Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35793873

ABSTRACT

BACKGROUND: Despite the prominent role of innate immunity in the antitumor response, little is known about the myeloid composition of human non-small cell lung cancer (NSCLC) with respect to histology and molecular subtype. We used multiplexed quantitative immunofluorescence (QIF) to measure the distribution and clinical significance of major myeloid cell subsets in large retrospective NSCLC collections. METHODS: We established a QIF panel to map major myeloid cell subsets in fixed human NSCLC including 4',6-Diamidino-2-Phenylindole for all cells, pancytokeratin for tumor-epithelial cells, CD68 for M1-like macrophages; and CD11b plus HLA-DR to interrogate mature and immature myeloid cell populations such as myeloid derived suppressor cells (MDSCs). We interrogated 793 NSCLCs represented in four tissue microarray-based cohorts: #1 (Yale, n=379) and #2 (Greece, n=230) with diverse NSCLC subtypes; #3 (Yale, n=138) with molecularly annotated lung adenocarcinomas (ADC); and #4 (Yale, n=46) with patient-matched NSCLC and morphologically-normal lung tissue. We examined associations between marker levels, myeloid cell profiles, clinicopathologic/molecular variables and survival. RESULTS: The levels of CD68+ M1 like macrophages were significantly lower and the fraction of CD11b+/HLA-DR- MDSC-like cells was prominently higher in tumor than in matched non-tumor lung tissues. HLA-DR was consistently higher in myeloid cells from tumors with elevated CD68 expression. Stromal CD11b was significantly higher in squamous cell carcinomas (SCC) than in ADC across the cohorts and EGFR-mutated lung ADCs displayed lower CD11b levels than KRAS-mutant tumors. Increased stromal CD68- and HLA-DR-expressing cells was associated with better survival in ADCs from two independent NSCLC cohorts. In SCC, increased stromal CD11b or HLA-DR expression was associated with a trend towards shorter 5-year survival. CONCLUSIONS: NSCLCs display an unfavorable myeloid immune contexture relative to non-tumor lung and exhibit distinct myeloid-cell profiles across histologies and presence of major oncogenic driver-mutations. Elevated M1-like stromal proinflammatory myeloid cells are prognostic in lung ADC, but not in SCC.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Adenocarcinoma of Lung/pathology , Carcinoma, Squamous Cell/pathology , HLA-DR Antigens/genetics , HLA-DR Antigens/metabolism , Humans , Myeloid Cells , Retrospective Studies
2.
J Immunother Cancer ; 10(6)2022 06.
Article in English | MEDLINE | ID: mdl-35649657

ABSTRACT

BACKGROUND: Tumor infiltrating lymphocytes (TILs) reflect adaptive antitumor immune responses in cancer and are generally associated with favorable prognosis. However, the relationships between TILs subsets and their spatial arrangement with clinical benefit from immune checkpoint inhibitors (ICI) in non-small cell lung cancer (NSCLC) remains less explored. METHODS: We used multiplexed quantitative immunofluorescence panels to determine the association of major TILs subpopulations, CD8+ cytotoxic T cells, CD4+ helper T cells and CD20+ B cells, and T cell exhaustion markers, programmed cell death protein-1 (PD-1),lymphocyte-activation gene 3 (LAG-3) and T cell immunoglobulin mucin-3 (TIM-3) with outcomes in a multi-institutional cohort of baseline tumor samples from 179 patients with NSCLC treated with ICI. The analysis of full-face tumor biopsies including numerous fields of view allowed a detailed spatial analysis and assessment of tumor immune heterogeneity using a multiparametric quadratic entropy metric (Rao's Q Index (RQI)). RESULTS: TILs were preferentially located in the stromal tissue areas surrounding tumor-cell nests and CD8+ T cells were the most abundant subset. Higher density of stromal CD8+ cytotoxic T cells was significantly associated with longer survival, and this effect was more prominent in programmed death ligand-1 (PD-L1) positive cases. The role of baseline T cell infiltration to stratify PD-L1 expressing cases was confirmed measuring the T cell receptor-burden in an independent NSCLC cohort studied with whole-exome DNA sequencing. High levels of LAG-3 on T cells or elevated RQI heterogeneity index were associated with worse survival in the cohort. CONCLUSION: Baseline T cell density and T cell exhaustion marker expression can stratify outcomes in PD-L1 positive patients with NSCLC treated with ICI. Spatial immune heterogeneity can be measured using the RQI and is associated with survival in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Lung Neoplasms/genetics , Lymphocytes, Tumor-Infiltrating , Programmed Cell Death 1 Receptor/metabolism
3.
Clin Cancer Res ; 27(10): 2837-2847, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33602682

ABSTRACT

PURPOSE: To analyze the distribution, associated immune contexture, and clinical significance of human leukocyte antigen (HLA) class-I and HLA class-II subunits in non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Using spatially resolved and quantitative multiplexed immunofluorescence we studied the tumor/stromal tissue distribution, cancer cell-specific defects, and clinicopathologic/survival associations of ß2 microglobulin (ß2M), HLA-A, and HLA-B,-C heavy chains, as well as HLA class-II ß chain in >700 immunotherapy-naïve NSCLCs from four independent cohorts. Genomic analysis of HLA genes in NSCLC was performed using two publicly available cohorts. RESULTS: Cancer cell-specific downregulation of HLA markers was identified in 30.4% of cases. ß2M was downregulated in 9.8% (70/714), HLA-A in 9% (65/722), HLA-B,-C in 12.1% (87/719), and HLA class-II in 17.7% (127/717) of evaluable samples. Concurrent downregulation of ß2M, HLA-B,-C, and HLA class-II was commonly identified. Deleterious mutations in HLA genes were detected in <5% of lung malignancies. Tumors with cancer cell-specific ß2M downregulation displayed reduced T cells and increased natural killer (NK)-cell infiltration. Samples with cancer cell HLA-A downregulation displayed modest increase in CD8+ T cells and NK-cell infiltration. Samples with cancer cell-selective HLA-B,-C or HLA class-II downregulation displayed reduced T cells and NK-cell infiltration. There was limited association of the markers with clinicopathologic variables and KRAS/EGFR mutations. Cancer cell-selective downregulation of the HLA subunits was associated with shorter overall survival. CONCLUSIONS: Our results reveal frequent and differential defects in HLA class-I and HLA class-II protein subunit expression in immunotherapy-naïve NSCLCs associated with distinct tumor microenvironment composition and patient survival.


Subject(s)
Alleles , Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class I/genetics , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/mortality , Computational Biology/methods , DNA Mutational Analysis , Fluorescent Antibody Technique/methods , Fluorescent Antibody Technique/standards , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/mortality , Mutation , Prognosis
4.
Clin Cancer Res ; 25(15): 4663-4673, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31053602

ABSTRACT

PURPOSE: To determine the tumor tissue/cell distribution, functional associations, and clinical significance of PD-1, LAG-3, and TIM-3 protein expression in human non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Using multiplexed quantitative immunofluorescence, we performed localized measurements of CD3, PD-1, LAG-3, and TIM-3 protein in >800 clinically annotated NSCLCs from three independent cohorts represented in tissue microarrays. Associations between the marker's expression and major genomic alterations were studied in The Cancer Genome Atlas NSCLC dataset. Using mass cytometry (CyTOF) analysis of leukocytes collected from 20 resected NSCLCs, we determined the levels, coexpression, and functional profile of PD-1, LAG-3, and TIM-3 expressing immune cells. Finally, we measured the markers in baseline samples from 90 patients with advanced NSCLC treated with PD-1 axis blockers and known response to treatment. RESULTS: PD-1, LAG-3, and TIM-3 were detected in tumor-infiltrating lymphocytes (TIL) from 55%, 41.5%, and 25.3% of NSCLC cases, respectively. These markers showed a prominent association with each other and limited association with major clinicopathologic variables and survival in patients not receiving immunotherapy. Expression of the markers was lower in EGFR-mutated adenocarcinomas and displayed limited association with tumor mutational burden. In single-cell CyTOF analysis, PD-1 and LAG-3 were predominantly localized on T-cell subsets/NKT cells, whereas TIM-3 expression was higher in NK cells and macrophages. Coexpression of PD-1, LAG-3, and TIM-3 was associated with prominent T-cell activation (CD69/CD137), effector function (Granzyme-B), and proliferation (Ki-67), but also with elevated levels of proapoptotic markers (FAS/BIM). LAG-3 and TIM-3 were present in TIL subsets lacking PD-1 expression and showed a distinct functional profile. In baseline samples from 90 patients with advanced NSCLC treated with PD-1 axis blockers, elevated LAG-3 was significantly associated with shorter progression-free survival. CONCLUSIONS: PD-1, LAG-3, and TIM-3 have distinct tissue/cell distribution, functional implications, and genomic correlates in human NSCLC. Expression of these immune inhibitory receptors in TILs is associated with prominent activation, but also with a proapoptotic T-cell phenotype. Elevated LAG-3 expression is associated with insensitivity to PD-1 axis blockade, suggesting independence of these immune evasion pathways.


Subject(s)
Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Hepatitis A Virus Cellular Receptor 2/metabolism , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/metabolism , Single-Cell Analysis/methods , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocyte Activation/immunology , Prognosis , Retrospective Studies , Survival Rate , Lymphocyte Activation Gene 3 Protein
5.
PLoS One ; 14(4): e0204387, 2019.
Article in English | MEDLINE | ID: mdl-30995246

ABSTRACT

Recent high-throughput-sequencing of cancer genomes has identified oncogenic mutations in the B-Raf genetic locus as one of the critical events in melanomagenesis. B-Raf encodes a serine/threonine kinase that regulates the MAPK/ERK kinase (MEK) and extracellular signal-regulated kinase (ERK) protein kinase cascade. In normal cells, the activity of B-Raf is tightly regulated and is required for cell growth and survival. B-Raf gain-of-function mutations in melanoma frequently lead to unrestrained growth, enhanced cell invasion and increased viability of cancer cells. Although it is clear that the invasive phenotypes of B-Raf mutated melanoma cells are stringently dependent on B-Raf-MEK-ERK activation, the downstream effector targets that are required for oncogenic B-Raf-mediated melanomagenesis are not well defined. miRNAs have regulatory functions towards the expression of genes that are important in carcinogenesis. We observed that miR-10b expression correlates with the presence of the oncogenic B-Raf (B-RafV600E) mutation in melanoma cells. While expression of miR-10b enhances anchorage-independent growth of B-Raf wild-type melanoma cells, miR-10b silencing decreases B-RafV600E cancer cell invasion in vitro. Importantly, the expression of miR-10b is required for B-RafV600E-mediated anchorage independent growth and invasion of melanoma cells in vitro. Taken together our results suggest that miR-10b is an important mediator of oncogenic B-RafV600E activity in melanoma.


Subject(s)
Gain of Function Mutation , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , MicroRNAs/biosynthesis , Proto-Oncogene Proteins B-raf/metabolism , RNA, Neoplasm/biosynthesis , Amino Acid Substitution , Cell Line, Tumor , Cell Survival , Humans , MAP Kinase Signaling System , Melanoma/genetics , Melanoma/pathology , MicroRNAs/genetics , Mutation, Missense , Neoplasm Invasiveness , Proto-Oncogene Proteins B-raf/genetics , RNA, Neoplasm/genetics
6.
Cell ; 176(1-2): 334-347.e12, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30580966

ABSTRACT

Lymphocyte-activation gene 3 (LAG-3) is an immune inhibitory receptor, with major histocompatibility complex class II (MHC-II) as a canonical ligand. However, it remains controversial whether MHC-II is solely responsible for the inhibitory function of LAG-3. Here, we demonstrate that fibrinogen-like protein 1 (FGL1), a liver-secreted protein, is a major LAG-3 functional ligand independent from MHC-II. FGL1 inhibits antigen-specific T cell activation, and ablation of FGL1 in mice promotes T cell immunity. Blockade of the FGL1-LAG-3 interaction by monoclonal antibodies stimulates tumor immunity and is therapeutic against established mouse tumors in a receptor-ligand inter-dependent manner. FGL1 is highly produced by human cancer cells, and elevated FGL1 in the plasma of cancer patients is associated with a poor prognosis and resistance to anti-PD-1/B7-H1 therapy. Our findings reveal an immune evasion mechanism and have implications for the design of cancer immunotherapy.


Subject(s)
Antigens, CD/metabolism , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Animals , Antigens, CD/immunology , Cell Line , Fibrinogen/immunology , Fibrinogen/metabolism , Genes, MHC Class II/genetics , Genes, MHC Class II/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Immunotherapy , Ligands , Liver/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Lymphocyte Activation Gene 3 Protein
7.
Clin Cancer Res ; 24(7): 1562-1573, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29203588

ABSTRACT

Purpose: Determine the localized expression pattern and clinical significance of VISTA/PD-1H in human non-small cell lung cancer (NSCLC).Experimental Design: Using multiplex quantitative immunofluorescence (QIF), we performed localized measurements of VISTA, PD-1, and PD-L1 protein in 758 stage I-IV NSCLCs from 3 independent cohorts represented in tissue microarray format. The targets were selectively measured in cytokeratin+ tumor epithelial cells, CD3+ T cells, CD4+ T-helper cells, CD8+ cytotoxic T cells, CD20+ B lymphocytes and CD68+ tumor-associated macrophages. We determined the association between the targets, clinicopathological/molecular variables and survival. Genomic analyses of lung cancer cases from TCGA were also performed.Results: VISTA protein was detected in 99% of NSCLCs with a predominant membranous/cytoplasmic staining pattern. Expression in tumor and stromal cells was seen in 21% and 98% of cases, respectively. The levels of VISTA were positively associated with PD-L1, PD-1, CD8+ T cells and CD68+ macrophages. VISTA expression was higher in T-lymphocytes than in macrophages; and in cytotoxic T cells than in T-helper cells. Elevated VISTA was associated with absence of EGFR mutations and lower mutational burden in lung adenocarcinomas. Presence of VISTA in tumor compartment predicted longer 5-year survival.Conclusions: VISTA is frequently expressed in human NSCLC and shows association with increased tumor-infiltrating lymphocytes, PD-1 axis markers, specific genomic alterations and outcome. These results support the immunomodulatory role of VISTA in human NSCLC and suggests its potential as therapeutic target. Clin Cancer Res; 24(7); 1562-73. ©2017 AACR.


Subject(s)
B7 Antigens/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Evaluation Studies as Topic , Female , Gene Expression Regulation, Neoplastic/immunology , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunologic Factors/metabolism , Immunotherapy/methods , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Male , Mutation/immunology , Mutation/physiology , Programmed Cell Death 1 Receptor/metabolism , Retrospective Studies
8.
Cancer Discov ; 7(12): 1420-1435, 2017 12.
Article in English | MEDLINE | ID: mdl-29025772

ABSTRACT

Mechanisms of acquired resistance to immune checkpoint inhibitors (ICI) are poorly understood. We leveraged a collection of 14 ICI-resistant lung cancer samples to investigate whether alterations in genes encoding HLA Class I antigen processing and presentation machinery (APM) components or interferon signaling play a role in acquired resistance to PD-1 or PD-L1 antagonistic antibodies. Recurrent mutations or copy-number changes were not detected in our cohort. In one case, we found acquired homozygous loss of B2M that caused lack of cell-surface HLA Class I expression in the tumor and a matched patient-derived xenograft (PDX). Downregulation of B2M was also found in two additional PDXs established from ICI-resistant tumors. CRISPR-mediated knockout of B2m in an immunocompetent lung cancer mouse model conferred resistance to PD-1 blockade in vivo, proving its role in resistance to ICIs. These results indicate that HLA Class I APM disruption can mediate escape from ICIs in lung cancer.Significance: As programmed death 1 axis inhibitors are becoming more established in standard treatment algorithms for diverse malignancies, acquired resistance to these therapies is increasingly being encountered. Here, we found that defective antigen processing and presentation can serve as a mechanism of such resistance in lung cancer. Cancer Discov; 7(12); 1420-35. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1355.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/metabolism , Lung Neoplasms/genetics , Humans , Lung Neoplasms/metabolism , Signal Transduction
9.
Oncotarget ; 7(18): 26925, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27532884

ABSTRACT

Present: Due to an error made by the authors while submitting a revision, Dr. Tuan Zea Tan was omitted from the list of authors.Corrected: Correct author list can be found below. Authors sincerely apologize for this oversight. Ila Datar1, Xiaoliang Qiu1, Hong Zhi Ma1, Miranda Yeung1, Shweta Aras1, Ivana de la Serna1, Fahd Al-Mulla2, Tuan Zea Tan3, Jean Paul Thiery3, Robert Trumbly1, Xuan Fan4, Hongjuan Cui4 and Kam C. Yeung1 1 Department of Biochemistry and Cancer Biology, University of Toledo, College of Medicine, Health Science Campus, Toledo, OH, USA 2 Kuwait University, Faculty of Medicine. P.O. Box 24923, Safat, Kuwait 3 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 4 State Key Laboratory Of Silkworm Genome Biology, Chongqing, China Original article: Oncotarget. 2015; 6(36): 39050-61. doi: 10.18632/oncotarget.5176.

10.
Clin Cancer Res ; 22(14): 3422-4, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27076625

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a complex process involved in metastasis. Immune evasion is required for tumor progression and is characterized by an ineffective antitumor immune response and upregulation of immune-suppressive signals. The coexistence of EMT and adaptive immune evasion opens the possibility of a mechanistic link between these processes. Clin Cancer Res; 22(14); 3422-4. ©2016 AACRSee related article by Lou et al., p. 3630.


Subject(s)
Epithelial-Mesenchymal Transition/immunology , Epithelial-Mesenchymal Transition/physiology , Immune Evasion/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Disease Progression , Gene Expression Regulation, Neoplastic/immunology , Humans , Lung/immunology , Lung/pathology , Up-Regulation/immunology
11.
Oncotarget ; 6(36): 39050-61, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26375811

ABSTRACT

Accumulating evidence suggests that presence of macrophages in the tumor microenvironment add to the invasive and tumor-promoting hallmarks of cancer cells by secreting angiogenic and growth factors. RKIP is a known metastasis suppressor and interferes with several steps of metastasis. However, the mechanistic underpinnings of its function as a broad metastasis suppressor remain poorly understood. Here, we establish a novel pathway for RKIP regulation of metastasis inhibition through the negative regulation of RANTES/CCL5 thereby limiting tumor macrophage infiltration and inhibition of angiogenesis. Using a combination of loss- and gain-of- function approaches, we show that RKIP hinders breast cancer cell invasion by inhibiting expression of the CC chemokine CCL5 in vitro. We also show that the expression levels of RKIP and CCL5 are inversely correlated among clinical human breast cancer samples. Using a mouse allograft breast cancer transplantation model, we highlight that ectopic expression of RKIP significantly decreases tumor vasculature, macrophage infiltration and lung metastases. Mechanistically, we demonstrate that the inhibition of the CCL5 expression is the cause of the observed effects resulting from RKIP expression. Taken together, our results underscore the significance of RKIP as important negative regulator of tumor microenvironment.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokine CCL5/biosynthesis , Macrophages/pathology , Phosphatidylethanolamine Binding Protein/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Cell Line, Tumor , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphatidylethanolamine Binding Protein/genetics , Tumor Microenvironment
12.
PLoS One ; 10(8): e0134494, 2015.
Article in English | MEDLINE | ID: mdl-26308852

ABSTRACT

Raf Kinase Inhibitory Protein or RKIP was initially identified as a Raf-1 binding protein using the yeast 2-hybrid screen. RKIP inhibits the activation phosphorylation of MEK by Raf-1 by competitively inhibiting the binding of MEK to Raf-1 and thus exerting an inhibitory effect on the Raf-MEK-Erk pathway. RKIP has been identified as a metastasis suppressor gene. Expression of RKIP is low in cancer metastases. Although primary tumor growth remains unaffected, re- expression of RKIP inhibits cancer metastasis. Mechanistically, RKIP constrains metastasis by inhibiting angiogenesis, local invasion, intravasation, and colonization. The molecular mechanism of how RKIP inhibits these individual steps remains undefined. In our present study, using an unbiased PCR based screening and by analyzing DNA microarray expression datasets we observe that the expression of multiple metalloproteases (MMPs) including MMP1, MMP3, MMP10 and MMP13 are negatively correlated with RKIP expression in breast cancer cell lines and clinical samples. Since expression of MMPs by cancer cells is important for cancer metastasis, we hypothesize that RKIP may mediate suppression of breast cancer metastasis by inhibiting multiple MMPs. We show that the expression signature of RKIP and MMPs is better at predicting high metastatic risk than the individual gene. Using a combination of loss- and gain-of-function approaches, we find that MMP13 is the cause of RKIP-mediated inhibition of local cancer invasion. Interestingly expression of MMP13 alone is not sufficient to reverse the inhibition of breast cancer cell metastasis to the lung due to the expression of RKIP. We find that RKIP negatively regulates MMP13 through the Erk2 signaling pathway and the repression of MMP13 by RKIP is transcription factor AP-1 independent. Together, our findings indicate that RKIP inhibits cancer cell invasion, in part, via MMP13 inhibition. These data also implicate RKIP in the regulation of MMP transcription, suggesting a potential mechanism by which RKIP inhibits tumor progression and metastasis.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Matrix Metalloproteinase 13/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Transcriptional Activation , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction
13.
Crit Rev Oncog ; 19(6): 417-30, 2014.
Article in English | MEDLINE | ID: mdl-25597352

ABSTRACT

Raf kinase inhibitory protein (RKIP) is known to modulate key signaling cascades and regulate normal physiological processes such as cellular proliferation, differentiation, and apoptosis. The expression of RKIP is found to be downregulated in several cancer metastases and the repressed RKIP expression can be reactivated on treatment with chemotherapeutic agents. RKIP is a proven tumor metastasis suppressor gene and investigating the mechanisms of transcriptional regulation of RKIP is therefore of immense clinical importance. In this review, we discuss the basal expression of RKIP in various tissues and the genetic aspects of the RKIP chromosomal locus including the structure of the RKIP promoter as well as gene regulatory elements such as enhancers. We also review the genetic and epigenetic modulation of RKIP transcription through EZH2, a component of the polycomb repressive complex 2 (PRC2) and sequence specific transcription factors (TFs) BACH1 and Snail. Emerging experimental evidence supports a unifying model in which both these TFs repress RKIP transcription in cancers by recruiting the EZH2 containing repressive complex to the proximal RKIP promoter. Finally, we review the known mechanisms employed by different types of chemotherapeutic agents to activate RKIP expression in cancer cells.


Subject(s)
Epigenesis, Genetic , Phosphatidylethanolamine Binding Protein/genetics , Animals , Enhancer Elements, Genetic , Gene Expression Regulation , Humans , Neoplasms/genetics , Neoplasms/pathology , Transcription, Genetic
14.
PLoS One ; 8(7): e69037, 2013.
Article in English | MEDLINE | ID: mdl-23874858

ABSTRACT

SOX10 is a Sry-related high mobility (HMG)-box transcriptional regulator that promotes differentiation of neural crest precursors into Schwann cells, oligodendrocytes, and melanocytes. Myelin, formed by Schwann cells in the peripheral nervous system, is essential for propagation of nerve impulses. SWI/SNF complexes are ATP dependent chromatin remodeling enzymes that are critical for cellular differentiation. It was recently demonstrated that the BRG1 subunit of SWI/SNF complexes activates SOX10 expression and also interacts with SOX10 to activate expression of OCT6 and KROX20, two transcriptional regulators of Schwann cell differentiation. To determine the requirement for SWI/SNF enzymes in the regulation of genes that encode components of myelin, which are downstream of these transcriptional regulators, we introduced SOX10 into fibroblasts that inducibly express dominant negative versions of the SWI/SNF ATPases, BRM or BRG1. Dominant negative BRM and BRG1 have mutations in the ATP binding site and inhibit gene activation events that require SWI/SNF function. Ectopic expression of SOX10 in cells derived from NIH 3T3 fibroblasts led to the activation of the endogenous Schwann cell specific gene, myelin protein zero (MPZ) and the gene that encodes myelin basic protein (MBP). Thus, SOX10 reprogrammed these cells into myelin gene expressing cells. Ectopic expression of KROX20 was not sufficient for activation of these myelin genes. However, KROX20 together with SOX10 synergistically activated MPZ and MBP expression. Dominant negative BRM and BRG1 abrogated SOX10 mediated activation of MPZ and MBP and synergistic activation of these genes by SOX10 and KROX20. SOX10 was required to recruit BRG1 to the MPZ locus. Similarly, in immortalized Schwann cells, BRG1 recruitment to SOX10 binding sites at the MPZ locus was dependent on SOX10 and expression of dominant negative BRG1 inhibited expression of MPZ and MBP in these cells. Thus, SWI/SNF enzymes cooperate with SOX10 to directly activate genes that encode components of peripheral myelin.


Subject(s)
Myelin Sheath/metabolism , SOXE Transcription Factors/metabolism , Animals , Cell Line , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone , DNA Helicases/genetics , DNA Helicases/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Flow Cytometry , Immunoblotting , Mice , Myelin Sheath/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Real-Time Polymerase Chain Reaction , SOXE Transcription Factors/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Int J Cell Biol ; 2012: 913242, 2012.
Article in English | MEDLINE | ID: mdl-22611400

ABSTRACT

Recent high-throughput-sequencing of the cancer genome has identified oncogenic mutations in BRaf genetic locus as one of the critical events in melanomagenesis. In normal cells, the activity of BRaf is tightly regulated. Gain-of-function mutations like those identified in melanoma frequently lead to enhanced cell-survival and unrestrained growth. The activating mutation of BRaf will also induce the cells to senesce. However, the mechanism by which the oncogenic BRaf induces the senescent barrier remains poorly defined. microRNAs have regulatory functions toward the expression of genes that are important in carcinogenesis. Here we show that expression of several microRNAs is altered when the oncogenic version of BRaf is introduced in cultured primary melanocytes and these cells undergo premature cellular senescence. These include eight microRNAs whose expression rates are significantly stimulated and three that are repressed. While most of the induced microRNAs have documented negative effects on cell cycle progression, one of the repressed microRNAs has proven oncogenic functions. Ectopic expression of some of these induced microRNAs increased the expression of senescence markers and induced growth arrest and senescence in primary melanocytes. Taken together, our results suggest that the change in microRNA expression rates may play a vital role in senescence induced by the oncogenic BRaf.

SELECTION OF CITATIONS
SEARCH DETAIL