Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Sci Adv ; 9(30): eadf6210, 2023 07 28.
Article En | MEDLINE | ID: mdl-37494452

KRAS/ERK pathway phosphorylates DICER1, causing its nuclear translocation, and phosphomimetic Dicer1 contributes to tumorigenesis in mice. Mechanisms through which phospho-DICER1 regulates tumor progression remain undefined. While DICER1 canonically regulates microRNAs (miRNA) and epithelial-to-mesenchymal transition (EMT), we found that phosphorylated nuclear DICER1 (phospho-nuclear DICER1) promotes late-stage tumor progression in mice with oncogenic Kras, independent of miRNAs and EMT. Instead, we observe that the murine AT2 tumor cells exhibit altered chromatin compaction, and cells from disorganized advanced tumors, but not localized tumors, express gastric genes. Collectively, this results in subpopulations of tumor cells transitioning from a restricted alveolar to a broader endodermal lineage state. In human LUADs, we observed expression of phospho-nuclear DICER1 in advanced tumors together with the expression of gastric genes. We define a multimeric chromatin-DICER1 complex composed of the Mediator complex subunit 12, CBX1, MACROH2A.1, and transcriptional regulators supporting the model that phospho-nuclear DICER1 leads to lineage reprogramming of AT2 tumor cells to mediate lung cancer progression.


Adenocarcinoma of Lung , Lung Neoplasms , MicroRNAs , Humans , Mice , Animals , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Chromatin/genetics , MicroRNAs/genetics , Adenocarcinoma of Lung/genetics , Lung Neoplasms/genetics , Ribonuclease III/genetics , Ribonuclease III/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism
2.
Mol Cancer Res ; 21(8): 779-794, 2023 08 01.
Article En | MEDLINE | ID: mdl-37255406

Epithelial-to-mesenchymal transition results in loss of specialized epithelial cell contacts and acquisition of mesenchymal invasive capacity. The transcription repressor zinc finger E-box-binding homeobox 1 (ZEB1) binds to E-boxes of gene promoter regions to suppress the expression of epithelial genes. ZEB1 has inconsistent molecular weights, which have been attributed to posttranslational modifications (PTM). We performed mass spectrometry and identified K811 acetylation as a novel PTM in ZEB1. To define the role of ZEB1 acetylation in regulating function, we generated ZEB1 acetyl-mimetic (K811Q) and acetyl-deficient (K811R) mutant-expressing non-small cell lung cancer cell lines (NSCLC). We demonstrate that the K811R ZEB1 (125 kDa) has a shorter protein half-life than wild-type (WT) ZEB1 and K811Q ZEB1 (∼225 kDa), suggesting that lack of ZEB1 acetylation in the lower molecular weight form affects protein stability. Further, the acetylated form of ZEB1 recruits the nucleosome remodeling and deacetylase (NuRD) complex to bind the promoter of its target genes mir200c-141 and SEMA3F. RNA-sequencing revealed that WT ZEB1 and K811Q ZEB1 downregulate the expression of epithelial genes to promote lung adenocarcinoma invasion and metastasis, whereas the K811R ZEB1 does not. Our findings establish that the K811 acetylation promotes ZEB1 protein stability, interaction with other protein complexes, and subsequent invasion/metastasis of lung adenocarcinoma via epithelial-to-mesenchymal transition. IMPLICATIONS: The molecular mechanisms by which ZEB1 is regulated by K811 acetylation to promote protein stability, NuRD complex and promoter interactions, and function are relevant to the development of treatment strategies to prevent and treat metastasis in patients with NSCLC.


Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Acetylation , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Protein Processing, Post-Translational , Adenocarcinoma of Lung/genetics , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics
3.
Mol Cancer Res ; 2023 May 12.
Article En | MEDLINE | ID: mdl-37171981

Epithelial-to-mesenchymal transition results in loss of specialized epithelial cell contacts and acquisition of mesenchymal invasive capacity. The transcription repressor zinc finger E-box-binding homeobox 1 (ZEB1) binds to E-boxes of gene promoter regions to suppress the expression of epithelial genes. ZEB1 has inconsistent molecular weights, which have been attributed to post-translational modifications (PTMs). We performed mass spectrometry and identified K811 acetylation as a novel PTM in ZEB1. To define the role of ZEB1 acetylation in regulating function, we generated ZEB1 acetyl-mimetic (K811Q) and acetyl-deficient (K811R) mutant-expressing non-small cell lung cancer cell lines (NSCLC). We demonstrate that the K811R ZEB1 (125 kDa) has a shorter protein half-life than wild-type (WT) ZEB1 and K811Q ZEB1 (&tilde225 kDa), suggesting that lack of ZEB1 acetylation in the lower molecular weight form affects protein stability. Further, the acetylated form of ZEB1 recruits the nucleosome remodeling and deacetylase (NuRD) complex to bind the promoter of its target genes mir200c-141 and SEMA3F. RNA-sequencing revealed that WT ZEB1 and K811Q ZEB1 downregulate the expression of epithelial genes to promote lung adenocarcinoma invasion and metastasis, while the K811R ZEB1 does not. Our findings establish that the K811 acetylation promotes ZEB1 protein stability, interaction with other protein complexes, and subsequent invasion/metastasis of lung adenocarcinoma via epithelial-to-mesenchymal transition. Implications: The molecular mechanisms by which ZEB1 is regulated by K811 acetylation to promote protein stability, NuRD complex and promoter interactions, and function are relevant to the development of treatment strategies to prevent and treat metastasis in NSCLC patients.

4.
Cell Rep ; 40(13): 111429, 2022 09 27.
Article En | MEDLINE | ID: mdl-36170810

Lung cancer is a highly aggressive and metastatic disease responsible for approximately 25% of all cancer-related deaths in the United States. Using high-throughput in vitro and in vivo screens, we have previously established Impad1 as a driver of lung cancer invasion and metastasis. Here we elucidate that Impad1 is a direct target of the epithelial microRNAs (miRNAs) miR-200 and miR∼96 and is de-repressed during epithelial-to-mesenchymal transition (EMT); thus, we establish a mode of regulation of the protein. Impad1 modulates Golgi apparatus morphology and vesicular trafficking through its interaction with a trafficking protein, Syt11. These changes in Golgi apparatus dynamics alter the extracellular matrix and the tumor microenvironment (TME) to promote invasion and metastasis. Inhibiting Impad1 or Syt11 disrupts the cancer cell secretome, regulates the TME, and reverses the invasive or metastatic phenotype. This work identifies Impad1 as a regulator of EMT and secretome-mediated changes during lung cancer progression.


Lung Neoplasms , MicroRNAs , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Synaptotagmins/metabolism , Tumor Microenvironment
5.
Genes Dev ; 36(9-10): 582-600, 2022 05 01.
Article En | MEDLINE | ID: mdl-35654454

One of the mechanisms by which cancer cells acquire hyperinvasive and migratory properties with progressive loss of epithelial markers is the epithelial-to-mesenchymal transition (EMT). We have previously reported that in different cancer types, including nonsmall cell lung cancer (NSCLC), the microRNA-183/96/182 cluster (m96cl) is highly repressed in cells that have undergone EMT. In the present study, we used a novel conditional m96cl mouse to establish that loss of m96cl accelerated the growth of Kras mutant autochthonous lung adenocarcinomas. In contrast, ectopic expression of the m96cl in NSCLC cells results in a robust suppression of migration and invasion in vitro, and tumor growth and metastasis in vivo. Detailed immune profiling of the tumors revealed a significant enrichment of activated CD8+ cytotoxic T lymphocytes (CD8+ CTLs) in m96cl-expressing tumors, and m96cl-mediated suppression of tumor growth and metastasis was CD8+ CTL-dependent. Using coculture assays with naïve immune cells, we show that m96cl expression drives paracrine stimulation of CD8+ CTL proliferation and function. Using tumor microenvironment-associated gene expression profiling, we identified that m96cl elevates the interleukin-2 (IL2) signaling pathway and results in increased IL2-mediated paracrine stimulation of CD8+ CTLs. Furthermore, we identified that the m96cl modulates the expression of IL2 in cancer cells by regulating the expression of transcriptional repressors Foxf2 and Zeb1, and thereby alters the levels of secreted IL2 in the tumor microenvironment. Last, we show that in vivo depletion of IL2 abrogates m96cl-mediated activation of CD8+ CTLs and results in loss of metastatic suppression. Therefore, we have identified a novel mechanistic role of the m96cl in the suppression of lung cancer growth and metastasis by inducing an IL2-mediated systemic CD8+ CTL immune response.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , Animals , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Interleukin-2/genetics , Interleukin-2/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , T-Lymphocytes, Cytotoxic , Tumor Microenvironment
6.
J Thorac Oncol ; 16(11): 1821-1839, 2021 11.
Article En | MEDLINE | ID: mdl-34274504

INTRODUCTION: Coronavirus disease 2019 is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which enters host cells through the cell surface proteins ACE2 and TMPRSS2. METHODS: Using a variety of normal and malignant models and tissues from the aerodigestive and respiratory tracts, we investigated the expression and regulation of ACE2 and TMPRSS2. RESULTS: We find that ACE2 expression is restricted to a select population of epithelial cells. Notably, infection with SARS-CoV-2 in cancer cell lines, bronchial organoids, and patient nasal epithelium induces metabolic and transcriptional changes consistent with epithelial-to-mesenchymal transition (EMT), including up-regulation of ZEB1 and AXL, resulting in an increased EMT score. In addition, a transcriptional loss of genes associated with tight junction function occurs with SARS-CoV-2 infection. The SARS-CoV-2 receptor, ACE2, is repressed by EMT through the transforming growth factor-ß, ZEB1 overexpression, and onset of EGFR tyrosine kinase inhibitor resistance. This suggests a novel model of SARS-CoV-2 pathogenesis in which infected cells shift toward an increasingly mesenchymal state, associated with a loss of tight junction components with acute respiratory distress syndrome-protective effects. AXL inhibition and ZEB1 reduction, as with bemcentinib, offer a potential strategy to reverse this effect. CONCLUSIONS: These observations highlight the use of aerodigestive and, especially, lung cancer model systems in exploring the pathogenesis of SARS-CoV-2 and other respiratory viruses and offer important insights into the potential mechanisms underlying the morbidity and mortality of coronavirus disease 2019 in healthy patients and patients with cancer alike.


COVID-19 , Lung Neoplasms , Bronchi , Humans , Lung , Peptidyl-Dipeptidase A , SARS-CoV-2
7.
bioRxiv ; 2021 Jan 28.
Article En | MEDLINE | ID: mdl-32577652

COVID-19 is an infectious disease caused by SARS-CoV-2, which enters host cells via the cell surface proteins ACE2 and TMPRSS2. Using a variety of normal and malignant models and tissues from the aerodigestive and respiratory tracts, we investigated the expression and regulation of ACE2 and TMPRSS2. We find that ACE2 expression is restricted to a select population of highly epithelial cells. Notably, infection with SARS-CoV-2 in cancer cell lines, bronchial organoids, and patient nasal epithelium, induces metabolic and transcriptional changes consistent with epithelial to mesenchymal transition (EMT), including upregulation of ZEB1 and AXL, resulting in an increased EMT score. Additionally, a transcriptional loss of genes associated with tight junction function occurs with SARS-CoV-2 infection. The SARS-CoV-2 receptor, ACE2, is repressed by EMT via TGFbeta, ZEB1 overexpression and onset of EGFR TKI inhibitor resistance. This suggests a novel model of SARS-CoV-2 pathogenesis in which infected cells shift toward an increasingly mesenchymal state, associated with a loss of tight junction components with acute respiratory distress syndrome-protective effects. AXL-inhibition and ZEB1-reduction, as with bemcentinib, offers a potential strategy to reverse this effect. These observations highlight the utility of aerodigestive and, especially, lung cancer model systems in exploring the pathogenesis of SARS-CoV-2 and other respiratory viruses, and offer important insights into the potential mechanisms underlying the morbidity and mortality of COVID-19 in healthy patients and cancer patients alike.

8.
Oncogene ; 39(43): 6719-6732, 2020 10.
Article En | MEDLINE | ID: mdl-32963352

Metastasis is the cause for 90% of cancer-related mortalities. Identification of genetic drivers promoting dissemination of tumor cells may provide opportunities for novel therapeutic strategies. We previously reported an in vivo gain-of-function screen that identified ~30 genes with a functional role in metastasis promotion and characterized detailed mechanistic functions of two hits. In this study, we characterized the contribution of one of the identified genes, MBIP (MAP3K12 binding inhibitory protein), towards driving tumor invasion and metastasis. We demonstrate that expression of MBIP significantly enhances the cellular proliferation, migration and invasion of NSCLC cells in vitro and metastasis in vivo. We functionally characterized that MBIP mediates activation of the JNK pathway and induces expression of matrix metalloproteinases (MMPs), which are necessary for the invasive and metastatic phenotype. Our findings establish a novel mechanistic role of MBIP as a driver of NSCLC progression and metastasis.


Carcinoma, Non-Small-Cell Lung/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/pathology , Matrix Metalloproteinases/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Models, Animal , Disease Progression , Female , Gain of Function Mutation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Male , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Xenograft Model Antitumor Assays
9.
Oncogene ; 39(37): 5979-5994, 2020 09.
Article En | MEDLINE | ID: mdl-32753652

Non-small cell lung cancer (NSCLC) is the deadliest form of cancer worldwide, due in part to its proclivity to metastasize. Identifying novel drivers of invasion and metastasis holds therapeutic potential for the disease. We conducted a gain-of-function invasion screen, which identified two separate hits, IMPAD1 and KDELR2, as robust, independent drivers of lung cancer invasion and metastasis. Given that IMPAD1 and KDELR2 are known to be localized to the ER-Golgi pathway, we studied their common mechanism of driving in vitro invasion and in vivo metastasis and demonstrated that they enhance Golgi-mediated function and secretion. Therapeutically inhibiting matrix metalloproteases (MMPs) suppressed both IMPAD1- and KDELR2-mediated invasion. The hits from this unbiased screen and the mechanistic validation highlight Golgi function as one of the key cellular features altered during invasion and metastasis.


Golgi Apparatus/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Phosphoric Monoester Hydrolases/genetics , Vesicular Transport Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Phosphoric Monoester Hydrolases/metabolism , Vesicular Transport Proteins/metabolism
10.
Nat Commun ; 10(1): 5125, 2019 11 12.
Article En | MEDLINE | ID: mdl-31719531

Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide, due in part to the propensity of lung cancer to metastasize. Aberrant epithelial-to-mesenchymal transition (EMT) is a proposed model for the initiation of metastasis. During EMT cell-cell adhesion is reduced allowing cells to dissociate and invade. Of the EMT-associated transcription factors, ZEB1 uniquely promotes NSCLC disease progression. Here we apply two independent screens, BioID and an Epigenome shRNA dropout screen, to define ZEB1 interactors that are critical to metastatic NSCLC. We identify the NuRD complex as a ZEB1 co-repressor and the Rab22 GTPase-activating protein TBC1D2b as a ZEB1/NuRD complex target. We find that TBC1D2b suppresses E-cadherin internalization, thus hindering cancer cell invasion and metastasis.


Cadherins/metabolism , Endocytosis , GTPase-Activating Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Co-Repressor Proteins/metabolism , Humans , Mice , Models, Biological , Neoplasm Metastasis , Protein Binding , rab GTP-Binding Proteins/metabolism
11.
Sci Transl Med ; 11(483)2019 03 13.
Article En | MEDLINE | ID: mdl-30867319

Mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitors have failed to show clinical benefit in Kirsten rat sarcoma (KRAS) mutant lung cancer due to various resistance mechanisms. To identify differential therapeutic sensitivities between epithelial and mesenchymal lung tumors, we performed in vivo small hairpin RNA screens, proteomic profiling, and analysis of patient tumor datasets, which revealed an inverse correlation between mitogen-activated protein kinase (MAPK) signaling dependency and a zinc finger E-box binding homeobox 1 (ZEB1)-regulated epithelial-to-mesenchymal transition. Mechanistic studies determined that MAPK signaling dependency in epithelial lung cancer cells is due to the scaffold protein interleukin-17 receptor D (IL17RD), which is directly repressed by ZEB1. Lung tumors in multiple Kras mutant murine models with increased ZEB1 displayed low IL17RD expression, accompanied by MAPK-independent tumor growth and therapeutic resistance to MEK inhibition. Suppression of ZEB1 function with miR-200 expression or the histone deacetylase inhibitor mocetinostat sensitized resistant cancer cells to MEK inhibition and markedly reduced in vivo tumor growth, showing a promising combinatorial treatment strategy for KRAS mutant cancers. In human lung tumor samples, high ZEB1 and low IL17RD expression correlated with low MAPK signaling, presenting potential markers that predict patient response to MEK inhibitors.


Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mutation/genetics , Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Interleukin-17/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Drug Resistance, Neoplasm , Epithelial Cells/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , MAP Kinase Signaling System , Mesoderm/pathology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasms/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
12.
Nat Commun ; 9(1): 2732, 2018 07 16.
Article En | MEDLINE | ID: mdl-30013058

Genetic aberrations driving pro-oncogenic and pro-metastatic activity remain an elusive target in the quest of precision oncology. To identify such drivers, we use an animal model of KRAS-mutant lung adenocarcinoma to perform an in vivo functional screen of 217 genetic aberrations selected from lung cancer genomics datasets. We identify 28 genes whose expression promoted tumor metastasis to the lung in mice. We employ two tools for examining the KRAS-dependence of genes identified from our screen: 1) a human lung cell model containing a regulatable mutant KRAS allele and 2) a lentiviral system permitting co-expression of DNA-barcoded cDNAs with Cre recombinase to activate a mutant KRAS allele in the lungs of mice. Mechanistic evaluation of one gene, GATAD2B, illuminates its role as a dual activity gene, promoting both pro-tumorigenic and pro-metastatic activities in KRAS-mutant lung cancer through interaction with c-MYC and hyperactivation of the c-MYC pathway.


Adenocarcinoma of Lung/genetics , GATA Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/therapy , Animals , Cell Line, Tumor , Female , GATA Transcription Factors/antagonists & inhibitors , GATA Transcription Factors/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , High-Throughput Screening Assays , Humans , Integrases/genetics , Integrases/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , Mice, Nude , Neoplasm Metastasis , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays
13.
Nat Commun ; 9(1): 2731, 2018 07 16.
Article En | MEDLINE | ID: mdl-30013069

Metastatic lung cancer is the leading cause of cancer-associated mortality worldwide, therefore necessitating novel approaches to identify specific genetic drivers for lung cancer progression and metastasis. We recently performed an in vivo gain-of-function genetic screen to identify driver genes of lung cancer metastasis. In the study reported here, we identify TMEM106B as a primary robust driver of lung cancer metastasis. Ectopic expression of TMEM106B could significantly promote the synthesis of enlarged vesicular lysosomes that are laden with elevated levels of active cathepsins. In a TFEB-dependent manner, TMEM106B could modulate the expression of lysosomal genes of the coordinated lysosomal expression and regulation (CLEAR) pathway in lung cancer cells and patient samples. We also demonstrate that TMEM106B-induced lysosomes undergo calcium-dependent exocytosis, thereby releasing active lysosomal cathepsins necessary for TMEM106B-mediated cancer cell invasion and metastasis in vivo, which could be therapeutically prevented by pharmacological inhibition of cathepsins. Further, in TCGA LUAD data sets, 19% of patients show elevated expression of TMEM106B, which predicts for poor disease-free and overall-survival.


Adenocarcinoma of Lung/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cathepsins/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Animals , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Calcium/metabolism , Cathepsins/antagonists & inhibitors , Cathepsins/metabolism , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Exocytosis , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Proteins/metabolism , Mice , Neoplasm Metastasis , Nerve Tissue Proteins/metabolism , Prognosis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolysis , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays
14.
Oncotarget ; 8(17): 28575-28587, 2017 Apr 25.
Article En | MEDLINE | ID: mdl-28212573

Small cell lung cancer (SCLC) is one of the most aggressive forms of cancer, with a 5-year survival <7%. A major barrier to progress is the absence of predictive biomarkers for chemotherapy and novel targeted agents such as PARP inhibitors. Using a high-throughput, integrated proteomic, transcriptomic, and genomic analysis of SCLC patient-derived xenografts (PDXs) and profiled cell lines, we identified biomarkers of drug sensitivity and determined their prevalence in patient tumors. In contrast to breast and ovarian cancer, PARP inhibitor response was not associated with mutations in homologous recombination (HR) genes (e.g., BRCA1/2) or HRD scores. Instead, we found several proteomic markers that predicted PDX response, including high levels of SLFN11 and E-cadherin and low ATM. SLFN11 and E-cadherin were also significantly associated with in vitro sensitivity to cisplatin and topoisomerase1/2 inhibitors (all commonly used in SCLC). Treatment with cisplatin or PARP inhibitors downregulated SLFN11 and E-cadherin, possibly explaining the rapid development of therapeutic resistance in SCLC. Supporting their functional role, silencing SLFN11 reduced in vitro sensitivity and drug-induced DNA damage; whereas ATM knockdown or pharmacologic inhibition enhanced sensitivity. Notably, SCLC with mesenchymal phenotypes (i.e., loss of E-cadherin and high epithelial-to-mesenchymal transition (EMT) signature scores) displayed striking alterations in expression of miR200 family and key SCLC genes (e.g., NEUROD1, ASCL1, ALDH1A1, MYCL1). Thus, SLFN11, EMT, and ATM mediate therapeutic response in SCLC and warrant further clinical investigation as predictive biomarkers.


Ataxia Telangiectasia Mutated Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Nuclear Proteins/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Ataxia Telangiectasia Mutated Proteins/metabolism , Biomarkers , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , Drug Resistance, Neoplasm/genetics , Gene Expression , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism
15.
PLoS One ; 7(6): e38561, 2012.
Article En | MEDLINE | ID: mdl-22701666

The desmosome anchors keratin filaments in epithelial cells leading to the formation of a tissue wide IF network. Loss of the desmosomal plaque protein plakophilin3 (PKP3) in HCT116 cells, leads to an increase in neoplastic progression and metastasis, which was accompanied by an increase in K8 levels. The increase in levels was due to an increase in the protein levels of the Phosphatase of Regenerating Liver 3 (PRL3), which results in a decrease in phosphorylation on K8. The increase in PRL3 and K8 protein levels could be reversed by introduction of an shRNA resistant PKP3 cDNA. Inhibition of K8 expression in the PKP3 knockdown clone S10, led to a decrease in cell migration and lamellipodia formation. Further, the K8 PKP3 double knockdown clones showed a decrease in colony formation in soft agar and decreased tumorigenesis and metastasis in nude mice. These results suggest that a stabilisation of K8 filaments leading to an increase in migration and transformation may be one mechanism by which PKP3 loss leads to tumor progression and metastasis.


Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic/physiology , Immediate-Early Proteins/metabolism , Keratin-8/metabolism , Neoplasm Metastasis/physiopathology , Neoplasms/metabolism , Plakophilins/deficiency , Protein Tyrosine Phosphatases/metabolism , Animals , Blotting, Western , Desmosomes/metabolism , Electrophoresis, Gel, Two-Dimensional , Fluorescence Resonance Energy Transfer , Gene Knockdown Techniques , HCT116 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Nude , Microscopy, Confocal , Microscopy, Fluorescence , Oligonucleotides/genetics , Phosphorylation , Plakophilins/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Cell Cycle ; 11(2): 361-6, 2012 Jan 15.
Article En | MEDLINE | ID: mdl-22189714

The KRAS-variant is a germline single nucleotide polymorphism (SNP) within the 3'UTR of the KRAS gene predicted to disrupt a complementary binding site (LCS6) for the let-7 microRNA (miRNA). The KRAS-variant is associated with increased risk of various cancers, including lung cancer, ovarian cancer and triple-negative breast cancer, and is associated with altered tumor biology in head and neck cancer, colon cancer and melanoma. To better understand the molecular pathways that may be regulated or affected by the presence of the KRAS-variant allele in cancer cells, we examined its prevalence in the NCI-60 panel of cell lines and sought to identify common features of the cell lines that carry the variant allele. This study provides a step forward towards understanding the molecular and pathological significance of the KRAS-variant.


3' Untranslated Regions , Alleles , MicroRNAs/metabolism , Open Reading Frames , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Binding Sites/genetics , Cell Line, Tumor , Epigenesis, Genetic , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Mutation , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins p21(ras)
17.
Mol Biol Cell ; 22(21): 4068-78, 2011 Nov.
Article En | MEDLINE | ID: mdl-21900500

Keratins are cytoplasmic intermediate filament proteins preferentially expressed by epithelial tissues in a site-specific and differentiation-dependent manner. The complex network of keratin filaments in stratified epithelia is tightly regulated during squamous cell differentiation. Keratin 14 (K14) is expressed in mitotically active basal layer cells, along with its partner keratin 5 (K5), and their expression is down-regulated as cells differentiate. Apart from the cytoprotective functions of K14, very little is known about K14 regulatory functions, since the K14 knockout mice show postnatal lethality. In this study, K14 expression was inhibited using RNA interference in cell lines derived from stratified epithelia to study the K14 functions in epithelial homeostasis. The K14 knockdown clones demonstrated substantial decreases in the levels of the K14 partner K5. These cells showed reduction in cell proliferation and delay in cell cycle progression, along with decreased phosphorylated Akt levels. K14 knockdown cells also exhibited enhanced levels of activated Notch1, involucrin, and K1. In addition, K14 knockdown AW13516 cells showed significant reduction in tumorigenicity. Our results suggest that K5 and K14 may have a role in maintenance of cell proliferation potential in the basal layer of stratified epithelia, modulating phosphatidylinositol 3-kinase/Akt-mediated cell proliferation and/or Notch1-dependent cell differentiation.


Cell Differentiation , Cell Proliferation , Epithelial Cells/physiology , Keratin-14/metabolism , Keratin-5/metabolism , Animals , Antigens, Differentiation/metabolism , Cell Adhesion , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Enzyme Activation , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Interphase , Keratin-14/genetics , Keratin-5/genetics , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasm Transplantation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, Notch1/metabolism , Signal Transduction
18.
J Cell Sci ; 124(Pt 12): 2096-106, 2011 Jun 15.
Article En | MEDLINE | ID: mdl-21610092

Keratins 8 and 18 (K8 and K18) are predominantly expressed in simple epithelial tissues and perform both mechanical and regulatory functions. Aberrant expression of K8 and K18 is associated with neoplastic progression and invasion in squamous cell carcinomas (SCCs). To understand the molecular basis by which K8 promotes neoplastic progression in oral SCC (OSCC), K8 expression was inhibited in AW13516 cells. The K8-knockdown clones showed a significant reduction in tumorigenic potential, which was accompanied by a reduction in cell motility, cell invasion, decreased fascin levels, alterations in the organization of the actin cytoskeleton and changes in cell shape. Furthermore, K8 knockdown led to a decrease in α6ß4 integrin levels and α6ß4-integrin-dependent signalling events, which have been reported to play an important role in neoplastic progression in epithelial tissues. Therefore, modulation of α6ß4 integrin signalling might be one of the mechanisms by which K8 and K18 promote malignant transformation and/or progression in OSCCs.


Carcinoma, Squamous Cell/metabolism , Integrin alpha6beta4/metabolism , Keratin-18/metabolism , Keratin-8/metabolism , Animals , Carcinoma, Squamous Cell/pathology , Cell Division/physiology , Cell Line, Tumor , Cell Movement/physiology , Disease Progression , Humans , Keratin-18/deficiency , Keratin-8/deficiency , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , Signal Transduction
19.
Cell Mol Life Sci ; 68(8): 1439-54, 2011 Apr.
Article En | MEDLINE | ID: mdl-20859650

A decrease in the levels of the desmosomal plaque protein, plakophilin3 (PKP3), leads to a decrease in desmosome size and cell-cell adhesion. To test the hypothesis that PKP3 is required for desmosome formation, the recruitment of desmosomal components to the cell surface was studied in the PKP3 knockdown clones. The PKP3 knockdown clones showed decreased cell border staining for multiple desmosomal proteins, when compared to vector controls, and did not form desmosomes in a calcium switch assay. Further analysis demonstrated that PKP3, plakoglobin (PG) and E-cadherin are present at the cell border at low concentrations of calcium. Loss of either PG or E-cadherin led to a decrease in the levels of PKP3 and other desmosomal proteins at the cell border. The results reported here are consistent with the model that PG and E-cadherin recruit PKP3 to the cell border to initiate desmosome formation.


Cadherins/metabolism , Desmosomes/metabolism , Plakophilins/metabolism , gamma Catenin/metabolism , Cell Adhesion , Cell Line , Fluorescent Antibody Technique , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , Humans , Microscopy, Confocal , Plakophilins/genetics
20.
J Biol ; 9(3): 20, 2010.
Article En | MEDLINE | ID: mdl-20359318

MicroRNAs (miRNAs) are small non-coding RNAs that regulate the expression of numerous target genes. Yet, while hundreds of miRNAs have been identified, little is known about their functions. In a recent report published in Silence, Zheng and colleagues demonstrate a technique for robust and specific knockdown of miRNA expression in Caenorhabditis elegans using modified antisense oligonucleotides, which could be utilized as a powerful tool for the study of regulation and function of miRNAs in vivo.


Caenorhabditis elegans/genetics , MicroRNAs/antagonists & inhibitors , RNA, Small Interfering/genetics , Animals , Cell Differentiation/genetics , Conserved Sequence , Gene Expression Regulation/drug effects , Genome, Helminth/drug effects , Longevity/genetics , MicroRNAs/analysis , MicroRNAs/metabolism , MicroRNAs/physiology , Oligonucleotides, Antisense/pharmacology
...