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










Publication year range
1.
Cancers (Basel) ; 13(19)2021 Oct 04.
Article in English | MEDLINE | ID: mdl-34638461

ABSTRACT

Immune checkpoint inhibitor (ICI) therapy has been a paradigm shift in the treatment of cancer. ICI therapy results in durable responses and survival benefit for a large number of tumor types. Osimertinib, a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) has shown great efficacy treating EGFR mutant lung cancers; however, all patients eventually develop resistance. ICI therapy has not benefitted EGFR mutant lung cancer. Herein, we employed stable isotope labeling by amino acids in cell culture (SILAC) quantitative mass spectrometry-based proteomics to investigate potential immune escape molecular mechanisms in osimertinib resistant EGFR mutant lung adenocarcinoma by interrogating the alterations in the human leukocyte antigen (HLA) Class I-presented immunopeptidome, Class I-interactome, and the whole cell proteome between isogenic osimertinib-sensitive and -resistant human lung adenocarcinoma cells. Our study demonstrates an overall reduction in HLA class I-presented immunopeptidome and downregulation of antigen presentation core complex (e.g., TAP1 and ERAP1/2) and immunoproteasome in osimertinib resistant lung adenocarcinoma cells. Several key components in autophagy pathway are differentially altered. S100 proteins and SLC3A2 may play critical roles in reduced antigen presentation. Our dataset also includes ~1000 novel HLA class I interaction partners and hundreds of Class I-presented immunopeptides in EGFR mutant lung adenocarcinoma. This large-scale unbiased proteomics study provides novel insights and potential mechanisms of immune evasion of EGFR mutant lung adenocarcinoma.

2.
Mol Cell Proteomics ; 20: 100136, 2021.
Article in English | MEDLINE | ID: mdl-34391887

ABSTRACT

Immune checkpoint inhibitors and adoptive lymphocyte transfer-based therapies have shown great therapeutic potential in cancers with high tumor mutational burden (TMB), such as melanoma, but not in cancers with low TMB, such as mutant epidermal growth factor receptor (EGFR)-driven lung adenocarcinoma. Precision immunotherapy is an unmet need for most cancers, particularly for cancers that respond inadequately to immune checkpoint inhibitors. Here, we employed large-scale MS-based proteogenomic profiling to identify potential immunogenic human leukocyte antigen (HLA) class I-presented peptides in melanoma and EGFR-mutant lung adenocarcinoma. Similar numbers of peptides were identified from both tumor types. Cell line and patient-specific databases (DBs) were constructed using variants identified from whole-exome sequencing. A de novo search algorithm was used to interrogate the HLA class I immunopeptidome MS data. We identified 12 variant peptides and several classes of tumor-associated antigen-derived peptides. We constructed a cancer germ line (CG) antigen DB with 285 antigens. This allowed us to identify 40 class I-presented CG antigen-derived peptides. The class I immunopeptidome comprised more than 1000 post-translationally modified (PTM) peptides representing 58 different PTMs, underscoring the critical role PTMs may play in HLA binding. Finally, leveraging de novo search algorithm and an annotated long noncoding RNA (lncRNA) DB, we developed a novel lncRNA-encoded peptide discovery pipeline to identify 44 lncRNA-derived peptides that are presented by class I. We validated tandem MS spectra of select variant, CG antigen, and lncRNA-derived peptides using synthetic peptides and performed HLA class I-binding assays to demonstrate binding to class I proteins. In summary, we provide direct evidence of HLA class I presentation of a large number of variant and tumor-associated peptides in both low and high TMB cancer. These results can potentially be useful for precision immunotherapies, such as vaccine or adoptive cell therapies in melanoma and EGFR-mutant lung cancers.


Subject(s)
Adenocarcinoma of Lung/metabolism , Antigens, Neoplasm/metabolism , Histocompatibility Antigens Class I/metabolism , Lung Neoplasms/metabolism , Melanoma/metabolism , Peptides/metabolism , Adenocarcinoma of Lung/genetics , Aged , Antigens, Neoplasm/genetics , Cell Line, Tumor , ErbB Receptors/genetics , Histocompatibility Antigens Class I/genetics , Humans , Lung Neoplasms/genetics , Male , Melanoma/genetics , Mutation , Peptides/genetics , Proteogenomics
3.
Cancer Res ; 81(11): 3051-3066, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33727228

ABSTRACT

Lung cancer is the leading cause of cancer mortality worldwide. The treatment of patients with lung cancer harboring mutant EGFR with orally administered EGFR tyrosine kinase inhibitors (TKI) has been a paradigm shift. Osimertinib and rociletinib are third-generation irreversible EGFR TKIs targeting the EGFR T790M mutation. Osimertinib is the current standard of care for patients with EGFR mutations due to increased efficacy, lower side effects, and enhanced brain penetrance. Unfortunately, all patients develop resistance. Genomic approaches have primarily been used to interrogate resistance mechanisms. Here we characterized the proteome and phosphoproteome of a series of isogenic EGFR-mutant lung adenocarcinoma cell lines that are either sensitive or resistant to these drugs, comprising the most comprehensive proteomic dataset resource to date to investigate third generation EGFR TKI resistance in lung adenocarcinoma. Unbiased global quantitative mass spectrometry uncovered alterations in signaling pathways, revealed a proteomic signature of epithelial-mesenchymal transition, and identified kinases and phosphatases with altered expression and phosphorylation in TKI-resistant cells. Decreased tyrosine phosphorylation of key sites in the phosphatase SHP2 suggests its inhibition, resulting in subsequent inhibition of RAS/MAPK and activation of PI3K/AKT pathways. Anticorrelation analyses of this phosphoproteomic dataset with published drug-induced P100 phosphoproteomic datasets from the Library of Integrated Network-Based Cellular Signatures program predicted drugs with the potential to overcome EGFR TKI resistance. The PI3K/MTOR inhibitor dactolisib in combination with osimertinib overcame resistance both in vitro and in vivo. Taken together, this study reveals global proteomic alterations upon third generation EGFR TKI resistance and highlights potential novel approaches to overcome resistance. SIGNIFICANCE: Global quantitative proteomics reveals changes in the proteome and phosphoproteome in lung cancer cells resistant to third generation EGFR TKIs, identifying the PI3K/mTOR inhibitor dactolisib as a potential approach to overcome resistance.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Drug Resistance, Neoplasm , Imidazoles/pharmacology , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteome/metabolism , Quinolines/pharmacology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Phosphatidylinositol 3-Kinases/chemistry , Phosphoproteins/analysis , Proteome/analysis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Cells, Cultured
4.
Cell Rep Med ; 1(1)2020 04 21.
Article in English | MEDLINE | ID: mdl-32483558

ABSTRACT

Clonal evolution of osimertinib-resistance mechanisms in EGFR mutant lung adenocarcinoma is poorly understood. Using multi-region whole-exome and RNA sequencing of prospectively collected pre- and post-osimertinib-resistant tumors, including at rapid autopsies, we identify a likely mechanism driving osimertinib resistance in all patients analyzed. The majority of patients acquire two or more resistance mechanisms either concurrently or in temporal sequence. Focal copy-number amplifications occur subclonally and are spatially and temporally separated from common resistance mutations such as EGFR C797S. MET amplification occurs in 66% (n = 6/9) of first-line osimertinib-treated patients, albeit spatially heterogeneous, often co-occurs with additional acquired focal copy-number amplifications and is associated with early progression. Noteworthy osimertinib-resistance mechanisms discovered include neuroendocrine differentiation without histologic transformation, PD-L1, KRAS amplification, and ESR1-AKAP12, MKRN1-BRAF fusions. The subclonal co-occurrence of acquired genomic alterations upon osimertinib resistance will likely require targeting multiple resistance mechanisms by combination therapies.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Carcinoma, Non-Small-Cell Lung , Clonal Evolution , Drug Resistance, Neoplasm/genetics , Lung Neoplasms , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Clonal Evolution/drug effects , Clonal Evolution/genetics , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , Female , Genetic Heterogeneity/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Protein Kinase Inhibitors/therapeutic use , Exome Sequencing , Young Adult
5.
Oncogene ; 39(4): 877-890, 2020 01.
Article in English | MEDLINE | ID: mdl-31570787

ABSTRACT

Barrett's esophagus (BE) is associated with reflux and is implicated the development of esophageal adenocarcinoma (EAC). Apoptosis induces cell death through mitochondrial outer membrane permeabilization (MOMP), which is considered an irreversible step in apoptosis. Activation of MOMP to levels that fail to reach the apoptotic threshold may paradoxically promote cancer-a phenomenon called "Minority MOMP." We asked whether reflux-induced esophageal carcinogenesis occurred via minority MOMP and whether compensatory resistance mechanisms prevented cell death during this process. We exposed preneoplastic, hTERT-immortalized Barrett's cell, CP-C and CP-A, to the oncogenic bile acid, deoxycholic acid (DCA), for 1 year. Induction of minority MOMP was tested via comet assay, CyQuant, annexin V, JC-1, cytochrome C subcellular localization, caspase 3 activation, and immunoblots. We used bcl-2 homology domain-3 (BH3) profiling to test the mitochondrial apoptotic threshold. One-year exposure of Barrett's cells to DCA induced a malignant phenotype noted by clone and tumor formation. DCA promoted minority MOMP noted by minimal release of cytochrome C and limited caspase 3 activation, which resulted in DNA damage without apoptosis. Upregulation of the antiapoptotic protein, Mcl-1, ROS generation, and NF-κB activation occurred in conjunction with minority MOMP. Inhibition of ROS blocked minority MOMP and Mcl-1 upregulation. Knockdown of Mcl-1 shifted minority MOMP to complete MOMP as noted by dynamic BH3 profiling and increased apoptosis. Minority MOMP contributes to DCA induced carcinogenesis in preneoplastic BE. Mcl-1 provided a balance within the mitochondria that induced resistance complete MOMP and cell death. Targeting Mcl-1 may be a therapeutic strategy in EAC.


Subject(s)
Apoptosis , Barrett Esophagus/pathology , Bile Acids and Salts/pharmacology , Carcinogenesis/pathology , Esophageal Neoplasms/pathology , Mitochondria/pathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Barrett Esophagus/drug therapy , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line , Cell Membrane Permeability , Cytochromes c/metabolism , DNA Damage , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophagus/drug effects , Esophagus/metabolism , Esophagus/pathology , Gastrointestinal Agents/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Signal Transduction
6.
J Thorac Oncol ; 15(3): 457-461, 2020 03.
Article in English | MEDLINE | ID: mdl-31783178

ABSTRACT

INTRODUCTION: Children and young adults diagnosed with malignant mesothelioma may have unique genetic characteristics. In this study, we evaluated for the presence of the anaplastic lymphoma kinase (ALK) translocations in these patients. METHODS: In a prospective study of mesothelioma natural history (ClinicalTrials.gov number NCT01950572), we assessed for the presence of the ALK translocation in patients younger than 40 years, irrespective of the site of disease. The presence of this translocation was assessed by means of fluorescence in situ hybridization (FISH). If the patients tested positive for the ALK translocation, both immunohistochemistry and RNA sequencing were performed on the tumor specimen. RESULTS: Between September 2013 and December 2018, 373 patients were enrolled in the mesothelioma natural history study, of which 32 patients were 40 years old or younger at the time of their mesothelioma diagnosis. There were 25 patients with peritoneal mesothelioma, five with pleural mesothelioma, one with pericardial mesothelioma, and one with bicompartmental mesothelioma. Presence of an ALK translocation by FISH was seen in two of the 32 patients (6%) with mesothelioma. Both patients, a 14-year-old female and a 27-year-old male, had peritoneal mesothelioma and had no history of asbestos exposure, prior radiation therapy, or predisposing germline mutations. Neither had detectable ALK expression by immunohistochemistry. RNA sequencing revealed the presence of an STRN fusion partner in the female patient but failed to identify any fusion protein in the male patient. CONCLUSIONS: Young patients with peritoneal mesothelioma should be evaluated for the presence of ALK translocations. Presence of this translocation should be assessed by FISH and these patients could potentially benefit from tyrosine kinase inhibitors targeting ALK.


Subject(s)
Lung Neoplasms , Mesothelioma , Adolescent , Adult , Anaplastic Lymphoma Kinase/genetics , Child , Female , Gene Rearrangement , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Male , Mesothelioma/genetics , Prospective Studies , Receptor Protein-Tyrosine Kinases/genetics , Young Adult
7.
Sci Rep ; 9(1): 16992, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31740700

ABSTRACT

Insulin signaling governs many processes including glucose homeostasis and metabolism, and is therapeutically used to treat hyperglycemia in diabetes. We demonstrated that insulin-induced Akt activation enhances the sensitivity to TGF-ß by directing an increase in cell surface TGF-ß receptors from a pool of intracellular TGF-ß receptors. Consequently, increased autocrine TGF-ß signaling in response to insulin participates in insulin-induced angiogenic responses of endothelial cells. With TGF-ß signaling controlling many cell responses, including differentiation and extracellular matrix deposition, and pathologically promoting fibrosis and cancer cell dissemination, we addressed to which extent autocrine TGF-ß signaling participates in insulin-induced gene responses of human endothelial cells. Transcriptome analyses of the insulin response, in the absence or presence of a TGF-ß receptor kinase inhibitor, revealed substantial positive and negative contributions of autocrine TGF-ß signaling in insulin-responsive gene responses. Furthermore, insulin-induced responses of many genes depended on or resulted from autocrine TGF-ß signaling. Our analyses also highlight extensive contributions of autocrine TGF-ß signaling to basal gene expression in the absence of insulin, and identified many novel TGF-ß-responsive genes. This data resource may aid in the appreciation of the roles of autocrine TGF-ß signaling in normal physiological responses to insulin, and implications of therapeutic insulin usage.


Subject(s)
Gene Expression Profiling/methods , Human Umbilical Vein Endothelial Cells/drug effects , Insulin/pharmacology , Receptors, Transforming Growth Factor beta/genetics , Smad Proteins/genetics , Transforming Growth Factor beta/pharmacology , Benzamides/pharmacology , Cells, Cultured , Dioxoles/pharmacology , Gene Ontology , High-Throughput Nucleotide Sequencing/methods , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoglycemic Agents/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Smad Proteins/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
8.
Proc Natl Acad Sci U S A ; 116(18): 9008-9013, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30975761

ABSTRACT

Survival from malignant mesothelioma, particularly pleural mesothelioma, is very poor. For patients with breast, ovarian, or prostate cancers, overall survival is associated with increased sensitivity to platinum chemotherapy due to loss-of-function mutations in DNA repair genes. The goal of this project was to evaluate, in patients with malignant mesothelioma, the relationship between inherited loss-of-function mutations in DNA repair and other tumor suppressor genes and overall survival following platinum chemotherapy. Patients with histologically confirmed malignant mesothelioma were evaluated for inherited mutations in tumor suppressor genes. Survival was evaluated with respect to genotype and site of mesothelioma. Among 385 patients treated with platinum chemotherapy, median overall survival was significantly longer for patients with loss-of-function mutations in any of the targeted genes compared with patients with no such mutation (P = 0.0006). The effect of genotype was highly significant for patients with pleural mesothelioma (median survival 7.9 y versus 2.4 y, P = 0.0012), but not for patients with peritoneal mesothelioma (median survival 8.2 y versus 5.4 y, P = 0.47). Effect of patient genotype on overall survival, measured at 3 y, remained independently significant after adjusting for gender and age at diagnosis, two other known prognostic factors. Patients with pleural mesothelioma with inherited mutations in DNA repair and other tumor suppressor genes appear to particularly benefit from platinum chemotherapy compared with patients without inherited mutations. These patients may also benefit from other DNA repair targeted therapies such as poly-ADP ribose polymerase (PARP) inhibitors.


Subject(s)
Mesothelioma/genetics , Mesothelioma/mortality , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA Repair/genetics , Female , Genetic Predisposition to Disease/genetics , Germ-Line Mutation , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Mesothelioma/drug therapy , Mesothelioma, Malignant , Middle Aged , Platinum/therapeutic use , Pleural Neoplasms/genetics , Pleural Neoplasms/mortality , Survival Analysis , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Young Adult
9.
Cell Rep ; 26(10): 2651-2666.e6, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30840888

ABSTRACT

Intratumor mutational heterogeneity has been documented in primary non-small-cell lung cancer. Here, we elucidate mechanisms of tumor evolution and heterogeneity in metastatic thoracic tumors (lung adenocarcinoma and thymic carcinoma) using whole-exome and transcriptome sequencing, SNP array for copy-number alterations (CNAs), and mass-spectrometry-based quantitative proteomics of metastases obtained by rapid autopsy. APOBEC mutagenesis, promoted by increased expression of APOBEC3 region transcripts and associated with a high-risk APOBEC3 germline variant, correlated with mutational tumor heterogeneity. TP53 mutation status was associated with APOBEC hypermutator status. Interferon pathways were enriched in tumors with high APOBEC mutagenesis and IFN-γ-induced expression of APOBEC3B in lung adenocarcinoma cells, suggesting that the immune microenvironment may promote mutational heterogeneity. CNAs occurring late in tumor evolution correlated with downstream transcriptomic and proteomic heterogeneity, although global proteomic heterogeneity was significantly greater than transcriptomic and CNA heterogeneity. These results illustrate key mechanisms underlying multi-dimensional heterogeneity in metastatic thoracic tumors.


Subject(s)
Cytidine Deaminase/genetics , Thoracic Neoplasms/genetics , APOBEC Deaminases , DNA Copy Number Variations , Genetic Heterogeneity , Germ-Line Mutation , Humans , Mutagenesis , Neoplasm Metastasis , Proteogenomics/methods , Thoracic Neoplasms/pathology
10.
Mol Cell Proteomics ; 18(4): 622-641, 2019 04.
Article in English | MEDLINE | ID: mdl-30617155

ABSTRACT

Lung cancer is the leading cause of cancer death in both men and women. Tumor heterogeneity is an impediment to targeted treatment of all cancers, including lung cancer. Here, we sought to characterize tumor proteome and phosphoproteome changes by longitudinal, prospective collection of tumor tissue from an exceptional responder lung adenocarcinoma patient who survived with metastatic lung adenocarcinoma for over seven years while undergoing HER2-directed therapy in combination with chemotherapy. We employed "Super-SILAC" and TMT labeling strategies to quantify the proteome and phosphoproteome of a lung metastatic site and eight distinct metastatic progressive lymph nodes collected during these seven years, including five lymph nodes procured at autopsy. We identified specific signaling networks enriched in lung compared with the lymph node metastatic sites. We correlated the changes in protein abundance with changes in copy number alteration (CNA) and transcript expression. ERBB2/HER2 protein expression was higher in lung, consistent with a higher degree of ERBB2 amplification in lung compared with the lymph node metastatic sites. To further interrogate the mass spectrometry data, a patient-specific database was built by incorporating all the somatic and germline variants identified by whole genome sequencing (WGS) of genomic DNA from the lung, one lymph node metastatic site and blood. An extensive validation pipeline was built to confirm variant peptides. We validated 360 spectra corresponding to 55 germline and 6 somatic variant peptides. Targeted MRM assays revealed two novel variant somatic peptides, CDK12-G879V and FASN-R1439Q, expressed in lung and lymph node metastatic sites, respectively. The CDK12-G879V mutation likely results in a nonfunctional CDK12 kinase and chemotherapy susceptibility in lung metastatic sites. Knockdown of CDK12 in lung adenocarcinoma cells increased chemotherapy sensitivity which was rescued by wild type, but not CDK12-G879V expression, consistent with the complete resolution of the lung metastatic sites in this patient.


Subject(s)
Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Cyclin-Dependent Kinases/genetics , Mass Spectrometry/methods , Mutation/genetics , Proteomics , Adenocarcinoma of Lung/metabolism , Cell Line, Tumor , DNA Copy Number Variations/genetics , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Male , Middle Aged , Mutant Proteins/metabolism , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Peptides/metabolism , Phosphoproteins/metabolism , Phosphorylation , Reproducibility of Results
11.
Proc Natl Acad Sci U S A ; 115(50): 12805-12810, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30455306

ABSTRACT

Noncoding RNAs have substantial effects in host-virus interactions. Circular RNAs (circRNAs) are novel single-stranded noncoding RNAs which can decoy other RNAs or RNA-binding proteins to inhibit their functions. The role of circRNAs is largely unknown in the context of Kaposi's sarcoma herpesvirus (KSHV). We hypothesized that circRNAs influence viral infection by inhibiting host and/or viral factors. Transcriptome analysis of KSHV-infected primary endothelial cells and a B cell line identified human circRNAs that are differentially regulated upon infection. We confirmed the expression changes with divergent PCR primers and RNase R treatment of specific circRNAs. Ectopic expression of hsa_circ_0001400, a circRNA induced by infection, suppressed expression of key viral latent gene LANA and lytic gene RTA in KSHV de novo infections. Since human herpesviruses express noncoding RNAs like microRNAs, we searched for viral circRNAs encoded in the KSHV genome. We performed circRNA-Seq analysis with RNase R-treated, circRNA-enriched RNA from KSHV-infected cells. We identified multiple circRNAs encoded by the KSHV genome that are expressed in KSHV-infected endothelial cells and primary effusion lymphoma (PEL) cells. The KSHV circRNAs are located within ORFs of viral lytic genes, are up-regulated upon the induction of the lytic cycle, and alter cell growth. Viral circRNAs were also detected in lymph nodes from patients of KSHV-driven diseases such as PEL, Kaposi's sarcoma, and multicentric Castleman's disease. We revealed new host-virus interactions of circRNAs: human antiviral circRNAs are activated in response to KSHV infection, and viral circRNA expression is induced in the lytic phase of infection.


Subject(s)
Herpesvirus 8, Human/genetics , RNA/genetics , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , B-Lymphocytes/virology , Castleman Disease/genetics , Castleman Disease/virology , Cell Line , Endothelial Cells/virology , Gene Expression Profiling/methods , Gene Expression Regulation, Viral/genetics , Genes, Viral/genetics , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , MicroRNAs/genetics , Open Reading Frames/genetics , RNA, Circular , RNA, Viral/genetics
12.
Mol Cancer Ther ; 17(9): 2013-2023, 2018 09.
Article in English | MEDLINE | ID: mdl-29934340

ABSTRACT

In 2017, an estimated 17,000 individuals were diagnosed with esophageal adenocarcinoma (EAC), and less than 20% will survive 5 years. Positron emission tomography avidity is indicative of high glucose utilization and is nearly universal in EAC. TXNIP blocks glucose uptake and exhibits proapoptotic functions. Higher expression in EAC has been associated with improved disease-specific survival, lack of lymph node involvement, reduced perineural invasion, and increased tumor differentiation. We hypothesized that TXNIP may act as a tumor suppressor that sensitizes EAC cells to standard chemotherapeutics. EAC cell lines and a Barrett epithelial cell line were used. qRT-PCR, immunoblot, and immunofluorescence techniques evaluated gene expression. TXNIP was stably overexpressed or knocked down using lentiviral RNA transduction techniques. Murine xenograft methods examined growth following overexpression of TXNIP. Apoptosis and DNA damage were measured by annexin V and γH2AX assays. Activation of the intrinsic apoptosis was quantitated with green fluorescence protein-caspase 3 reporter assay. In cultured cells and an esophageal tissue array, TXNIP expression was higher in Barrett epithelia and normal tissue compared with EAC. Constitutive overexpression of TXNIP decreased proliferation, clonogenicity, and tumor xenograft growth. TXNIP overexpression increased, whereas knockdown abrogated, DNA damage and apoptosis following cisplatin treatment. An HDAC inhibitor, entinostat (currently in clinical trials), upregulated TXNIP and synergistically increased cisplatin-mediated DNA damage and apoptosis. TXNIP is a tumor suppressor that is downregulated in EACC. Its reexpression dramatically sensitizes these cells to cisplatin. Our findings support phase I/II evaluation of "priming" strategies to enhance the efficacy of conventional chemotherapeutics in EAC. Mol Cancer Ther; 17(9); 2013-23. ©2018 AACR.


Subject(s)
Adenocarcinoma/drug therapy , Apoptosis/drug effects , Benzamides/pharmacology , Carrier Proteins/genetics , DNA Damage , Esophageal Neoplasms/drug therapy , Pyridines/pharmacology , Xenograft Model Antitumor Assays , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cisplatin/administration & dosage , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice, Nude , Transcriptional Activation/drug effects
13.
Nucleic Acids Res ; 45(20): 12039-12053, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29036671

ABSTRACT

CRISPR/Cas9 is a powerful gene editing tool for gene knockout studies and functional genomic screens. Successful implementation of CRISPR often requires Cas9 to elicit efficient target knockout in a population of cells. In this study, we investigated the role of several key factors, including variation in target copy number, inherent potency of sgRNA guides, and expression level of Cas9 and sgRNA, in determining CRISPR knockout efficiency. Using isogenic, clonal cell lines with variable copy numbers of an EGFP transgene, we discovered that CRISPR knockout is relatively insensitive to target copy number, but is highly dependent on the potency of the sgRNA guide sequence. Kinetic analysis revealed that most target mutation occurs between 5 and 10 days following Cas9/sgRNA transduction, while sgRNAs with different potencies differ by their knockout time course and by their terminal-phase knockout efficiency. We showed that prolonged, low level expression of Cas9 and sgRNA often fails to elicit target mutation, particularly if the potency of the sgRNA is also low. Our findings provide new insights into the behavior of CRISPR/Cas9 in mammalian cells that could be used for future improvement of this platform.


Subject(s)
CRISPR-Cas Systems , Gene Dosage , Gene Knockout Techniques/methods , Green Fluorescent Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Cell Line, Tumor , Endonucleases/genetics , Endonucleases/metabolism , Flow Cytometry , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Kinetics , Mutation , Polymerase Chain Reaction , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Reproducibility of Results , Transgenes/genetics
14.
Mol Cell Proteomics ; 16(5): 891-910, 2017 05.
Article in English | MEDLINE | ID: mdl-28331001

ABSTRACT

Mutations in the Epidermal growth factor receptor (EGFR) kinase domain, such as the L858R missense mutation and deletions spanning the conserved sequence 747LREA750, are sensitive to tyrosine kinase inhibitors (TKIs). The gatekeeper site residue mutation, T790M accounts for around 60% of acquired resistance to EGFR TKIs. The first generation EGFR TKIs, erlotinib and gefitinib, and the second generation inhibitor, afatinib are FDA approved for initial treatment of EGFR mutated lung adenocarcinoma. The predominant biomarker of EGFR TKI responsiveness is the presence of EGFR TKI-sensitizing mutations. However, 30-40% of patients with EGFR mutations exhibit primary resistance to these TKIs, underscoring the unmet need of identifying additional biomarkers of treatment response. Here, we sought to characterize the dynamics of tyrosine phosphorylation upon EGFR TKI treatment of mutant EGFR-driven human lung adenocarcinoma cell lines with varying sensitivity to EGFR TKIs, erlotinib and afatinib. We employed stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative mass spectrometry to identify and quantify tyrosine phosphorylated peptides. The proportion of tyrosine phosphorylated sites that had reduced phosphorylation upon erlotinib or afatinib treatment correlated with the degree of TKI-sensitivity. Afatinib, an irreversible EGFR TKI, more effectively inhibited tyrosine phosphorylation of a majority of the substrates. The phosphosites with phosphorylation SILAC ratios that correlated with the TKI-sensitivity of the cell lines include sites on kinases, such as EGFR-Y1197 and MAPK7-Y221, and adaptor proteins, such as SHC1-Y349/350, ERRFI1-Y394, GAB1-Y689, STAT5A-Y694, DLG3-Y705, and DAPP1-Y139, suggesting these are potential biomarkers of TKI sensitivity. DAPP1, is a novel target of mutant EGFR signaling and Y-139 is the major site of DAPP1 tyrosine phosphorylation. We also uncovered several off-target effects of these TKIs, such as MST1R-Y1238/Y1239 and MET-Y1252/1253. This study provides unique insight into the TKI-mediated modulation of mutant EGFR signaling, which can be applied to the development of biomarkers of EGFR TKI response.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Phosphotyrosine/metabolism , Protein Kinase Inhibitors/therapeutic use , Proteomics/methods , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Afatinib , Cell Line, Tumor , Cluster Analysis , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Humans , Isotope Labeling , Lung Neoplasms/pathology , Mass Spectrometry , Mutation/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Reproducibility of Results , Signal Transduction/drug effects , Tyrosine/metabolism
15.
Nucleic Acids Res ; 45(11): e101, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28334828

ABSTRACT

CRISPR/Cas9-based gene knockout libraries have emerged as a powerful tool for functional screens. We present here a set of pre-designed human and mouse sgRNA sequences that are optimized for both high on-target potency and low off-target effect. To maximize the chance of target gene inactivation, sgRNAs were curated to target both 5΄ constitutive exons and exons that encode conserved protein domains. We describe here a robust and cost-effective method to construct multiple small sized CRISPR library from a single oligo pool generated by array synthesis using parallel oligonucleotide retrieval. Together, these resources provide a convenient means for individual labs to generate customized CRISPR libraries of variable size and coverage depth for functional genomics application.


Subject(s)
CRISPR-Cas Systems , Gene Knockout Techniques , Oligonucleotides/genetics , Animals , Exons , Gene Library , Humans , Mice
17.
Cold Spring Harb Mol Case Stud ; 2(6): a001263, 2016 11.
Article in English | MEDLINE | ID: mdl-27900369

ABSTRACT

We used next-generation sequencing to identify somatic alterations in multiple metastatic sites from an "exceptional responder" lung adenocarcinoma patient during his 7-yr course of ERBB2-directed therapies. The degree of heterogeneity was unprecedented, with ∼1% similarity between somatic alterations of the lung and lymph nodes. One novel translocation, PLAG1-ACTA2, present in both sites, up-regulated ACTA2 expression. ERBB2, the predominant driver oncogene, was amplified in both sites, more pronounced in the lung, and harbored an L869R mutation in the lymph node. Functional studies showed increased proliferation, migration, metastasis, and resistance to ERBB2-directed therapy because of L869R mutation and increased migration because of ACTA2 overexpression. Within the lung, a nonfunctional CDK12, due to a novel G879V mutation, correlated with down-regulation of DNA damage response genes, causing genomic instability, and sensitivity to chemotherapy. We propose a model whereby a subclone metastasized early from the primary site and evolved independently in lymph nodes.


Subject(s)
Adenocarcinoma/genetics , Lung Neoplasms/genetics , Receptor, ErbB-2/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Down-Regulation , Gene Expression Regulation, Neoplastic/genetics , Genes, erbB-2/genetics , Genomics , High-Throughput Nucleotide Sequencing/methods , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Mutation , Neoplasm Metastasis/genetics , Receptor, ErbB-2/metabolism , Treatment Outcome
18.
Mol Cell ; 64(3): 549-564, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27746021

ABSTRACT

In advanced stages of cancers, TGF-ß promotes tumor progression in conjunction with inputs from receptor tyrosine kinase pathways. However, mechanisms that underpin the signaling cooperation and convert TGF-ß from a potent growth inhibitor to a tumor promoter are not fully understood. We report here that TGF-ß directly regulates alternative splicing of cancer stem cell marker CD44 through a phosphorylated T179 of SMAD3-mediated interaction with RNA-binding protein PCBP1. We show that TGF-ß and EGF respectively induce SMAD3 and PCBP1 to colocalize in SC35-positive nuclear speckles, and the two proteins interact in the variable exon region of CD44 pre-mRNA to inhibit spliceosome assembly in favor of expressing the mesenchymal isoform CD44s over the epithelial isoform CD44E. We further show that the SMAD3-mediated alternative splicing is essential to the tumor-promoting role of TGF-ß and has a global influence on protein products of genes instrumental to epithelial-to-mesenchymal transition and metastasis.


Subject(s)
Alternative Splicing/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Hyaluronan Receptors/genetics , Lung Neoplasms/genetics , Smad3 Protein/genetics , Animals , Cell Line, Tumor , DNA-Binding Proteins , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Exons , Female , Gene Expression Profiling , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Hyaluronan Receptors/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA-Binding Proteins , Signal Transduction , Smad3 Protein/metabolism , Threonine/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
19.
PLoS Genet ; 12(10): e1006384, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27776121

ABSTRACT

We have asked how the common S34F mutation in the splicing factor U2AF1 regulates alternative splicing in lung cancer, and why wild-type U2AF1 is retained in cancers with this mutation. A human lung epithelial cell line was genetically modified so that U2AF1S34F is expressed from one of the two endogenous U2AF1 loci. By altering levels of mutant or wild-type U2AF1 in this cell line and by analyzing published data on human lung adenocarcinomas, we show that S34F-associated changes in alternative splicing are proportional to the ratio of S34F:wild-type gene products and not to absolute levels of either the mutant or wild-type factor. Preferential recognition of specific 3' splice sites in S34F-expressing cells is largely explained by differential in vitro RNA-binding affinities of mutant versus wild-type U2AF1 for those same 3' splice sites. Finally, we show that lung adenocarcinoma cell lines bearing U2AF1 mutations do not require the mutant protein for growth in vitro or in vivo. In contrast, wild-type U2AF1 is required for survival, regardless of whether cells carry the U2AF1S34F allele. Our results provide mechanistic explanations of the magnitude of splicing changes observed in U2AF1-mutant cells and why tumors harboring U2AF1 mutations always retain an expressed copy of the wild-type allele.


Subject(s)
Adenocarcinoma/genetics , Alternative Splicing/genetics , Cell Proliferation/genetics , Lung Neoplasms/genetics , Splicing Factor U2AF/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Mice , Mutant Proteins/genetics , Mutation , Nucleotide Motifs/genetics , RNA, Messenger/biosynthesis , Splicing Factor U2AF/biosynthesis , Transcriptome/genetics , Xenograft Model Antitumor Assays
20.
Methods Mol Biol ; 1176: 45-58, 2014.
Article in English | MEDLINE | ID: mdl-25030918

ABSTRACT

Pooled shRNA library is a powerful, rapid, and cost-effective technology to carry out functional genomic screens in mammalian cells. This approach has been applied extensively to identify genetic dependencies in cancer cells that might be exploited for therapeutic purposes. In this chapter we provide a detailed protocol for using the Hannon-Elledge miR30-based library to conduct dropout screens in cancer cell lines. This protocol is readily adaptable to other pooled shRNA libraries and should facilitate the functional annotation of the human genome.


Subject(s)
Gene Library , Genes, Lethal , MicroRNAs/genetics , Mutation , Neoplasms/genetics , RNA, Small Interfering/genetics , Animals , Cell Line , Cell Line, Tumor , Computational Biology/methods , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...