Try a new search

Format these results:

Searched for:

person:logans02

Total Results:

69


Design of Peptoid-peptide Macrocycles to Inhibit the β-catenin TCF Interaction in Prostate Cancer

Schneider, Jeffrey A; Craven, Timothy W; Kasper, Amanda C; Yun, Chi; Haugbro, Michael; Briggs, Erica M; Svetlov, Vladimir; Nudler, Evgeny; Knaut, Holger; Bonneau, Richard; Garabedian, Michael J; Kirshenbaum, Kent; Logan, Susan K
New chemical inhibitors of protein-protein interactions are needed to propel advances in molecular pharmacology. Peptoids are peptidomimetic oligomers with the capability to inhibit protein-protein interactions by mimicking protein secondary structure motifs. Here we report the in silico design of a macrocycle primarily composed of peptoid subunits that targets the β-catenin:TCF interaction. The β-catenin:TCF interaction plays a critical role in the Wnt signaling pathway which is over-activated in multiple cancers, including prostate cancer. Using the Rosetta suite of protein design algorithms, we evaluate how different macrocycle structures can bind a pocket on β-catenin that associates with TCF. The in silico designed macrocycles are screened in vitro using luciferase reporters to identify promising compounds. The most active macrocycle inhibits both Wnt and AR-signaling in prostate cancer cell lines, and markedly diminishes their proliferation. In vivo potential is demonstrated through a zebrafish model, in which Wnt signaling is potently inhibited.
PMID: 30352998
ISSN: 2041-1723
CID: 3384682

Evaluating the effects of HIPK2 inhibition on castration-resistant prostate cancer cells [Meeting Abstract]

Camacho-Hernandez, E M; Thomas, P; Ledet, R; Garabedian, M J; Logan, S K
Prostate cancer is the second leading cause of death in men in the United States. It depends on the signaling by the androgen receptor (AR), which is activated by testosterone. AR signaling promotes normal prostate development as well as cancer. Current treatments for prostate cancer include prostatectomy and androgen deprivation therapy. Although androgen deprivation is effective in halting prostate cancer growth, a group of men are often diagnosed with castration resistant prostate cancer (CRPC), which is refractory to hormone deprivation. Therefore, new targets are needed to stop prostate cancer growth. In this study, we investigated whether Homeodomain-interacting protein kinase 2 (HIPK2) affects AR signaling and prostate cancer cell growth. To do this, we utilized doxycycline-induced shRNA-mediated silencing of HIPK2 expression in 22Rv1 and LNCaP-95, two cell lines that are refractory to androgen deprivation, and demonstrated substantial knockdown of both HIPK2 mRNA and protein. In addition, we observed significant changes in AR target gene expression. To determine the effects of HIPK2 knockdown on AR subcellular localization, we fractionated control and doxycycline-treated 22Rv1 and LNCaP-95 cells. We observed modest changes in chromatin-bound AR upon HIPK2 depletion, suggesting that HIPK2 influences AR chromatin occupancy and in turn AR-dependent transcription. We also tested HIPK2 catalytic inhibitors by examining their effects on 22Rv1 and LNCaP-95 cellular proliferation, and phosphorylation a HIPK2 substrate, SIAH2. We found that HIPK2 inhibitors modestly affected the proliferation and substrate phosphorylation. Thus, HIPK2 can modulate AR activity, highlighting the role of HIPK2 as a new drug target for prostate cancer
EMBASE:622543890
ISSN: 1530-6860
CID: 3160442

Revisiting the role of Wnt/beta-catenin signaling in prostate cancer

Schneider, Jeffrey A; Logan, Susan K
The androgen receptor (AR) is a widely accepted therapeutic target in prostate cancer and multiple studies indicate that the AR and Wnt/beta-catenin pathways intersect. Recent genome-wide analysis of prostate cancer metastases illustrate the importance of the Wnt/beta-catenin pathway in prostate cancer and compel us to reexamine the interaction of the AR and Wnt/beta-catenin signaling pathways. This review includes newer areas of interest such as non-canonical Wnt signaling and the role of Wnts in prostate cancer stem cells. The effort to develop Wnt modulating therapeutics, both biologics and small molecules, is also discussed.
PMCID:5550366
PMID: 28189566
ISSN: 1872-8057
CID: 2449002

Long interspersed nuclear element-1 expression and retrotransposition in prostate cancer cells

Briggs, Erica M; Ha, Susan; Mita, Paolo; Brittingham, Gregory; Sciamanna, Ilaria; Spadafora, Corrado; Logan, Susan K
Background/UNASSIGNED:Long Interspersed Nuclear Element-1 (LINE-1) is an autonomous retrotransposon that generates new genomic insertions through the retrotransposition of a RNA intermediate. Expression of LINE-1 is tightly repressed in most somatic tissues to prevent DNA damage and ensure genomic integrity. However, the reactivation of LINE-1 has been documented in cancer and the role of LINE-1 protein expression and retrotransposition has become of interest in the development, progression, and adaptation of many epithelial neoplasms, including prostate cancer. Results/UNASSIGNED:Here, we examined endogenous LINE-1 protein expression and localization in a panel of prostate cancer cells and observed a diverse range of LINE-1 expression patterns between cell lines. Subcellular localization of LINE-1 proteins, ORF1p and ORF2p, revealed distinct expression patterns. ORF1p, a nucleic acid chaperone that binds LINE-1 mRNA, was predominantly expressed in the cytoplasm, with minor localization in the nucleus. ORF2p, containing endonuclease and reverse transcriptase domains, exhibited punctate foci in the nucleus and also displayed co-localization with PCNA and γH2AX. Using a retrotransposition reporter assay, we found variations in LINE-1 retrotransposition between cell lines. Conclusions/UNASSIGNED:Overall, our findings reveal new insight into the expression and retrotransposition of LINE-1 in prostate cancer. The prostate cancer cells we investigated provide a unique model for investigating endogenous LINE-1 activity and provide a functional model for studying LINE-1 mechanisms in prostate cancer.
PMCID:5753491
PMID: 29308092
ISSN: 1759-8753
CID: 2905782

UXT is required for spermatogenesis in mice

Schafler, Eric D; Thomas, Phillip A; Ha, Susan; Wang, Yu; Bermudez-Hernandez, Keria; Tang, Zuojian; Fenyo, David; Vigodner, Margarita; Logan, Susan K
Male mammals must simultaneously produce prodigious numbers of sperm and maintain an adequate reserve of stem cells to ensure continuous production of gametes throughout life. Failures in the mechanisms responsible for balancing germ cell differentiation and spermatogonial stem cell (SSC) self-renewal can result in infertility. We discovered a novel requirement for Ubiquitous Expressed Transcript (UXT) in spermatogenesis by developing the first knockout mouse model for this gene. Constitutive deletion of Uxt is embryonic lethal, while conditional knockout in the male germline results in a Sertoli cell-only phenotype during the first wave of spermatogenesis that does not recover in the adult. This phenotype begins to manifest between 6 and 7 days post-partum, just before meiotic entry. Gene expression analysis revealed that Uxt deletion downregulates the transcription of genes governing SSC self-renewal, differentiation, and meiosis, consistent with its previously defined role as a transcriptional co-factor. Our study has revealed the first in vivo function for UXT in the mammalian germline as a regulator of distinct transcriptional programs in SSCs and differentiating spermatogonia.
PMCID:5896988
PMID: 29649254
ISSN: 1932-6203
CID: 3036952

Role of the Unconventional Prefoldin Proteins URI and UXT in Transcription Regulation

Thomas, Phillip A; Mita, Paolo; Ha, Susan; Logan, Susan K
The Unconventional prefoldin RPB5 interacting protein (URI), also known as RPB5-Mediating Protein (RMP) has been shown to play several regulatory roles in different cellular compartments including the mitochondria, as a phosphatase binding protein; in the cytoplasm, as a chaperone-like protein; and in the nucleus, as a transcriptional regulator through binding to RPB5 and RNA polymerase II (polII). This chapter focuses on the role URI plays in transcriptional regulation in the prostate cell. In prostate cells, URI is tightly bound to another prefoldin-like protein called UXT, a known androgen receptor (AR) cofactor. Part of a multiprotein complex, URI and UXT act as transcriptional repressors, and URI regulates KAP1 through PP2A phosphatase activity. The discovery of the interaction of URI and UXT with KAP1, AR, and PP2A, as well as the numerous interactions between URI and components of the R2TP/prefoldin-like complex, RPB5, and nuclear proteins involved in DNA damage response, chromatin remodeling and gene transcription, reveal a pleiotropic effect of the URI/UXT complex on nuclear processes. The mechanisms by which URI/UXT affect transcription, chromatin structure and regulation, and genome stability, remain to be elucidated but will be of fundamental importance considering the many processes affected by alterations of URI/UXT and other prefoldins and prefoldin-like proteins.
PMID: 30484154
ISSN: 0065-2598
CID: 3500632

Chromatin-associated protein SIN3B prevents prostate cancer progression by inducing senescence

Bainor, Anthony J; Deng, Fang-Ming; Wang, Yu; Lee, Peng; Cantor, David J; Logan, Susan K; David, Gregory
Distinguishing between indolent and aggressive prostate adenocarcinoma (PCa) remains a priority to accurately identify patients who need therapeutic intervention. SIN3B has been implicated in the initiation of senescence in vitro Here we show that in a mouse model of prostate cancer, SIN3B provides a barrier to malignant progression. SIN3B was required for PTEN-induced cellular senescence and prevented progression to invasive PCa. Furthermore, SIN3B was downregulated in human PCa correlating with upregulation of its target genes. Our results suggest a tumor suppressor function for SIN3B that limits PCa progression, with potential implications for the use of SIN3B and its target genes as candidate diagnostic markers to distinguish indolent from aggressive disease.
PMCID:5626631
PMID: 28807943
ISSN: 1538-7445
CID: 2670812

URI Regulates KAP1 Phosphorylation and Transcriptional Repression Via PP2A Phosphatase in Prostate Cancer Cells

Mita, Paolo; Savas, Jeffrey N; Briggs, Erica M; Ha, Susan; Gnanakkan, Veena; Yates, John R 3rd; Robins, Diane M; David, Gregory; Boeke, Jef D; Garabedian, Michael J; Logan, Susan K
URI is an unconventional prefoldin, RNA polymerase II interactor that functions as a transcriptional repressor, and is part of a larger nuclear protein complex. The components of this complex and the mechanism of transcriptional repression have not been characterized. Here we show that the KRAB-associated protein 1 (KAP1) and the protein phosphatase PP2A interact with URI. Mechanistically, we show that KAP1 phosphorylation is decreased following recruitment of PP2A by URI. We functionally characterize the novel URI-KAP1-PP2A complex, demonstrating a role of URI in retrotransposon repression, a key function previously demonstrated for the KAP1-SETDB1 complex. Microarray analysis of annotated transposons revealed a selective increase in the transcription of LINE-1 and L1PA2 retroelements upon knockdown of URI. These data unveil a new nuclear function of URI and identify a novel post-transcriptional regulation of KAP1 protein that may have important implications in reactivation of transposable elements in prostate cancer cells.
PMCID:5207251
PMID: 27780869
ISSN: 1083-351x
CID: 2288712

Multivalent peptoid conjugates suppress enzalutamide-resistant prostate cancer cellular proliferation

Wang, Yu; Dehigaspitiya, Dilani C; Levine, Paul M; Profit, Adam A; Haugbro, Michael; Imberg-Kazdan, Keren; Logan, Susan K; Kirshenbaum, Kent; Garabedian, Michael J
Development of resistance to anti-androgens for treating advanced prostate cancer is a growing concern, and extends to recently developed therapeutics, including enzalutamide. Therefore, new strategies to block androgen receptor (AR) function in prostate cancer are required. Here we report the characterization of a multivalent conjugate presenting two bioactive ethisterone ligands arrayed as spatially defined pendant groups on a peptoid oligomer. The conjugate, named Multivalent Peptoid Conjugate 6 (MPC6), suppressed the proliferation of multiple AR-expressing prostate cancer cell lines including those that failed to respond to enzalutamide and ARN509. The structure-activity relationships of MPC6 variants were evaluated, revealing that increased spacing between ethisterone moieties and changes in peptoid topology eliminated its anti-proliferative effect, suggesting that both ethisterone ligand presentation and scaffold characteristics contribute to MPC6 activity. Mechanistically, MPC6 blocked AR coactivator-peptide interaction, and prevented AR intermolecular interactions. Protease sensitivity assays suggested that the MPC6-bound AR induced a receptor conformation distinct from that of dihydrotestosterone- or enzalutamide-bound AR. Pharmacological studies revealed that MPC6 was metabolically stable and displayed a low plasma clearance rate. Notably, MPC6 treatment reduced tumor growth and decreased Ki67 and AR expression in mouse xenograft models of enzalutamide-resistant LNCaP-abl cells. Thus, MPC6 represents a new class of compounds with the potential to combat treatment-resistant prostate cancer.
PMCID:5010535
PMID: 27488525
ISSN: 1538-7445
CID: 2199512

Dynein axonemal heavy chain 8 promotes androgen receptor activity and associates with prostate cancer progression

Wang, Yu; Ledet, Russell J; Imberg-Kazdan, Keren; Logan, Susan K; Garabedian, Michael J
To gain insight into cellular factors regulating AR action that could promote castration resistant prostate cancer (CRPC), we performed a genome-wide RNAi screen for factors that promote ligand-independent AR transcriptional activity and integrated clinical databases for candidate genes that are positively associated with prostate cancer metastasis and recurrence. From this analysis, we identified Dynein Axonemal Heavy Chain 8 (DNAH8) as an AR regulator that displayed higher mRNA expression in metastatic than in primary tumors, and showed high expression in patients with poor prognosis. Axonemal dyneins function in cellular motility, but the function of DNAH8 in prostate cancer or other cell types has not been reported. DNAH8 is on chromosome 6q21.2, a cancer-associated amplicon, and is primarily expressed in prostate and testis. Its expression is higher in primary tumors compared to normal prostate, and is further increased in metastatic prostate cancers. Patients expressing high levels of DNAH8 have a greater risk of relapse and a poor prognosis after prostatectomy. Depletion of DNAH8 in prostate cancer cells suppressed AR transcriptional activity and proliferation. Androgen treatment increased DNAH8 mRNA expression, and AR bound the DNAH8 promoter sequence indicating DNAH8 is an AR target gene. Thus, DNAH8 is a new regulator of AR associated with metastatic tumors and poor prognosis.
PMCID:5226506
PMID: 27363033
ISSN: 1949-2553
CID: 2167082