Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 204
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 179(1): 8-12, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31519311
2.
Mol Cell ; 83(14): 2398-2416.e12, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37402365

ABSTRACT

Nuclear receptor-binding SET-domain protein 1 (NSD1), a methyltransferase that catalyzes H3K36me2, is essential for mammalian development and is frequently dysregulated in diseases, including Sotos syndrome. Despite the impacts of H3K36me2 on H3K27me3 and DNA methylation, the direct role of NSD1 in transcriptional regulation remains largely unknown. Here, we show that NSD1 and H3K36me2 are enriched at cis-regulatory elements, particularly enhancers. NSD1 enhancer association is conferred by a tandem quadruple PHD (qPHD)-PWWP module, which recognizes p300-catalyzed H3K18ac. By combining acute NSD1 depletion with time-resolved epigenomic and nascent transcriptomic analyses, we demonstrate that NSD1 promotes enhancer-dependent gene transcription by facilitating RNA polymerase II (RNA Pol II) pause release. Notably, NSD1 can act as a transcriptional coactivator independent of its catalytic activity. Moreover, NSD1 enables the activation of developmental transcriptional programs associated with Sotos syndrome pathophysiology and controls embryonic stem cell (ESC) multilineage differentiation. Collectively, we have identified NSD1 as an enhancer-acting transcriptional coactivator that contributes to cell fate transition and Sotos syndrome development.


Subject(s)
Nuclear Proteins , Sotos Syndrome , Animals , Humans , Nuclear Proteins/metabolism , Chromatin , Sotos Syndrome/genetics , Sotos Syndrome/metabolism , Histone Methyltransferases/genetics , Transcription Factors/genetics , Cell Differentiation/genetics , Mammals/metabolism , Histone-Lysine N-Methyltransferase/genetics
3.
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
4.
Cell ; 159(1): 163-175, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25201529

ABSTRACT

The prostate gland consists of basal and luminal cells arranged as pseudostratified epithelium. In tissue recombination models, only basal cells reconstitute a complete prostate gland, yet murine lineage-tracing experiments show that luminal cells generate basal cells. It has remained challenging to address the molecular details of these transitions and whether they apply to humans, due to the lack of culture conditions that recapitulate prostate gland architecture. Here, we describe a 3D culture system that supports long-term expansion of primary mouse and human prostate organoids, composed of fully differentiated CK5+ basal and CK8+ luminal cells. Organoids are genetically stable, reconstitute prostate glands in recombination assays, and can be experimentally manipulated. Single human luminal and basal cells give rise to organoids, yet luminal-cell-derived organoids more closely resemble prostate glands. These data support a luminal multilineage progenitor cell model for prostate tissue and establish a robust, scalable system for mechanistic studies.


Subject(s)
Organ Culture Techniques , Organoids , Prostate/cytology , Androgens/metabolism , Humans , Male , Stem Cells/cytology , Stem Cells/metabolism
5.
Cell ; 159(1): 176-187, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25201530

ABSTRACT

The lack of in vitro prostate cancer models that recapitulate the diversity of human prostate cancer has hampered progress in understanding disease pathogenesis and therapy response. Using a 3D organoid system, we report success in long-term culture of prostate cancer from biopsy specimens and circulating tumor cells. The first seven fully characterized organoid lines recapitulate the molecular diversity of prostate cancer subtypes, including TMPRSS2-ERG fusion, SPOP mutation, SPINK1 overexpression, and CHD1 loss. Whole-exome sequencing shows a low mutational burden, consistent with genomics studies, but with mutations in FOXA1 and PIK3R1, as well as in DNA repair and chromatin modifier pathways that have been reported in advanced disease. Loss of p53 and RB tumor suppressor pathway function are the most common feature shared across the organoid lines. The methodology described here should enable the generation of a large repertoire of patient-derived prostate cancer lines amenable to genetic and pharmacologic studies.


Subject(s)
Culture Techniques , Organoids , Prostatic Neoplasms/pathology , Heterografts , Humans , Male , Neoplasm Metastasis/pathology , Organoids/pathology , Pharmacology/methods , Tumor Suppressor Proteins/metabolism
6.
Cell ; 155(6): 1309-22, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24315100

ABSTRACT

The treatment of advanced prostate cancer has been transformed by novel antiandrogen therapies such as enzalutamide. Here, we identify induction of glucocorticoid receptor (GR) expression as a common feature of drug-resistant tumors in a credentialed preclinical model, a finding also confirmed in patient samples. GR substituted for the androgen receptor (AR) to activate a similar but distinguishable set of target genes and was necessary for maintenance of the resistant phenotype. The GR agonist dexamethasone was sufficient to confer enzalutamide resistance, whereas a GR antagonist restored sensitivity. Acute AR inhibition resulted in GR upregulation in a subset of prostate cancer cells due to relief of AR-mediated feedback repression of GR expression. These findings establish a mechanism of escape from AR blockade through expansion of cells primed to drive AR target genes via an alternative nuclear receptor upon drug exposure.


Subject(s)
Androgen Antagonists/therapeutic use , Androgen Receptor Antagonists/therapeutic use , Drug Resistance, Neoplasm , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Receptors, Glucocorticoid/metabolism , Animals , Benzamides , Disease Models, Animal , Gene Expression Regulation , Heterografts , Humans , Male , Mice , Neoplasm Transplantation , Nitriles , Phenylthiohydantoin/therapeutic use , Receptors, Androgen/metabolism , Transcriptome
7.
Cell ; 148(6): 1089-98, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22424221

ABSTRACT

During the past decade, cancer drug development has shifted from a focus on cytotoxic chemotherapies to drugs that target specific molecular alterations in tumors. Although these drugs dramatically shrink tumors, the responses are temporary. Research is now focused on overcoming drug resistance, a frequent cause of treatment failure. Here we reflect on analogous challenges faced by researchers in infectious diseases. We compare and contrast the resistance mechanisms arising in cancer and infectious diseases and discuss how approaches for overcoming viral and bacterial infections, such as HIV and tuberculosis, are instructive for developing a more rational approach for cancer therapy. In particular, maximizing the effect of the initial treatment response, which often requires synergistic combination therapy, is foremost among these approaches. A remaining challenge in both fields is identifying drugs that eliminate drug-tolerant "persister" cells (infectious disease) or tumor-initiating/stem cells (cancer) to prevent late relapse and shorten treatment duration.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Combined Modality Therapy , Drug Resistance, Microbial , HIV Infections/drug therapy , Humans , Treatment Failure , Tuberculosis/drug therapy
8.
Proc Natl Acad Sci U S A ; 121(28): e2322203121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968122

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 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 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 current and future antigen-directed therapies, its delineation may yield signatures for patient selection in clinical trials, potentially across distinct cancer types.


Subject(s)
Single-Cell Analysis , Male , Humans , Single-Cell Analysis/methods , Animals , Mice , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Antigens, Surface/metabolism , Antigens, Surface/genetics , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/drug therapy , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/drug therapy , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy
10.
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
11.
Cell ; 137(5): 796-8, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19490885

ABSTRACT

Kinase inhibitors have ushered in the era of targeted therapy, but their utility to date is primarily limited to cancers bearing oncogenic kinase mutations. Two papers in this issue (Luo et al., 2009; Scholl et al., 2009) could change this landscape by uncovering kinase-specific vulnerabilities in tumors with RAS mutations.


Subject(s)
Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , ras Proteins/metabolism , Cell Line, Tumor , Humans , Mutation , ras Proteins/genetics
12.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Article in English | MEDLINE | ID: mdl-34353917

ABSTRACT

The increasing complexity of different cell types revealed by single-cell analysis of tissues presents challenges in efficiently elucidating their functions. Here we show, using prostate as a model tissue, that primary organoids and freshly isolated epithelial cells can be CRISPR edited ex vivo using Cas9-sgRNA (guide RNA) ribotnucleoprotein complex technology, then orthotopically transferred in vivo into immunocompetent or immunodeficient mice to generate cancer models with phenotypes resembling those seen in traditional genetically engineered mouse models. Large intrachromosomal (∼2 Mb) or multigenic deletions can be engineered efficiently without the need for selection, including in isolated subpopulations to address cell-of-origin questions.


Subject(s)
Chromosome Deletion , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Prostate/cytology , Adaptor Proteins, Signal Transducing/metabolism , Animals , CRISPR-Associated Protein 9/genetics , Epithelial Cells , Genes, Tumor Suppressor , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Organoids , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Guide, Kinetoplastida , Ribonucleoproteins/genetics , Transcriptional Regulator ERG/genetics , Xenograft Model Antitumor Assays
14.
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
15.
Proc Natl Acad Sci U S A ; 117(15): 8584-8592, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32220959

ABSTRACT

The androgen receptor (AR) is a type I nuclear hormone receptor and the primary drug target in prostate cancer due to its role as a lineage survival factor in prostate luminal epithelium. In prostate cancer, the AR cistrome is reprogrammed relative to normal prostate epithelium and particularly in cancers driven by oncogenic ETS fusion genes. The molecular basis for this change has remained elusive. Using purified proteins, we report a minimal cell-free system that demonstrates interdomain cooperativity between the ligand (LBD) and DNA binding domains (DBD) of AR, and its autoinhibition by the N terminus of AR. Furthermore, we identify ERG as a cofactor that activates AR's ability to bind DNA in both high and lower affinity contexts through direct interaction within a newly identified AR-interacting motif (AIM) in the ETS domain, independent of ERG's own DNA binding ability. Finally, we present evidence that this interaction is conserved among ETS factors whose expression is altered in prostate cancer. Our work highlights, at a biochemical level, how tumor-initiating ETS translocations result in reprogramming of the AR cistrome.


Subject(s)
DNA/metabolism , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Receptors, Androgen/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA/genetics , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/genetics , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Transcriptional Regulator ERG/chemistry , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism , Tumor Cells, Cultured
16.
Proc Natl Acad Sci U S A ; 116(23): 11428-11436, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31061129

ABSTRACT

Heterogeneity in the genomic landscape of metastatic prostate cancer has become apparent through several comprehensive profiling efforts, but little is known about the impact of this heterogeneity on clinical outcome. Here, we report comprehensive genomic and transcriptomic analysis of 429 patients with metastatic castration-resistant prostate cancer (mCRPC) linked with longitudinal clinical outcomes, integrating findings from whole-exome, transcriptome, and histologic analysis. For 128 patients treated with a first-line next-generation androgen receptor signaling inhibitor (ARSI; abiraterone or enzalutamide), we examined the association of 18 recurrent DNA- and RNA-based genomic alterations, including androgen receptor (AR) variant expression, AR transcriptional output, and neuroendocrine expression signatures, with clinical outcomes. Of these, only RB1 alteration was significantly associated with poor survival, whereas alterations in RB1, AR, and TP53 were associated with shorter time on treatment with an ARSI. This large analysis integrating mCRPC genomics with histology and clinical outcomes identifies RB1 genomic alteration as a potent predictor of poor outcome, and is a community resource for further interrogation of clinical and molecular associations.


Subject(s)
Prostatic Neoplasms, Castration-Resistant/genetics , Aged , Androstenes/therapeutic use , Benzamides , Biomarkers, Tumor/genetics , Drug Resistance, Neoplasm/genetics , Genomics/methods , Humans , Male , Middle Aged , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/genetics , Treatment Outcome
17.
Genes Dev ; 28(16): 1800-14, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25128497

ABSTRACT

One-year survival rates for newly diagnosed hepatocellular carcinoma (HCC) are <50%, and unresectable HCC carries a dismal prognosis owing to its aggressiveness and the undruggable nature of its main genetic drivers. By screening a custom library of shRNAs directed toward known drug targets in a genetically defined Myc-driven HCC model, we identified cyclin-dependent kinase 9 (Cdk9) as required for disease maintenance. Pharmacological or shRNA-mediated CDK9 inhibition led to robust anti-tumor effects that correlated with MYC expression levels and depended on the role that both CDK9 and MYC exert in transcription elongation. Our results establish CDK9 inhibition as a therapeutic strategy for MYC-overexpressing liver tumors and highlight the relevance of transcription elongation in the addiction of cancer cells to MYC.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cyclin-Dependent Kinase 9/metabolism , Liver Neoplasms/enzymology , Proto-Oncogene Proteins c-myc/metabolism , Transcription Elongation, Genetic/physiology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression , Gene Library , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Small Interfering/metabolism
18.
Nature ; 471(7339): 523-6, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21430781

ABSTRACT

Human lung adenocarcinomas with activating mutations in EGFR (epidermal growth factor receptor) often respond to treatment with EGFR tyrosine kinase inhibitors (TKIs), but the magnitude of tumour regression is variable and transient. This heterogeneity in treatment response could result from genetic modifiers that regulate the degree to which tumour cells are dependent on mutant EGFR. Through a pooled RNA interference screen, we show that knockdown of FAS and several components of the NF-κB pathway specifically enhanced cell death induced by the EGFR TKI erlotinib in EGFR-mutant lung cancer cells. Activation of NF-κB through overexpression of c-FLIP or IKK (also known as CFLAR and IKBKB, respectively), or silencing of IκB (also known as NFKBIA), rescued EGFR-mutant lung cancer cells from EGFR TKI treatment. Genetic or pharmacologic inhibition of NF-κB enhanced erlotinib-induced apoptosis in erlotinib-sensitive and erlotinib-resistant EGFR-mutant lung cancer models. Increased expression of the NF-κB inhibitor IκB predicted for improved response and survival in EGFR-mutant lung cancer patients treated with EGFR TKI. These data identify NF-κB as a potential companion drug target, together with EGFR, in EGFR-mutant lung cancers and provide insight into the mechanisms by which tumour cells escape from oncogene dependence.


Subject(s)
ErbB Receptors/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutant Proteins/metabolism , NF-kappa B/metabolism , Signal Transduction , fas Receptor/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride , Genes, erbB-1/genetics , Humans , I-kappa B Proteins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Models, Biological , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutation/genetics , NF-kappa B/antagonists & inhibitors , Quinazolines/pharmacology , Quinazolines/therapeutic use , RNA Interference , Signal Transduction/drug effects , fas Receptor/antagonists & inhibitors
19.
Proc Natl Acad Sci U S A ; 111(41): 14876-81, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25271328

ABSTRACT

MAGUK Inverted 2 (MAGI-2) is a PTEN-interacting scaffold protein implicated in cancer on the basis of rare, recurrent genomic translocations and deletions in various tumors. In the renal glomerulus, MAGI-2 is exclusively expressed in podocytes, specialized cells forming part of the glomerular filter, where it interacts with the slit diaphragm protein nephrin. To further explore MAGI-2 function, we generated Magi-2-KO mice through homologous recombination by targeting an exon common to all three alternative splice variants. Magi-2 null mice presented with progressive proteinuria as early as 2 wk postnatally, which coincided with loss of nephrin expression in the glomeruli. Magi-2-null kidneys revealed diffuse podocyte foot process effacement and focal podocyte hypertrophy by 3 wk of age, as well as progressive podocyte loss. By 5.5 wk, coinciding with a near-complete loss of podocytes, Magi-2-null mice developed diffuse glomerular extracapillary epithelial cell proliferations, and died of renal failure by 3 mo of age. As confirmed by immunohistochemical analysis, the proliferative cell populations in glomerular lesions were exclusively composed of activated parietal epithelial cells (PECs). Our results reveal that MAGI-2 is required for the integrity of the kidney filter and podocyte survival. Moreover, we demonstrate that PECs can be activated to form glomerular lesions resembling a noninflammatory glomerulopathy with extensive extracapillary proliferation, sometimes resembling crescents, following rapid and severe podocyte loss.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Guanylate Kinases/metabolism , Kidney/pathology , Animals , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Immunohistochemistry , Inflammation/pathology , Kidney/metabolism , Kidney Glomerulus/blood supply , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Membrane Proteins/metabolism , Mice, Knockout , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Podocytes/metabolism , Podocytes/pathology , Proteinuria/metabolism , Proteinuria/pathology , Survival Analysis , Up-Regulation
20.
Proc Natl Acad Sci U S A ; 111(30): 11139-44, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024180

ABSTRACT

Primary prostate cancer is the most common malignancy in men but has highly variable outcomes, highlighting the need for biomarkers to determine which patients can be managed conservatively. Few large prostate oncogenome resources currently exist that combine the molecular and clinical outcome data necessary to discover prognostic biomarkers. Previously, we found an association between relapse and the pattern of DNA copy number alteration (CNA) in 168 primary tumors, raising the possibility of CNA as a prognostic biomarker. Here we examine this question by profiling an additional 104 primary prostate cancers and updating the initial 168 patient cohort with long-term clinical outcome. We find that CNA burden across the genome, defined as the percentage of the tumor genome affected by CNA, was associated with biochemical recurrence and metastasis after surgery in these two cohorts, independent of the prostate-specific antigen biomarker or Gleason grade, a major existing histopathological prognostic variable in prostate cancer. Moreover, CNA burden was associated with biochemical recurrence in intermediate-risk Gleason 7 prostate cancers, independent of prostate-specific antigen or nomogram score. We further demonstrate that CNA burden can be measured in diagnostic needle biopsies using low-input whole-genome sequencing, setting the stage for studies of prognostic impact in conservatively treated cohorts.


Subject(s)
Biomarkers, Tumor/genetics , DNA Copy Number Variations , DNA, Neoplasm/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Biopsy, Needle , Disease-Free Survival , Follow-Up Studies , Humans , Male , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL