Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38645034

ABSTRACT

Targeting cell surface molecules using radioligand and antibody-based therapies has yielded considerable success across cancers. However, it remains unclear how the expression of putative lineage markers, particularly cell surface molecules, varies in the process of lineage plasticity, wherein tumor cells alter their identity and acquire new oncogenic properties. A notable example of lineage plasticity is the transformation of prostate adenocarcinoma (PRAD) to neuroendocrine prostate cancer (NEPC)--a growing resistance mechanism that results in the loss of responsiveness to androgen blockade and portends dismal patient survival. To understand how lineage markers vary across the evolution of lineage plasticity in prostate cancer, we applied single cell analyses to 21 human prostate tumor biopsies and two genetically engineered mouse models, together with tissue microarray analysis (TMA) on 131 tumor samples. Not only did we observe a higher degree of phenotypic heterogeneity in castrate-resistant PRAD and NEPC than previously anticipated, but also found that the expression of molecules targeted therapeutically, namely PSMA, STEAP1, STEAP2, TROP2, CEACAM5, and DLL3, varied within a subset of gene-regulatory networks (GRNs). We also noted that NEPC and small cell lung cancer (SCLC) subtypes shared a set of GRNs, indicative of conserved biologic pathways that may be exploited therapeutically across tumor types. While this extreme level of transcriptional heterogeneity, particularly in cell surface marker expression, may mitigate the durability of clinical responses to novel antigen-directed therapies, its delineation may yield signatures for patient selection in clinical trials, potentially across distinct cancer types.

2.
bioRxiv ; 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38645223

ABSTRACT

Lineage plasticity is a recognized hallmark of cancer progression that can shape therapy outcomes. The underlying cellular and molecular mechanisms mediating lineage plasticity remain poorly understood. Here, we describe a versatile in vivo platform to identify and interrogate the molecular determinants of neuroendocrine lineage transformation at different stages of prostate cancer progression. Adenocarcinomas reliably develop following orthotopic transplantation of primary mouse prostate organoids acutely engineered with human-relevant driver alterations (e.g., Rb1-/-; Trp53-/-; cMyc+ or Pten-/-; Trp53-/-; cMyc+), but only those with Rb1 deletion progress to ASCL1+ neuroendocrine prostate cancer (NEPC), a highly aggressive, androgen receptor signaling inhibitor (ARSI)-resistant tumor. Importantly, we show this lineage transition requires a native in vivo microenvironment not replicated by conventional organoid culture. By integrating multiplexed immunofluorescence, spatial transcriptomics and PrismSpot to identify cell type-specific spatial gene modules, we reveal that ASCL1+ cells arise from KRT8+ luminal epithelial cells that progressively acquire transcriptional heterogeneity, producing large ASCL1+;KRT8- NEPC clusters. Ascl1 loss in established NEPC results in transient tumor regression followed by recurrence; however, Ascl1 deletion prior to transplantation completely abrogates lineage plasticity, yielding adenocarcinomas with elevated AR expression and marked sensitivity to castration. The dynamic feature of this model reveals the importance of timing of therapies focused on lineage plasticity and offers a platform for identification of additional lineage plasticity drivers.

3.
Article in English | MEDLINE | ID: mdl-37734867

ABSTRACT

In the last decade, organoid technology has become a cornerstone in cancer research. Organoids are long-term primary cell cultures, usually of epithelial origin, grown in a three-dimensional (3D) protein matrix and a fully defined medium. Organoids can be derived from many organs and cancer types and sites, encompassing both murine and human tissues. Importantly, they can be established from various stages during tumor evolution and recapitulate with high accuracy patient genomics and phenotypes in vitro, offering a platform for personalized medicine. Additionally, organoids are remarkably amendable for experimental manipulation. Taken together, these features make organoids a powerful tool with applications in basic cancer research and personalized medicine. Here, we will discuss the origins of organoid culture, applications in cancer research, and how cancer organoids can synergize with other models of cancer to drive basic discoveries as well as to translate these toward clinical solutions.

4.
Sci Transl Med ; 15(707): eadf7006, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37531417

ABSTRACT

In lung and prostate adenocarcinomas, neuroendocrine (NE) transformation to an aggressive derivative resembling small cell lung cancer (SCLC) is associated with poor prognosis. We previously described dependency of SCLC on the nuclear transporter exportin 1. Here, we explored the role of exportin 1 in NE transformation. We observed up-regulated exportin 1 in lung and prostate pretransformation adenocarcinomas. Exportin 1 was up-regulated after genetic inactivation of TP53 and RB1 in lung and prostate adenocarcinoma cell lines, accompanied by increased sensitivity to the exportin 1 inhibitor selinexor in vitro. Exportin 1 inhibition prevented NE transformation in different TP53/RB1-inactivated prostate adenocarcinoma xenograft models that acquire NE features upon treatment with the aromatase inhibitor enzalutamide and extended response to the EGFR inhibitor osimertinib in a lung cancer transformation patient-derived xenograft (PDX) model exhibiting combined adenocarcinoma/SCLC histology. Ectopic SOX2 expression restored the enzalutamide-promoted NE phenotype on adenocarcinoma-to-NE transformation xenograft models despite selinexor treatment. Selinexor sensitized NE-transformed lung and prostate small cell carcinoma PDXs to standard cytotoxics. Together, these data nominate exportin 1 inhibition as a potential therapeutic target to constrain lineage plasticity and prevent or treat NE transformation in lung and prostate adenocarcinoma.


Subject(s)
Adenocarcinoma , Lung Neoplasms , Prostatic Neoplasms , SOXB1 Transcription Factors , Small Cell Lung Carcinoma , Humans , Male , Adenocarcinoma/pathology , Down-Regulation , Lung Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Small Cell Lung Carcinoma/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Animals , Exportin 1 Protein
5.
Science ; 377(6611): 1180-1191, 2022 09 09.
Article in English | MEDLINE | ID: mdl-35981096

ABSTRACT

Drug resistance in cancer is often linked to changes in tumor cell state or lineage, but the molecular mechanisms driving this plasticity remain unclear. Using murine organoid and genetically engineered mouse models, we investigated the causes of lineage plasticity in prostate cancer and its relationship to antiandrogen resistance. We found that plasticity initiates in an epithelial population defined by mixed luminal-basal phenotype and that it depends on increased Janus kinase (JAK) and fibroblast growth factor receptor (FGFR) activity. Organoid cultures from patients with castration-resistant disease harboring mixed-lineage cells reproduce the dependency observed in mice by up-regulating luminal gene expression upon JAK and FGFR inhibitor treatment. Single-cell analysis confirms the presence of mixed-lineage cells with increased JAK/STAT (signal transducer and activator of transcription) and FGFR signaling in a subset of patients with metastatic disease, with implications for stratifying patients for clinical trials.


Subject(s)
Cell Plasticity , Drug Resistance, Neoplasm , ErbB Receptors , Janus Kinases , Prostatic Neoplasms , STAT Transcription Factors , Androgen Antagonists , Animals , Humans , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/genetics , Janus Kinases/metabolism , Male , Mice , Neoplasms, Experimental , Organoids , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
6.
Mol Cell ; 82(11): 2021-2031.e5, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35447082

ABSTRACT

The androgen receptor (AR) is a nuclear receptor that governs gene expression programs required for prostate development and male phenotype maintenance. Advanced prostate cancers display AR hyperactivation and transcriptome expansion, in part, through AR amplification and interaction with oncoprotein cofactors. Despite its biological importance, how AR domains and cofactors cooperate to bind DNA has remained elusive. Using single-particle cryo-electron microscopy, we isolated three conformations of AR bound to DNA, showing that AR forms a non-obligate dimer, with the buried dimer interface utilized by ancestral steroid receptors repurposed to facilitate cooperative DNA binding. We identify novel allosteric surfaces which are compromised in androgen insensitivity syndrome and reinforced by AR's oncoprotein cofactor, ERG, and by DNA-binding motifs. Finally, we present evidence that this plastic dimer interface may have been adopted for transactivation at the expense of DNA binding. Our work highlights how fine-tuning AR's cooperative interactions translate to consequences in development and disease.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Cryoelectron Microscopy , DNA/metabolism , Dimerization , Humans , Male , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcriptional Activation
7.
Cancer Gene Ther ; 29(6): 793-802, 2022 06.
Article in English | MEDLINE | ID: mdl-34135475

ABSTRACT

Treatment of castration-resistant prostate cancer remains a challenging clinical problem. Despite the promising effects of immunotherapy in other solid cancers, prostate cancer has remained largely unresponsive. Oncolytic viruses represent a promising therapeutic avenue, as oncolytic virus treatment combines tumour cell lysis with activation of the immune system and mounting of effective anti-tumour responses. Mammalian Orthoreoviruses are non-pathogenic human viruses with a preference of lytic replication in human tumour cells. In this study, we evaluated the oncolytic efficacy of the bioselected oncolytic reovirus mutant jin-3 in multiple human prostate cancer models. The jin-3 reovirus displayed efficient infection, replication, and anti-cancer responses in 2D and 3D prostate cancer models, as well as in ex vivo cultured human tumour slices. In addition, the jin-3 reovirus markedly reduced the viability and growth of human cancer cell lines and patient-derived xenografts. The infection induced the expression of mediators of immunogenic cell death, interferon-stimulated genes, and inflammatory cytokines. Taken together, our data demonstrate that the reovirus mutant jin-3 displays tumour tropism, and induces potent oncolytic and immunomodulatory responses in human prostate cancer models. Therefore, jin-3 reovirus represents an attractive candidate for further development as oncolytic agent for treatment of patients with aggressive localised or advanced prostate cancer.


Subject(s)
Mammalian orthoreovirus 3 , Oncolytic Virotherapy , Oncolytic Viruses , Prostatic Neoplasms , Reoviridae , Animals , Cell Line, Tumor , Humans , Male , Mammals , Oncolytic Viruses/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Reoviridae/genetics
8.
Cancer Cell ; 38(4): 534-550.e9, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32888433

ABSTRACT

Mutations in the pioneer transcription factor FOXA1 are a hallmark of estrogen receptor-positive (ER+) breast cancers. Examining FOXA1 in ∼5,000 breast cancer patients identifies several hotspot mutations in the Wing2 region and a breast cancer-specific mutation SY242CS, located in the third ß strand. Using a clinico-genomically curated cohort, together with breast cancer models, we find that FOXA1 mutations associate with a lower response to aromatase inhibitors. Mechanistically, Wing2 mutations display increased chromatin binding at ER loci upon estrogen stimulation, and an enhanced ER-mediated transcription without changes in chromatin accessibility. In contrast, SY242CS shows neomorphic properties that include the ability to open distinct chromatin regions and activate an alternative cistrome and transcriptome. Structural modeling predicts that SY242CS confers a conformational change that mediates stable binding to a non-canonical DNA motif. Taken together, our results provide insights into how FOXA1 mutations perturb its function to dictate cancer progression and therapeutic response.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Chromatin/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Mutation, Missense , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromatin/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/chemistry , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , MCF-7 Cells , Mice, Nude , Models, Molecular , Protein Domains , Xenograft Model Antitumor Assays/methods
10.
Cancer Cell ; 38(2): 279-296.e9, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32679108

ABSTRACT

Despite the development of second-generation antiandrogens, acquired resistance to hormone therapy remains a major challenge in treating advanced prostate cancer. We find that cancer-associated fibroblasts (CAFs) can promote antiandrogen resistance in mouse models and in prostate organoid cultures. We identify neuregulin 1 (NRG1) in CAF supernatant, which promotes resistance in tumor cells through activation of HER3. Pharmacological blockade of the NRG1/HER3 axis using clinical-grade blocking antibodies re-sensitizes tumors to hormone deprivation in vitro and in vivo. Furthermore, patients with castration-resistant prostate cancer with increased tumor NRG1 activity have an inferior response to second-generation antiandrogen therapy. This work reveals a paracrine mechanism of antiandrogen resistance in prostate cancer amenable to clinical testing using available targeted therapies.


Subject(s)
Androgen Antagonists/pharmacology , Drug Resistance, Neoplasm/genetics , Neuregulin-1/genetics , Prostatic Neoplasms/genetics , Tumor Microenvironment/genetics , Animals , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kaplan-Meier Estimate , Male , Mice, SCID , Neuregulin-1/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/prevention & control , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays/methods
11.
Science ; 368(6490): 497-505, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32355025

ABSTRACT

Androgen deprivation is the cornerstone of prostate cancer treatment. It results in involution of the normal gland to ~90% of its original size because of the loss of luminal cells. The prostate regenerates when androgen is restored, a process postulated to involve stem cells. Using single-cell RNA sequencing, we identified a rare luminal population in the mouse prostate that expresses stemlike genes (Sca1 + and Psca +) and a large population of differentiated cells (Nkx3.1 +, Pbsn +). In organoids and in mice, both populations contribute equally to prostate regeneration, partly through androgen-driven expression of growth factors (Nrg2, Rspo3) by mesenchymal cells acting in a paracrine fashion on luminal cells. Analysis of human prostate tissue revealed similar differentiated and stemlike luminal subpopulations that likewise acquire enhanced regenerative potential after androgen ablation. We propose that prostate regeneration is driven by nearly all persisting luminal cells, not just by rare stem cells.


Subject(s)
Androgens/metabolism , Prostate/physiology , Prostate/surgery , Prostatic Neoplasms/surgery , Regeneration , Androgen Antagonists/therapeutic use , Androgen-Binding Protein/genetics , Animals , Antigens, Neoplasm/genetics , Ataxin-1/genetics , Cell Differentiation/genetics , GPI-Linked Proteins/genetics , Gene Expression , Homeodomain Proteins/genetics , Humans , Male , Mesenchymal Stem Cells/physiology , Mice , Neoplasm Proteins/genetics , Nerve Growth Factors/genetics , Organ Size , Organoids/metabolism , Organoids/physiology , Prostate/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Regeneration/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Thrombospondins/genetics , Transcription Factors/genetics
12.
J Vis Exp ; (152)2019 10 24.
Article in English | MEDLINE | ID: mdl-31710046

ABSTRACT

Presented here is a protocol to study pharmacodynamics, stem cell potential, and cancer differentiation in prostate epithelial organoids. Prostate organoids are androgen responsive, three-dimensional (3D) cultures grown in a defined medium that resembles the prostatic epithelium. Prostate organoids can be established from wild-type and genetically engineered mouse models, benign human tissue, and advanced prostate cancer. Importantly, patient derived organoids closely resemble tumors in genetics and in vivo tumor biology. Moreover, organoids can be genetically manipulated using CRISPR/Cas9 and shRNA systems. These controlled genetics make the organoid culture attractive as a platform for rapidly testing the effects of genotypes and mutational profiles on pharmacological responses. However, experimental protocols must be specifically adapted to the 3D nature of organoid cultures to obtain reproducible results. Described here are detailed protocols for performing seeding assays to determine organoid formation capacity. Subsequently, this report shows how to perform drug treatments and analyze pharmacological response via viability measurements, protein isolation, and RNA isolation. Finally, the protocol describes how to prepare organoids for xenografting and subsequent in vivo growth assays using subcutaneous grafting. These protocols yield highly reproducible data and are widely applicable to 3D culture systems.


Subject(s)
Organoids/metabolism , Prostate/physiopathology , Animals , Cell Differentiation , Disease Models, Animal , Genotype , Humans , Male , Mice , Mutation
13.
Nat Med ; 25(10): 1607-1614, 2019 10.
Article in English | MEDLINE | ID: mdl-31591597

ABSTRACT

Rectal cancer (RC) is a challenging disease to treat that requires chemotherapy, radiation and surgery to optimize outcomes for individual patients. No accurate model of RC exists to answer fundamental research questions relevant to patients. We established a biorepository of 65 patient-derived RC organoid cultures (tumoroids) from patients with primary, metastatic or recurrent disease. RC tumoroids retained molecular features of the tumors from which they were derived, and their ex vivo responses to clinically relevant chemotherapy and radiation treatment correlated with the clinical responses noted in individual patients' tumors. Upon engraftment into murine rectal mucosa, human RC tumoroids gave rise to invasive RC followed by metastasis to lung and liver. Importantly, engrafted tumors displayed the heterogenous sensitivity to chemotherapy observed clinically. Thus, the biology and drug sensitivity of RC clinical isolates can be efficiently interrogated using an organoid-based, ex vivo platform coupled with in vivo endoluminal propagation in animals.


Subject(s)
Chemoradiotherapy , Organoids/pathology , Rectal Neoplasms/drug therapy , Rectal Neoplasms/radiotherapy , Animals , Fluorouracil/pharmacology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/radiotherapy , Liver Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Lung Neoplasms/secondary , Mice , Neoplasm Metastasis , Organoids/drug effects , Organoids/radiation effects , Rectal Neoplasms/pathology
14.
Nature ; 571(7765): 408-412, 2019 07.
Article in English | MEDLINE | ID: mdl-31243370

ABSTRACT

Mutations in the transcription factor FOXA1 define a unique subset of prostate cancers but the functional consequences of these mutations and whether they confer gain or loss of function is unknown1-9. Here, by annotating the landscape of FOXA1 mutations from 3,086 human prostate cancers, we define two hotspots in the forkhead domain: Wing2 (around 50% of all mutations) and the highly conserved DNA-contact residue R219 (around 5% of all mutations). Wing2 mutations are detected in adenocarcinomas at all stages, whereas R219 mutations are enriched in metastatic tumours with neuroendocrine histology. Interrogation of the biological properties of wild-type FOXA1 and fourteen FOXA1 mutants reveals gain of function in mouse prostate organoid proliferation assays. Twelve of these mutants, as well as wild-type FOXA1, promoted an exaggerated pro-luminal differentiation program, whereas two different R219 mutants blocked luminal differentiation and activated a mesenchymal and neuroendocrine transcriptional program. Assay for transposase-accessible chromatin using sequencing (ATAC-seq) of wild-type FOXA1 and representative Wing2 and R219 mutants revealed marked, mutant-specific changes in open chromatin at thousands of genomic loci and exposed sites of FOXA1 binding and associated increases in gene expression. Of note, ATAC-seq peaks in cells expressing R219 mutants lacked the canonical core FOXA1-binding motifs (GTAAAC/T) but were enriched for a related, non-canonical motif (GTAAAG/A), which was preferentially activated by R219-mutant FOXA1 in reporter assays. Thus, FOXA1 mutations alter its pioneering function and perturb normal luminal epithelial differentiation programs, providing further support for the role of lineage plasticity in cancer progression.


Subject(s)
Cell Differentiation/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Mutation , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Lineage , Chromatin/genetics , Chromatin/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/chemistry , Humans , Male , Mice , Mice, Inbred NOD , Nucleotide Motifs , Organoids/cytology , Organoids/metabolism
16.
Nat Commun ; 9(1): 2404, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29921838

ABSTRACT

A major hurdle in the study of rare tumors is a lack of existing preclinical models. Neuroendocrine prostate cancer is an uncommon and aggressive histologic variant of prostate cancer that may arise de novo or as a mechanism of treatment resistance in patients with pre-existing castration-resistant prostate cancer. There are few available models to study neuroendocrine prostate cancer. Here, we report the generation and characterization of tumor organoids derived from needle biopsies of metastatic lesions from four patients. We demonstrate genomic, transcriptomic, and epigenomic concordance between organoids and their corresponding patient tumors. We utilize these organoids to understand the biologic role of the epigenetic modifier EZH2 in driving molecular programs associated with neuroendocrine prostate cancer progression. High-throughput organoid drug screening nominated single agents and drug combinations suggesting repurposing opportunities. This proof of principle study represents a strategy for the study of rare cancer phenotypes.


Subject(s)
Neuroendocrine Tumors/genetics , Organoids/metabolism , Prostate/metabolism , Prostatic Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Epigenomics/methods , Gene Expression Profiling/methods , Genomics/methods , Humans , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Organoids/pathology , Phenotype , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
17.
Elife ; 62017 09 11.
Article in English | MEDLINE | ID: mdl-28891793

ABSTRACT

In prostate cancer, resistance to the antiandrogen enzalutamide (Enz) can occur through bypass of androgen receptor (AR) blockade by the glucocorticoid receptor (GR). In contrast to fixed genomic alterations, here we show that GR-mediated antiandrogen resistance is adaptive and reversible due to regulation of GR expression by a tissue-specific enhancer. GR expression is silenced in prostate cancer by a combination of AR binding and EZH2-mediated repression at the GR locus, but is restored in advanced prostate cancers upon reversion of both repressive signals. Remarkably, BET bromodomain inhibition resensitizes drug-resistant tumors to Enz by selectively impairing the GR signaling axis via this enhancer. In addition to revealing an underlying molecular mechanism of GR-driven drug resistance, these data suggest that inhibitors of broadly active chromatin-readers could have utility in nuanced clinical contexts of acquired drug resistance with a more favorable therapeutic index.


Subject(s)
Androgen Antagonists/pharmacology , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Animals , Azepines , Benzamides , Cell Line, Tumor , Chromatin Immunoprecipitation/methods , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Resistance, Neoplasm/drug effects , Heterografts , Humans , Male , Metabolism, Inborn Errors/metabolism , Mice , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/deficiency , Sequence Analysis , Signal Transduction/drug effects , Triazoles
18.
Nature ; 546(7660): 671-675, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28614298

ABSTRACT

Half of all prostate cancers are caused by the TMPRSS2-ERG gene-fusion, which enables androgens to drive expression of the normally silent E26 transformation-specific (ETS) transcription factor ERG in prostate cells. Recent genomic landscape studies of such cancers have reported recurrent point mutations and focal deletions of another ETS member, the ETS2 repressor factor ERF. Here we show these ERF mutations cause decreased protein stability and mostly occur in tumours without ERG upregulation. ERF loss recapitulates the morphological and phenotypic features of ERG gain in normal mouse prostate cells, including expansion of the androgen receptor transcriptional repertoire, and ERF has tumour suppressor activity in the same genetic background of Pten loss that yields oncogenic activity by ERG. In the more common scenario of ERG upregulation, chromatin immunoprecipitation followed by sequencing indicates that ERG inhibits the ability of ERF to bind DNA at consensus ETS sites both in normal and in cancerous prostate cells. Consistent with a competition model, ERF overexpression blocks ERG-dependent tumour growth, and ERF loss rescues TMPRSS2-ERG-positive prostate cancer cells from ERG dependency. Collectively, these data provide evidence that the oncogenicity of ERG is mediated, in part, by competition with ERF and they raise the larger question of whether other gain-of-function oncogenic transcription factors might also inactivate endogenous tumour suppressors.


Subject(s)
Carcinogenesis/genetics , Mutation , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/genetics , Androgens/metabolism , Animals , Cell Line, Tumor , Genes/genetics , Humans , Male , Mice , Prostate/metabolism , Protein Stability , Receptors, Androgen/metabolism , Repressor Proteins/deficiency , Repressor Proteins/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/metabolism , Signal Transduction , Transcriptional Regulator ERG/deficiency , Transcriptional Regulator ERG/metabolism , Transcriptome/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
19.
Science ; 355(6320): 84-88, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28059768

ABSTRACT

Some cancers evade targeted therapies through a mechanism known as lineage plasticity, whereby tumor cells acquire phenotypic characteristics of a cell lineage whose survival no longer depends on the drug target. We use in vitro and in vivo human prostate cancer models to show that these tumors can develop resistance to the antiandrogen drug enzalutamide by a phenotypic shift from androgen receptor (AR)-dependent luminal epithelial cells to AR-independent basal-like cells. This lineage plasticity is enabled by the loss of TP53 and RB1 function, is mediated by increased expression of the reprogramming transcription factor SOX2, and can be reversed by restoring TP53 and RB1 function or by inhibiting SOX2 expression. Thus, mutations in tumor suppressor genes can create a state of increased cellular plasticity that, when challenged with antiandrogen therapy, promotes resistance through lineage switching.


Subject(s)
Androgen Antagonists/therapeutic use , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Retinoblastoma Binding Proteins/genetics , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Benzamides , Cell Line, Tumor , Cell Lineage , Cell Plasticity , Humans , Male , Nitriles , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/genetics , SOXB1 Transcription Factors/genetics
20.
Proc Natl Acad Sci U S A ; 113(37): E5399-407, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27573849

ABSTRACT

Leucine-rich repeat-containing G-protein coupled receptor 5-positive (Lgr5(+)) stem cells reside at crypt bottoms of the small and large intestine. Small intestinal Paneth cells supply Wnt3, EGF, and Notch signals to neighboring Lgr5(+) stem cells. Whereas the colon lacks Paneth cells, deep crypt secretory (DCS) cells are intermingled with Lgr5(+) stem cells at crypt bottoms. Here, we report regenerating islet-derived family member 4 (Reg4) as a marker of DCS cells. To investigate a niche function, we eliminated DCS cells by using the diphtheria-toxin receptor gene knocked into the murine Reg4 locus. Ablation of DCS cells results in loss of stem cells from colonic crypts and disrupts gut homeostasis and colon organoid growth. In agreement, sorted Reg4(+) DCS cells promote organoid formation of single Lgr5(+) colon stem cells. DCS cells can be massively produced from Lgr5(+) colon stem cells in vitro by combined Notch inhibition and Wnt activation. We conclude that Reg4(+) DCS cells serve as Paneth cell equivalents in the colon crypt niche.


Subject(s)
Colonic Neoplasms/metabolism , Neoplasm Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/metabolism , Animals , Colon/cytology , Colon/growth & development , Colon/metabolism , Colonic Neoplasms/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Intestine, Small/cytology , Intestine, Small/metabolism , Mice , Neoplasm Proteins/metabolism , Organoids/growth & development , Organoids/metabolism , Pancreatitis-Associated Proteins , Paneth Cells/cytology , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/genetics , Stem Cell Niche/genetics , Stem Cells/cytology , Wnt Signaling Pathway/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...