Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Thorac Oncol ; 19(2): 273-284, 2024 02.
Article in English | MEDLINE | ID: mdl-37717856

ABSTRACT

INTRODUCTION: Morphologic and molecular data for staging of multifocal lung squamous cell carcinomas (LSCCs) are limited. In this study, whole exome sequencing (WES) was used as the gold standard to determine whether multifocal LSCC represented separate primary lung cancers (SPLCs) or intrapulmonary metastases (IPMs). Genomic profiles were compared with the comprehensive morphologic assessment. METHODS: WES was performed on 20 tumor pairs of multifocal LSCC and matched normal lymph nodes using the Illumina NovaSeq6000 S4-Xp (Illumina, San Diego, CA). WES clonal and subclonal analysis data were compared with histologic assessment by 16 thoracic pathologists. In addition, the immune gene profiling of the study cases was characterized by the HTG EdgeSeq Precision Immuno-Oncology Panel. RESULTS: By WES data, 11 cases were classified as SPLC and seven cases as IPM. Two cases were technically suboptimal. Analysis revealed marked genomic and immunogenic heterogeneity, but immune gene expression profiles highly correlated with mutation profiles. Tumors classified as IPM have a large number of shared mutations (ranging from 33.5% to 80.7%). The agreement between individual morphologic assessments for each case and WES was 58.3%. One case was unanimously interpreted morphologically as IPM and was in agreement with WES. In a further 17 cases, the number of pathologists whose morphologic interpretation was in agreement with WES ranged from two (one case) to 15 pathologists (one case) per case. Pathologists showed a fair interobserver agreement in the morphologic staging of multiple LSCCs, with an overall kappa of 0.232. CONCLUSIONS: Staging of multifocal LSCC based on morphologic assessment is unreliable. Comprehensive genomic analyses should be adopted for the staging of multifocal LSCC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Humans , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Genomics , Lung/pathology
2.
J Thorac Oncol ; 18(10): 1290-1302, 2023 10.
Article in English | MEDLINE | ID: mdl-37702631

ABSTRACT

INTRODUCTION: Pathologic response has been proposed as an early clinical trial end point of survival after neoadjuvant treatment in clinical trials of NSCLC. The International Association for the Study of Lung Cancer (IASLC) published recommendations for pathologic evaluation of resected lung cancers after neoadjuvant therapy. The aim of this study was to assess pathologic response interobserver reproducibility using IASLC criteria. METHODS: An international panel of 11 pulmonary pathologists reviewed hematoxylin and eosin-stained slides from the lung tumors of resected NSCLC from 84 patients who received neoadjuvant immune checkpoint inhibitors in six clinical trials. Pathologic response was assessed for percent viable tumor, necrosis, and stroma. For each slide, tumor bed area was measured microscopically, and pre-embedded formulas calculated unweighted and weighted major pathologic response (MPR) averages to reflect variable tumor bed proportion. RESULTS: Unanimous agreement among pathologists for MPR was observed in 68 patients (81%), and inter-rater agreement (IRA) was 0.84 (95% confidence interval [CI]: 0.76-0.92) and 0.86 (95% CI: 0.79-0.93) for unweighted and weighted averages, respectively. Overall, unweighted and weighted methods did not reveal significant differences in the classification of MPR. The highest concordance by both methods was observed for cases with more than 95% viable tumor (IRA = 0.98, 95% CI: 0.96-1) and 0% viable tumor (IRA = 0.94, 95% CI: 0.89-0.98). The most common reasons for discrepancies included interpretations of tumor bed, presence of prominent stromal inflammation, distinction between reactive and neoplastic pneumocytes, and assessment of invasive mucinous adenocarcinoma. CONCLUSIONS: Our study revealed excellent reliability in cases with no residual viable tumor and good reliability for MPR with the IASLC recommended less than or equal to 10% cutoff for viable tumor after neoadjuvant therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/pathology , Neoadjuvant Therapy/methods , Reproducibility of Results , Carcinoma, Non-Small-Cell Lung/pathology , Lung/pathology
4.
Clin Cancer Res ; 28(11): 2461-2473, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35394499

ABSTRACT

PURPOSE: Our understanding of the immunopathology of resectable non-small cell lung cancer (NSCLC) is still limited. Here, we explore immune programs that inform of tumor immunity and response to neoadjuvant chemotherapy and chemoimmunotherapy in localized NSCLC. EXPERIMENTAL DESIGN: Targeted immune gene sequencing using the HTG Precision Immuno-Oncology panel was performed in localized NSCLCs from three cohorts based on treatment: naïve (n = 190), neoadjuvant chemotherapy (n = 38), and neoadjuvant chemoimmunotherapy (n = 21). Tumor immune microenvironment (TIME) phenotypes were based on the location of CD8+ T cells (inflamed, cold, excluded), tumoral PD-L1 expression (<1% and ≥1%), and tumor-infiltrating lymphocytes (TIL). Immune programs and signatures were statistically analyzed on the basis of tumoral PD-L1 expression, immune phenotypes, and pathologic response and were cross-compared across the three cohorts. RESULTS: PD-L1-positive tumors exhibited increased signature scores for various lymphoid and myeloid cell subsets (P < 0.05). TIME phenotypes exhibited disparate frequencies by stage, PD-L1 expression, and mutational burden. Inflamed and PD-L1+/TILs+ NSCLCs displayed overall significantly heightened levels of immune signatures, with the excluded group representing an intermediate state. A cytotoxic T-cell signature was associated with favorable survival in neoadjuvant chemotherapy-treated NSCLCs (P < 0.05). Pathologic response to chemoimmunotherapy was positively associated with higher expression of genes involved in immune activation, chemotaxis, as well as T and natural killer cells (P < 0.05 for all). Among the three cohorts, chemoimmunotherapy-treated NSCLCs exhibited the highest scores for various immune cell subsets including T effector and B cells (P < 0.05). CONCLUSIONS: Our findings highlight immune gene programs that may underlie host tumor immunity and response to neoadjuvant chemotherapy and chemoimmunotherapy in resectable NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , B7-H1 Antigen , Biomarkers, Tumor/therapeutic use , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lymphocytes, Tumor-Infiltrating , Neoadjuvant Therapy , Prognosis , Tumor Microenvironment/genetics
5.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34155143

ABSTRACT

A chromosome 1q21.3 region that is frequently amplified in diverse cancer types encodes phosphatidylinositol (PI)-4 kinase IIIß (PI4KIIIß), a key regulator of secretory vesicle biogenesis and trafficking. Chromosome 1q21.3-amplified lung adenocarcinoma (1q-LUAD) cells rely on PI4KIIIß for Golgi-resident PI-4-phosphate (PI4P) synthesis, prosurvival effector protein secretion, and cell viability. Here, we show that 1q-LUAD cells subjected to prolonged PI4KIIIß antagonist treatment acquire tolerance by activating an miR-218-5p-dependent competing endogenous RNA network that up-regulates PI4KIIα, which provides an alternative source of Golgi-resident PI4P that maintains prosurvival effector protein secretion and cell viability. These findings demonstrate an addiction to Golgi-resident PI4P synthesis in a genetically defined subset of cancers.


Subject(s)
Adenocarcinoma of Lung/genetics , Chromosomes, Human, Pair 1/genetics , Gene Amplification , Golgi Apparatus/metabolism , Phosphatidylinositol Phosphates/biosynthesis , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/metabolism , Cell Line, Tumor , Enzyme Activation , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol Phosphates/antagonists & inhibitors , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation/genetics
6.
Cell Rep ; 35(3): 109009, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882319

ABSTRACT

Cancer cells function as primary architects of the tumor microenvironment. However, the molecular features of cancer cells that govern stromal cell phenotypes remain unclear. Here, we show that cancer-associated fibroblast (CAF) heterogeneity is driven by lung adenocarcinoma (LUAD) cells at either end of the epithelial-to-mesenchymal transition (EMT) spectrum. LUAD cells that have high expression of the EMT-activating transcription factor ZEB1 reprogram CAFs through a ZEB1-dependent secretory program and direct CAFs to the tips of invasive projections through a ZEB1-driven CAF repulsion process. The EMT, in turn, sensitizes LUAD cells to pro-metastatic signals from CAFs. Thus, CAFs respond to contextual cues from LUAD cells to promote metastasis.


Subject(s)
Adenocarcinoma of Lung/genetics , Cancer-Associated Fibroblasts/metabolism , Epithelial Cells/metabolism , Kidney Neoplasms/genetics , Lung Neoplasms/genetics , Mesenchymal Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/secondary , Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Cell Communication , Cell Line, Tumor , Cell Movement , Cell Proliferation , Discoidin Domain Receptor 2/genetics , Discoidin Domain Receptor 2/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Transgenic , Signal Transduction , Tumor Microenvironment/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
7.
Commun Biol ; 4(1): 482, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33875777

ABSTRACT

Cancer cells are a major source of enzymes that modify collagen to create a stiff, fibrotic tumor stroma. High collagen lysyl hydroxylase 2 (LH2) expression promotes metastasis and is correlated with shorter survival in lung adenocarcinoma (LUAD) and other tumor types. LH2 hydroxylates lysine (Lys) residues on fibrillar collagen's amino- and carboxy-terminal telopeptides to create stable collagen cross-links. Here, we show that electrostatic interactions between the LH domain active site and collagen determine the unique telopeptidyl lysyl hydroxylase (tLH) activity of LH2. However, CRISPR/Cas-9-mediated inactivation of tLH activity does not fully recapitulate the inhibitory effect of LH2 knock out on LUAD growth and metastasis in mice, suggesting that LH2 drives LUAD progression, in part, through a tLH-independent mechanism. Protein homology modeling and biochemical studies identify an LH2 isoform (LH2b) that has previously undetected collagen galactosylhydroxylysyl glucosyltransferase (GGT) activity determined by a loop that enhances UDP-glucose-binding in the GLT active site and is encoded by alternatively spliced exon 13 A. CRISPR/Cas-9-mediated deletion of exon 13 A sharply reduces the growth and metastasis of LH2b-expressing LUADs in mice. These findings identify a previously unrecognized collagen GGT activity that drives LUAD progression.


Subject(s)
Adenocarcinoma of Lung/physiopathology , Disease Progression , Glucosyltransferases/metabolism , Lung Neoplasms/physiopathology , Animals , Mice
8.
Cancers (Basel) ; 13(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669241

ABSTRACT

Lung cancer is consistently the leading cause of cancer-related death worldwide, and it ranks as the second most frequent type of new cancer cases diagnosed in the United States, both in males and females. One subtype of lung cancer, small cell lung carcinoma (SCLC), is an aggressive, poorly differentiated, and high-grade neuroendocrine carcinoma that accounts for 13% of all lung carcinomas. SCLC is the most frequent neuroendocrine lung tumor, and it is commonly presented as an advanced stage disease in heavy smokers. Due to its clinical presentation, it is typically diagnosed in small biopsies or cytology specimens, with routine immunostaining only. However, immunohistochemistry markers are extremely valuable in demonstrating neuroendocrine features of SCLC and supporting its differential diagnosis. The 2015 WHO classification grouped all pulmonary neuroendocrine carcinomas in one category and maintained the SCLC combined variant that was previously recognized. In this review, we explore multiple aspects of the pathologic features of this entity, as well as clinically relevant immunohistochemistry markers expression and its molecular characteristics. In addition, we will focus on characteristics of the tumor microenvironment, and the latest pathogenesis findings to better understand the new therapeutic options in the current era of personalized therapy.

9.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32931483

ABSTRACT

Therapeutic strategies designed to target TP53-deficient cancer cells remain elusive. Here, we showed that TP53 loss initiated a pharmacologically actionable secretory process that drove lung adenocarcinoma (LUAD) progression. Molecular, biochemical, and cell biological studies showed that TP53 loss increased the expression of Golgi reassembly and stacking protein 55 kDa (G55), a Golgi stacking protein that maintains Golgi organelle integrity and is part of a GOLGIN45 (G45)-myosin IIA-containing protein complex that activates secretory vesicle biogenesis in the Golgi. TP53 loss activated G55-dependent secretion by relieving G55 and myosin IIA from miR-34a-dependent silencing. G55-dependent secreted proteins enhanced the proliferative and invasive activities of TP53-deficient LUAD cells and promoted angiogenesis and CD8+ T cell exhaustion in the tumor microenvironment. A small molecule that blocks G55-G45 interactions impaired secretion and reduced TP53-deficient LUAD growth and metastasis. These results identified a targetable secretory vulnerability in TP53-deficient LUAD cells.


Subject(s)
Adenocarcinoma of Lung/metabolism , Golgi Apparatus/metabolism , Lung Neoplasms/metabolism , Tumor Suppressor Protein p53/deficiency , Vesicular Transport Proteins/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Cell Line, Tumor , Golgi Apparatus/genetics , Golgi Apparatus/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , Tumor Suppressor Protein p53/metabolism , Vesicular Transport Proteins/genetics
10.
Matrix Biol ; 91-92: 8-18, 2020 09.
Article in English | MEDLINE | ID: mdl-32442601

ABSTRACT

Tumor progression is marked by dense collagenous matrix accumulations that dynamically reorganize to accommodate a growing and invasive tumor mass. Cancer-associated fibroblasts (CAFs) play an essential role in matrix remodeling and influence other processes in the tumor microenvironment, including angiogenesis, immunosuppression, and invasion. These findings have spawned efforts to elucidate CAF functionality at the single-cell level. Here, we will discuss how those efforts have impacted our understanding of the ways in which CAFs govern matrix remodeling and the influence of matrix remodeling on the development of an immunosuppressive tumor microenvironment.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Extracellular Matrix Proteins/immunology , Extracellular Matrix/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Cancer-Associated Fibroblasts/pathology , Cell Lineage/genetics , Cell Lineage/immunology , Cytokines/genetics , Cytokines/immunology , Disease Progression , Extracellular Matrix/chemistry , Extracellular Matrix Proteins/genetics , Fibrosis , Gene Expression Regulation, Neoplastic , Humans , Immunity, Innate , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Signal Transduction , Single-Cell Analysis , Stromal Cells/immunology , Stromal Cells/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
11.
Sci Transl Med ; 12(527)2020 01 22.
Article in English | MEDLINE | ID: mdl-31969487

ABSTRACT

Heightened secretion of protumorigenic effector proteins is a feature of malignant cells. Yet, the molecular underpinnings and therapeutic implications of this feature remain unclear. Here, we identify a chromosome 1q region that is frequently amplified in diverse cancer types and encodes multiple regulators of secretory vesicle biogenesis and trafficking, including the Golgi-dedicated enzyme phosphatidylinositol (PI)-4-kinase IIIß (PI4KIIIß). Molecular, biochemical, and cell biological studies show that PI4KIIIß-derived PI-4-phosphate (PI4P) synthesis enhances secretion and accelerates lung adenocarcinoma progression by activating Golgi phosphoprotein 3 (GOLPH3)-dependent vesicular release from the Golgi. PI4KIIIß-dependent secreted factors maintain 1q-amplified cancer cell survival and influence prometastatic processes in the tumor microenvironment. Disruption of this functional circuitry in 1q-amplified cancer cells with selective PI4KIIIß antagonists induces apoptosis and suppresses tumor growth and metastasis. These results support a model in which chromosome 1q amplifications create a dependency on PI4KIIIß-dependent secretion for cancer cell survival and tumor progression.


Subject(s)
Adenocarcinoma of Lung/metabolism , Chromosomes, Human, Pair 1/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adenocarcinoma of Lung/genetics , Animals , Chromosomes, Human, Pair 1/genetics , Enzyme-Linked Immunosorbent Assay , Golgi Apparatus/metabolism , Humans , In Vitro Techniques , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , X-Ray Microtomography
12.
Matrix Biol Plus ; 8: 100047, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33543040

ABSTRACT

Collagenous stromal accumulations predict a worse clinical outcome in a variety of malignancies. Better tools are needed to elucidate the way in which collagen influences cancer cells. Here, we report a method to generate collagenous matrices that are deficient in key post-translational modifications and evaluate cancer cell behaviors on those matrices. We utilized genetic and biochemical approaches to inhibit lysine hydroxylation and glucosylation on collagen produced by MC-3T3-E1 murine osteoblasts (MC cells). Seeded onto MC cell-derived matrix surface, multicellular aggregates containing lung adenocarcinoma cells alone or in combination with cancer-associated fibroblasts dissociated with temporal and spatial patterns that were influenced by collagen modifications. These findings demonstrate the feasibility of generating defined collagen matrices that are suitable for cell culture studies.

13.
Nat Commun ; 9(1): 2719, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29988035

ABSTRACT

In the originally published version of this Article, financial support was not fully acknowledged. The PDF and HTML versions of the Article have now been corrected to also include support from the National Institutes of Health grant T32GM008280 to Sarah Alvarado.

14.
Nat Commun ; 9(1): 512, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29410444

ABSTRACT

Collagen lysyl hydroxylases (LH1-3) are Fe2+- and 2-oxoglutarate (2-OG)-dependent oxygenases that maintain extracellular matrix homeostasis. High LH2 levels cause stable collagen cross-link accumulations that promote fibrosis and cancer progression. However, developing LH antagonists will require structural insights. Here, we report a 2 Å crystal structure and X-ray scattering on dimer assemblies for the LH domain of L230 in Acanthamoeba polyphaga mimivirus. Loop residues in the double-stranded ß-helix core generate a tail-to-tail dimer. A stabilizing hydrophobic leucine locks into an aromatic tyrosine-pocket on the opposite subunit. An active site triad coordinates Fe2+. The two active sites flank a deep surface cleft that suggest dimerization creates a collagen-binding site. Loss of Fe2+-binding disrupts the dimer. Dimer disruption and charge reversal in the cleft increase Km and reduce LH activity. Ectopic L230 expression in tumors promotes collagen cross-linking and metastasis. These insights suggest inhibitor targets for fibrosis and cancer.


Subject(s)
Iron/chemistry , Mimiviridae/enzymology , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/chemistry , Protein Multimerization , Viral Proteins/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Cell Line, Tumor , Collagen/chemistry , Collagen/metabolism , Crystallography, X-Ray , Enzyme Stability , Humans , Iron/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Mimiviridae/genetics , Mutation , Neoplasm Metastasis , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Protein Binding , Scattering, Small Angle , Sequence Homology, Amino Acid , Transplantation, Heterologous , Viral Proteins/genetics , Viral Proteins/metabolism , X-Ray Diffraction
15.
Sci Rep ; 7(1): 6478, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28744021

ABSTRACT

Cancer-associated fibroblasts (CAFs) regulate diverse intratumoral biological programs and can promote or inhibit tumorigenesis, but those CAF populations that negatively impact the clinical outcome of lung cancer patients have not been fully elucidated. Because Thy-1 (CD90) marks CAFs that promote tumor cell invasion in a murine model of KrasG12D-driven lung adenocarcinoma (KrasLA1), here we postulated that human lung adenocarcinomas containing Thy-1+ CAFs have a worse prognosis. We first examined the location of Thy-1+ CAFs within human lung adenocarcinomas. Cells that co-express Thy-1 and α-smooth muscle actin (αSMA), a CAF marker, were located on the tumor periphery surrounding collectively invading tumor cells and in perivascular regions. To interrogate a human lung cancer database for the presence of Thy-1+ CAFs, we isolated Thy-1+ CAFs and normal lung fibroblasts (LFs) from the lungs of KrasLA1 mice and wild-type littermates, respectively, and performed global proteomic analysis on the murine CAFs and LFs, which identified 425 proteins that were differentially expressed. Used as a probe to identify Thy-1+ CAF-enriched tumors in a compendium of 1,586 lung adenocarcinomas, the presence of the 425-gene signature predicted a significantly shorter survival. Thus, Thy-1 marks a CAF population that adversely impacts clinical outcome in human lung cancer.


Subject(s)
Adenocarcinoma of Lung/pathology , Cancer-Associated Fibroblasts/pathology , Lung Neoplasms/pathology , Thy-1 Antigens/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Animals , Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Mice, Mutant Strains , Phosphoproteins/genetics , Phosphoproteins/metabolism , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors , YAP-Signaling Proteins
16.
Oncotarget ; 8(1): 3-14, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27980214

ABSTRACT

The ubiquitin-like modifier interferon-stimulated gene 15 (ISG15) is implicated in both oncogenic and tumor suppressive programs. Yet, few ISGylation substrates are known and functionally validated in cancer biology. We previously found specific oncoproteins were substrates of ISGylation and were stabilized by the ISG15-specific deubiquitinase (DUB) ubiquitin specific peptidase 18 (USP18). Using reverse-phase protein arrays (RPPAs), this study reports that engineered loss of the DUB USP18 destabilized the tumor suppressor protein phosphatase and tensin homologue (PTEN) in both murine and human lung cancer cell lines. In contrast, engineered gain of USP18 expression in these same lung cancer cell lines stabilized PTEN protein. Using the protein synthesis inhibitor cycloheximide (CHX), USP18 knockdown was shown to destabilize PTEN whereas USP18 overexpression stabilized PTEN protein. Interestingly, repression of USP18 decreased cytoplasmic PTEN relative to nuclear PTEN protein levels. We sought to identify mechanisms engaged in this PTEN protein destabilization using immunoprecipitation assays and found ISG15 directly conjugated with PTEN. To confirm translational relevance of this work, USP18 and PTEN immunohistochemical expression were compared in comprehensive lung cancer arrays. There was a significant (P < 0.0001) positive correlation and association between PTEN and USP18 protein expression profiles in human lung cancers. Taken together, this study identified PTEN as a previously unrecognized substrate of the ISGylation post-translational modification pathway. The deconjugase USP18 serves as a novel regulator of PTEN stability. This indicates inhibition of ISGylation is therapeutically relevant in cancers.


Subject(s)
Cytokines/metabolism , Endopeptidases/metabolism , PTEN Phosphohydrolase/metabolism , Ubiquitins/metabolism , Animals , Cell Line, Tumor , Cytokines/genetics , Endopeptidases/genetics , Gene Knockout Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , PTEN Phosphohydrolase/genetics , Protein Processing, Post-Translational , Protein Stability , Ubiquitin Thiolesterase , Ubiquitination , Ubiquitins/genetics
17.
Sci Rep ; 5: 17806, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26632449

ABSTRACT

How several signaling pathways are coordinated to generate complex organs through regulation of tissue growth and patterning is a fundamental question in developmental biology. The larval trachea of Drosophila is composed of differentiated functional cells and groups of imaginal tracheoblasts that build the adult trachea during metamorphosis. Air sac primordium cells (ASP) are tracheal imaginal cells that form the dorsal air sacs that supply oxygen to the flight muscles of the Drosophila adult. The ASP emerges from the tracheal branch that connects to the wing disc by the activation of both Bnl-FGF/Btl and EGFR signaling pathways. Together, these pathways promote cell migration and proliferation. In this study we demonstrate that Vein (vn) is the EGF ligand responsible for the activation of the EGFR pathway in the ASP. We also find that the Bnl-FGF/Btl pathway regulates the expression of vn through the transcription factor PointedP2 (PntP2). Furthermore, we show that the FGF target gene escargot (esg) attenuates EGFR signaling at the tip cells of the developing ASP, reducing their mitotic rate to allow proper migration. Altogether, our results reveal a link between Bnl-FGF/Btl and EGFR signaling and provide novel insight into how the crosstalk of these pathways regulates migration and growth.


Subject(s)
Air Sacs/growth & development , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Fibroblast Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neuregulins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Air Sacs/cytology , Air Sacs/metabolism , Animals , Animals, Genetically Modified , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Larva , Male , Nerve Tissue Proteins/genetics , Neuregulins/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Signal Transduction/genetics , Transcription Factors/genetics
18.
Cancer Res ; 74(4): 1190-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24371225

ABSTRACT

KRAS phosphorylation has been reported recently to modulate the activity of mutant KRAS protein in vitro. In this study, we defined S181 as a specific phosphorylation site required to license the oncogenic function of mutant KRAS in vivo. The phosphomutant S181A failed to induce tumors in mice, whereas the phosphomimetic mutant S181D exhibited an enhanced tumor formation capacity, compared with the wild-type KRAS protein. Reduced growth of tumors composed of cells expressing the nonphosphorylatable KRAS S181A mutant was correlated with increased apoptosis. Conversely, increased growth of tumors composed of cells expressing the phosphomimetic KRAS S181D mutant was correlated with increased activation of AKT and ERK, two major downstream effectors of KRAS. Pharmacologic treatment with PKC inhibitors impaired tumor growth associated with reduced levels of phosphorylated KRAS and reduced effector activation. In a panel of human tumor cell lines expressing various KRAS isoforms, we showed that KRAS phosphorylation was essential for survival and tumorigenic activity. Furthermore, we identified phosphorylated KRAS in a panel of primary human pancreatic tumors. Taken together, our findings establish that KRAS requires S181 phosphorylation to manifest its oncogenic properties, implying that its inhibition represents a relevant target to attack KRAS-driven tumors.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Serine/metabolism , ras Proteins/metabolism , Animals , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Humans , Mice , Mice, Knockout , Mice, Nude , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics
19.
J Extracell Vesicles ; 22013 Sep 24.
Article in English | MEDLINE | ID: mdl-24223259

ABSTRACT

Extracellular vesicles (EVs), including exosomes, have been widely recognized for their role in intercellular communication of the immune response system. In the past few years, significance has been given to exosomes in the induction and modulation of cell-fate-inducing signalling pathways, such as the Hedgehog (Hh), Wnts, Notch, transforming growth factor (TGF-ß), epidermal growth factor (EGF) and fibroblast growth factor (FGF) pathways, placing them in the wider context of development and also of cancer. These protein families induce signalling cascades responsible for tissue specification, homeostasis and maintenance. Exosomes contribute to cell-fate signal secretion, and vice versa exosome secretion can be induced by these proteins. Interestingly, exosomes can also transfer their mRNA to host cells or modulate the signalling pathways directly by the removal of downstream effector molecules from the cell. Surprisingly, much of what we know about the function of exosomes in cell determination is gathered from pathological transformed cancer cells and wound healing while data about their biogenesis and biology in normal developing and adult tissue lag behind. In this report, we will summarize some of the published literature and point to current advances and questions in this fast-developing topic. In a brief foray, we will also update and shortly discuss their potential in diagnosis and targeted cancer treatment.

SELECTION OF CITATIONS
SEARCH DETAIL