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










Publication year range
1.
Biochemistry ; 63(1): 1-8, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38086054

ABSTRACT

CBP/p300 is a master transcriptional coactivator that regulates gene activation by interacting with multiple transcriptional activators. Dysregulation of protein-protein interactions (PPIs) between the CBP/p300 KIX domain and its activators is implicated in a number of cancers, including breast, leukemia, and colorectal cancer. However, KIX is typically considered "undruggable" because of its shallow binding surfaces lacking both significant topology and promiscuous binding profiles. We previously reported a dual-targeting peptide (MybLL-tide) that inhibits the KIX-Myb interaction with excellent specificity and potency. Here, we demonstrate a branched, second-generation analogue, CREBLL-tide, that inhibits the KIX-CREB PPI with higher potency and selectivity. Additionally, the best of these CREBLL-tide analogues shows excellent and selective antiproliferation activity in breast cancer cells. These results indicate that CREBLL-tide is an effective tool for assessing the role of KIX-activator interactions in breast cancer and expanding the dual-targeting strategy for inhibiting KIX and other coactivators that contain multiple binding surfaces.


Subject(s)
Breast Neoplasms , CREB-Binding Protein , Humans , Female , Binding Sites , Ligands , CREB-Binding Protein/chemistry , Transcription Factors/metabolism , Protein Binding , Transcriptional Activation , Breast Neoplasms/drug therapy
2.
JCI Insight ; 8(15)2023 08 08.
Article in English | MEDLINE | ID: mdl-37440313

ABSTRACT

Lysine-specific demethylase 1 (LSD1) is a histone demethylase that promotes stemness and cell survival in cancers such as prostate cancer. Most prostate malignancies are adenocarcinomas with luminal differentiation. However, some tumors undergo cellular reprogramming to a more lethal subset termed neuroendocrine prostate cancer (NEPC) with neuronal differentiation. The frequency of NEPC is increasing since the widespread use of potent androgen receptor signaling inhibitors. Currently, there are no effective treatments for NEPC. We previously determined that LSD1 promotes survival of prostate adenocarcinoma tumors. However, the role of LSD1 in NEPC is unknown. Here, we determined that LSD1 is highly upregulated in NEPC versus adenocarcinoma patient tumors. LSD1 suppression with RNAi or allosteric LSD1 inhibitors - but not catalytic inhibitors - reduced NEPC cell survival. RNA-Seq analysis revealed that LSD1 represses pathways linked to luminal differentiation, and TP53 was the top reactivated pathway. We confirmed that LSD1 suppressed the TP53 pathway by reducing TP53 occupancy at target genes while LSD1's catalytic function was dispensable for this effect. Mechanistically, LSD1 inhibition disrupted LSD1-HDAC interactions, increasing histone acetylation at TP53 targets. Finally, LSD1 inhibition suppressed NEPC tumor growth in vivo. These findings suggest that blocking LSD1's noncatalytic function may be a promising treatment strategy for NEPC.


Subject(s)
Adenocarcinoma , Prostatic Neoplasms , Humans , Male , Adenocarcinoma/genetics , Cell Line, Tumor , Histone Demethylases/genetics , Prostatic Neoplasms/pathology , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Adv Nanobiomed Res ; 3(4)2023 Apr.
Article in English | MEDLINE | ID: mdl-37234365

ABSTRACT

Brain metastases are the most lethal progression event, in part because the biological processes underpinning brain metastases are poorly understood. There is a paucity of realistic models of metastasis, as current in vivo murine models are slow to manifest metastasis. We set out to delineate metabolic and secretory modulators of brain metastases by utilizing two models consisting of in vitro microfluidic devices: 1) a blood brain niche (BBN) chip that recapitulates the blood-brain-barrier and niche; and 2) a migration chip that assesses cell migration. We report secretory cues provided by the brain niche that attract metastatic cancer cells to colonize the brain niche region. Astrocytic Dkk-1 is increased in response to brain-seeking breast cancer cells and stimulates cancer cell migration. Brain-metastatic cancer cells under Dkk-1 stimulation increase gene expression of FGF-13 and PLCB1. Further, extracellular Dkk-1 modulates cancer cell migration upon entering the brain niche.

4.
Nat Commun ; 13(1): 5345, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36109521

ABSTRACT

The androgen receptor (AR) signaling inhibitor enzalutamide (enza) is one of the principal treatments for metastatic castration-resistant prostate cancer (CRPC). Several emergent enza clinical resistance mechanisms have been described, including lineage plasticity in which the tumors manifest reduced dependency on the AR. To improve our understanding of enza resistance, herein we analyze the transcriptomes of matched biopsies from men with metastatic CRPC obtained prior to treatment and at progression (n = 21). RNA-sequencing analysis demonstrates that enza does not induce marked, sustained changes in the tumor transcriptome in most patients. However, three patients' progression biopsies show evidence of lineage plasticity. The transcription factor E2F1 and pathways linked to tumor stemness are highly activated in baseline biopsies from patients whose tumors undergo lineage plasticity. We find a gene signature enriched in these baseline biopsies that is strongly associated with poor survival in independent patient cohorts and with risk of castration-induced lineage plasticity in patient-derived xenograft models, suggesting that tumors harboring this gene expression program may be at particular risk for resistance mediated by lineage plasticity and poor outcomes.


Subject(s)
E2F1 Transcription Factor , Prostatic Neoplasms, Castration-Resistant , Androgen Receptor Antagonists/pharmacology , Benzamides , Biopsy , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , E2F1 Transcription Factor/metabolism , Humans , Male , Nitriles , Phenylthiohydantoin , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
5.
Front Endocrinol (Lausanne) ; 13: 926585, 2022.
Article in English | MEDLINE | ID: mdl-35909568

ABSTRACT

The androgen receptor (AR) signaling pathway is critical for growth and differentiation of prostate cancer cells. For that reason, androgen deprivation therapy with medical or surgical castration is the principal treatment for metastatic prostate cancer. More recently, new potent AR signaling inhibitors (ARSIs) have been developed. These drugs improve survival for men with metastatic castration-resistant prostate cancer (CRPC), the lethal form of the disease. However, ARSI resistance is nearly universal. One recently appreciated resistance mechanism is lineage plasticity or switch from an AR-driven, luminal differentiation program to an alternate differentiation program. Importantly, lineage plasticity appears to be increasing in incidence in the era of new ARSIs, strongly implicating AR suppression in this process. Lineage plasticity and shift from AR-driven tumors occur on a continuum, ranging from AR-expressing tumors with low AR activity to AR-null tumors that have activation of alternate differentiation programs versus the canonical luminal program found in AR-driven tumors. In many cases, AR loss coincides with the activation of a neuronal program, most commonly exemplified as therapy-induced neuroendocrine prostate cancer (t-NEPC). While genetic events clearly contribute to prostate cancer lineage plasticity, it is also clear that epigenetic events-including chromatin modifications and DNA methylation-play a major role. Many epigenetic factors are now targetable with drugs, establishing the importance of clarifying critical epigenetic factors that promote lineage plasticity. Furthermore, epigenetic marks are readily measurable, demonstrating the importance of clarifying which measurements will help to identify tumors that have undergone or are at risk of undergoing lineage plasticity. In this review, we discuss the role of AR pathway loss and activation of a neuronal differentiation program as key contributors to t-NEPC lineage plasticity. We also discuss new epigenetic therapeutic strategies to reverse lineage plasticity, including those that have recently entered clinical trials.


Subject(s)
Carcinoma, Neuroendocrine , Prostatic Neoplasms , Androgen Antagonists/therapeutic use , Carcinoma, Neuroendocrine/pathology , Epigenesis, Genetic , Humans , Male , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
6.
Front Oncol ; 11: 712041, 2021.
Article in English | MEDLINE | ID: mdl-34513691

ABSTRACT

Metastases are the leading cause of death in cancer patients. RhoC, a member of the Rho GTPase family, has been shown to facilitate metastasis of aggressive breast cancer cells by influencing motility, invasion, and chemokine secretion, but as yet there is no integrated model of the precise mechanism of how RhoC promotes metastasis. A common phenotypic characteristic of metastatic cells influenced by these mechanisms is dysregulation of cell-cell junctions. Thus, we set out to study how RhoA- and RhoC-GTPase influence the cell-cell junctions in aggressive breast cancers. We demonstrate that CRISPR-Cas9 knockout of RhoC in SUM 149 and MDA 231 breast cancer cells results in increased normalization of junctional integrity denoted by junction protein expression/colocalization. In functional assessments of junction stability, RhoC knockout cells have increased barrier integrity and increased cell-cell adhesion compared to wild-type cells. Whole exome RNA sequencing and targeted gene expression profiling demonstrate decreased expression of Type I interferon-stimulated genes in RhoC knockout cells compared to wild-type, and subsequent treatment with interferon-alpha resulted in significant increases in adhesion and decreases in invasiveness of wild-type cells and a dampened response to interferon-alpha stimulation with respect to adhesion and invasiveness in RhoC knockout cells. We delineate a key role of RhoC-GTPase in modulation of junctions and response to interferon, which supports inhibition of RhoC as a potential anti-invasion therapeutic strategy.

7.
J Am Chem Soc ; 143(25): 9297-9302, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34137598

ABSTRACT

Inhibitors of transcriptional protein-protein interactions (PPIs) have high value both as tools and for therapeutic applications. The PPI network mediated by the transcriptional coactivator Med25, for example, regulates stress-response and motility pathways, and dysregulation of the PPI networks contributes to oncogenesis and metastasis. The canonical transcription factor binding sites within Med25 are large (∼900 Å2) and have little topology, and thus, they do not present an array of attractive small-molecule binding sites for inhibitor discovery. Here we demonstrate that the depsidone natural product norstictic acid functions through an alternative binding site to block Med25-transcriptional activator PPIs in vitro and in cell culture. Norstictic acid targets a binding site comprising a highly dynamic loop flanking one canonical binding surface, and in doing so, it both orthosterically and allosterically alters Med25-driven transcription in a patient-derived model of triple-negative breast cancer. These results highlight the potential of Med25 as a therapeutic target as well as the inhibitor discovery opportunities presented by structurally dynamic loops within otherwise challenging proteins.


Subject(s)
Lactones/pharmacology , Mediator Complex/metabolism , Protein Binding/drug effects , Salicylates/pharmacology , Transcription, Genetic/drug effects , Allosteric Regulation , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Mediator Complex/chemistry , Molecular Dynamics Simulation , Protein Domains , Transcription Factors/metabolism
8.
Clin Cancer Res ; 27(17): 4923-4936, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34145028

ABSTRACT

PURPOSE: Lineage plasticity in prostate cancer-most commonly exemplified by loss of androgen receptor (AR) signaling and a switch from a luminal to alternate differentiation program-is now recognized as a treatment resistance mechanism. Lineage plasticity is a spectrum, but neuroendocrine prostate cancer (NEPC) is the most virulent example. Currently, there are limited treatments for NEPC. Moreover, the incidence of treatment-emergent NEPC (t-NEPC) is increasing in the era of novel AR inhibitors. In contradistinction to de novo NEPC, t-NEPC tumors often express the AR, but AR's functional role in t-NEPC is unknown. Furthermore, targetable factors that promote t-NEPC lineage plasticity are also unclear. EXPERIMENTAL DESIGN: Using an integrative systems biology approach, we investigated enzalutamide-resistant t-NEPC cell lines and their parental, enzalutamide-sensitive adenocarcinoma cell lines. The AR is still expressed in these t-NEPC cells, enabling us to determine the role of the AR and other key factors in regulating t-NEPC lineage plasticity. RESULTS: AR inhibition accentuates lineage plasticity in t-NEPC cells-an effect not observed in parental, enzalutamide-sensitive adenocarcinoma cells. Induction of an AR-repressed, lineage plasticity program is dependent on activation of the transcription factor E2F1 in concert with the BET bromodomain chromatin reader BRD4. BET inhibition (BETi) blocks this E2F1/BRD4-regulated program and decreases growth of t-NEPC tumor models and a subset of t-NEPC patient tumors with high activity of this program in a BETi clinical trial. CONCLUSIONS: E2F1 and BRD4 are critical for activating an AR-repressed, t-NEPC lineage plasticity program. BETi is a promising approach to block this program.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , E2F1 Transcription Factor/drug effects , E2F1 Transcription Factor/physiology , Nitriles/therapeutic use , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/drug therapy , Proteins/antagonists & inhibitors , Cell Line, Tumor , Humans , Male
9.
Eur Urol ; 80(1): 71-81, 2021 07.
Article in English | MEDLINE | ID: mdl-33785255

ABSTRACT

CONTEXT: In addition to genetic alterations, epigenetic alterations play a crucial role during prostate cancer progression. A better understanding of the epigenetic factors that promote prostate cancer progression may lead to the design of rational therapeutic strategies to target prostate cancer more effectively. OBJECTIVE: To systematically review recent literature on the role of epigenetic factors in prostate cancer and highlight key preclinical and translational data with epigenetic therapies. EVIDENCE ACQUISITION: We performed a systemic literature search in PubMed. At the request of the editors, we limited our search to articles published between January 2015 and August 2020 in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. Clinical trials targeting epigenetic factors were retrieved from clinicaltrials.gov. EVIDENCE SYNTHESIS: We retrieved 1451 articles, and 62 were finally selected for review. Twelve additional foundational studies outside this time frame were also included. Findings from both preclinical and clinical studies were reviewed and summarized. We also discuss 12 ongoing clinical studies with epigenetic targeted therapies. CONCLUSIONS: Epigenetic mechanisms impact prostate cancer progression. Understanding the role of specific epigenetic factors is critical to determine how we may improve prostate cancer treatment and modulate resistance to standard therapies. Recent preclinical studies and ongoing or completed clinical studies with epigenetic therapies provide a useful roadmap for how to best deploy epigenetic therapies clinically to target prostate cancer. PATIENT SUMMARY: Epigenetics is a process by which gene expression is regulated without changes in the DNA sequence itself. Oftentimes, epigenetic changes influence cellular behavior and contribute to cancer development or progression. Understanding how epigenetic changes occur in prostate cancer is the first step toward therapeutic targeting in patients. Importantly, laboratory-based studies and recently completed and ongoing clinical trials suggest that drugs targeting epigenetic factors are promising. More work is necessary to determine whether this class of drugs will add to our existing treatment arsenal in prostate cancer.


Subject(s)
Pharmaceutical Preparations , Prostatic Neoplasms , Biomarkers , DNA Methylation , Epigenesis, Genetic , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics
10.
SLAS Technol ; 26(2): 200-208, 2021 04.
Article in English | MEDLINE | ID: mdl-33183152

ABSTRACT

It is often desirable to evaluate the ability of cells to move in an unrestricted manner in multiple directions without chemical gradients. By combining the standard radial migration assay with injection-molded gaskets and a rigid fixture, we have developed a highly reliable and sensitive method for observing and measuring radial cell migration. This method is adapted for use on high-throughput automated imaging systems. The use of injection-molded gaskets enables low-cost replacement of cell-wetted components. Moreover, the design enables secondary placement of attractants and co-cultures. This device and its enhanced throughput permit the use of therapeutic screening to evaluate phenotypic responses, for example, cancer cell migration response due to drugs or chemical signals. This approach is orthogonal to other 2D cell migration applications, such as scratch wound assays, although here we offer a noninvasive, enhanced-throughput device, which currently is not commercially available but is easily constructed. The proposed device is a systematic, reliable, rapid application to monitor phenotypic responses to chemotherapeutic screens, genetic alterations (e.g., RNAi and CRISPR), supplemental regimens, and other approaches, offering a reliable methodology to survey unbiased and noninvasive cell migration.


Subject(s)
Neoplasms , Biological Assay , Cell Movement , Humans
11.
Neoplasia ; 22(6): 253-262, 2020 06.
Article in English | MEDLINE | ID: mdl-32403054

ABSTRACT

Neuroendocrine prostate cancer (NEPC) is the most virulent form of prostate cancer. Importantly, our recent work examining metastatic biopsy samples demonstrates NEPC is increasing in frequency. In contrast to prostate adenocarcinomas that express a luminal gene expression program, NEPC tumors express a neuronal gene expression program. Despite this distinction, the diagnosis of NEPC is often challenging, demonstrating an urgent need to identify new biomarkers and therapeutic targets. Our prior work demonstrated that the histone demethylase LSD1 (KDM1A) is important for survival of prostate adenocarcinomas, but little was known about LSD1's role in NEPC. Recently, a neural-specific transcript variant of LSD1-LSD1+8a-was discovered and demonstrated to activate neuronal gene expression in neural cells. The splicing factor SRRM4 was previously shown to promote LSD1+8a splicing in neuronal cells, and SRRM4 promotes NEPC differentiation and cell survival. Therefore, we sought to determine if LSD1+8a might play a role in NEPC and whether LSD1+8a splicing was linked to SRRM4. To investigate a potential role for LSD1+8a in NEPC, we examined a panel of prostate adenocarcinoma and NEPC patient-derived xenografts and metastatic biopsies. LSD1+8a was expressed exclusively in NEPC samples and correlated significantly with elevated expression of SRRM4. Using SRRM4-overexpressing cell lines, we determined that SRRM4 mediates alternative splicing of LSD1+8a. Finally, using gain of function studies, we confirmed that LSD1+8a and SRRM4 co-regulate target genes distinct from canonical LSD1. Our findings suggest further study of the interplay between SRRM4 and LSD1+8a and mechanisms by which LSD1+8a regulates gene expression in NEPC is warranted.


Subject(s)
Histone Demethylases/genetics , Nerve Tissue Proteins/genetics , Neuroendocrine Tumors/genetics , Prostatic Neoplasms/genetics , Alternative Splicing/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Epigenomics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Neuroendocrine Tumors/pathology
12.
Breast Cancer Res Treat ; 179(2): 337-347, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31655920

ABSTRACT

PURPOSE: There is a need for biomarkers of drug efficacy for targeted therapies in triple-negative breast cancer (TNBC). As a step toward this, we identify multi-omic molecular determinants of anti-TNBC efficacy in cell lines for a panel of oncology drugs. METHODS: Using 23 TNBC cell lines, drug sensitivity scores (DSS3) were determined using a panel of investigational drugs and drugs approved for other indications. Molecular readouts were generated for each cell line using RNA sequencing, RNA targeted panels, DNA sequencing, and functional proteomics. DSS3 values were correlated with molecular readouts using a FDR-corrected significance cutoff of p* < 0.05 and yielded molecular determinant panels that predict anti-TNBC efficacy. RESULTS: Six molecular determinant panels were obtained from 12 drugs we prioritized based on their efficacy. Determinant panels were largely devoid of DNA mutations of the targeted pathway. Molecular determinants were obtained by correlating DSS3 with molecular readouts. We found that co-inhibiting molecular correlate pathways leads to robust synergy across many cell lines. CONCLUSIONS: These findings demonstrate an integrated method to identify biomarkers of drug efficacy in TNBC where DNA predictions correlate poorly with drug response. Our work outlines a framework for the identification of novel molecular determinants and optimal companion drugs for combination therapy based on these correlates.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/etiology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Computational Biology/methods , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Female , Gene Expression Profiling , Humans , Mutation , Proteomics , Treatment Outcome , Triple Negative Breast Neoplasms/metabolism
13.
PLoS One ; 14(9): e0220973, 2019.
Article in English | MEDLINE | ID: mdl-31536495

ABSTRACT

In breast cancer, tumor hypoxia has been linked to poor prognosis and increased metastasis. Hypoxia activates transcriptional programs in cancer cells that lead to increased motility and invasion, as well as various metabolic changes. One of these metabolic changes, an increase in glycogen metabolism, has been further associated with protection from reactive oxygen species damage that may lead to premature senescence. Here we report that breast cancer cells significantly increase glycogen stores in response to hypoxia. We found that knockdown of the brain isoform of an enzyme that catalyzes glycogen breakdown, glycogen phosphorylase B (PYGB), but not the liver isoform, PYGL, inhibited glycogen utilization in estrogen receptor negative and positive breast cancer cells; whereas both independently inhibited glycogen utilization in the normal-like breast epithelial cell line MCF-10A. Functionally, PYGB knockdown and the resulting inhibition of glycogen utilization resulted in significantly decreased wound-healing capability in MCF-7 cells and a decrease in invasive potential of MDA-MB-231 cells. Thus, we identify PYGB as a novel metabolic target with potential applications in the management and/or prevention of metastasis in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Glycogen/metabolism , Hypoxia/metabolism , Phenotype , Phosphorylase b/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Metabolic Networks and Pathways , Neoplasm Metastasis , Neoplasm Staging , Phosphorylase b/genetics , Protein Isoforms , RNA Interference , RNA, Small Interfering/genetics
14.
Front Oncol ; 9: 456, 2019.
Article in English | MEDLINE | ID: mdl-31214501

ABSTRACT

Tumor associated macrophages (TAMs) are increasingly recognized as major contributors to the metastatic progression of breast cancer and enriched levels of TAMs often correlate with poor prognosis. Despite our current advances it remains unclear which subset of M2-like macrophages have the highest capacity to enhance the metastatic program and which mechanisms regulate this process. Effective targeting of macrophages that aid cancer progression requires knowledge of the specific mechanisms underlying their pro-metastatic actions, as to avoid the anticipated toxicities from generalized targeting of macrophages. To this end, we set out to understand the relationship between the regulation of tumor secretions by Rho-GTPases, which were previously demonstrated to affect them, macrophage differentiation, and the converse influence of macrophages on cancer cell phenotype. Our data show that IL-4/IL-13 in vitro differentiated M2a macrophages significantly increase migratory and invasive potential of breast cancer cells at a greater rate than M2b or M2c macrophages. Our previous work demonstrated that the Rho-GTPases are potent regulators of macrophage-induced migratory responses; therefore, we examined M2a-mediated responses in RhoA or RhoC knockout breast cancer cell models. We find that both RhoA and RhoC regulate migration and invasion in MDA-MB-231 and SUM-149 cells following stimulation with M2a conditioned media. Secretome analysis of M2a conditioned media reveals high levels of vascular endothelial growth factor (VEGF) and chemokine (C-C motif) ligand 18 (CCL-18). Results from our functional assays reveal that M2a TAMs synergistically utilize VEGF and CCL-18 to promote migratory and invasive responses. Lastly, we show that pretreatment with ROCK inhibitors Y-276332 or GSK42986A attenuated VEGF/CCL-18 and M2a-induced migration and invasion. These results support Rho-GTPase signaling regulates downstream responses induced by TAMs, offering a novel approach for the prevention of breast cancer metastasis by anti-RhoA/C therapies.

15.
J Cell Sci ; 131(8)2018 04 26.
Article in English | MEDLINE | ID: mdl-29588397

ABSTRACT

Defective endocytosis and vesicular trafficking of signaling receptors has recently emerged as a multifaceted hallmark of malignant cells. Clathrin-coated pits (CCPs) display highly heterogeneous dynamics on the plasma membrane where they can take from 20 s to over 1 min to form cytosolic coated vesicles. Despite the large number of cargo molecules that traffic through CCPs, it is not well understood whether signaling receptors activated in cancer, such as epidermal growth factor receptor (EGFR), are regulated through a specific subset of CCPs. The signaling lipid phosphatidylinositol (3,4,5)-trisphosphate [PI(3,4,5)P3], which is dephosphorylated by phosphatase and tensin homolog (PTEN), is a potent tumorigenic signaling lipid. By using total internal reflection fluorescence microscopy and automated tracking and detection of CCPs, we found that EGF-bound EGFR and PTEN are enriched in a distinct subset of short-lived CCPs that correspond with clathrin-dependent EGF-induced signaling. We demonstrated that PTEN plays a role in the regulation of CCP dynamics. Furthermore, increased PI(3,4,5)P3 resulted in higher proportion of short-lived CCPs, an effect that recapitulates PTEN deletion. Altogether, our findings provide evidence for the existence of short-lived 'signaling-capable' CCPs.


Subject(s)
Coated Pits, Cell-Membrane/metabolism , ErbB Receptors/metabolism , PTEN Phosphohydrolase/genetics , Humans , Signal Transduction
16.
Sci Rep ; 6: 39190, 2016 12 19.
Article in English | MEDLINE | ID: mdl-27991524

ABSTRACT

Inflammatory breast cancer (IBC) is the most lethal form of breast cancer. All IBC patients have lymph node involvement and one-third of patients already have distant metastasis at diagnosis. This propensity for metastasis is a hallmark of IBC distinguishing it from less lethal non-inflammatory breast cancers (nIBC). Genetic profiling studies have been conducted to differentiate IBC from nIBC, but no IBC cancer-cell-specific gene signature has been identified. We hypothesized that a tumor-extrinsic factor, notably tumor-associated macrophages, promotes and contributes to IBC's extreme metastatic phenotype. To this end, we studied the effect of macrophage-conditioned media (MCM) on IBC. We show that two IBC cell lines are hyper-responsive to MCM as compared to normal-like breast and aggressive nIBC cell lines. We further interrogated IBC's hyper-responsiveness to MCM using a microfluidic migration device, which permits individual cell migration path tracing. We found the MCM "primes" the IBC cells' cellular machinery to become extremely migratory in response to a chemoattractant. We determined that interleukins -6, -8, and -10 within the MCM are sufficient to stimulate this enhanced IBC migration effect, and that the known metastatic oncogene, RhoC GTPase, is necessary for the enhanced migration response.


Subject(s)
Inflammatory Breast Neoplasms/pathology , Macrophages/metabolism , rhoC GTP-Binding Protein/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Chemotactic Factors/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Female , Humans , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/metabolism , Interleukin-10/analysis , Interleukin-10/metabolism , Interleukin-10/pharmacology , Interleukin-6/analysis , Interleukin-6/metabolism , Interleukin-6/pharmacology , Interleukin-8/analysis , Interleukin-8/metabolism , Interleukin-8/pharmacology , Macrophages/cytology , Microfluidics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects
17.
J Biol Chem ; 291(26): 13715-29, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129239

ABSTRACT

Inflammatory breast cancer (IBC) is an extremely lethal cancer that rapidly metastasizes. Although the molecular attributes of IBC have been described, little is known about the underlying metabolic features of the disease. Using a variety of metabolic assays, including (13)C tracer experiments, we found that SUM149 cells, the primary in vitro model of IBC, exhibit metabolic abnormalities that distinguish them from other breast cancer cells, including elevated levels of N-acetylaspartate, a metabolite primarily associated with neuronal disorders and gliomas. Here we provide the first evidence of N-acetylaspartate in breast cancer. We also report that the oncogene RhoC, a driver of metastatic potential, modulates glutamine and N-acetylaspartate metabolism in IBC cells in vitro, revealing a novel role for RhoC as a regulator of tumor cell metabolism that extends beyond its well known role in cytoskeletal rearrangement.


Subject(s)
Aspartic Acid/analogs & derivatives , Glutamine/metabolism , Inflammatory Breast Neoplasms/metabolism , Neoplasm Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Aspartic Acid/biosynthesis , Aspartic Acid/genetics , Cell Line, Tumor , Female , Glutamine/genetics , Humans , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/pathology , Neoplasm Proteins/genetics , rho GTP-Binding Proteins/genetics , rhoC GTP-Binding Protein
18.
Mol Endocrinol ; 24(6): 1165-74, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20308527

ABSTRACT

The androgen receptor (AR) mediates the effect of androgens through its transcriptional function during both normal prostate development and in the emergence and progression of prostate cancer. AR is known to assemble coactivator complexes at target promoters to facilitate transcriptional activation in response to androgens. Here we identify the ATP-dependent chromatin remodeling factor chromodomain helicase DNA-binding protein 8 (CHD8) as a novel coregulator of androgen-responsive transcription. We demonstrate that CHD8 directly associates with AR and that CHD8 and AR simultaneously localize to the TMPRSS2 enhancer after androgen treatment. In the LNCaP cell line, reduction of CHD8 levels by small interfering RNA treatment severely diminishes androgen-dependent activation of the TMPRSS2 gene. We demonstrate that the recruitment of AR to the TMPRSS2 promoter in response to androgen treatment requires CHD8. Finally, CHD8 facilitates androgen-stimulated proliferation of LNCaP cells, emphasizing the physiological importance of CHD8. Taken together, we present evidence of a functional role for CHD8 in AR-mediated transcriptional regulation of target genes.


Subject(s)
Androgens/pharmacology , Chromatin Assembly and Disassembly/drug effects , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Protein Binding/drug effects , Protein Transport/drug effects , Receptors, Androgen/metabolism , Response Elements/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Transcription Factors/genetics
19.
FEBS Lett ; 584(4): 689-93, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20085832

ABSTRACT

Chromodomain, helicase, DNA-binding protein 8 (CHD8) is an ATP-dependent chromatin remodeling enzyme that has been demonstrated to exist within a large protein complex which includes WDR5, Ash2L, and RbBP5, members of the Mixed Lineage Leukemia (MLL) histone modifying complexes. Here we show that CHD8 relocalizes to the promoter of the MLL regulated gene HOXA2 upon gene activation. Depletion of CHD8 enhances HOXA2 expression under activating conditions. Furthermore, depletion of CHD8 results in a loss of the WDR5/Ash2L/RbBP5 subcomplex, and consequently H3K4 trimethylation, at the HOXA2 promoter. These studies suggest that CHD8 alters HOXA2 gene expression and regulates the recruitment of chromatin modifying enzymes.


Subject(s)
Adenosine Triphosphate/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Blotting, Western , Cell Line, Tumor , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lysine/metabolism , Methylation , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Tretinoin/pharmacology
20.
Gene ; 400(1-2): 16-24, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17601686

ABSTRACT

Telomerase is essential for maintaining telomere length and chromosome stability in most eukaryotic organisms. The telomerase ribonucleoprotein complex consists of two essential components, the catalytic telomerase reverse transcriptase protein (TERT) and the intrinsic telomerase RNA. The sea squirts, as urochordates, occupy a key position in the phylogenetic tree of the chordates: they diverged from the other chordates just before the lineage of vertebrates, and thus provide special insight into the origin and evolution of vertebrate genes. Here, we report the cloning and characterization of TERT genes from two sea squirts, Ciona intestinalis and Ciona savignyi. The C. intestinalis TERT (CinTERT) gene encodes 907 amino acids and consists of 17 exons, which are similar to vertebrate TERT genes. The C. savignyi TERT (CsaTERT) gene encodes 843 amino acids, but surprisingly does not contain any introns. Both Ciona TERTs contain all of the reverse transcriptase (RT) motifs (1, 2, A, B, C, D, and E) that are typically present in telomerase and viral RTs. Interestingly, the alignment of Ciona and vertebrate TERT sequences reveals a previously unknown motif, named motif 3, located between motifs 2 and A. The Ciona TERT gene is expressed in all tissues analyzed except the brain and heart. This is the first report of the TERT gene in invertebrate chordates.


Subject(s)
Telomerase/genetics , Urochordata/enzymology , Urochordata/genetics , Amino Acid Sequence , Animals , Base Sequence , Gene Expression , Molecular Sequence Data , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Telomerase/chemistry , Vertebrates/genetics
SELECTION OF CITATIONS
SEARCH DETAIL