ebook img

DTIC ADA593709: Regulation of TRAIL-Medicated Apoptosis in Prostate Cancer by Overexpression of XIAP PDF

13.3 MB·English
Save to my drive
Quick download
Download
Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.

Preview DTIC ADA593709: Regulation of TRAIL-Medicated Apoptosis in Prostate Cancer by Overexpression of XIAP

AD_________________ Award Number: DAMD17-02-1-0023 TITLE: Regulation of TRAIL-Medicated Apoptosis in Prostate Cancer by Overexpression of XIAP PRINCIPAL INVESTIGATOR: Benjamin Bonavida, Ph.D. CONTRACTING ORGANIZATION: The University of California Los Angeles, CA 90024-1406 REPORT DATE: January 2006 TYPE OF REPORT: Final PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland 21702-5012 DISTRIBUTION STATEMENT: Approved for Public Release; Distribution Unlimited The views, opinions and/or findings contained in this report are those of the author(s) and should not be construed as an official Department of the Army position, policy or decision unless so designated by other documentation. Form Approved REPORT DOCUMENTATION PAGE OMB No. 0704-0188 Public reporting burden for this collection of information is estimated to average 1 hour per response, including the time for reviewing instructions, searching existing data sources, gathering and maintaining the data needed, and completing and reviewing this collection of information. Send comments regarding this burden estimate or any other aspect of this collection of information, including suggestions for reducing this burden to Department of Defense, Washington Headquarters Services, Directorate for Information Operations and Reports (0704-0188), 1215 Jefferson Davis Highway, Suite 1204, Arlington, VA 22202- 4302. Respondents should be aware that notwithstanding any other provision of law, no person shall be subject to any penalty for failing to comply with a collection of information if it does not display a currently valid OMB control number. PLEASE DO NOT RETURN YOUR FORM TO THE ABOVE ADDRESS. 1. REPORT DATE (DD-MM-YYYY) 2. REPORT TYPE 3. DATES COVERED (From - To) 01-01-2006 Final 15 DEC 2001 - 14 DEC 2005 4. TITLE AND SUBTITLE 5a. CONTRACT NUMBER Regulation of TRAIL-Medicated Apoptosis in Prostate Cancer by Overexpression of XIAP 5b. GRANT NUMBER DAMD17-02-1-0023 5c. PROGRAM ELEMENT NUMBER 6. AUTHOR(S) 5d. PROJECT NUMBER Benjamin Bonavida, Ph.D. 5e. TASK NUMBER 5f. WORK UNIT NUMBER E-mail: [email protected] 7. PERFORMING ORGANIZATION NAME(S) AND ADDRESS(ES) 8. PERFORMING ORGANIZATION REPORT NUMBER The University of California Los Angeles, CA 90024-1406 9. SPONSORING / MONITORING AGENCY NAME(S) AND ADDRESS(ES) 10. SPONSOR/MONITOR’S ACRONYM(S) U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland 21702-5012 11. SPONSOR/MONITOR’S REPORT NUMBER(S) 12. DISTRIBUTION / AVAILABILITY STATEMENT Approved for Public Release; Distribution Unlimited 13. SUPPLEMENTARY NOTES 14. ABSTRACT Patients with prostate cancer (CaP) develop resistance to conventional therapies and alternative therapies, such as immunotherapy, are being actively considered. TRAIL is selectively cytotoxic to tumor cells and minimally cytotoxic to normal tissues and is a candidate for immunotherapy. CaP cells, however, are resistant to TRAIL due to antiapoptotic mechanisms such as overexpression of XIAP. This proposal investigated the mechanism by which XIAP regulates resistance to TRAIL and the findings demonstrate that TRAIL resistance is regulated by the expression of the TRAIL receptor DR5. The regulation of DR5 was found to be under the control of the transcription repressor YY1. Inhibition of NF-κB inhibited both YY1 and XIAP and sensitized the cells with TRAIL-induced apoptosis. We demonstrate that YY1 regulates the transcription of DR5 via a YY1 DNA-binding site on the DR5 promoter. The clinical importance of XIAP expression in prostate cancer tissue was examined in tissue microarrays and we demonstrate its prognostic significance in prostate cancer patients. This also correlated with the overexpression of YY1 which also showed its prognostic significance in prostate cancer patients. In addition, we have demonstrated that cytokines derived from prostate cancer and/or from the tumor microenvironment regulate the constitutive activation of NF-κB and downstream XIAP and YY1 and regulate resistance to TRAIL. Overall, our findings identify new therapeutic targets for TRAIL. 15. SUBJECT TERMS TRAIL, signaling, XIAP, apoptosis, NF-κB, sensitization, Bcl-xL, Immunotherapy, cytokines, drugs 16. SECURITY CLASSIFICATION OF: 17. LIMITATION 18. NUMBER 19a. NAME OF RESPONSIBLE PERSON OF ABSTRACT OF PAGES USAMRMC a. REPORT b. ABSTRACT c. THIS PAGE 19b. TELEPHONE NUMBER (include area U U U UU 363 code) Standard Form 298 (Rev. 8-98) Prescribed by ANSI Std. Z39.18 Table of Contents Introduction…………………………………………………………….…………....4 Body…………………………………………………………………………………….6 Key Research Accomplishments………………………………………….………14 Reportable Outcomes……………………………………………………………….16 Conclusions…………………………………………………………………………..19 References……………………………………………………………………………20 Appendices……………………………………………………………………………23 Introduction Prostate cancer (CaP) is the most common malignancy and age-related cause of cancer deaths in American males. Treatments for metastatic prostate cancer include hormonal ablation, chemotherapy, and combination therapies. These treatments are aimed at inhibiting tumor growth and at also inducing apoptosis. Unfortunately, hormonal therapy is always followed by the relapse of an aggressive androgen-independent disease that is insensitive to further hormonal manipulation or to treatment with conventional chemotherapeutic drugs. The underlying mechanisms by which the prostate tumor cells develop resistance to hormonal/drug-mediated effects are poorly understood. It is probable that the tumor cells acquire mechanisms that resist androgen ablation/drug-mediated apoptosis. Failure to eradicate advanced resistant tumors with conventional therapies has led to the exploration of novel therapeutic approaches such as immunotherapy. Immunotherapy is generally aimed at the generation of anti-tumor cytotoxic lymphocytes that can recognize and eradicate the drug-resistant tumor cells. Also, immunotherapy is predicated on the notion that all drug-resistant tumors should succumb to cytotoxic lymphocyte- mediated killing. Tumors that develop anti-apoptotic mechanisms to escape drugs/radiation- mediated apoptosis may also develop cross-resistance to apoptosis mediated by cytotoxic lymphocytes. We and others have reported that many types of drug-resistant tumor cells including CaP cells are also resistant to immune-related cytotoxic mechanism of killing. Thus, the development of immune resistance by drug-refractory tumors may hinder the therapeutic effect of immunotherapy. However, if the tumor can be sensitized to overcome resistance, the combination of sensitization and immunotherapy may be more efficacious. The underlying molecular mechanisms of sensitization to immunotherapy are therefore important to explore for the identification of gene targets involved in resistance and hence develop new means to modify these targets and reversing immune resistance. The inhibitor of apoptosis proteins (IAPs) represent a family of endogenous caspase inhibitors that share a conserved structure known as the BIR domains (Reed, 2001). Eight encoding IAPs genes are found in the human genome, and some of these are overexpressed in cancers (Ferreira et al., 2001; Hofmann et al., 2002; Tamm et al., 2000). These proteins are involved in maintaining tumor cell survival or for regulating resistance to apoptosis induction by various therapeutics (Bilim et al., 2003). The X-chromosome linked IAP (XIAP) is the best characterized of the IAP family members in terms of its caspase inhibitory mechanism. The XIAP protein contains three BIR domains. XIAP levels have been shown to be pathologically elevated in many acute and chronic leukemia, prostate cancer, lung cancer, and other tumor (Byrd et al., 2002; Ferreira et al., 2001; Hofmann et al., 2002; Schimmer et al., 2003; Seligson et al., 2006). Schimmer et al., (2004) have recently reported that small molecule antagonists of apoptosis suppress XIAP function and exhibit a broad anti-tumor activity and such molecules are considered as targets for cancer therapy. The TNF ligand superfamily serves as an important role in the host immune defenses against cancer as an inducer of apoptosis in tumor cells. Apo2L/TRAIL recently has drawn interest as a potential effective anti-tumor therapeutic agent in a variety of cell lines since it is selectively cytotoxic against transformed cells and not against the majority of normal cells. Our recent findings have demonstrated that drug-resistant CaP cells are resistant to TRAIL and also demonstrated that chemotherapeutic drugs (e.g. Act D, ADR and VP-16) sensitize CaP cells to TRAIL-mediated apoptosis (Zisman et al, 2001; Ng et al., 2002). Unlike a recent study showing 4 FLIP overexpression is responsible for TRAIL (Fulda et al., 2000), we have also identified a member of inhibitor of apoptosis proteins (IAPs), XIAP, as responsible for resistance in CaP. Therefore, this project is designed to investigate the role and regulation of XIAP overexpression in CaP in order to find new means to inhibit its expression by new agents and reverse TRAIL resistance for clinical use. Our proposed studies are relevant to the success of CaP immunotherapy. Accumulating evidence suggest overexpression of IAPs, particularly XIAP, is a cause of apoptosis dysfunction in cancer cells (Holcik et al., 2000; Kitada et al., 2000). For instance, Tamm et al. (2000) reported the relevance of XIAP in vivo responses to cytarabine in AML. Patients with lower XIAP protein had significantly longer survival and a tendency towards longer remission duration than those with higher levels of XIAP. In prostate cancer, we identified XIAP as and important anti-apoptotic gene product that regulates TRAIL apoptosis. In this grant application, we have proposed to investigate the following aims: 1) The role of XIAP in protecting CaP cells from TRAIL-mediated-apoptosis. Examination of the direct role of XIAP by transfection utilizing the newly reported IAP inhibitor Smac/DIABLO and by XIAP- antisense. The sensitivity to TRAIL and signaling pathway for apoptosis in transfectants and normal CaP will be compared. Further, we proposed to correlate the expression of both XIAP and Smac/ DIABLO in freshly derived normal, benign, and human prostate tumor cells at different stages of the disease and establish correlations with prognosis. 2) The role of constitutively activated NFκB (survival factor) in the regulation of both resistance to TRAIL and XIAP-expression. We proposed to examine the role of NFκB in the regulation of XIAP expression and TRAIL sensitivity. 3) The roles of constitutive and exogenous TNF-α and IL-6 in the regulation of NFκB, XIAP expression, and sensitivity to TRAIL. 5 Body The following tasks were proposed for investigation: Task 1: The role of XIAP in protecting CaP cells from TRAIL-mediated apoptosis Task 2: Regulation of XIAP by NFκB and NFκB regulation of XIAP Task 3: To determine the role of endogenous TNF-α and IL-6 in the regulation of XIAP and resistance to TRAIL We have investigated all of the above proposed tasks and several reports emanated from our findings. Further, we have made several novel findings and have identified new gene products that regulate TRAIL sensitivity in prostate cancer cells. The following summarizes our progress to date. We have proposed to examine in Task 1 the direct role of XIAP in the regulation of TRAIL resistance by transfection experiments utilizing the newly reported IAP inhibitor, Smac/DIABLO and by XIAP antisense. We have also proposed to examine the sensitivity and signaling to TRAIL in the transfectants. We have completed the studies with the transfection of CaP cells with Smac/DIABLO and the studies have been reported in two publications (Ng, et al. 2002; Ng and Bonavida, 2002a). Briefly, the studies provided novel findings in the mechanism of XIAP regulation of TRAIL resistance. Noteworthy, we show in CaP cells that TRAIL signaling results in the loss of mitochondrial potential, the release of cytochrome c and Smac/DIABLO into the cytoplasm in the absence of caspase activation due to overexpression of XIAP. However, inhibition of XIAP by transfection with Smac/DIABLO (or treatment with Actinomycin D, which inhibits XIAP expression) allowed the TRAIL signaling to proceed to activation of caspase 9 and 3 and induction of apoptosis. These findings demonstrated that two complementary signals are involved to overcome prostate cancer as well as possibly other cancers to TRAIL-induced apoptosis. The two signal model has been proposed in the studies that we have reported and has also been detailed in a recent review that was published in Advances in Cancer Research (Ng and Bonavida, 2002b). In Task 1 we have also proposed to examine the expression of XIAP and Smac/DIABLO in freshly derived normal, benign, and prostate tumor cells at different stages and grades and correlation with prognosis. We have examined the expression of Smac/DIABLO in tumor cell lines by immunohistochemistry (IHC). We have established the optimal conditions and specificity of the antibody against Smac/DIABLO and demonstrated successfully that the antibody is applicable for IHC in addition to its usage for Western blot in our publications. Due to limitations in CaP tissue microarrays, we first examined the clinical role of XIAP overexpresssion in such microarrrays. Prognostic significance of XIAP expression in CaP tissue microarrays Our findings have established that overexpression of XIAP in CaP is associated with both chemoresistance and immune resistance to TRAIL-induced apoptosis. We hypothesize that overexpression of XIAP in human CaP tissues may be associated with resistance and thus, may be of prognostic clinical significance. We examined the expression of XIAP by immunohistochemistry using CaP tissue microarrays and the data were statistically analyzed. The findings demonstrate that overexpression of XIAP is of prognostic significance in a subset of patients with prostate cancer. A manuscript has been completed and will be submitted shortly (Seligson et al., 2006; appendix). The abstract is highlighted below. 6 Abstract Objective: The X-linked Inhibitor of Apoptosis (XIAP), a member of the family proteins, has been linked to tumor cell survival and drug resistance by direct blockade of caspase-mediated extrinsic apoptotic pathways. Thus, XIAP status may help predict prostate cancer recurrence and clinical response to therapies relying on unencumbered apoptotic machinery. It is therefore important to validate the foundational protein expression patterns of XIAP and examine its prognostic implications in human prostate cancer. Methods: Immunohistochemistry was performed on tissue microarrays constructed from paraffin embedded primary prostate cancer specimens from 226 hormone naïve patients who underwent radical retropubic prostatectomy. 223 cases provided informative epithelium for XIAP analysis encompassing 1,107 total tissue microarray spots including morphologically normal prostate (NL; n=252), benign prostatic hyperplasia (BPH; n=122), prostatic intraepithelial neoplasia (PIN; n=48) and invasive prostate cancer (Cancer; n=685). XIAP expression was scored in a semi-quantitative fashion using an integrated intensity measure (0.0-3.0). The protein expression distribution was examined across the spectrum of epithelial tissues and its association with standard clinicopathological covariates and tumor recurrence was examined in 192 outcome- informative patients. Results: The mean XIAP expression was significantly higher in prostate cancer (intensity = 1.32) compared to PIN (intensity = 1.08; p=0.019), normal (intensity = 0.78; p<0.0001), and BPH (intensity = 0.57; p<0.0001). 69% of BPH stained negatively to weakly (intensity <1.0), 53% of normal, 37% of PIN and only 26% of prostate carcinomas. With XIAP expression dichotomized at an intensity of 1.8, XIAP is an independent predictor of tumor recurrence in multivariate Cox proportional hazards analysis in all patients (P = 0.0025; HR = 8.92; 95% CI = 2.16-38.86), as well as after substratifying by Gleason score (P = 0.010; HR = 6.61; 95% CI = 1.57-27.89 for high Gleason score [7-10] cases). In patient substrata with low Gleason score tumors [2-6], no patients (0%) with an XIAP intensity > 1.8 (n=23) experienced tumor recurrence, while 26% with low XIAP (n=89) recurred. Patients with high grade or non-organ confined tumors with high XIAP have a lower risk of recurrence as a group than any patients whose tumors express low XIAP, even those of low grade or that are organ confined. These data are consistent with findings in a recent report (Krajewska 2003). Conclusions: XIAP is expressed at higher levels in prostate cancers compared to matched normal tissues. High XIAP expression is strongly associated with a reduced risk of tumor recurrence, and is not directly associated with Gleason score, tumor stage, capsular involvement or preoperative PSA status, suggesting that it is a novel prognosticator and a potential target for prostate cancer diagnosis and therapy. Based on these results, patients with lower XIAP expression in tumors are most in need of therapeutic intervention and may also be most responsiveness to chemotherapeutic and death receptor targeted therapies. In task 2 and 3 we have proposed to investigate the role of NF-κB and TNF-α in the regulation of XIAP. 1. Role of constitutive NF-κB activity and downstream anti-apoptotic gene expression (XIAP and Bcl- ) in the regulation of TRAIL resistance. xL We and others have also demonstrated that prostate cancer cell lines exhibit constitutively active nuclear factor kappa B (NF-κB) (Suh et al., 2002; Huerta-Yepez et al., 2004). NF-κB regulates 7 the transcription of many anti-apoptotic gene products, including XIAP and Bcl- . We xL examined the role and mechanism of NF-κB-induced resistance to TRAIL apoptosis. We used the nitric oxide donor DETANONOate and the NF-κB inhibitior Bay 11-7085 to inhibit NF-κB activity, and treated PC-3 cells resulted in downstream inhibition of both XIAP and Bcl- xL expression. The inhibition of NF-κB resulted in sensitization to TRAIL apoptosis. Further, the role of Bcl- in the regulation of TRAIL resistance was corroborated by the use of the chemical xL inhibitor 2-methoxyantimycin A which sensitized PC-3 cells to TRAIL-induced apoptosis. We further examined the apoptotic-signaling pathways following treatment of PC-3 cells with the combination of NF-κB inhibitors and TRAIL, and demonstrate that the combination, but not single agents alone, activate the mitochondrial pathway and the activation of caspases 9 and 3 and the induction of apoptosis. The above findings have been recently reported (Huerta-Yepez et al., 2004). 2. Regulation of prostate carcinoma cell line resistance to TRAIL via upregulation of DR5 expression. We have previously reported that treatment of CaP with nitric oxide donors or inhibitors of NF-κB resulted in sensitization to TRAIL-induced apoptosis (Huerta-Yepez et al., 2004). We examined the mechanism of resistance by delineating putative transcription factors, aside from NF-κB, that may be involved in DR5 transcription. We demonstrate, by using various constructs of the DR5 promoter, that deletion in the promoter of a region with a putative YY1 DNA binding site resulted in significant augmentation of luciferase activity and suggested that YY1 may negatively regulate DR5 transcription. This finding was corroborated by demonstrating that mutation in the DNA binding site also resulted in augmentation of luciferase activity. In addition, inhibition of YY1 via chemical modification with nitric oxide or by transfection with sIRNA for YY1 resulted in both upregulation of DR5 expression and sensitization to TRAIL-induced apoptosis. Altogether, these findings demonstrate that inhibition of NF-κB, which results in downstream inhibition of YY1 and XIAP, results in inhibition of YY1 activity, leading to upregulation of DR5 and subsequent sensitization to TRAIL. The findings are in a manuscript that will soon be submitted for publication (Huerta-Yepez et al., 2006). The abstract is highlighted below. Abstract Most tumors are resistant to TRAIL and need to be sensitized to undergo apoptosis. We have recently reported that TRAIL-resistant human prostate carcinoma cell lines can be sensitized by various NF-κB inhibitors (Huerta-Yepez et al., 2004), and sensitization correlated with upregulation of DR5 expression. We hypothesized that a gene product(s) regulated by NF- κB with DR5 repressor activity may be responsible for the DR5 regulation. Inhibition of NF-κB activity resulted in significant upregulation of DR5 expression and sensitized prostate tumor cells to TRAIL-mediated apoptosis and synergy is achieved. Treatment of PC-3 cells with NO inhibited both NF-κB and YY1 DNA-binding activity and also inhibited YY1 expression. Treatment of PC-3 cells with YY1 siRNA resulted in upregulation of DR5 expression and sensitization to TRAIL-induced apoptosis. The direct role of YY1 in the regulation of DR5 expression was examined in an DR5 luciferase reporter system (pDR5). Two constructs were generated, the pDR5/-605 construct with a deletion in the promoter region containing the putative YY1 DNA-binding region (-1224 to -605) and a construct pDR5-YY1 with a mutation of the YY1 DNA-binding site. Transfection of PC-3 cells with these two constructs resulted in 8 significant (3-fold) augmentation of luciferase activity over baseline suggesting the repressor activity of YY1. The present findings demonstrate that YY1 negatively regulates DR5 transcription and expression and hence, regulates resistance to TRAIL-induced apoptosis. Inhibitors of YY1 expression and/or activity in combination with TRAIL may be useful in the treatment of TRAIL-resistant tumor cells. 3. Chemical modification of the transcription repressor YY1 by nitric oxide: mechanism of NO- induced upregulation of DR5 and sensitization to TRAIL-induced apoptosis The above findings in section 1 (above) and 3 (below) have clearly demonstrated that DR5 transcription is negatively regulated by the overexpression of the transcription repressor YY1 in CaP and overexpression of YY1 is regulated by the constitutively activated NFkB activity which also regulates XIAP. In addition to the transcription regulation of YY1 by NO donors and by drugs, we demonstrate that NO also directly modifies YY1 via S-nitrosylation and thus, preventing its DNA binding activity and transcription DR5 repressor activity. These studies have been recently published (Hongo et al., 2005) and the abstract is presented below. Abstract Treatment of several prostate cancer (CaP) cell lines (PC-3, CL-1, and DU-145) with the nitric oxide (NO) donor DETA/NONOate upregulated Fas expression and sensitized the CaP cells to the Fas ligand CH-11 agonist monoclonal antibody-induced apoptosis. Previous findings demonstrated that the transcription repressor Yin Yang 1 (YY1), which is inhibited by NO, negatively regulates Fas transcription [H.J. Garban, B. Bonavida, Nitric oxide inhibits the transcription repressor Yin-Yang 1 binding activity at the silencer region of the Fas promoter: a pivotal role for nitric oxide in the upregulation of Fas gene expression in human tumor cells, J. Immunol. 167 (2001) 75-81]. YY1 is a zinc finger protein and thus, we hypothesized that NO inhibits YY1 activity via S-nitrosation of critical cysteines residues coordinated by Zn2+. Treatment of PC-3 cells with DETA/NONOate inhibited the constitutive DNA-binding activity of YY1 as assessed by EMSA. Further, treatment with DETA/NONOate resulted in S-nitrosation of YY1 as detected by two different methods. The DAN-based method examined NO-treated tumor- derived cell lysates that were immunoprecipitated with an anti-YY1 specific antibody and the NO released was determined quantitatively by fluorometry. The second method consisted of immunoprecipitation of the tumor cell lysates by an anti-SNO cysteine antibody and the immunoprecipitate was immunoblotted with anti-YY1 antibody. Both methods revealed significant S-nitrosation of YY1 by DETA/NONOate treatment over control untreated cells. The S-nitrosation of YY1 was further corroborated by immunohistochemistry using dual color immunofluorescence. The direct role of YY1 in the negative regulation of Fas expression was demonstrated by transfection of cells with siRNA YY1. The transfectants exhibited upregulation of Fas expression in the absence of treatment with DETA/NONOate and were sensitized to CH- 11-induced apoptosis. Altogether, these findings reveal that NO inhibits YY1 DNA-binding activity through S-nitrosation and consequently results in upregulation of Fas expression and tumor cell sensitization to Fas-induced apoptosis. 4. Role of chemotherapeutic drugs in the sensitization of CaP to TRAIL-induced apoptosis Previous studies have reported that treatment of tumor cell lines with chemotherapeutic drugs such as adriamycin or CDDP resulted in upregulation of DR5 expression and sensitization to TRAIL-induced apoptosis. The mechanism underlying the drug-induced regulation of DR5 9 expression is not known. Our studies with the chemotherapeutic drugs corroborate the above findings. In addition, we demonstrate that treatment of CaP with CDDP resulted in inhibition of NF-κB and YY1 activities, resulting in upregulation of DR5. We demonstrate that inhibitors of XIAP, NF-κB and YY1 mimic CDDP-induced effects. In addition, we demonstrate that YY1 is a transcription repressor and negatively regulates DR5 expression and thus, its inhibition by CDDP, reverses the repression and upregulates DR5 transcription and sensitizes CaP to TRAIL-induced apoptosis. Further studies, as demonstrated above for NO-mediated effects using reporter systems, confirmed the role of CDDP-induced inhibition of YY1 in the regulation of DR5 expression and sensitivity to TRAIL. A manuscript has been prepared and will soon be submitted for publication (Baritaki et al., 2006; Appendix). The abstract of this publication in highlighted below. Abstract Cancer patients initially respond to treatment with chemotherapy, however, recurrences occur and the tumors become refractory to further chemotherapy. Immunotherapy is currently being investigated as an alternative to overcome drug resistance. TRAIL, a member of the TNF family, has been shown to kill sensitive tumor cells with minimal toxicity to normal tissues and is a new candidate for immunotherapy. Many drug-resistant tumor cells are also resistant to TRAIL and such tumors require sensitization to reverse TRAIL resistance. We and others have reported that several sensitizing agents (ex. VP-16, CDDP, ADR, chemical inhibitors, etc.) in combination with TRAIL result in reversal of resistance to TRAIL apoptosis. Sensitization correlated with the upregulation of DR5 expression. This study examined the mechanism underlying the upregulation of DR5 expression. We hypothesize that the sensitizing agents may inhibit a transcription repressor acting at the DR5 promoter. Treatment of drug resistant PC-3 tumor cells with drugs (example CDDP, vp-16, adriamycin, vincristine) sensitized the tumor cells to TRAIL-induced apoptosis and apoptosis correlated with upregulation of DR5 expression and inhibition of YY1. By examining the promoter of DR5, we detected the presence of one putative binding site for the transcription repressor YY1. We examined whether YY1 negatively regulates DR5 transcription and whether YY1 inhibition by the drug upregulates DR5 expression. We used PC-3 cells transfected with a luciferase reporter system (pDR5 WT) and plasmids in which the YY1 binding site was either deleted (pDR5 -605) and/or mutated (pDR5/YY1 mutant). The findings revealed that the baseline reporter activity was significantly augmented in cells transfected with either the deleted or mutated plasmids. In addition, CDDP treatment augmented the luciferase activity in the WT reporter system, whereas there was no augmentation in the deleted or mutant transfected cells. The direct role of YY1 in the upregulation of DR5 expression and sensitization to TRAIL-induced apoptosis was demonstrated in cells treated with siRNA YY1. The findings demonstrate that drug-induced upregulation of DR5 and sensitization to TRAIL is mediated through inhibition of the transcription repressor YY1. Inhibition of YY1 correlated with sensitization to TRAIL-induced apoptosis. 5. Role of XIAP and Bcl- expression in the regulation of PC-3 resistance to CDDP-induced xL apoptosis We have found that prostate cancer tumor cell lines (PC-3, CL-1, LNCaP) are resistant to CDDP-mediated apoptosis. We examined whether the resistance is due in part to the constitutive activation of NF-κB and downstream regulation of XIAP and Bcl- expression similar to the xL resistance observed against TRAIL. We also hypothesized that tumor-derived cytokines (e.g. 10

See more

The list of books you might like

Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.