Browsing by Author "Chen, Ming"
Results Per Page
Sort Options
Item Open Access Abi1 loss drives prostate tumorigenesis through activation of EMT and non-canonical WNT signaling(Cell Communication and Signaling, 2019-12) Nath, Disharee; Li, Xiang; Mondragon, Claudia; Post, Dawn; Chen, Ming; White, Julie R; Hryniewicz-Jankowska, Anita; Caza, Tiffany; Kuznetsov, Vladimir A; Hehnly, Heidi; Jamaspishvili, Tamara; Berman, David M; Zhang, Fan; Kung, Sonia HY; Fazli, Ladan; Gleave, Martin E; Bratslavsky, Gennady; Pandolfi, Pier Paolo; Kotula, LeszekItem Open Access An aberrant SREBP-dependent lipogenic program promotes metastatic prostate cancer.(Nature genetics, 2018-02) Chen, Ming; Zhang, Jiangwen; Sampieri, Katia; Clohessy, John G; Mendez, Lourdes; Gonzalez-Billalabeitia, Enrique; Liu, Xue-Song; Lee, Yu-Ru; Fung, Jacqueline; Katon, Jesse M; Menon, Archita Venugopal; Webster, Kaitlyn A; Ng, Christopher; Palumbieri, Maria Dilia; Diolombi, Moussa S; Breitkopf, Susanne B; Teruya-Feldstein, Julie; Signoretti, Sabina; Bronson, Roderick T; Asara, John M; Castillo-Martin, Mireia; Cordon-Cardo, Carlos; Pandolfi, Pier PaoloLipids, either endogenously synthesized or exogenous, have been linked to human cancer. Here we found that PML is frequently co-deleted with PTEN in metastatic human prostate cancer (CaP). We demonstrated that conditional inactivation of Pml in the mouse prostate morphs indolent Pten-null tumors into lethal metastatic disease. We identified MAPK reactivation, subsequent hyperactivation of an aberrant SREBP prometastatic lipogenic program, and a distinctive lipidomic profile as key characteristic features of metastatic Pml and Pten double-null CaP. Furthermore, targeting SREBP in vivo by fatostatin blocked both tumor growth and distant metastasis. Importantly, a high-fat diet (HFD) induced lipid accumulation in prostate tumors and was sufficient to drive metastasis in a nonmetastatic Pten-null mouse model of CaP, and an SREBP signature was highly enriched in metastatic human CaP. Thus, our findings uncover a prometastatic lipogenic program and lend direct genetic and experimental support to the notion that a Western HFD can promote metastasis.Item Open Access CCDC62/ERAP75 functions as a coactivator to enhance estrogen receptor beta-mediated transactivation and target gene expression in prostate cancer cells.(Carcinogenesis, 2009-05) Chen, Ming; Ni, Jing; Chang, Hong-Chiang; Lin, Chen-Yong; Muyan, Mesut; Yeh, ShuyuanHuman prostate cancer (PCa) and prostate epithelial cells predominantly express estrogen receptor (ER) beta, but not ERalpha. ERbeta might utilize various ER coregulators to mediate the E2-signaling pathway in PCa. Here, we identified coiled-coil domain containing 62 (CCDC62)/ERAP75 as a novel ER coactivator. CCDC62/ERAP75 is widely expressed in PCa cell lines and has low expression in MCF7 cells. Both in vitro and in vivo interaction assays using mammalian two-hybrid, glutathione S-transferase pull-down and coimmunoprecipitation methods proved that ERbeta can interact with the C-terminus of CCDC62/ERAP75 via the ligand-binding domain. The first LXXLL motif within CCDC62/ERAP75 is required for the interaction between ERbeta and CCDC62/ERAP75. Electrophoretic mobility shift assay showed that CCDC62/ERAP75 can be recruited by the estrogen response element-ER complex in the presence of ligand. Furthermore, a chromatin immunoprecipitation assay demonstrated the hormone-dependent recruitment of CCDC62/ERAP75 within the promoter of the estrogen-responsive gene cyclin D1. In addition, using silencing RNA (siRNA) against endogeneous CCDC62/ERAP75, we demonstrated that inhibition of endogenous CCDC62/ERAP75 results in the suppression of ERbeta-mediated transactivation as well as target gene expression in LNCaP cells. More importantly, using the tet-on overexpression system, we showed that induced expression of CCDC62/ERAP75 can enhance the E2-regulated cyclin D1 expression and cell growth in LNCaP cells. Together, our results revealed the role of CCDC62/ERAP75 as a novel coactivator in PCa cells that can modulate ERbeta transactivation and receptor function.Item Open Access Compound haploinsufficiency of Dok2 and Dusp4 promotes lung tumorigenesis.(The Journal of clinical investigation, 2019-01) Chen, Ming; Zhang, Jiangwen; Berger, Alice H; Diolombi, Moussa S; Ng, Christopher; Fung, Jacqueline; Bronson, Roderick T; Castillo-Martin, Mireia; Thin, Tin Htwe; Cordon-Cardo, Carlos; Plevin, Robin; Pandolfi, Pier PaoloRecurrent broad-scale heterozygous deletions are frequently observed in human cancer. Here we tested the hypothesis that compound haploinsufficiency of neighboring genes at chromosome 8p promotes tumorigenesis. By targeting the mouse orthologs of human DOK2 and DUSP4 genes, which were co-deleted in approximately half of human lung adenocarcinomas, we found that compound-heterozygous deletion of Dok2 and Dusp4 in mice resulted in lung tumorigenesis with short latency and high incidence, and that their co-deletion synergistically activated MAPK signaling and promoted cell proliferation. Conversely, restoration of DOK2 and DUSP4 in lung cancer cells suppressed MAPK activation and cell proliferation. Importantly, in contrast to downregulation of DOK2 or DUSP4 alone, concomitant downregulation of DOK2 and DUSP4 was associated with poor survival in human lung adenocarcinoma. Therefore, our findings lend in vivo experimental support to the notion that compound haploinsufficiency, due to broad-scale chromosome deletions, constitutes a driving force in tumorigenesis.Item Open Access Defects of prostate development and reproductive system in the estrogen receptor-alpha null male mice.(Endocrinology, 2009-01) Chen, Ming; Hsu, Iawen; Wolfe, Andrew; Radovick, Sally; Huang, KuoHsiang; Yu, Shengqiang; Chang, Chawnshang; Messing, Edward M; Yeh, ShuyuanThe estrogen receptor-alpha knockout (ERalphaKO, ERalpha-/-) mice were generated via the Cre-loxP system by mating floxed ERalpha mice with beta-actin (ACTB)-Cre mice. The impact of ERalpha gene deletion in the male reproductive system was investigated. The ACTB-Cre/ERalpha(-/-) male mice are infertile and have lost 90% of epididymal sperm when compared with wild-type mice. Serum testosterone levels in ACTB-Cre/ERalpha(-/-) male mice are 2-fold elevated. The ACTB-Cre/ERalpha(-/-) testes consist of atrophic and degenerating seminiferous tubules with less cellularity in the disorganized seminiferous epithelia. Furthermore, the ventral and dorsal-lateral prostates of ACTB-Cre/ERalpha(-/-) mice display reduced branching morphogenesis. Loss of ERalpha could also be responsible for the decreased fibroblast proliferation and changes in the stromal content. In addition, we found bone morphogenetic protein, a mesenchymal inhibitor of prostatic branching morphogenesis, is significantly up-regulated in the ACTB-Cre/ERalpha(-/-) prostates. Collectively, these results suggest that ERalpha is required for male fertility, acts through a paracrine mechanism to regulate prostatic branching morphogenesis, and is involved in the proliferation and differentiation of prostatic stromal compartment.Item Open Access Deregulated PP1α phosphatase activity towards MAPK activation is antagonized by a tumor suppressive failsafe mechanism.(Nature communications, 2018-01-15) Chen, Ming; Wan, Lixin; Zhang, Jiangwen; Zhang, Jinfang; Mendez, Lourdes; Clohessy, John G; Berry, Kelsey; Victor, Joshua; Yin, Qing; Zhu, Yuan; Wei, Wenyi; Pandolfi, Pier PaoloThe mitogen-activated protein kinase (MAPK) pathway is frequently aberrantly activated in advanced cancers, including metastatic prostate cancer (CaP). However, activating mutations or gene rearrangements among MAPK signaling components, such as Ras and Raf, are not always observed in cancers with hyperactivated MAPK. The mechanisms underlying MAPK activation in these cancers remain largely elusive. Here we discover that genomic amplification of the PPP1CA gene is highly enriched in metastatic human CaP. We further identify an S6K/PP1α/B-Raf signaling pathway leading to activation of MAPK signaling that is antagonized by the PML tumor suppressor. Mechanistically, we find that PP1α acts as a B-Raf activating phosphatase and that PML suppresses MAPK activation by sequestering PP1α into PML nuclear bodies, hence repressing S6K-dependent PP1α phosphorylation, 14-3-3 binding and cytoplasmic accumulation. Our findings therefore reveal a PP1α/PML molecular network that is genetically altered in human cancer towards aberrant MAPK activation, with important therapeutic implications.Item Open Access DOK2 inhibits EGFR-mutated lung adenocarcinoma.(PloS one, 2013-01) Berger, Alice H; Chen, Ming; Morotti, Alessandro; Janas, Justyna A; Niki, Masaru; Bronson, Roderick T; Taylor, Barry S; Ladanyi, Marc; Van Aelst, Linda; Politi, Katerina; Varmus, Harold E; Pandolfi, Pier PaoloSomatic mutations in the EGFR proto-oncogene occur in ~15% of human lung adenocarcinomas and the importance of EGFR mutations for the initiation and maintenance of lung cancer is well established from mouse models and cancer therapy trials in human lung cancer patients. Recently, we identified DOK2 as a lung adenocarcinoma tumor suppressor gene. Here we show that genomic loss of DOK2 is associated with EGFR mutations in human lung adenocarcinoma, and we hypothesized that loss of DOK2 might therefore cooperate with EGFR mutations to promote lung tumorigenesis. We tested this hypothesis using genetically engineered mouse models and find that loss of Dok2 in the mouse accelerates lung tumorigenesis initiated by oncogenic EGFR, but not that initiated by mutated Kras. Moreover, we find that DOK2 participates in a negative feedback loop that opposes mutated EGFR; EGFR mutation leads to recruitment of DOK2 to EGFR and DOK2-mediated inhibition of downstream activation of RAS. These data identify DOK2 as a tumor suppressor in EGFR-mutant lung adenocarcinoma.Item Open Access Endosome and INPP4B.(Oncotarget, 2016-01) Chew, Chen Li; Chen, Ming; Pandolfi, Pier PaoloItem Open Access Heat shock factor 1 directly regulates transsulfuration pathway to promote prostate cancer proliferation and survival(Communications Biology) Hauck, J Spencer; Moon, David; Jiang, Xue; Wang, Mu-En; Zhao, Yue; Xu, Lingfan; Quang, Holly; Butler, William; Chen, Ming; Macias, Everardo; Gao, Xia; He, Yiping; Huang, JiaotiAbstractThere are limited therapeutic options for patients with advanced prostate cancer (PCa). We previously found that heat shock factor 1 (HSF1) expression is increased in PCa and is an actionable target. In this manuscript, we identify that HSF1 regulates the conversion of homocysteine to cystathionine in the transsulfuration pathway by altering levels of cystathionine-β-synthase (CBS). We find that HSF1 directly binds the CBS gene and upregulates CBS mRNA levels. Targeting CBS decreases PCa growth and induces tumor cell death while benign prostate cells are largely unaffected. Combined inhibition of HSF1 and CBS results in more pronounced inhibition of PCa cell proliferation and reduction of transsulfuration pathway metabolites. Combination of HSF1 and CBS knockout decreases tumor size for a small cell PCa xenograft mouse model. Our study thus provides new insights into the molecular mechanism of HSF1 function and an effective therapeutic strategy against advanced PCa.Item Open Access In Vivo Role of INPP4B in Tumor and Metastasis Suppression through Regulation of PI3K-AKT Signaling at Endosomes.(Cancer discovery, 2015-07) Li Chew, Chen; Lunardi, Andrea; Gulluni, Federico; Ruan, Daniel T; Chen, Ming; Salmena, Leonardo; Nishino, Michiya; Papa, Antonella; Ng, Christopher; Fung, Jacqueline; Clohessy, John G; Sasaki, Junko; Sasaki, Takehiko; Bronson, Roderick T; Hirsch, Emilio; Pandolfi, Pier PaoloThe phosphatases PTEN and INPP4B have been proposed to act as tumor suppressors by antagonizing PI3K-AKT signaling and are frequently dysregulated in human cancer. Although PTEN has been extensively studied, little is known about the underlying mechanisms by which INPP4B exerts its tumor-suppressive function and its role in tumorigenesis in vivo. Here, we show that a partial or complete loss of Inpp4b morphs benign thyroid adenoma lesions in Pten heterozygous mice into lethal and metastatic follicular-like thyroid cancer (FTC). Importantly, analyses of human thyroid cancer cell lines and specimens reveal INPP4B downregulation in FTC. Mechanistically, we find that INPP4B, but not PTEN, is enriched in the early endosomes of thyroid cancer cells, where it selectively inhibits AKT2 activation and in turn tumor proliferation and anchorage-independent growth. We therefore identify INPP4B as a novel tumor suppressor in FTC oncogenesis and metastasis through localized regulation of the PI3K-AKT pathway at the endosomes.Although both PTEN and INPP4B can inhibit PI3K-AKT signaling through their lipid phosphatase activities, here we demonstrate lack of an epistatic relationship between the two tumor suppressors. Instead, the qualitative regulation of PI3K-AKT2 signaling by INPP4B provides a mechanism for their cooperation in suppressing thyroid tumorigenesis and metastasis.Item Open Access Interplay between c-Src and the APC/C co-activator Cdh1 regulates mammary tumorigenesis(Nature Communications, 2019-12) Han, Tao; Jiang, Shulong; Zheng, Hong; Yin, Qing; Xie, Mengyu; Little, Margaret R; Yin, Xiu; Chen, Ming; Song, Su Jung; Beg, Amer A; Pandolfi, Pier Paolo; Wan, LixinItem Open Access Lipid nanoparticle-based mRNA vaccines: a new frontier in precision oncology(Precision Clinical Medicine) Jacob, Eden M; Huang, Jiaoti; Chen, MingAbstract The delivery of lipid nanoparticle (LNP)-based mRNA therapeutics has captured the attention of the vaccine research community as an innovative and versatile tool for treating a variety of human malignancies. mRNA vaccines are now in the limelight as an alternative to conventional vaccines owing to their high precision, low-cost, rapid manufacture, and superior safety profile. Multiple mRNA vaccine platforms have been developed to target several types of cancer, and many have demonstrated encouraging results in animal models and human trials. The effectiveness of these new mRNA vaccines depends on the efficacy and stability of the antigen(s) of interest generated and the reliability of their delivery to antigen-presenting cells (APCs), especially dendritic cells (DCs). In this review, we provide a detailed overview of mRNA vaccines and their delivery strategies and consider future directions and challenges in advancing and expanding this promising vaccine platform to widespread therapeutic use against cancer.Item Open Access Loss of epithelial oestrogen receptor α inhibits oestrogen-stimulated prostate proliferation and squamous metaplasia via in vivo tissue selective knockout models.(The Journal of pathology, 2012-01) Chen, Ming; Yeh, Chiuan-Ren; Chang, Hong-Chiang; Vitkus, Spencer; Wen, Xing-Qiao; Bhowmick, Neil A; Wolfe, Andrew; Yeh, ShuyuanSquamous metaplasia (SQM) is a specific phenotype in response to oestrogen in the prostate and oestrogen receptor (ER) α is required to mediate this response. Previous studies utilizing tissue recombination with seminal vesicle (SV) mesenchyme and prostatic ductal tips from wild type and ERαKO mice suggested that both epithelial and stromal ERα are necessary for SQM. However, tissue recombination is conducted in the renal capsule of immune-deficient mice, in which the microenvironment is different from normal prostate microenvironment in the intact mice. Furthermore, whether the requirement of stromal ERα in the SV for developing SQM is the same as in the prostate is unknown. Therefore, there is a clear need to evaluate the respective roles of ERα in prostate epithelial versus stromal compartments in the intact mouse. Here we generated a mouse model that has selectively lost ERα in either stromal (FSP-ERαKO) or epithelial prostate cells (pes-ERαKO) to determine the requirements of ERα for oestrogen-stimulated prostate proliferation and SQM. Our results indicated that FSP-ERαKO prostates develop full and uniform SQM, which suggests that loss of the majority (~65%) of stromal ERα will not influence oestrogen-mediated SQM. In contrast, loss of epithelial ERα inhibits oestrogen-mediated prostate growth and SQM evidenced by decreasing cytokertin 10 positive squamous cell stratification and differentiation, by reduced ERα protein expression in SQM compared to wild type mice ERα, and by the presence of normal proliferative activities in the oestrogen-treated pes-ERαKO prostates. These in vivo results suggest that epithelial ERα is required for oestrogen-mediated proliferative response and could be an appropriate target for preventing aberrant oestrogen signalling in the prostate.Item Open Access Loss of LDAH associated with prostate cancer and hearing loss.(Human molecular genetics, 2018-12) Currall, Benjamin B; Chen, Ming; Sallari, Richard C; Cotter, Maura; Wong, Kristen E; Robertson, Nahid G; Penney, Kathryn L; Lunardi, Andrea; Reschke, Markus; Hickox, Ann E; Yin, Yanbo; Wong, Garrett T; Fung, Jacqueline; Brown, Kerry K; Williamson, Robin E; Sinnott-Armstrong, Nicholas A; Kammin, Tammy; Ivanov, Andrew; Zepeda-Mendoza, Cinthya J; Shen, Jun; Quade, Bradley J; Signoretti, Sabina; Arnos, Kathleen S; Banks, Alexander S; Patsopoulos, Nikolaos; Liberman, M Charles; Kellis, Manolis; Pandolfi, Pier Paolo; Morton, Cynthia CGreat strides in gene discovery have been made using a multitude of methods to associate phenotypes with genetic variants, but there still remains a substantial gap between observed symptoms and identified genetic defects. Herein, we use the convergence of various genetic and genomic techniques to investigate the underpinnings of a constellation of phenotypes that include prostate cancer (PCa) and sensorineural hearing loss (SNHL) in a human subject. Through interrogation of the subject's de novo, germline, balanced chromosomal translocation, we first identify a correlation between his disorders and a poorly annotated gene known as lipid droplet associated hydrolase (LDAH). Using data repositories of both germline and somatic variants, we identify convergent genomic evidence that substantiates a correlation between loss of LDAH and PCa. This correlation is validated through both in vitro and in vivo models that show loss of LDAH results in increased risk of PCa and, to a lesser extent, SNHL. By leveraging convergent evidence in emerging genomic data, we hypothesize that loss of LDAH is involved in PCa and other phenotypes observed in support of a genotype-phenotype association in an n-of-one human subject.Item Open Access Nuclear PTEN Regulates Thymidylate Biosynthesis and Cellular Sensitivity to Antifolate Treatment(2023) Loh, Zoe NathaniaMetabolic reprogramming contributes to tumorigenesis and holds significant promise for cancer therapy. The PTEN tumor suppressor governs a variety of biological processes, including metabolism, by acting on distinct molecular targets in different subcellular compartments. In the cytoplasm, PTEN regulates a plethora of metabolic processes through antagonizing the PI3K/AKT/mTORC1 pathway. However, the metabolic regulation of PTEN in the nucleus remain undefined. Using a gain-of-function approach to examine the metabolic consequences of PTEN targeted to different sub-cellular compartments in human prostate cancer cell lines, we reveal a nuclear function for PTEN in controlling de novo thymidylate biosynthesis and may also open novel therapeutic avenues for targeting nuclear-excluded PTEN prostate cancer cells with anti-folate cancer treatment. The first four chapters of this dissertation are introductory information that outlines the role of PTEN in cancer, the importance of metabolic compartmentalization, the fundamentals of pyrimidine biosynthesis pathways, and the novelty of anti-folate cancer treatments. Chapter 1 explains the role of PTEN as a tumor suppressor and continues on to discuss its role at the cell membrane as a metabolic regulator. The gap in knowledge in the field is understanding the role nuclear PTEN plays as a metabolic regulator. This is important as nuclear PTEN has been shown to be associated with more aggressive cancer phenotypes. Chapter 2 focuses on the background of metabolic compartmentalization and how, by strategically placing genes and metabolites spatiotemporally, the cell is able to execute key molecular mechanisms more efficiently. In this chapter, we highlight where the current field is with metabolic compartmentalization, the advantages of compartmentalization and how utilization of this knowledge can be used for definitive therapeutics. Chapter 3 focuses on pyrimidine biosynthesis and thymidylate biosynthesis. Thymidylate is synthesized de novo by thymidylate synthase (TYMS), with the enzymes dihydrofolate reductase (DHFR) and methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) or serine hydroxymethyltransferase (SHMT) which are required to regenerate 5, 10-methylenetetrahydrofolate. MTHFD1 is the primary source of 5,10-methylenetetrhydrofolate generation, and therefore its proper function in the nucleus ensures the functioning of de novo thymidylate biosynthesis. Using Mass-Spectrometry, we discovered that MTHFD1 is a top candidate protein interacting with PTEN in human prostate cancer cells. This is the key focus of this paper and will be important background for Chapter 7 and 8 which delve into the thymidylate pathway and the potential role of nuclear PTEN. A deeper understanding of nuclear PTEN’s regulation of MTHFD1 may, in turn, open new therapeutic avenues for anti-folate cancer treatment tailored to PTEN sub-cellular localization. This brings us to Chapter 4, which focuses on the current market of anti-folate treatment and the importance of precision medicine and combinatorial treatment. Chapter 5 is the start of our project, it is the basis of the remainder chapters and what allowed us to elucidate the importance of nuclear PTEN. To explore the role of PTEN as not just a tumor suppressor, but as a metabolic regulator, we first sought to generate overexpression cell lines with PTEN localized to various subcellular compartments. As explained in chapter 1 and 2, we discuss how in the cytosol inactive PTEN can be recruited to the plasma membrane where it functions as a lipid phosphatase to suppress the activation of the proto-oncogenic phosphoinositide 3-kinase (PI3K)–AKT-mTOR signaling pathway. In the nucleus, PTEN acts to induce cell cycle arrest and maintain genomic stability. However, the role of PTEN in metabolism is incompletely understood. It is clearly understood that each subcellular compartment harbors specific metabolic activities and PTEN is present in different subcellular locations where it performs distinct functions acting on specific effectors. To explore this, we used a gain-of-function approach to examine the metabolic consequences of PTEN targeted to different sub-cellular compartments. Plasmids were generated using site-directed mutagenesis and PCR. We used the vector pTRIPZ, which is an inducible TET-ON system. This was necessary as overexpression of a tumor suppressor in cancer cell lines, if left permanently on, leads to slow cell growth and expulsion of the plasmid by inherent cancer cell mechanisms. We generated a vector plasmid, which was used as the baseline for all experiments, a wildtype PTEN plasmid, which was “normal” PTEN and PTEN had the ability to localize to the membrane or nucleus as it pleased, a cytoplasmic membrane plasmid, which localized PTEN permanently to the membrane, nuclear PTEN, which localized PTEN permanently to the nucleus, and mutant C124S PTEN, which generates a catalytically silent PTEN variant preventing its role at the membrane and its involvement in the PI3K-AKT-mTOR pathway. All of the plasmids were transfected as a lentivirus into PC3 and C4-2 prostate cancer cells. Both cell lines are PTEN-null and C4-2 represents early stage prostate cancer and is AR positive, while PC3 cells represent later stage metastatic prostate cancer and are AR negative. Prostate cancer is one of the leading causes of morbidity and mortality in the world. Identification of novel therapeutics to combat this deadly disease is still needed today, despite advances in PSA testing, molecular diagnostics, and androgen-deprivation therapy treatments. Inactivation of PTEN by deletion or mutation is found in 20% of primary prostate tumor samples and over 50% of castration-resistant tumors. This highlights the importance of using prostate cancer cell lines as a model system. Cell lines were confirmed for PTEN localization by 3 methods: Western blotting, subcellular fractionation assays, and immunofluorescence assays. Confirmation of all 5 cell lines in both PC3 and C4-2 systems sets up the system used in the next couple chapters. Chapter 6 focuses on the metabolic profiling of our cell lines and highlights the big data generated opening up multiple avenues to be explored. It emphasizes the potential role of PTEN as not only a tumor suppressor, but as a metabolic regulator. Unbiased metabolic profiling was performed on PC3 and C4-2 overexpression cell lines. A polar metabolomics profiling platform using selected reaction monitoring with the 5500 QTRAP hybrid triple quadrupole mass spectrometer, created by Dr John Asara, was utilized for metabolomics data generation. Using LC MS/MS for polar metabolite profiling the platform is a single normal phase hydrophilic interaction liquid chromatographic run (HILIC) and has a short mass spectra acquisition time of only 15 minutes. The platform allows for over 250 metabolic compounds to be targeted without chromatographically scheduled selected reaction monitoring. Lastly, all analysis of data is done through Metaboanalyst, and output files are generated in R command. Large data sets were generated for each cell type and allowed for multiple avenues of research for this dissertation. Looking at a heat map of all 5 cell lines, it was clear nuclear PTEN stood out and followed a different and opposite pattern to that of wildtype and cytoplasmic membrane PTEN. This led us to compare nuclear PTEN to vector PTEN null cell lines and determine which pathways were enriched. Pyrimidine biosynthesis was at the top of the list, which leads us to chapter 7. Chapter 7 focuses on our analysis of pyrimidine biosynthesis and the potential role nuclear PTEN plays in it. The absence of nuclear PTEN is associate with more aggressive disease in patients and therefore can serve as a useful biomarker and potential therapeutic avenue. While purine metabolism happens exclusively in the cytoplasm, pyrimidine metabolism occurs in the cytoplasm, mitochondria, and nucleus. Most notable from our data, in relation to pyrimidine pathways, was the upregulation of dTMP levels. dTMP, as mentioned in chapter 3, is produced from dUMP and both metabolites participate in thymidine biosynthesis. Nuclear localization of the dTMP biosynthesis pathway is a critical factor for allowing the pathway to play its role in DNA synthesis and proper cell division. This pathway utilizes folate one-carbon metabolism of which the enzyme MTHFD1 is an important facilitator of the pathway. Previous data shows, through mass spectrometric analysis, that MTHFD1 was identified as a PTEN interacting protein. This information suggests a potential role that PTEN plays in thymidylate biosynthesis. This chapter aims to highlight the potential roles PTEN plays in thymidylate biosynthesis and pave the way for novel therapeutic treatment. Chapter 8 discusses the findings in chapter 7 and their ability to contribute to the metabolic therapeutic field. We utilized crystal violet assays to measure cell growth over a 6 day time period and treated cells with both an anti-folate and anti-PI3K drug to maximize efficacy against nuclear PTEN-null cancer patients. Pharmaceuticals that target enzymes in folate-dependent dTMP biosynthesis are developed as anti-cancer drugs. 5-fluorouracil (5-FU) is the most widely used anti-folate cancer drug and focuses on inhibition of TYMS. A full discussion of 5-FU’s pathway is discussed in chapter 4. The information and literature lead us to hypothesize that presence of nuclear-PTEN would affect 5-FU efficacy and combinatorial therapeutics would be a novel treatment method. In combination with 5-FU we used Buparlisib, an oral PI3K inhibitor, to inhibit tumor growth via the PI3K pathway which has been shown to promote castration and chemotherapy resistance. We sought to determine if BKM used in combination with 5-FU could be a potential therapeutic option for patients with nuclear PTEN loss. Previous data shows evidence of a protective effect of nuclear PTEN on drug treatment suggesting novel precision therapeutic treatment for loss of nuclear PTEN patients. Chapter 9 concludes our findings and provides combinatorial therapeutic pathways for metastatic cancer cell lines that have PTEN deletions and or mutations. The chapter also explores future directions for the work, highlights the contributions to the field of PTEN and metabolism, and recognizes PTEN as a metabolic regulator and gene that is vital in cancer tumorigenesis progression, applicable pan-cancer.
Item Open Access Nuclear PTEN Regulates Thymidylate Biosynthesis in Human Prostate Cancer Cell Lines(Metabolites) Loh, Zoe N; Wang, Mu-En; Wan, Changxin; Asara, John M; Ji, Zhicheng; Chen, MingThe phosphatase and tensin homologue deleted on chromosome 10 (PTEN) tumor suppressor governs a variety of biological processes, including metabolism, by acting on distinct molecular targets in different subcellular compartments. In the cytosol, inactive PTEN can be recruited to the plasma membrane where it dimerizes and functions as a lipid phosphatase to regulate metabolic processes mediated by the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin complex 1 (mTORC1) pathway. However, the metabolic regulation of PTEN in the nucleus remains undefined. Here, using a gain-of-function approach to targeting PTEN to the plasma membrane and nucleus, we show that nuclear PTEN contributes to pyrimidine metabolism, in particular de novo thymidylate (dTMP) biosynthesis. PTEN appears to regulate dTMP biosynthesis through interaction with methylenetetrahydrofolate dehydrogenase 1 (MTHFD1), a key enzyme that generates 5,10-methylenetetrahydrofolate, a cofactor required for thymidylate synthase (TYMS) to catalyze deoxyuridylate (dUMP) into dTMP. Our findings reveal a nuclear function for PTEN in controlling dTMP biosynthesis and may also have implications for targeting nuclear-excluded PTEN prostate cancer cells with antifolate drugs.Item Open Access Ornithine Decarboxylase Is Sufficient for Prostate Tumorigenesis via Androgen Receptor Signaling.(The American journal of pathology, 2016-12) Shukla-Dave, Amita; Castillo-Martin, Mireia; Chen, Ming; Lobo, Jose; Gladoun, Nataliya; Collazo-Lorduy, Ana; Khan, Faisal M; Ponomarev, Vladimir; Yi, Zhengzi; Zhang, Weijia; Pandolfi, Pier P; Hricak, Hedvig; Cordon-Cardo, CarlosIncreased polyamine synthesis is known to play an important role in prostate cancer. We aimed to explore its functional significance in prostate tumor initiation and its link to androgen receptor (AR) signaling. For this purpose, we generated a new cell line derived from normal epithelial prostate cells (RWPE-1) with overexpression of ornithine decarboxylase (ODC) and used it for in vitro and in vivo experiments. We then comprehensively analyzed the expression of the main metabolic enzymes of the polyamine pathway and spermine abundance in 120 well-characterized cases of human prostate cancer and high-grade prostate intraepithelial neoplasia (HGPIN). Herein, we show that the ODC-overexpressing prostate cells underwent malignant transformation, revealing that ODC is sufficient for de novo tumor initiation in 94% of injected mice. This oncogenic capacity was acquired through alteration of critical signaling networks, including AR, EIF2, and mTOR/MAPK. RNA silencing experiments revealed the link between AR signaling and polyamine metabolism. Human prostate cancers consistently demonstrated up-regulation of the main polyamine enzymes analyzed (ODC, polyamine oxidase, and spermine synthase) and reduction of spermine. This phenotype was also dominant in HGPIN, rendering it a new biomarker of malignant transformation. In summary, we report that ODC plays a key role in prostate tumorigenesis and that the polyamine pathway is altered as early as HGPIN.Item Open Access Pills of PTEN? In and out for tumor suppression.(Cell research, 2013-10) Papa, Antonella; Chen, Ming; Pandolfi, Pier PaoloThe tumor-suppressive activity of PTEN has always been attributed to its endogenous intracellular function. Recently two different groups have demonstrated that PTEN is secreted/exported into the extracellular environment for uptake by recipient cells, and functions as a tumor suppressor in a cell non-autonomous manner.Item Open Access Preclinical and Coclinical Studies in Prostate Cancer.(Cold Spring Harbor perspectives in medicine, 2018-04-02) Chen, Ming; Pandolfi, Pier PaoloMen who develop metastatic castration-resistant prostate cancer (mCRPC) will invariably succumb to their disease. Thus there remains a pressing need for preclinical testing of new drugs and drug combinations for late-stage prostate cancer (PCa). Insights from the mCRPC genomic landscape have revealed that, in addition to sustained androgen receptor (AR) signaling, there are other actionable molecular alterations and distinct molecular subclasses of PCa; however, the rate at which this knowledge translates into patient care via current preclinical testing is painfully slow and inefficient. Here, we will highlight the issues involved and discuss a new translational platform, "the co-clinical trial project," to expedite current preclinical studies and optimize clinical trial and experimental drug testing. With this platform, in vivo preclinical and early clinical studies are closely aligned, enabling in vivo testing of drugs using genetically engineered mouse models (GEMMs) in defined genetic contexts to personalize individual therapies. We will discuss the principles and essential components of this novel paradigm, representative success stories and future therapeutic options for mCRPC that should be explored.Item Open Access RB1-deficient prostate tumor growth and metastasis are vulnerable to ferroptosis induction via the E2F/ACSL4 axis.(The Journal of clinical investigation, 2023-03) Wang, Mu-En; Chen, Jiaqi; Lu, Yi; Bawcom, Alyssa R; Wu, Jinjin; Ou, Jianhong; Asara, John M; Armstrong, Andrew J; Wang, Qianben; Li, Lei; Wang, Yuzhuo; Huang, Jiaoti; Chen, MingInactivation of the RB1 tumor suppressor gene is common in several types of therapy-resistant cancers, including metastatic castration-resistant prostate cancer, and predicts poor clinical outcomes. Effective therapeutic strategies against RB1-deficient cancers, however, remain elusive. Here we showed that RB1-loss/E2F activation sensitized cancer cells to ferroptosis, a form of regulated cell death driven by iron-dependent lipid peroxidation, by upregulating expression of ACSL4 and enriching ACSL4-dependent arachidonic acid-containing phospholipids, which are key components of ferroptosis execution. ACSL4 appeared to be a direct E2F target gene and was critical to RB1 loss-induced sensitization to ferroptosis. Importantly, using cell line-derived xenografts and genetically engineered tumor models, we demonstrated that induction of ferroptosis in vivo by JKE-1674, a highly selective and stable GPX4 inhibitor, blocked RB1-deficient prostate tumor growth and metastasis and led to improved survival of the mice. Thus, our findings uncover an RB/E2F/ACSL4 molecular axis that governs ferroptosis, and also suggest a promising approach for the treatment of RB1-deficient malignancies.