying noticeable systemic toxicity. TNP[Prodrug-4] promises a formulation that could potentially make DM1 treatment available for wider clinical applications with a long-term goal for better patient outcomes.


Subject(s)
Maytansine , Multiple Myeloma , Nanoparticles , Prodrugs , Humans , Prodrugs/chemistry , Multiple Myeloma/drug therapy , Maytansine/therapeutic use , Maytansine/pharmacology , Nanoparticles/chemistry , Liposomes , Peptides , Cell Line, Tumor
9.
Front Oncol ; 12: 988119, 2022.
Article in English | MEDLINE | ID: mdl-36212456

ABSTRACT

Aquaporins (AQPs) are a family of small transmembrane proteins that selectively transport water and other small molecules and ions following an osmotic gradient across cell plasma membranes. This enables them to regulate numerous functions including water homeostasis, fat metabolism, proliferation, migration, and adhesion. Previous structural and functional studies highlight a strong biological relationship between AQP protein expression, localization, and key biological functions in normal and cancer tissues, where aberrant AQP expression correlates with tumorigenesis and metastasis. In this review, we discuss the roles of AQP1, AQP3, AQP4, AQP5, and AQP7 in breast cancer progression and metastasis, including the role of AQPs in the tumor microenvironment, to highlight potential contributions of stromal-derived to epithelial-derived AQPs to breast cancer. Emerging evidence identifies AQPs as predictors of response to cancer therapy and as targets for increasing their sensitivity to treatment. However, these studies have not evaluated the requirements for protein structure on AQP function within the context of breast cancer. We also examine how AQPs contribute to a patient's response to cancer treatment, existing AQP inhibitors and how AQPs could serve as novel predictive biomarkers of therapy response in breast cancer. Future studies also should evaluate AQP redundancy and compensation as mechanisms used to overcome aberrant AQP function. This review highlights the need for additional research into how AQPs contribute molecularly to therapeutic resistance and by altering the tumor microenvironment.

10.
Nature ; 436(7047): 123-7, 2005 Jul 07.
Article in English | MEDLINE | ID: mdl-16001073

ABSTRACT

The tumour microenvironment can be a potent carcinogen, not only by facilitating cancer progression and activating dormant cancer cells, but also by stimulating tumour formation. We have previously investigated stromelysin-1/matrix metalloproteinase-3 (MMP-3), a stromal enzyme upregulated in many breast tumours, and found that MMP-3 can cause epithelial-mesenchymal transition (EMT) and malignant transformation in cultured cells, and genomically unstable mammary carcinomas in transgenic mice. Here we explain the molecular pathways by which MMP-3 exerts these effects: exposure of mouse mammary epithelial cells to MMP-3 induces the expression of an alternatively spliced form of Rac1, which causes an increase in cellular reactive oxygen species (ROS). The ROS stimulate the expression of the transcription factor Snail and EMT, and cause oxidative damage to DNA and genomic instability. These findings identify a previously undescribed pathway in which a component of the breast tumour microenvironment alters cellular structure in culture and tissue structure in vivo, leading to malignant transformation.


Subject(s)
Cell Differentiation , Epithelial Cells/metabolism , Genomic Instability/genetics , Matrix Metalloproteinase 3/metabolism , Mesoderm/metabolism , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/metabolism , Alternative Splicing/genetics , Animals , Cell Line , Cell Transformation, Neoplastic , DNA Damage , Epithelial Cells/cytology , Epithelial Cells/enzymology , Humans , Matrix Metalloproteinase 3/genetics , Mesoderm/cytology , Mesoderm/enzymology , Mice , Mice, Transgenic , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , rac1 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/metabolism
11.
Mol Biol Cell ; 32(10): 1009-1019, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33689396

ABSTRACT

Metastasis is responsible for over 90% of cancer-related deaths, and bone is the most common site for breast cancer metastasis. Metastatic breast cancer cells home to trabecular bone, which contains hematopoietic and stromal lineage cells in the marrow. As such, it is crucial to understand whether bone or marrow cells enhance breast cancer cell migration toward the tissue. To this end, we quantified the migration of MDA-MB-231 cells toward human bone in two- and three-dimensional (3D) environments. First, we found that the cancer cells cultured on tissue culture plastic migrated toward intact trabecular bone explants at a higher rate than toward marrow-deficient bone or devitalized bone. Leptin was more abundant in conditioned media from the cocultures with intact explants, while higher levels of IL-1ß, IL-6, and TNFα were detected in cultures with both intact bone and cancer cells. We further verified that the cancer cells migrated into bone marrow using a bioreactor culture system. Finally, we studied migration toward bone in 3D gelatin. Migration speed did not depend on stiffness of this homogeneous gel, but many more dendritic-shaped cancer cells oriented and migrated toward bone in stiffer gels than softer gels, suggesting a coupling between matrix mechanics and chemotactic signals.


Subject(s)
Bone Marrow/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Movement , Chemotactic Factors/metabolism , Bioreactors , Cell Culture Techniques , Chemokines/metabolism , Culture Media, Conditioned , Cytokines/metabolism , Hydrogels , Neoplasm Metastasis , Neoplastic Cells, Circulating/metabolism , Tumor Cells, Cultured
12.
J Control Release ; 322: 530-541, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32276005

ABSTRACT

Here, we report rationally engineered peptide-targeted liposomal doxorubicin nanoparticles that have an enhanced selectivity for HER2-positive breast tumor cells with high purity, reproducibility, and precision in controlling stoichiometry of targeting peptides. To increase HER2-positive tumor cell selective drug delivery, we optimized the two most important design parameters, peptide density and linker length, via systematic evaluations of their effects on both in vitro cellular uptake and in vivo tumor accumulation and cellular uptake. The optimally designed nanoparticles were finally evaluated for their tumor inhibition efficacy using in vivo MMTV-neu transplantation mouse model. In vitro, we demonstrated that ~1% peptide density and EG8 linker were optimal parameters for targeted nanoparticle formulations to enhance HER2-positive cancer cellular uptake while preventing non-selectivity. In vivo results demonstrated that at 0.5% peptide density, enhancement of tumor cell uptake over non-targeted nanoparticles was ~2.7 fold and ~3.4 fold higher for targeted nanoparticles with EG8 and EG18 linker, respectively, while their accumulation levels at tumor tissue were similar to the non-targeted nanoparticles. These results were consistent with in vivo efficacy outcomes that ~90% tumor growth inhibition was achieved by Dox-loaded HER2 receptor targeted nanoparticles, TNPHER2pep, over control while all nanoparticle formulations minimized overall systemic toxicity relative to free Dox. This study highlights the significance of understanding and optimizing the effects of liposomal nanoparticle design parameters for enhancement of tumor selectivity to achieve improved in vivo therapeutic outcomes.


Subject(s)
Breast Neoplasms , Nanoparticles , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Doxorubicin/therapeutic use , Drug Delivery Systems , Female , Humans , Mice , Peptides/therapeutic use , Reproducibility of Results
13.
Sci Rep ; 10(1): 1460, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31996718

ABSTRACT

SERS (surface-enhanced Raman scattering) enhances the Raman signals, but the plasmonic effects are sensitive to the chemical environment and the coupling between nanoparticles, resulting in large and variable backgrounds, which make signal matching and analyte identification highly challenging. Removing background is essential, but existing methods either cannot fit the strong fluctuation of the SERS spectrum or do not consider the spectra's shape change across time. Here we present a new statistical approach named SABARSI that overcomes these difficulties by combining information from multiple spectra. Further, after efficiently removing the background, we have developed the first automatic method, as a part of SABARSI, for detecting signals of molecules and matching signals corresponding to identical molecules. The superior efficiency and reproducibility of SABARSI are shown on two types of experimental datasets.

14.
J Hematol Oncol ; 13(1): 145, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33138841

ABSTRACT

BACKGROUND: Drug-loaded nanoparticles have established their benefits in the fight against multiple myeloma; however, ligand-targeted nanomedicine has yet to successfully translate to the clinic due to insufficient efficacies reported in preclinical studies. METHODS: In this study, liposomal nanoparticles targeting multiple myeloma via CD38 or CD138 receptors are prepared from pre-synthesized, purified constituents to ensure increased consistency over standard synthetic methods. These nanoparticles are then tested both in vitro for uptake to cancer cells and in vivo for accumulation at the tumor site and uptake to tumor cells. Finally, drug-loaded nanoparticles are tested for long-term efficacy in a month-long in vivo study by tracking tumor size and mouse health. RESULTS: The targeted nanoparticles are first optimized in vitro and show increased uptake and cytotoxicity over nontargeted nanoparticles, with CD138-targeting showing superior enhancement over CD38-targeted nanoparticles. However, biodistribution and tumor suppression studies established CD38-targeted nanoparticles to have significantly increased in vivo tumor accumulation, tumor cell uptake, and tumor suppression over both nontargeted and CD138-targeted nanoparticles due to the latter's poor selectivity. CONCLUSION: These results both highlight a promising cancer treatment option in CD38-targeted nanoparticles and emphasize that targeting success in vitro does not necessarily translate to success in vivo.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , Liposomes/metabolism , Multiple Myeloma/drug therapy , Syndecan-1/metabolism , ADP-ribosyl Cyclase 1/chemistry , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Humans , Liposomes/chemistry , Male , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Docking Simulation , Multiple Myeloma/metabolism , Peptides/chemistry , Peptides/metabolism , Syndecan-1/chemistry , Tissue Distribution
15.
Cancer Res ; 80(19): 4071-4086, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32631905

ABSTRACT

The complex yet interrelated connections between cancer metabolism, gene expression, and oncogenic driver genes have the potential to identify novel biomarkers and drug targets with prognostic and therapeutic value. Here we effectively integrated metabolomics and gene expression data from breast cancer mouse models through a novel unbiased correlation-based network analysis. This approach identified 35 metabolite and 34 gene hubs with the most network correlations. These hubs have prognostic value and are likely integral to tumor metabolism and breast cancer. The gene hub Aquaporin-7 (Aqp7), a water and glycerol channel, was identified as a novel regulator of breast cancer. AQP7 was prognostic of overall survival in patients with breast cancer. In mouse breast cancer models, reduced expression of Aqp7 caused reduced primary tumor burden and lung metastasis. Metabolomics and complex lipid profiling of cells and tumors with reduced Aqp7 revealed significantly altered lipid metabolism, glutathione metabolism, and urea/arginine metabolism compared with controls. These data identify AQP7 as a critical regulator of metabolic and signaling responses to environmental cellular stresses in breast cancer, highlighting AQP7 as a potential cancer-specific therapeutic vulnerability. SIGNIFICANCE: Aquaporin-7 is identified as a critical regulator of nutrient availability and signaling that responds to cellular stresses, making it an attractive therapeutic target in breast cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/19/4071/F1.large.jpg.


Subject(s)
Aquaporins/genetics , Aquaporins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Adipocytes/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Carbohydrate Metabolism , Cell Movement , Female , Gene Expression Regulation, Neoplastic , Glycolipids/metabolism , Glycolysis , Humans , Inositol/analogs & derivatives , Inositol/metabolism , Lipids/biosynthesis , Lipids/genetics , Lung Neoplasms/secondary , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/physiology , Nitric Oxide/metabolism , Prognosis
16.
J Clin Invest ; 130(11): 5721-5737, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32721948

ABSTRACT

Women with dense breasts have an increased lifetime risk of malignancy that has been attributed to a higher epithelial density. Quantitative proteomics, collagen analysis, and mechanical measurements in normal tissue revealed that stroma in the high-density breast contains more oriented, fibrillar collagen that is stiffer and correlates with higher epithelial cell density. microRNA (miR) profiling of breast tissue identified miR-203 as a matrix stiffness-repressed transcript that is downregulated by collagen density and reduced in the breast epithelium of women with high mammographic density. Culture studies demonstrated that ZNF217 mediates a matrix stiffness- and collagen density-induced increase in Akt activity and mammary epithelial cell proliferation. Manipulation of the epithelium in a mouse model of mammographic density supported a causal relationship between stromal stiffness, reduced miR-203, higher levels of the murine homolog Zfp217, and increased Akt activity and mammary epithelial proliferation. ZNF217 was also increased in the normal breast epithelium of women with high mammographic density, correlated positively with epithelial proliferation and density, and inversely with miR-203. The findings identify ZNF217 as a potential target toward which preexisting therapies, such as the Akt inhibitor triciribine, could be used as a chemopreventive agent to reduce cancer risk in women with high mammographic density.


Subject(s)
Breast Neoplasms , Mammary Glands, Human , Oncogene Proteins/metabolism , Trans-Activators/metabolism , Adult , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Double-Blind Method , Female , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mice , MicroRNAs/metabolism , Middle Aged , Proto-Oncogene Proteins c-akt/metabolism , RNA, Neoplasm/metabolism , Risk Factors
17.
Nat Commun ; 10(1): 4404, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31562303

ABSTRACT

Bone is one of the most common sites for metastasis across cancers. Cancer cells that travel through the vasculature and invade new tissues can remain in a non-proliferative dormant state for years before colonizing the metastatic site. Switching from dormancy to colonization is the rate-limiting step of bone metastasis. Here we develop an ex vivo co-culture method to grow cancer cells in mouse bones to assess cancer cell proliferation using healthy or cancer-primed bones. Profiling soluble factors from conditioned media identifies the chemokine CXCL5 as a candidate to induce metastatic colonization. Additional studies using CXCL5 recombinant protein suggest that CXCL5 is sufficient to promote breast cancer cell proliferation and colonization in bone, while inhibition of its receptor CXCR2 with an antagonist blocks proliferation of metastatic cancer cells. This study suggests that CXCL5 and CXCR2 inhibitors may have efficacy in treating metastatic bone tumors dependent on the CXCL5/CXCR2 axis.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Chemokine CXCL5/metabolism , Receptors, Interleukin-8B/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chemokine CXCL5/antagonists & inhibitors , Chemokine CXCL5/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mice, Transgenic , Middle Aged , Phenylurea Compounds/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Cell Death Dis ; 10(3): 180, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30792401

ABSTRACT

Potassium ion channels are critical in the regulation of cell motility. The acquisition of cell motility is an essential parameter of cancer metastasis. However, the role of K+ channels in cancer metastasis has been poorly studied. High expression of the hG1 gene, which encodes for Kv11.1 channel associates with good prognosis in estrogen receptor-negative breast cancer (BC). We evaluated the efficacy of the Kv11.1 activator NS1643 in arresting metastasis in a triple negative breast cancer (TNBC) mouse model. NS1643 significantly reduces the metastatic spread of breast tumors in vivo by inhibiting cell motility, reprogramming epithelial-mesenchymal transition via attenuation of Wnt/ß-catenin signaling and suppressing cancer cell stemness. Our findings provide important information regarding the clinical relevance of potassium ion channel expression in breast tumors and the mechanisms by which potassium channel activity can modulate tumor biology. Findings suggest that Kv11.1 activators may represent a novel therapeutic approach for the treatment of metastatic estrogen receptor-negative BC. Ion channels are critical factor for cell motility but little is known about their role in metastasis. Stimulation of the Kv11.1 channel suppress the metastatic phenotype in TNBC. This work could represent a paradigm-shifting approach to reducing mortality by targeting a pathway that is central to the development of metastases.


Subject(s)
ERG1 Potassium Channel/metabolism , Epithelial-Mesenchymal Transition , Triple Negative Breast Neoplasms/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cresols/pharmacology , Cresols/therapeutic use , ERG1 Potassium Channel/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , MCF-7 Cells , Mice , Neoplasm Metastasis , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Transplantation, Heterologous , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
19.
Nat Commun ; 10(1): 2860, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253784

ABSTRACT

Lacking targetable molecular drivers, triple-negative breast cancer (TNBC) is the most clinically challenging subtype of breast cancer. In this study, we reveal that Death Effector Domain-containing DNA-binding protein (DEDD), which is overexpressed in > 60% of TNBCs, drives a mitogen-independent G1/S cell cycle transition through cytoplasm localization. The gain of cytosolic DEDD enhances cyclin D1 expression by interacting with heat shock 71 kDa protein 8 (HSC70). Concurrently, DEDD interacts with Rb family proteins and promotes their proteasome-mediated degradation. DEDD overexpression renders TNBCs vulnerable to cell cycle inhibition. Patients with TNBC have been excluded from CDK 4/6 inhibitor clinical trials due to the perceived high frequency of Rb-loss in TNBCs. Interestingly, our study demonstrated that, irrespective of Rb status, TNBCs with DEDD overexpression exhibit a DEDD-dependent vulnerability to combinatorial treatment with CDK4/6 inhibitor and EGFR inhibitor in vitro and in vivo. Thus, our study provided a rationale for the clinical application of CDK4/6 inhibitor combinatorial regimens for patients with TNBC.


Subject(s)
DNA-Binding Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Lapatinib/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , DNA-Binding Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/genetics , ErbB Receptors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Receptor, ErbB-2/antagonists & inhibitors , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Triple Negative Breast Neoplasms/metabolism
20.
Cancer Metab ; 6: 5, 2018.
Article in English | MEDLINE | ID: mdl-29619217

ABSTRACT

BACKGROUND: The complex yet interrelated connections between cancer metabolism and oncogenic driver genes are relatively unexplored but have the potential to identify novel biomarkers and drug targets with prognostic and therapeutic value. The goal of this study was to identify global metabolic profiles of breast tumors isolated from multiple transgenic mouse models and to identify unique metabolic signatures driven by these oncogenes. METHODS: Using mass spectrometry (GC-MS, LC-MS/MS, and capillary zone electrophoresis (CZE)-MS platforms), we quantified and compared the levels of 374 metabolites in breast tissue from normal and transgenic mouse breast cancer models overexpressing a panel of oncogenes (PyMT, PyMT-DB, Wnt1, Neu, and C3-TAg). We also compared the mouse metabolomics data to published human metabolomics data already linked to clinical data. RESULTS: Through analysis of our metabolomics data, we identified metabolic differences between normal and tumor breast tissues as well as metabolic differences unique to each initiating oncogene. We also quantified the metabolic profiles of the mammary fat pad versus mammary epithelium by CZE-MS/MS. However, the differences between the tissues did not account for the majority of the metabolic differences between the normal mammary gland and breast tumor tissues. Therefore, the differences between the cohorts were unlikely due to cellular heterogeneity. Of the mouse models used in this study, C3-TAg was the only cohort with a tumor metabolic signature composed of ten metabolites that had significant prognostic value in breast cancer patients. Gene expression analysis identified candidate genes that may contribute to the metabolic reprogramming. CONCLUSIONS: This study identifies oncogene-induced metabolic reprogramming within mouse breast tumors and compares the results to that of human breast tumors, providing a unique look at the relationship between and clinical value of oncogene initiation and metabolism during breast cancer.

SELECTION OF CITATIONS
SEARCH DETAIL