Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Salma Siddiqui is active.

Publication


Featured researches published by Salma Siddiqui.


Clinical Cancer Research | 2009

Nuclear versus cytoplasmic localization of filamin A in prostate cancer: immunohistochemical correlation with metastases.

Roble Bedolla; Yu Wang; Alfredo Asuncion; Karim Chamie; Salma Siddiqui; Maria Mudryj; Thomas J. Prihoda; Javed Siddiqui; Arul M. Chinnaiyan; Rohit Mehra; Ralph W. deVere White; Paramita M. Ghosh

Purpose: We previously showed that nuclear localization of the actin-binding protein, filamin A (FlnA), corresponded to hormone-dependence in prostate cancer. Intact FlnA (280 kDa, cytoplasmic) cleaved to a 90 kDa fragment which translocated to the nucleus in hormone-naïve cells, whereas in hormone-refractory cells, FlnA was phosphorylated, preventing its cleavage and nuclear translocation. We have examined whether FlnA localization determines a propensity to metastasis in advanced androgen-independent prostate cancer. Experimental Design: We examined, by immunohistochemistry, FlnA localization in paraffin-embedded human prostate tissue representing different stages of progression. Results were correlated with in vitro studies in a cell model of prostate cancer. Results: Nuclear FlnA was significantly higher in benign prostate (0.6612 ± 0.5888), prostatic intraepithelial neoplasia (PIN; 0.6024 ± 0.4620), and clinically localized cancers (0.69134 ± 0.5686) compared with metastatic prostate cancers (0.3719 ± 0.4992, P = 0.0007). Cytoplasmic FlnA increased from benign prostate (0.0833 ± 0.2677), PIN (0.1409 ± 0.2293), localized cancers (0.3008 ± 0.3762, P = 0.0150), to metastases (0.7632 ± 0.4414, P < 0.00001). Logistic regression of metastatic versus nonmetastatic tissue yielded the area under the receiver operating curve as 0.67 for nuclear-FlnA, 0.79 for cytoplasmic-FlnA, and 0.82 for both, indicating that metastasis correlates with cytoplasmic to nuclear translocation. In vitro studies showed that cytoplasmic localization of FlnA induced cell invasion whereas nuclear translocation of the protein inhibited it. FlnA dephosphorylation with the protein kinase A inhibitor H-89 facilitated FlnA nuclear translocation, resulting in decreased invasiveness and AR transcriptional activity, and induced sensitivity to androgen withdrawal in hormone-refractory cells. Conclusions: The data presented in this study indicate that in prostate cancer, metastasis correlates with cytoplasmic localization of FlnA and may be prevented by cleavage and subsequent nuclear translocation of this protein.


Cancer Research | 2010

Nrdp1-mediated regulation of ErbB3 expression by the androgen receptor in androgen-dependent but not castrate-resistant prostate cancer cells.

Liqun Chen; Salma Siddiqui; Swagata Bose; Benjamin A. Mooso; Alfredo Asuncion; Roble Bedolla; Ruth L. Vinall; Clifford G. Tepper; Regina Gandour-Edwards; Xu-Bao Shi; Xiao Hua Lu; Javed Siddiqui; Arul M. Chinnaiyan; Rohit Mehra; Ralph W. deVere White; Kermit L. Carraway; Paramita M. Ghosh

Patients with advanced prostate cancer (PCa) are initially susceptible to androgen withdrawal (AW), but ultimately develop resistance to this therapy (castration-resistant PCa, CRPC). Here, we show that AW can promote CRPC development by increasing the levels of the receptor tyrosine kinase ErbB3 in androgen-dependent PCa, resulting in AW-resistant cell cycle progression and increased androgen receptor (AR) transcriptional activity. CRPC cell lines and human PCa tissue overexpressed ErbB3, whereas downregulation of ErbB3 prevented CRPC cell growth. Investigation of the mechanism by which AW augments ErbB3, using normal prostate-derived pRNS-1-1 cells, and androgen-dependent PCa lines LNCaP, PC346C, and CWR22 mouse xenografts, revealed that the AR suppresses ErbB3 protein levels, whereas AW relieves this suppression, showing for the first time the negative regulation of ErbB3 by AR. We show that AR activation promotes ErbB3 degradation in androgen-dependent cells, and that this effect is mediated by AR-dependent transcriptional upregulation of neuregulin receptor degradation protein-1 (Nrdp1), an E3 ubiquitin ligase that targets ErbB3 for degradation but whose role in PCa has not been previously examined. Therefore, AW decreases Nrdp1 expression, promoting ErbB3 protein accumulation, and leading to AR-independent proliferation. However, in CRPC sublines of LNCaP and CWR22, which strongly overexpress the AR, ErbB3 levels remain elevated due to constitutive suppression of Nrdp1, which prevents AR regulation of Nrdp1. Our observations point to a model of CRPC development in which progression of PCa to castration resistance is associated with the inability of AR to transcriptionally regulate Nrdp1, and predict that inhibition of ErbB3 during AW may impair CRPC development.


Endocrine-related Cancer | 2012

Enhancing the effectiveness of androgen deprivation in prostate cancer by inducing Filamin A nuclear localization

Benjamin A. Mooso; Ruth L. Vinall; Clifford G. Tepper; Rosalinda M. Savoy; Jean P. Cheung; Sheetal Singh; Salma Siddiqui; Yu Wang; Roble Bedolla; Anthony Martinez; Maria Mudryj; Hsing Jien Kung; Ralph W. deVere White; Paramita M. Ghosh

As prostate cancer (CaP) is regulated by androgen receptor (AR) activity, metastatic CaP is treated with androgen deprivation therapy (ADT). Despite initial response, patients on ADT eventually progress to castration-resistant CaP (CRPC), which is currently incurable. We previously showed that cleavage of the 280 kDa structural protein Filamin A (FlnA) to a 90 kDa fragment, and nuclear localization of the cleaved product, sensitized CRPC cells to ADT. Hence, treatment promoting FlnA nuclear localization would enhance androgen responsiveness. Here, we show that FlnA nuclear localization induced apoptosis in CRPC cells during ADT, identifying it as a treatment tool in advanced CaP. Significantly, the natural product genistein combined polysaccharide (GCP) had a similar effect. Investigation of the mechanism of GCP-induced apoptosis showed that GCP induced FlnA cleavage and nuclear localization and that apoptosis resulting from GCP treatment was mediated by FlnA nuclear localization. Two main components of GCP are genistein and daidzein: the ability of GCP to induce G2 arrest was due to genistein whereas sensitivity to ADT stemmed from daidzein; hence, both were needed to mediate GCPs effects. FlnA cleavage is regulated by its phosphorylation; we show that ADT enhanced FlnA phosphorylation, which prevented its cleavage, whereas GCP inhibited FlnA phosphorylation, thereby sensitizing CaP cells to ADT. In a mouse model of CaP recurrence, GCP, but not vehicle, impeded relapse following castration, indicating that GCP, when administered with ADT, interrupted the development of CRPC. These results demonstrate the efficacy of GCP in promoting FlnA nuclear localization and enhancing androgen responsiveness in CaP.


Endocrine-related Cancer | 2015

Transcription of Nrdp1 by the androgen receptor is regulated by nuclear filamin A in prostate cancer

Rosalinda M. Savoy; Liqun Chen; Salma Siddiqui; Frank Melgoza; Blythe Durbin-Johnson; Christiana Drake; Maitreyee K. Jathal; Swagata Bose; Thomas M. Steele; Benjamin A. Mooso; Leandro S. D'Abronzo; William H. Fry; Kermit L. Carraway; Maria Mudryj; Paramita M. Ghosh

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.


Oncogene | 2017

The androgen receptor is a negative regulator of eIF4E phosphorylation at S209: implications for the use of mTOR inhibitors in advanced prostate cancer

Leandro S. D'Abronzo; Swagata Bose; M E Crapuchettes; R E Beggs; Ruth L. Vinall; Clifford Tepper; Salma Siddiqui; Maria Mudryj; Frank Melgoza; Blythe Durbin-Johnson; R. W. DeVere White; Paramita M. Ghosh

The antiandrogen bicalutamide is widely used in the treatment of advanced prostate cancer (PCa) in many countries, but its effect on castration-resistant PCa (CRPC) is limited. We previously showed that resistance to bicalutamide results from activation of mechanistic target of rapamycin (mTOR). Interestingly, clinical trials testing combinations of the mTOR inhibitor RAD001 with bicalutamide were effective in bicalutamide-naïve CRPC patients, but not in bicalutamide-pretreated ones. Here we investigate causes for their difference in response. Evaluation of CRPC cell lines identified resistant vs sensitive in vitro models, and revealed that increased eIF4E(S209) phosphorylation is associated with resistance to the combination. We confirmed using a human-derived tumor xenograft mouse model that bicalutamide pre-treatment is associated with an increase in eIF4E(S209) phosphorylation. Thus, AR suppressed eukaryotic initiation factor 4E (eIF4E) phosphorylation, while the use of antiandrogens relieved this suppression, thereby triggering its increase. Additional investigation in human prostatectomy samples showed that increased eIF4E phosphorylation strongly correlated with the cell proliferation marker Ki67. Small interfering RNA-mediated knockdown (k/d) of eIF4E-sensitized CRPC cells to RAD001+bicalutamide, whereas eIF4E overexpression induced resistance. Inhibition of eIF4E phosphorylation by treatment with CGP57380 (an inhibitor of mitogen-activated protein kinase-interacting serine–threonine kinases MAP kinase-interacting kinase 1 (Mnk1/2), the eIF4E upstream kinase) or inhibitors of extracellular signal-regulated kinase 1/2 (ERK1/2), the upstream kinase-regulating Mnk1/2, also sensitized CRPC cells to RAD001+bicalutamide. Examination of downstream targets of eIF4E-mediated translation, including survivin, demonstrated that eIF4E(S209) phosphorylation increased cap-independent translation, whereas its inhibition restored cap-dependent translation, which could be inhibited by mTOR inhibitors. Thus, our results demonstrate that while combinations of AR and mTOR inhibitors were effective in suppressing tumor growth by inhibiting both AR-induced transcription and mTOR-induced cap-dependent translation, pre-treatment with AR antagonists including bicalutamide increased eIF4E phosphorylation that induced resistance to combinations of AR and mTOR inhibitors by inducing cap-independent translation. We conclude that this resistance can be overcome by inhibiting eIF4E phosphorylation with Mnk1/2 or ERK1/2 inhibitors.


Cancer Research | 2016

Abstract 1303: In vivo analysis of EGFR family signalling as a bypass mechanism in prostate cancer

Maitreyee K. Jathal; Thomas M. Steele; Salma Siddiqui; Benjamin A. Mooso; Leandro S. D’Abronzo; Christiana Drake; Paramita M. Ghosh

Background: Prostate cancers (PCa) rely on androgenic ligands and the androgen receptor (AR) for their growth and survival, making AR inhibition a predominant therapeutic strategy for these tumors. Some prostate tumors however fail this therapy due to ‘bypass’ mechanisms that emerge as a result of prolonged AR targeting. This in vivo study attempted to assess the expression and activation of the epidermal growth factor receptor (EGFR) family (whose role is well-documented in PCa) in response to androgen deprivation therapy (ADT). Methods: Nude mice were implanted (s.c.) with CWR22 tumors (human-patient-derived, androgen-dependent ‘AD’) and its castration-resistant (‘CR’) subline CWR22-Rv1 (relapsed CWR22). Androgen deprivation (i.e. AR inhibition) was achieved by surgical or ‘sham’ castration of mice. Tumors were analyzed (immunohistochemistry/immunoblot) for EGFR/ErbB2/ErbB3/AR proteins and proliferative/apoptotic markers. Results: Castration caused significant tumor regression in AD but not CR tumors. in vitro viability assays demonstrated that castration (mimicked by using charcoal-stripped serum, ‘css’) did not slow down CR cells to the same degree as it did AD cells. At baseline, intratumoral EGFR protein was unchanged in R22 tumors, ErbB2 levels decreased and ErbB3 protein increased in Rv1 tumors. Castration increased ErbB3 but not EGFR or ErbB2 proteins in CWR22 tumors. Phosphorylated forms of these receptors were generally difficult to detect but there was more phosphorylated ErbB3 protein in Rv1 tumors. Downstream of the EGFR family, there was less phosphorylated Erk but not Akt protein in CWR22-Rv1 tumors. Castration decreased Erk protein in AD tumors but increased it in CR tumors. Immunohistochemical quantification revealed that cytoplasmic EGFR and ErbB3 proteins were elevated in CR tumors but reduced in AD tumors. Castration greatly decreased Ki-67 staining in AD but not in CR tumors while the number of TUNEL-positive nuclei and intensity of PARP staining decreased in castrated CR but not in AD tumors. ErbB3 and AR proteins were significantly correlated with DNA damage and proliferation in CWR22 tumors but only nuclear AR levels and proliferation were significantly correlated in CR tumors. Conclusions: We conclude that androgen deprivation therapy may alter EGFR and ErbB3 protein levels and localization in androgen-dependent and castration-resistant tumors. The EGFR family is typically activated at the cell surface hence their presence and activity there, in response to castration, may initiate signalling pathways encouraging tumor cell proliferation and survival. Citation Format: Maitreyee K. Jathal, Thomas M. Steele, Salma Siddiqui, Benjamin A. Mooso, Leandro S. D’Abronzo, Christiana M. Drake, Paramita M. Ghosh. In vivo analysis of EGFR family signalling as a bypass mechanism in prostate cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1303.


Oncotarget | 2018

XPO1 inhibition by selinexor induces potent cytotoxicity against high grade bladder malignancies

Han Bit Baek; Alan P. Lombard; Stephen J. Libertini; Aleida Fernandez-Rubio; Ruth L. Vinall; Regina Gandour-Edwards; Rachel M. Nakagawa; Kathleen D. Vidallo; Kristine Nishida; Salma Siddiqui; Hiromi I. Wettersten; Yosef Landesman; Robert H. Weiss; Paramita M. Ghosh; Maria Mudryj

Treatment options for high grade urothelial cancers are limited and have remained largely unchanged for several decades. Selinexor (KPT-330), a first in class small molecule that inhibits the nuclear export protein XPO1, has shown efficacy as a single agent treatment for numerous different malignancies, but its efficacy in limiting bladder malignancies has not been tested. In this study we assessed selinexor-dependent cytotoxicity in several bladder tumor cells and report that selinexor effectively reduced XPO1 expression and limited cell viability in a dose dependent manner. The decrease in cell viability was due to an induction of apoptosis and cell cycle arrest. These results were recapitulated in in vivo studies where selinexor decreased tumor growth. Tumors treated with selinexor expressed lower levels of XPO1, cyclin A, cyclin B, and CDK2 and increased levels of RB and CDK inhibitor p27, a result that is consistent with growth arrest. Cells expressing wildtype RB, a potent tumor suppressor that promotes growth arrest and apoptosis, were most susceptible to selinexor. Cell fractionation and immunofluorescence studies showed that selinexor treatment increased nuclear RB levels and mechanistic studies revealed that RB ablation curtailed the response to the drug. Conversely, limiting CDK4/6 dependent RB phosphorylation by palbociclib was additive with selinexor in reducing bladder tumor cell viability, confirming that RB activity has a role in the response to XPO1 inhibition. These results provide a rationale for XPO1 inhibition as a novel strategy for the treatment of bladder malignancies.


Cancer Research | 2016

Abstract 4676: Overcoming EGFR-induced resistance to enzalutamide in castration-resistant prostate cancer

Thomas M. Steele; Maitreyee K. Jathal; Salma Siddiqui; Paramita M. Ghosh

Background: The androgen receptor (AR) remains a major therapeutic target in patients with castration-resistant prostate cancer (CRPC). Enzalutamide, an AR inhibitor that is FDA-approved for patients with CRPC, prevents ligand induced AR transcriptional activity, but some initial responders eventually become resistant to the drug. The expression and activity of the EGFR/ErbB family of receptor tyrosine kinases also increases in CRPC patients. The present work was undertaken to determine whether the activation of EGFR family (EGFR/ErbB2/ErbB3/ErbB4) may be responsible for enzalutamide resistance and whether treatment with receptor tyrosine kinase inhibitors would overcome this effect. Methods: Human-patient-derived androgen-dependent and CRPC prostate tumor cells were grown in 10% Fetal Bovine Serum (FBS). Protein expression and phosphorylation status of the EGFR family were determined by immunoblotting techniques. MTT assays were used to determine the viability of cells treated with enzalutamide, lapatinib (HER2/EGFR inhibitor), erlotinib (EGFR inhibitor), or dacomitinib (pan-ErbB inhibitor). A Luciferase Assay kit (Roche) was used to determine AR transcriptional activity. Lapatinib was obtained from LC Laboratories while erlotinib and dacomitinib were obtained from Selleck Chemicals. Enzalutamide was kindly provided by Medivation Inc. Results: In viability assays, erlotinib and dacomitinib were more effective than lapatinib in sensitizing CRPC cells to enzalutamide. Enzalutamide suppressed AR transcriptional activity, either alone or in combination with lapatinib, erlotinib, or dacomitinib. Erlotinib, lapatinib, and dacomitinib equally inhibited EGFR and ErbB3 phosphorylation. However, in EGF stimulated cells, erlotinib and dacomitinib—but not lapatinib—suppressed ERK 1/2 phosphorylation at Tyr202/Thr204, indicating a stark difference in downstream inhibition and potentially proliferation. Conclusions: The above results indicate that ERK 1/2 may play an important role in reducing the efficacy of enzalutamide by itself and in combination with lapatinib. Furthermore, lapatinib9s inability to prevent ERK phosphorylation upon EGF stimulation may play a role in its ineffectiveness in prostate cancer. Our preliminary preclinical data indicate that co-administration of enzalutamide with an EGFR targeted inhibitor may be a suitable therapeutic approach towards overcoming enzalutamide resistance in vitro. We are testing whether ERK 1/2 or MEK may be effective in reducing this ERK specific enzalutamide resistance. These strategies may potentially prolong enzalutamide sensitivity in CRPC patients. Citation Format: Thomas M. Steele, Maitreyee K. Jathal, Salma Siddiqui, Paramita M. Ghosh. Overcoming EGFR-induced resistance to enzalutamide in castration-resistant prostate cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4676.


Cancer Research | 2015

Abstract 5051: Androgen receptor-mediated regulation of p14ARF transcription in prostate tumor cells

Maria Mudryj; Salma Siddiqui; Stephen J. Libertini; Alan P. Lombard; Benjamin A. Mooso; Leandro S. D'Abronzo; Frank Melgoza; Alexander D. Borowsky; Christiana Drake; LiHong Qi; Paramita M. Ghosh

The p14ARF tumor suppressor is often deleted or silenced in malignancies. Prostate tumors are an exception, where p14 expression is elevated. To understand this phenomenon, we assessed the expression of p53 pathway members, which are most effected by p14ARF. The expression of androgen receptor (AR), a pivotal prostate cancer regulator, which is also affected by p14arf and MDM2 was analyzed as well. The studies used archival prostate tumor tissues obtained from prostatectomies performed at the Veterans Affairs-Northern California Health Care System in Mather California between 1996 and 2002 to better define the relationship between these interrelated networks. A prostate tumor tissue array consisting of 78 tumors of differing stages and grades was constructed to evaluate correlations between multiple parameters. Immunohistochemical studies assessed expression of the proliferation marker Ki67, p53, MDM2, MDM4, p14ARF, and the AR in the nuclear and cytoplasmic compartments of tumor and adjacent cells. p53, MDM4, p14ARF and AR were detected in nuclear and cytoplasmic compartments of tumor and non-tumor cells, but were predominantly nuclear. MDM2 expression was primarily cytoplasmic in tumor cells. Multivariate analysis of the immunohistochemical markers identified a strong correlation between expression of p14ARF and AR. Studies utilizing the prostate CWR22 xenograft and LNCaP cell line models revealed that castration or androgen deprivation resulted in reduced p14arf levels and that this effect correlated with a precipitous decline in E2F1-3a levels. In a reciprocal analysis, RB ablation enhanced p14ARF transcription, arguing that the E2F/RB pathway mediates AR-dependent p14ARF expression. However, we also identified an AR binding site located ∼40 KB upstream of the p14ARF gene. Chromatin immunoprecipitation (ChIP) studies showed that in prostate cells this site was bound by AR. ChIP studies also revealed E2F1 and E2F3 were present at the p14ARF promoter. Together, the studies argue p14ARF is a direct transcriptional target of AR and that AR and E2F collaborate to promote p14ARF expression. . Citation Format: Maria Mudryj, Salma Siddiqui, Stephen J. Libertini, Alan P. Lombard, Benjamin Mooso, Leandro D9Abronzo, Frank Melgoza, Alexander Borowsky, Christiana Drake, LiHong Qi, Paramita M. Ghosh. Androgen receptor-mediated regulation of p14ARF transcription in prostate tumor cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5051. doi:10.1158/1538-7445.AM2015-5051


Cancer Research | 2014

Abstract 4438: Changes in Nrdp1 regulation of ErbB3 in androgen-dependent vs. independent prostate cancer

Rosalinda M. Savoy; Salma Siddiqui; William H. Fry; Kermit L. Carraway; Paramita M. Ghosh

In prostate cancer, the androgen receptor (AR) is a major regulator of gene transcription. We previously demonstrated that the E3 ubiquitin ligase Nrdp1 is transcriptionally regulated by the AR [Chen, L., Cancer Res, 2010]. The effect of the AR on ErbB3 is mediated by Nrdp1, which regulates the degradation of ErbB3. Therefore, we investigated the function of the various domains of Nrdp1 on ErbB3 in prostate cancer. ErbB3 is a major factor in cell growth and proliferation so finding ways to regulate it would be beneficial for patient survival. We found that the Nrdp1 protein had two splice variants, a full length 36kDa protein and a C-terminal 28kDa protein. In addition, the full-length protein remained in the cytoplasm of LNCaP cells, which are androgen dependent, while the 28kDa protein went into the nucleus of C4-2 cells, an androgen independent subline of LNCaP cells. To further investigate the subcellular difference and role of these two splice variants on ErbB3, we used 4 flag tagged vectors that contain either full-length Nrdp1 (Nrdp1), an N-terminal Nrdp1 containing amino acids 1-169 (Zn), a C-terminal Nrdp1 containing amino acids 169-317 (32) , and an Nrdp1 lacking the coiled coil domain (ΔCC). We found that the ΔCC vector caused an increase of cell proliferation in LNCaP cells, an androgen dependent cell line, with no to little change in cell proliferation in its androgen independent sub cell lines, LNCaP R273H (LNCaP stably transfected with a mutated p53), C4-2, and C4-2B, and PC3 and PC3 WT-AR cells. Despite this, we found that the ΔCC protein caused a decrease of ErbB3 protein in all cell lines. We also performed subcellular fractionation on these cell lines transfected with the Nrdp1 flag vectors. All of the Nrdp1 proteins were found in the cytoplasm of the cell lines. In addition, the ΔCC protein was also found in the nuclear fraction of LNCaP R273H, C4-2, and C4-2B, while in LNCaP cells the Zn protein was also found in the nuclear fraction and in PC3 cells the 32 protein was also found in the nuclear fraction. In LNCaP cells, ErbB3 was found in the nucleus when transfected with Nrdp1 protein, Zn protein, and 32 protein, but found in the cytoplasm when transfected with the ΔCC protein. However, in LNCaP R273H, C4-2, and C4-2B cell lines, ErbB3 was found in the nucleus when transfected with all of the Nrdp1 vectors. Based on available data, we concluded that the coiled coil domain is important for ErbB3 protein regulation, but it had opposing effects in castration sensitive and resistant cells. In castration sensitive LNCaP cells, the coiled coil domain of Nrdp1 likely suppresses ErbB3-mediated cell proliferation by preventing ErbB3 translocation from the nucleus to the cytoplasm. However, in castration resistant cells, the coiled coil domain prevents the degradation of ErbB3 by Nrdp1. Further studies are required to determine the mechanism of action of the Nrdp1 coiled coil domain in these two phenotypes. Citation Format: Rosalinda M. Savoy, Salma Siddiqui, William H. Fry, Kermit L. Carraway, Paramita Ghosh. Changes in Nrdp1 regulation of ErbB3 in androgen-dependent vs. independent prostate cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4438. doi:10.1158/1538-7445.AM2014-4438

Collaboration


Dive into the Salma Siddiqui's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Mudryj

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ruth L. Vinall

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Frank Melgoza

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Swagata Bose

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge