Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
iScience ; 27(2): 108984, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38327800

ABSTRACT

Olaparib is a pioneering PARP inhibitor (PARPi) approved for treating castration-resistant prostate cancer (CRPC) tumors harboring DNA repair defects, but clinical resistance has been documented. To study acquired resistance, we developed Olaparib-resistant (OlapR) cell lines through chronic Olaparib treatment of LNCaP and C4-2B cell lines. Here, we found that IGFBP3 is highly expressed in acquired (OlapR) and intrinsic (Rv1) models of Olaparib resistance. We show that IGFBP3 expression promotes Olaparib resistance by enhancing DNA repair capacity through activation of EGFR and DNA-PKcs. IGFBP3 depletion enhances efficacy of Olaparib by promoting DNA damage accumulation and subsequently, cell death in resistant models. Mechanistically, we show that silencing IGFBP3 or EGFR expression reduces cell viability and resensitizes OlapR cells to Olaparib treatment. Inhibition of EGFR by Gefitinib suppressed growth of OlapR cells and improved Olaparib sensitivity, thereby phenocopying IGFBP3 inhibition. Collectively, our results highlight IGFBP3 and EGFR as critical mediators of Olaparib resistance.

2.
J Biol Chem ; 299(8): 104973, 2023 08.
Article in English | MEDLINE | ID: mdl-37380074

ABSTRACT

Prostate cancer is initially regulated by the androgen receptor (AR), a ligand-activated, transcription factor, and is in a hormone-dependent state (hormone-sensitive prostate cancer (HSPC)), but eventually becomes androgen-refractory (castration-resistant prostate cancer (CRPC)) because of mechanisms that bypass the AR, including by activation of ErbB3, a member of the epidermal growth factor receptor family. ErbB3 is synthesized in the cytoplasm and transported to the plasma membrane for ligand binding and dimerization, where it regulates downstream signaling, but nuclear forms are reported. Here, we demonstrate in prostatectomy samples that ErbB3 nuclear localization is observed in malignant, but not benign prostate, and that cytoplasmic (but not nuclear) ErbB3 correlated positively with AR expression but negatively with AR transcriptional activity. In support of the latter, androgen depletion upregulated cytoplasmic, but not nuclear ErbB3, while in vivo studies showed that castration suppressed ErbB3 nuclear localization in HSPC, but not CRPC tumors. In vitro treatment with the ErbB3 ligand heregulin-1ß (HRG) induced ErbB3 nuclear localization, which was androgen-regulated in HSPC but not in CRPC. In turn, HRG upregulated AR transcriptional activity in CRPC but not in HSPC cells. Positive correlation between ErbB3 and AR expression was demonstrated in AR-null PC-3 cells where stable transfection of AR restored HRG-induced ErbB3 nuclear transport, while AR knockdown in LNCaP reduced cytoplasmic ErbB3. Mutations of ErbB3's kinase domain did not affect its localization but was responsible for cell viability in CRPC cells. Taken together, we conclude that AR expression regulated ErbB3 expression, its transcriptional activity suppressed ErbB3 nuclear translocation, and HRG binding to ErbB3 promoted it.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Receptors, Androgen , Humans , Male , Androgens/metabolism , Cell Line, Tumor , Ligands , Neuregulin-1/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
3.
Am J Clin Exp Urol ; 10(5): 299-310, 2022.
Article in English | MEDLINE | ID: mdl-36313205

ABSTRACT

Resistance to androgen receptor (AR) targeted therapies remains as the main reason for most prostate cancer related deaths. Lineage plasticity resulting in altered, treatment insensitive prostate tumor cell phenotypes such neuroendocrine differentiated prostate cancer is a common manifestation within resistant tumors upon AR-targeted therapies. The mechanisms responsible for lineage plasticity in prostate cancer remain incompletely understood. Here we demonstrate that the enzalutamide resistant MDVR cell line possesses lineage plastic characteristics associated with overexpression of the Wnt transporter Wntless (WLS). Furthermore, we present evidence that overexpression of WLS is common in varying cell line models of lineage plastic prostate cancer, is higher in neuroendocrine patient samples, and positively correlates with the neuroendocrine marker SYP in clinical data. Targeting WLS in lineage plastic cellular models reduces viability and represses lineage plasticity associated gene expression. Our study provides insight into the importance of WLS to the development of lethal resistant prostate cancer and provides a potential target for the treatment of advanced disease.

4.
Commun Med (Lond) ; 2: 118, 2022.
Article in English | MEDLINE | ID: mdl-36159187

ABSTRACT

Background: Treatment-emergent neuroendocrine prostate cancer (NEPC) after androgen receptor (AR) targeted therapies is an aggressive variant of prostate cancer with an unfavorable prognosis. The underlying mechanisms for early neuroendocrine differentiation are poorly defined and diagnostic and prognostic biomarkers are needed. Methods: We performed transcriptomic analysis on the enzalutamide-resistant prostate cancer cell line C4-2B MDVR and NEPC patient databases to identify neural lineage signature (NLS) genes. Correlation of NLS genes with clinicopathologic features was determined. Cell viability was determined in C4-2B MDVR and H660 cells after knocking down ARHGEF2 using siRNA. Organoid viability of patient-derived xenografts was measured after knocking down ARHGEF2. Results: We identify a 95-gene NLS representing the molecular landscape of neural precursor cell proliferation, embryonic stem cell pluripotency, and neural stem cell differentiation, which may indicate an early or intermediate stage of neuroendocrine differentiation. These NLS genes positively correlate with conventional neuroendocrine markers such as chromogranin and synaptophysin, and negatively correlate with AR and AR target genes in advanced prostate cancer. Differentially expressed NLS genes stratify small-cell NEPC from prostate adenocarcinoma, which are closely associated with clinicopathologic features such as Gleason Score and metastasis status. Higher ARGHEF2, LHX2, and EPHB2 levels among the 95 NLS genes correlate with a shortened survival time in NEPC patients. Furthermore, downregulation of ARHGEF2 gene expression suppresses cell viability and markers of neuroendocrine differentiation in enzalutamide-resistant and neuroendocrine cells. Conclusions: The 95 neural lineage gene signatures capture an early molecular shift toward neuroendocrine differentiation, which could stratify advanced prostate cancer patients to optimize clinical treatment and serve as a source of potential therapeutic targets in advanced prostate cancer.

5.
Mol Cancer Ther ; 21(10): 1594-1607, 2022 10 07.
Article in English | MEDLINE | ID: mdl-35930737

ABSTRACT

The next-generation antiandrogen drugs such as enzalutamide and abiraterone extend survival times and improve quality of life in patients with advanced prostate cancer. However, resistance to both drugs occurs frequently through mechanisms that are incompletely understood. Wnt signaling, particularly through Wnt5a, plays vital roles in promoting prostate cancer progression and induction of resistance to enzalutamide and abiraterone. Development of novel strategies targeting Wnt5a to overcome resistance is an urgent need. In this study, we demonstrated that Wnt5a/FZD2-mediated noncanonical Wnt pathway is overexpressed in enzalutamide-resistant prostate cancer. In patient databases, both the levels of Wnt5a and FZD2 expression are upregulated upon the development of enzalutamide resistance and correlate with higher Gleason score, biochemical recurrence, and metastatic status, and with shortened disease-free survival duration. Blocking Wnt5a/FZD2 signal transduction not only diminished the activation of noncanonical Wnt signaling pathway, but also suppressed the constitutively activated androgen receptor (AR) and AR variants. Furthermore, we developed a novel bioengineered BERA-Wnt5a siRNA construct and demonstrated that inhibition of Wnt5a expression by the BERA-Wnt5a siRNA significantly suppressed tumor growth and enhanced enzalutamide treatment in vivo. These results indicate that Wnt5a/FZD2 signal pathway plays a critical role in promoting enzalutamide resistance, and targeting this pathway by BERA-Wnt5a siRNA can be developed as a potential therapy to treat advanced prostate cancer.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Androgen Antagonists/pharmacology , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Frizzled Receptors/therapeutic use , Humans , Male , Nitriles/therapeutic use , Phenylthiohydantoin , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Small Interfering/therapeutic use , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Wnt Signaling Pathway , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism
6.
Mol Cancer Ther ; 21(4): 677-685, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35086956

ABSTRACT

PARP inhibition represents the dawn of precision medicine for treating prostate cancer. Despite this advance, questions remain regarding the use of PARP inhibitors (PARPi) for the treatment of this disease, including (i) how specifically do PARPi-sensitive tumor cells respond to treatment, and (ii) how does PARPi resistance develop? To address these questions, we characterized response to olaparib in sensitive LNCaP and C4-2B cells and developed two olaparib-resistant derivative cell line models from each, termed LN-OlapR and 2B-OlapR, respectively. OlapR cells possess distinct morphology from parental cells and display robust resistance to olaparib and other clinically relevant PARPis, including rucaparib, niraparib, and talazoparib. In LNCaP and C4-2B cells, we found that olaparib induces massive DNA damage, leading to activation of the G2-M checkpoint, activation of p53, and cell-cycle arrest. Furthermore, our data suggest that G2-M checkpoint activation leads to both cell death and senescence associated with p21 activity. In contrast, both LN-OlapR and 2B-OlapR cells do not arrest at G2-M and display a markedly blunted response to olaparib treatment. Interestingly, both OlapR cell lines harbor increased DNA damage relative to parental cells, suggesting that OlapR cells accumulate and manage persistent DNA damage during acquisition of resistance, likely through augmenting DNA repair capacity. Further impairing DNA repair through CDK1 inhibition enhances DNA damage, induces cell death, and sensitizes OlapR cells to olaparib treatment. Our data together further our understanding of PARPi treatment and provide a cellular platform system for the study of response and resistance to PARP inhibition.


Subject(s)
Phthalazines , Prostatic Neoplasms , Cell Cycle Checkpoints , Cell Line, Tumor , Humans , Male , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics
7.
Mol Cancer Ther ; 20(10): 2061-2070, 2021 10.
Article in English | MEDLINE | ID: mdl-34326198

ABSTRACT

Docetaxel and cabazitaxel-based taxane chemotherapy are critical components in the management of advanced prostate cancer. However, their efficacy is hindered due to de novo presentation with or the development of resistance. Characterizing models of taxane-resistant prostate cancer will lead to creation of strategies to overcome insensitivity. We have previously characterized docetaxel-resistant C4-2B and DU145 cell line derivatives, TaxR and DU145-DTXR, respectively. In the present study, we characterize cabazitaxel-resistant derivative cell lines created from chronic cabazitaxel exposure of TaxR and DU145-DTXR cells, CabR and CTXR, respectively. We show that CabR and CTXR cells are robustly resistant to both taxanes but retain sensitivity to antiandrogens. Both CabR and CTXR cells possess increased expression of ABCB1, which is shown to mediate resistance to treatment. Interestingly, we also present evidence for coordinated overexpression of additional genes present within the 7q21.12 gene locus where ABCB1 resides. This locus, known as the ABCB1 amplicon, has been demonstrated to be amplified in multidrug-resistant tumor cells, but little is known regarding its role in prostate cancer. We show that two ABCB1-amplicon genes other than ABCB1, RUNDC3B and DBF4, promote cellular viability and treatment resistance in taxane-resistant prostate cancer models. We present evidence that coordinated amplification of ABCB1-amplicon genes is common in a subset of prostate cancer patients. These data together suggest that ABCB1-amplicon activation plays a critical role in taxane resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms, Castration-Resistant/drug therapy , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Docetaxel/administration & dosage , Humans , Male , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Taxoids/administration & dosage , Tumor Cells, Cultured
8.
Clin Cancer Res ; 26(22): 6064-6074, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32928794

ABSTRACT

PURPOSE: Most patients with prostate cancer receiving enzalutamide or abiraterone develop resistance. Clinical evidence indicates that serum levels of dehydroepiandrosterone sulfate (DHEAS) and biologically active DHEA remain in the high range despite antiandrogen treatment. The conversion of DHEAS into DHEA by steroid sulfatase (STS) may contribute to sustained intracrine androgen synthesis. Here, we determine the contribution of STS to treatment resistance and explore the potential of targeting STS to overcome resistance in prostate cancer. EXPERIMENTAL DESIGN: STS expression was examined in patients and cell lines. In vitro, STS activity and expression were modulated using STS-specific siRNA or novel STS inhibitors (STSi). Cell growth, colony formation, androgen production, and gene expression were examined. RNA-sequencing analysis was conducted on VCaP cells treated with STSi. Mice were treated with STSis with or without enzalutamide to determine their effects in vivo. RESULTS: STS is overexpressed in patients with castration-resistant prostate cancer (CRPC) and resistant cells. STS overexpression increases intracrine androgen synthesis, cell proliferation, and confers resistance to enzalutamide and abiraterone. Inhibition of STS using siRNA suppresses prostate cancer cell growth. Targeting STS activity using STSi inhibits STS activity, suppresses androgen receptor transcriptional activity, and reduces the growth of resistant C4-2B and VCaP prostate cancer cells. STSis significantly suppress resistant VCaP tumor growth, decrease serum PSA levels, and enhance enzalutamide treatment in vitro and in vivo. CONCLUSIONS: These studies suggest that STS drives intracrine androgen synthesis and prostate cancer proliferation. Targeting STS represents a therapeutic strategy to treat CRPC and improve second-generation antiandrogen therapy.


Subject(s)
Androgens/biosynthesis , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms/drug therapy , Steryl-Sulfatase/genetics , Androgen Antagonists/pharmacology , Androgens/genetics , Androstenes/adverse effects , Androstenes/pharmacology , Benzamides/adverse effects , Benzamides/pharmacology , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone Sulfate/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Neoplasm Staging , Nitriles/adverse effects , Nitriles/pharmacology , Phenylthiohydantoin/adverse effects , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , RNA-Seq
9.
Mol Cancer Ther ; 19(8): 1708-1718, 2020 08.
Article in English | MEDLINE | ID: mdl-32430485

ABSTRACT

The next-generation antiandrogen drugs, XTANDI (enzalutamide), ZYTIGA (abiraterone acetate), ERLEADA (apalutamide) and NUBEQA (darolutamide) extend survival times and improve quality of life in patients with advanced prostate cancer. Despite these advances, resistance occurs frequently and there is currently no definitive cure for castration-resistant prostate cancer. Our previous studies identified that similar mechanisms of resistance to enzalutamide or abiraterone occur following treatment and cross-resistance exists between these therapies in advanced prostate cancer. Here, we show that enzalutamide- and abiraterone-resistant prostate cancer cells are further cross-resistant to apalutamide and darolutamide. Mechanistically, we have determined that the AKR1C3/AR-V7 axis confers this cross-resistance. Knockdown of AR-V7 in enzalutamide-resistant cells resensitize cells to apalutamide and darolutamide treatment. Furthermore, targeting AKR1C3 resensitizes resistant cells to apalutamide and darolutamide treatment through AR-V7 inhibition. Chronic apalutamide treatment in C4-2B cells activates the steroid hormone biosynthesis pathway and increases AKR1C3 expression, which confers resistance to enzalutamide, abiraterone, and darolutamide. In conclusion, our results suggest that apalutamide and darolutamide share similar resistant mechanisms with enzalutamide and abiraterone. The AKR1C3/AR-V7 complex confers cross-resistance to second-generation androgen receptor-targeted therapies in advanced prostate cancer.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/metabolism , Alternative Splicing , Androgen Receptor Antagonists/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/chemistry , Aldo-Keto Reductase Family 1 Member C3/genetics , Androgen Receptor Antagonists/classification , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Humans , Male , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Tumor Cells, Cultured
10.
Am J Clin Exp Urol ; 7(4): 203-214, 2019.
Article in English | MEDLINE | ID: mdl-31511827

ABSTRACT

BACKGROUND: De-regulation of Wnt signaling pathways has been shown to be associated with progression of castration-resistant prostate cancer and more recently, studies indicate that both canonical and non-canonical Wnt pathways may mediate resistance to anti-androgen therapies such as enzalutamide. However, the mechanisms by which Wnt signaling is altered in prostate cancer remain poorly understood. Wnt pathway function begins with Wnt biogenesis and secretion from Wnt signal sending cells. While previous studies have investigated downstream mechanisms of Wnt pathway alterations in prostate cancer, little is known on the role of Wnt secretion mediating proteins. Wntless (WLS) is thought to be essential for the secretion of all Wnts. In this study, we sought to understand the role of WLS in prostate cancer. METHODS: RNA-seq and gene set enrichment analysis were used to understand expression profile changes in enzalutamide-resistant C4-2B-MDVR (MDVR) cells versus parental C4-2B cells. Quantitative-PCR and western blot were used to confirm RNA-seq data and to assess expression changes of gene targets of interest. Rv1 cells were used as a separate model of enzalutamide-resistant prostate cancer. RNAi was used to inhibit WLS expression. Cell viability, colony formation, and PSA ELISA assays were used to assess cell growth and survival. RESULTS: Transcriptomic profiling revealed enriched Wnt pathway signatures in MDVR versus parental C4-2B cells. We further show that MDVR cells upregulate Wnt signaling and overexpress WLS. Inhibition of WLS decreases Wnt signaling, markedly attenuates prostate cancer cell viability, induces apoptosis, and re-sensitizes enzalutamide-resistant cells to enzalutamide treatment. Lastly, we show that inhibition of WLS reduces AR and AR-variants expression and downstream signaling. CONCLUSIONS: Our findings support a role for WLS in the progression of prostate cancer to a treatment-resistant state. Further efforts to understand Wnt signaling pathway alterations in this disease may lead to the development of novel treatments.

11.
Mol Cancer Ther ; 18(10): 1875-1886, 2019 10.
Article in English | MEDLINE | ID: mdl-31308078

ABSTRACT

The mechanisms resulting in resistance to next-generation antiandrogens in castration-resistant prostate cancer are incompletely understood. Numerous studies have determined that constitutively active androgen receptor (AR) signaling or full-length AR bypass mechanisms may contribute to the resistance. Previous studies established that AKR1C3 and AR-V7 play important roles in enzalutamide and abiraterone resistance. In the present study, we found that AKR1C3 increases AR-V7 expression in resistant prostate cancer cells through enhancing protein stability via activation of the ubiquitin-mediated proteasome pathway. AKR1C3 reprograms AR signaling in enzalutamide-resistant prostate cancer cells. In addition, bioinformatical analysis of indomethacin-treated resistant cells revealed that indomethacin significantly activates the unfolded protein response, p53, and apoptosis pathways, and suppresses cell-cycle, Myc, and AR/ARV7 pathways. Targeting AKR1C3 with indomethacin significantly decreases AR/AR-V7 protein expression in vitro and in vivo through activation of the ubiquitin-mediated proteasome pathway. Our results suggest that the AKR1C3/AR-V7 complex collaboratively confers resistance to AR-targeted therapies in advanced prostate cancer.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/metabolism , Alternative Splicing/genetics , Drug Resistance, Neoplasm , Molecular Targeted Therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Administration, Oral , Animals , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Indomethacin/administration & dosage , Indomethacin/pharmacology , Male , Mice, SCID , Neoplasm Staging , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/drug therapy , Protein Binding/drug effects , Protein Stability/drug effects , Signal Transduction/drug effects , Steroids/biosynthesis
12.
Br J Cancer ; 121(3): 237-248, 2019 07.
Article in English | MEDLINE | ID: mdl-31209328

ABSTRACT

BACKGROUND: Despite overexpression of the ErbB (EGFR/HER2/ErbB3/ErbB4) family in castration-resistant prostate cancer (CRPC), some inhibitors of this family, including the dual EGFR/HER2 inhibitor lapatinib, failed in Phase II clinical trials. Hence, we investigated mechanisms of lapatinib resistance to determine whether alternate ErbB inhibitors can succeed. METHODS: The CWR22 human tumour xenograft and its CRPC subline 22Rv1 and sera from lapatinib-treated CRPC patients from a previously reported Phase II trial were used to study lapatinib resistance. Mechanistic studies were conducted in LNCaP, C4-2 and 22Rv1 cell lines. RESULTS: Lapatinib increased intratumoral HER2 protein, which encouraged resistance to this treatment in mouse models. Sera from CRPC patients following lapatinib treatment demonstrated increased HER2 levels. Investigation of the mechanism of lapatinib-induced HER2 increase revealed that lapatinib promotes HER2 protein stability, leading to membrane localisation, EGFR/HER2 heterodimerisation and signalling, elevating cell viability. Knockdown of HER2 and ErbB3, but not EGFR, sensitised CRPC cells to lapatinib. At equimolar concentrations, the recently FDA-approved pan-ErbB inhibitor dacomitinib decreased HER2 protein stability, prevented ErbB membrane localisation (despite continued membrane integrity) and EGFR/HER2 heterodimerisation, thereby decreasing downstream signalling and increasing apoptosis. CONCLUSIONS: Targeting the EGFR axis using the irreversible pan-ErbB inhibitor dacomitinib is a viable therapeutic option for CRPC.


Subject(s)
Lapatinib/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Quinazolinones/therapeutic use , Receptor, ErbB-2/biosynthesis , Animals , Cell Line, Tumor , Disease Models, Animal , ErbB Receptors/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Prostatic Neoplasms, Castration-Resistant/metabolism , Protein Multimerization , Receptor, ErbB-2/blood , Receptor, ErbB-2/chemistry
13.
Transl Oncol ; 12(7): 871-878, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31075528

ABSTRACT

Castration-resistant prostate cancer remains as an incurable disease. Exploiting DNA damage repair defects via inhibition of poly (ADP-ribose) polymerase (PARP) is becoming an attractive therapeutic option. The TOPARP-A clinical trial demonstrated that the PARP inhibitor olaparib may be an effective strategy for treating prostate cancer. However, several unanswered questions regarding the use of olaparib remain: 1) How do we best stratify patients for olaparib treatment? 2) Where do we place olaparib in the treatment sequence paradigm? 3) Is there cross-resistance between olaparib and currently used therapies? Here, we tested putative cross-resistance between current therapies and olaparib in treatment-resistant castration-resistant prostate cancer models. Docetaxel-resistant cells exhibited robust resistance to olaparib which could be attributed to blunted PARP trapping in response to olaparib treatment. Upregulated ABCB1 mediates cross-resistance between taxanes and olaparib, which can be overcome through decreasing ABCB1 expression or inhibiting ABCB1 using elacridar or enzalutamide. We also show that combining olaparib with enzalutamide is more effective in olaparib-sensitive cells than either single agent. Our results demonstrate that cross-resistance between olaparib and other therapies could blunt response to treatment and highlight the need to develop strategies to maximize olaparib efficacy.

14.
Neoplasia ; 20(6): 563-573, 2018 06.
Article in English | MEDLINE | ID: mdl-29730477

ABSTRACT

Prostate cancer (PCa) progression involves a shift from endocrine to paracrine and eventually autocrine control resulting from alterations in molecular mechanisms in the cells. Deregulation of RNA translation is crucial for tumor cells to grow and proliferate; therefore, overactivation of the translation machinery is often observed in cancer. The two most important signal transduction pathways regulating PCa progression are PI3K/Akt/mTOR and Ras/MAPK. These two pathways converge on the eukaryotic translation initiation factor 4E (eIF4E) which binds to the protein scaffold eIF4G upon mechanistic target of rapamycin (mTOR) activation and is phosphorylated by the mitogen-activated protein kinase (MAPK) interacting protein kinases (Mnk1/2). This review describes the role of eIF4E in mRNA translation initiation mediated by its binding to the methylated 5' terminal structure (m7G-cap) of many mRNAs, and the ability of many tumor cells to bypass this mechanism. Hormonal therapy and chemotherapy are two of the most prevalent therapies used in patients with advanced PCa, and studies have implicated a role for eIF4E phosphorylation in promoting resistance to both these therapies. It appears that eIF4E phosphorylation enhances the rate of translation of oncogene mRNAs to increase tumorigenicity.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Phosphorylation/genetics , Prostatic Neoplasms/genetics , Humans , Male , Mitogen-Activated Protein Kinases/genetics , RNA, Messenger/genetics , Signal Transduction/genetics
15.
Methods Mol Biol ; 1655: 319-334, 2018.
Article in English | MEDLINE | ID: mdl-28889394

ABSTRACT

The epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases (RTK) consists of four members: EGFR1/ErbB1/HER1, ErbB2/HER2, ErbB3/HER3, and HER4/ErbB4. Signaling through these receptors regulates many key cellular activities, such as cell division, migration, adhesion, differentiation, and apoptosis. The ErbB family has been shown to be overexpressed in different types of cancers and is a target of several inhibitors already in clinical trials. ErbB3 lacks a functional tyrosine kinase domain and therefore has not been as extensively studied as the other members of this family, but its importance in activating downstream pathways, such as the PI3K/Akt pathway, makes this RTK a worthy investigation target, especially in urothelial carcinoma where the PI3K/Akt pathway is vital for progression. In recent times, ErbB3 overexpression has been linked to drug resistance and progression of various diseases, especially cancer. ErbB3 levels in the serum were shown in many cases to be reflective of its role in disease progression, and therefore detection of serum ErbB3 levels during treatment may be of importance.Here we describe two methods for detecting ErbB3 protein in serum from patients who have undergone a clinical trial, utilizing two well-established methods in molecular biology-western blotting and ELISA, focusing on sample preparation and troubleshooting.


Subject(s)
Biomarkers, Tumor , Receptor, ErbB-3/blood , Blotting, Western/methods , Disease Management , Enzyme-Linked Immunosorbent Assay , Humans , Neoplasms/blood , Neoplasms/diagnosis , Neoplasms/therapy , Protein Isoforms , Receptor, ErbB-3/genetics
16.
Endocr Relat Cancer ; 22(3): 369-86, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25759396

ABSTRACT

Prostate cancer (PCa) progression is regulated by the androgen receptor (AR); however, patients undergoing androgen-deprivation therapy (ADT) for disseminated PCa eventually develop castration-resistant PCa (CRPC). Results of previous studies indicated that AR, a transcription factor, occupies distinct genomic loci in CRPC compared with hormone-naïve PCa; however, the cause of this distinction was unknown. The E3 ubiquitin ligase Nrdp1 is a model AR target modulated by androgens in hormone-naïve PCa but not in CRPC. Using Nrdp1, we investigated how AR switches transcription programs during CRPC progression. The proximal Nrdp1 promoter contains an androgen response element (ARE); we demonstrated AR binding to this ARE in androgen-sensitive PCa. Analysis of hormone-naive human prostatectomy specimens revealed correlation between Nrdp1 and AR expression, supporting AR regulation of NRDP1 levels in androgen-sensitive tissue. However, despite sustained AR levels, AR binding to the Nrdp1 promoter and Nrdp1 expression were suppressed in CRPC. Elucidation of the suppression mechanism demonstrated correlation of NRDP1 levels with nuclear localization of the scaffolding protein filamin A (FLNA) which, as we previously showed, is itself repressed following ADT in many CRPC tumors. Restoration of nuclear FLNA in CRPC stimulated AR binding to Nrdp1 ARE, increased its transcription, and augmented NRDP1 protein expression and responsiveness to ADT, indicating that nuclear FLNA controls AR-mediated androgen-sensitive Nrdp1 transcription. Expression of other AR-regulated genes lost in CRPC was also re-established by nuclear FLNA. Thus, our results indicate that nuclear FLNA promotes androgen-dependent AR-regulated transcription in PCa, while loss of nuclear FLNA in CRPC alters the AR-regulated transcription program.


Subject(s)
Filamins/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Cell Line, Tumor , Disease Progression , Filamins/metabolism , Heterografts , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Transcription, Genetic , Transfection , Ubiquitin-Protein Ligases/biosynthesis
17.
Environ Sci Technol ; 46(11): 6081-7, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22587496

ABSTRACT

The health condition of out-migrating juvenile salmonids can influence migration success. Physical damage, pathogenic infection, contaminant exposure, and immune system status can affect survival probability. The present study is part of a wider investigation of out-migration success in juvenile steelhead (Oncorhynchus mykiss) and focuses on the application of molecular profiling to assess sublethal effects of environmental stressors in field-collected fish. We used a suite of genes in O. mykiss to specifically assess responses that could be directly related to steelhead health condition during out-migration. These biomarkers were used on juvenile steelhead captured in the Snake River, a tributary of the Columbia River, in Washington, USA, and were applied on gill and anterior head kidney tissue to assess immune system responses, pathogen-defense (NRAMP, Mx, CXC), general stress (HSP70), metal-binding (metallothionein-A), and xenobiotic metabolism (Cyp1a1) utilizing quantitative polymerase chain reaction (PCR) technology. Upon capture, fish were ranked according to visual external physical conditions into good, fair, poor, and bad categories; gills and kidney tissues were then dissected and preserved for gene analyses. Transcription responses were tissue-specific for gill and anterior head kidney with less significant responses in gill tissue than in kidney. Significant differences between the condition ranks were attributed to NRAMP, MX, CXC, and Cyp1a1 responses. Gene profiling correlated gene expression with pathogen presence, and results indicated that gene profiling can be a useful tool for identifying specific pathogen types responsible for disease. Principal component analysis (PCA) further correlated these responses with specific health condition categories, strongly differentiating good, poor, and bad condition ranks. We conclude that molecular profiling is an informative and useful tool that could be applied to indicate and monitor numerous population-level parameters of management interest.


Subject(s)
Environmental Health , Environmental Monitoring , Gene Expression Profiling , Health , Oncorhynchus mykiss/genetics , Rivers , Transcription, Genetic , Animals , Cluster Analysis , Principal Component Analysis , Washington
18.
Fish Physiol Biochem ; 38(4): 1107-1116, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22252335

ABSTRACT

The striped bass (Morone saxatilis) supports a valuable recreational fishery and is among the most important piscivorous fish of the San Francisco Estuary. This species has suffered a significant decline in numbers over the past decades, and there is indication that contaminants are important contributors. Polycyclic aromatic hydrocarbons (PAHs) and polyhalogenated aromatic hydrocarbons (PHAHs) including PCBs and dioxins are widespread in the estuary, they typically bioaccumulate through trophic levels, reaching highest levels in top predators and are known to affect the fish health and development. The aim of this study was to investigate the dynamics of cytochrome P4501A (Cyp1a) induction simultaneously at different levels of biological organization (RNA transcription and protein synthesis) as a biomarker of exposure to PAHs and PHAHs. We utilized ß-naphthoflavone (BNF) as a model PAH to induce Cyp1a responses in juvenile striped bass in both dose-response and time-response assessments and determined Cyp1a mRNA and protein levels. Significant responses were measured in both systems at 10 mg ΒΝF kg⁻¹, a concentration used for time-response studies. Messenger RNA levels peaked at 6 h post-injection, while protein levels increased progressively with time, significantly peaking at 96 h post-injection; both remaining elevated throughout the duration of the test (8 days). Our data suggest that rapid induction of gene transcription following exposure and subsequent cumulative protein synthesis could provide a useful means of identifying temporal variants in exposure to Cyp1a inducers in Morone saxatilis. The potential application of this combined Cyp1a gene and protein biomarker in this species for field studies is discussed.


Subject(s)
Bass/metabolism , Cytochrome P-450 Enzyme System/metabolism , Fish Proteins/metabolism , beta-Naphthoflavone/toxicity , Animals , Biomarkers/metabolism , Dose-Response Relationship, Drug , Environmental Monitoring , Polycyclic Aromatic Hydrocarbons/toxicity , RNA, Messenger/metabolism , Time Factors , Xenobiotics/toxicity
19.
Aquat Toxicol ; 105(3-4): 369-77, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21820383

ABSTRACT

The delta smelt (Hypomesus transpacificus) is an endangered pelagic fish species endemic to the Sacramento-San Joaquin Estuary in Northern California, which acts as an indicator of ecosystem health in its habitat range. Interrogative tools are required to successfully monitor effects of contaminants upon the delta smelt, and to research potential causes of population decline in this species. We used microarray technology to investigate genome-wide effects in fish exposed to ammonia; one of multiple contaminants arising from wastewater treatment plants and agricultural runoff. A 4-day exposure of 57-day old juveniles resulted in a total ammonium (NH(4)(+)-N) median lethal concentration (LC50) of 13 mg/L, and a corresponding un-ionized ammonia (NH(3)) LC50 of 147 µg/L. Using the previously designed delta smelt microarray we assessed altered gene transcription in juveniles exposed to 10mg/L NH(4)(+)-N from this 4-day exposure. Over half of the responding genes were associated with membrane integrity and function, however, neurological and muscular function was also affected. Amongst the notable pathways affected by ammonium exposure, directly associated with cellular membranes, are energy metabolism through oxidative phosphorylation, cellular responses to environmental stimuli, highlighted through signal transduction and molecular interactions, cellular processes encompassing transport and catabolism, along with cell motility, development, communication and cell death. To assess these impacts further, key genes were selected as potential biomarkers and investigated using quantitative PCR analysis on fish exposed to 2.5, 5, 10, 20 and 40 mg/L NH(4)(+)-N. Quantitative PCR results indicate biphasic responses, pivoting around the estimated no-observed effect concentration (NOEC; 5.0mg/L NH(4)(+)-N) and below. Genes significantly affected by ammonia exposure include claudin-10, Keratin-15, Septin-3, Transmembrane protein 4, superfamily 4 (membrane), Tropomyosin, Myosin light chain, Calmodulin (muscular), Tubulin cofactor beta (neurological), Sirtuin-6 (development), and Rhesus associated type C glycoprotein 1 (gill- and skin-specific ammonium transporter). The quantitation of the ammonium transporter may highlight the capacity of delta smelt to contend with elevated levels of ammonia, the peak response of which may be indicative of short-term thresholds of tolerance. Our study supports the notion that exposure to ammonia results in cell membrane destabilization, potentially affecting membrane permeability, enhancing uptake and thus synergistic effects of multiple-contaminant exposure.


Subject(s)
Ammonia/toxicity , Osmeriformes/genetics , Transcriptome/drug effects , Water Pollutants, Chemical/toxicity , Animals , Biomarkers , Endangered Species , Gene Expression Profiling , Lethal Dose 50 , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Toxicity Tests, Acute
20.
Environ Toxicol Chem ; 30(2): 290-300, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21072851

ABSTRACT

The delta smelt (Hypomesus transpacificus) is an endangered pelagic fish species endemic to the Sacramento-San Joaquin estuary (CA, USA), and considered an indicator of ecosystem health. Copper is a contaminant of concern in Californian waterways that may affect the development and survival of this endangered species. The experimental combination of molecular biomarkers with higher level effects may allow for interpretation of responses in a functional context that can be used to predict detrimental outcomes caused by exposure. A delta smelt microarray was developed and applied to screen for candidate molecular biomarkers that may be used in monitoring programs. Functional classifications of microarray responses were used along with quantitative polymerase chain reaction determining effects upon neuromuscular, digestive, and immune responses in Cu-exposed delta smelt. Differences in sensitivity were measured between juveniles and larvae (median lethal concentration = 25.2 and 80.4 µg/L Cu(2+), respectively). Swimming velocity declined with higher exposure concentrations in a dose-dependent manner (r = -0.911, p < 0.05), though was not statistically significant to controls. Genes encoding for aspartoacylase, hemopexin, α-actin, and calcium regulation proteins were significantly affected by exposure and were functionally interpreted with measured swimming responses. Effects on digestion were measured by upregulation of chitinase and downregulation of amylase, whereas downregulation of tumor necrosis factor indicated a probable compromised immune system. Results from this study, and many others, support the use of functionally characterized molecular biomarkers to assess effects of contaminants in field scenarios. We thus propose that to attribute environmental relevance to molecular biomarkers, research should concentrate on their application in field studies with the aim of assisting monitoring programs.


Subject(s)
Copper/adverse effects , Endangered Species , Gene Expression Regulation , Osmeriformes/physiology , Water Pollutants, Chemical/adverse effects , Animals , California , Copper/toxicity , Genetic Markers , Larva/genetics , Larva/immunology , Larva/physiology , Microarray Analysis , Osmeriformes/genetics , Osmeriformes/immunology , Polymerase Chain Reaction , Swimming , Water/analysis , Water Pollutants, Chemical/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...