Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Viruses ; 15(2)2023 02 02.
Article in English | MEDLINE | ID: mdl-36851637

ABSTRACT

Epstein-Barr virus (EBV) latency patterns are well defined in EBV-associated epithelial, NK/T-cell, and B-cell malignancies, with links between latency stage and tumorigenesis deciphered in various studies. In vitro studies suggest that the oncogenic activity of EBV in T-cells might be somewhat different from that in EBV-tropic B lymphoid cells, prompting us to study this much less investigated viral gene expression pattern and its regulation in nine EBV+ peripheral T-cell lymphoma (PTCL) biopsies. Using frozen specimens, RT-PCR showed 6/7 cases with a latency II pattern of EBV gene expression. Analyses of EBNA1 promoter usage and CpG methylation status in these six cases showed that only Qp was used, while Cp, Wp, and Fp were all silent. However, the remaining case showed an exceptionally unique latency III type with lytic activation, as evidenced by EBV lytic clonality and confirmed by the full usage of Cp and Qp as well as weakly lytic Fp and Wp, fully unmethylated Cp and marginally unmethylated Wp. Further immunostaining of the eight cases revealed a few focally clustered LMP1+ cells in 7/8 cases, with rare isolated LMP1+ cells detected in another case. Double immunostaining confirmed that the LMP1+ cells were of the T-cell phenotype (CD3+). In 6/8 cases, sporadically scattered Zta+ cells were detected. Double staining of EBER-ISH with T-cell (CD45RO/UCHL1) or B-cell (CD20) markers confirmed that the vast majority of EBER+ cells were of the T-cell phenotype. Predominant type-A EBV variant and LMP1 30-bp deletion variant were present, with both F and f variants detected. In summary, the EBV gene expression pattern in PTCL was found to be mainly of latency II (BART+EBNA1(Qp)+LMP1+LMP2A+BZLF1+), similar to that previously reported in EBV-infected nasopharyngeal epithelial, NK/T-cell, and Hodgkin malignancies; however, fully lytic infection could also be detected in occasional cases. Rare cells with sporadic immediate-early gene expression were commonly detected in PTCL. These findings have implications for the future development of EBV-targeting therapeutics for this cancer.


Subject(s)
Epstein-Barr Virus Infections , Latent Infection , Lymphoma, T-Cell, Peripheral , Humans , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Gene Expression , Herpesvirus 4, Human/genetics , Lymphoma, T-Cell, Peripheral/genetics , Methylation , Promoter Regions, Genetic
3.
Theranostics ; 8(1): 61-77, 2018.
Article in English | MEDLINE | ID: mdl-29290793

ABSTRACT

Rationale: Oncogenic STAT3 signaling activation and 3p22-21.3 locus alteration are common in multiple tumors, especially carcinomas of the nasopharynx, esophagus and lung. Whether these two events are linked remains unclear. Our CpG methylome analysis identified a 3p22.2 gene, DLEC1, as a methylated target in esophageal squamous cell (ESCC), nasopharyngeal (NPC) and lung carcinomas. Thus, we further characterized its epigenetic abnormalities and functions. Methods: CpG methylomes were established by methylated DNA immunoprecipitation. Promoter methylation was analyzed by methylation-specific PCR and bisulfite genomic sequencing. DLEC1 expression and clinical significance were analyzed using TCGA database. DLEC1 functions were analyzed by transfections followed by various cell biology assays. Protein-protein interaction was assessed by docking, Western blot and immunoprecipitation analyses. Results: We defined the DLEC1 promoter within a CpG island and p53-regulated. DLEC1 was frequently downregulated in ESCC, lung and NPC cell lines and primary tumors, but was readily expressed in normal tissues and immortalized normal epithelial cells, with mutations rarely detected. DLEC1 methylation was frequently detected in ESCC tumors and correlated with lymph node metastasis, tumor recurrence and progression, with DLEC1 as the most frequently methylated among the established 3p22.2 tumor suppressors (RASSF1A, PLCD1 and ZMYND10/BLU). DLEC1 inhibits carcinoma cell growth through inducing cell cycle arrest and apoptosis, and also suppresses cell metastasis by reversing epithelial-mesenchymal transition (EMT) and cell stemness. Moreover, DLEC1 represses oncogenic signaling including JAK/STAT3, MAPK/ERK, Wnt/ß-catenin and AKT pathways in multiple carcinoma types. Particularly, DLEC1 inhibits IL-6-induced STAT3 phosphorylation in a dose-dependent manner. DLEC1 contains three YXXQ motifs and forms a protein complex with STAT3 via protein docking, which blocks STAT3-JAK2 interaction and STAT3 phosphorylation. IL-6 stimulation enhances the binding of DLEC1 with STAT3, which diminishes their interaction with JAK2 and further leads to decreased STAT3 phosphorylation. The YXXQ motifs of DLEC1 are crucial for its inhibition of STAT3 phosphorylation, and disruption of these motifs restores STAT3 phosphorylation through abolishing DLEC1 binding to STAT3. Conclusions: Our study demonstrates, for the first time, predominant epigenetic silencing of DLEC1 in ESCC, and a novel mechanistic link of epigenetic DLEC1 disruption with oncogenic STAT3 signaling in multiple carcinomas.


Subject(s)
Epigenomics/methods , Esophageal Neoplasms/genetics , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Aged , Alternative Splicing/genetics , Alternative Splicing/physiology , CpG Islands/genetics , DNA Methylation/genetics , DNA Methylation/physiology , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Middle Aged , Phosphorylation , STAT3 Transcription Factor/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
4.
Cell ; 171(2): 481-494.e15, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985567

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is the most common form of blood cancer and is characterized by a striking degree of genetic and clinical heterogeneity. This heterogeneity poses a major barrier to understanding the genetic basis of the disease and its response to therapy. Here, we performed an integrative analysis of whole-exome sequencing and transcriptome sequencing in a cohort of 1,001 DLBCL patients to comprehensively define the landscape of 150 genetic drivers of the disease. We characterized the functional impact of these genes using an unbiased CRISPR screen of DLBCL cell lines to define oncogenes that promote cell growth. A prognostic model comprising these genetic alterations outperformed current established methods: cell of origin, the International Prognostic Index comprising clinical variables, and dual MYC and BCL2 expression. These results comprehensively define the genetic drivers and their functional roles in DLBCL to identify new therapeutic opportunities in the disease.


Subject(s)
CRISPR-Cas Systems , Gene Expression Profiling , Lymphoma, Large B-Cell, Diffuse/genetics , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cells, Cultured , Exome , Female , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Male , Rituximab/administration & dosage
5.
J Exp Med ; 214(5): 1371-1386, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28424246

ABSTRACT

Enteropathy-associated T cell lymphoma (EATL) is a lethal, and the most common, neoplastic complication of celiac disease. Here, we defined the genetic landscape of EATL through whole-exome sequencing of 69 EATL tumors. SETD2 was the most frequently silenced gene in EATL (32% of cases). The JAK-STAT pathway was the most frequently mutated pathway, with frequent mutations in STAT5B as well as JAK1, JAK3, STAT3, and SOCS1 We also identified mutations in KRAS, TP53, and TERT Type I EATL and type II EATL (monomorphic epitheliotropic intestinal T cell lymphoma) had highly overlapping genetic alterations indicating shared mechanisms underlying their pathogenesis. We modeled the effects of SETD2 loss in vivo by developing a T cell-specific knockout mouse. These mice manifested an expansion of γδ T cells, indicating novel roles for SETD2 in T cell development and lymphomagenesis. Our data render the most comprehensive genetic portrait yet of this uncommon but lethal disease and may inform future classification schemes.


Subject(s)
Enteropathy-Associated T-Cell Lymphoma/physiopathology , Histone-Lysine N-Methyltransferase/physiology , Animals , DNA Copy Number Variations/genetics , Enteropathy-Associated T-Cell Lymphoma/classification , Enteropathy-Associated T-Cell Lymphoma/genetics , Female , Gene Expression Profiling , Gene Silencing , Humans , Male , Mice, Knockout , Middle Aged , Mutation/genetics , Sequence Analysis, DNA , T-Lymphocytes/physiology
6.
Cancer Discov ; 7(4): 369-379, 2017 04.
Article in English | MEDLINE | ID: mdl-28122867

ABSTRACT

Hepatosplenic T-cell lymphoma (HSTL) is a rare and lethal lymphoma; the genetic drivers of this disease are unknown. Through whole-exome sequencing of 68 HSTLs, we define recurrently mutated driver genes and copy-number alterations in the disease. Chromatin-modifying genes, including SETD2, INO80, and ARID1B, were commonly mutated in HSTL, affecting 62% of cases. HSTLs manifest frequent mutations in STAT5B (31%), STAT3 (9%), and PIK3CD (9%), for which there currently exist potential targeted therapies. In addition, we noted less frequent events in EZH2, KRAS, and TP53SETD2 was the most frequently silenced gene in HSTL. We experimentally demonstrated that SETD2 acts as a tumor suppressor gene. In addition, we found that mutations in STAT5B and PIK3CD activate critical signaling pathways important to cell survival in HSTL. Our work thus defines the genetic landscape of HSTL and implicates gene mutations linked to HSTL pathogenesis and potential treatment targets.Significance: We report the first systematic application of whole-exome sequencing to define the genetic basis of HSTL, a rare but lethal disease. Our work defines SETD2 as a tumor suppressor gene in HSTL and implicates genes including INO80 and PIK3CD in the disease. Cancer Discov; 7(4); 369-79. ©2017 AACR.See related commentary by Yoshida and Weinstock, p. 352This article is highlighted in the In This Issue feature, p. 339.


Subject(s)
DNA Helicases/genetics , Histone-Lysine N-Methyltransferase/genetics , Liver Neoplasms/genetics , Lymphoma, T-Cell/genetics , Splenic Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , ATPases Associated with Diverse Cellular Activities , Adolescent , Adult , Aged , Base Sequence , Child , Child, Preschool , DNA-Binding Proteins , Enhancer of Zeste Homolog 2 Protein , Exome/genetics , Female , Humans , Liver Neoplasms/complications , Liver Neoplasms/pathology , Lymphoma, T-Cell/complications , Lymphoma, T-Cell/pathology , Male , Middle Aged , Proto-Oncogene Proteins p21(ras) , Splenic Neoplasms/complications , Splenic Neoplasms/pathology , Transcription Factors , Tumor Suppressor Proteins/genetics , Young Adult
7.
Blood ; 125(10): 1589-600, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25612622

ABSTRACT

Nasal-type natural killer/T-cell lymphoma (NKTCL) is an aggressive disease characterized by frequent deletions on 6q, and constitutive activation of signal transducer and activator of transcription 3 (STAT3). Phosphorylation at Tyr705 activates STAT3, inducing dimerization, nuclear translocation, and DNA binding. In this study, we investigated whether receptor-type tyrosine-protein phosphatase κ (PTPRK), the only protein tyrosine phosphatase at 6q that contains a STAT3-specifying motif, negatively regulates STAT3 activation in NKTCL. PTPRK was highly expressed in normal NK cells but was underexpressed in 4 of 5 (80%) NKTCL cell lines and 15 of 27 (55.6%) primary tumors. Significantly, PTPRK protein expression was inversely correlated with nuclear phospho-STAT3(Tyr705) expression in NKTCL cell lines (P = .025) and tumors (P = .040). PTPRK restoration decreased nuclear phospho-STAT3(Tyr705) levels, whereas knockdown of PTPRK increased such levels in NKTCL cells. Phosphatase substrate-trapping mutant assays demonstrated the binding of PTPRK to STAT3, and phosphatase assays showed that PTPRK directly dephosphorylated phospho-STAT3(Tyr705). Restoration of PTPRK inhibited tumor cell growth and reduced the migration and invasion ability of NKTCL cells. Monoallelic deletion and promoter hypermethylation caused underexpression of PTPRK messenger RNA in NKTCL, and methylation of the PTPRK promoter significantly correlated with inferior overall survival (P = .049) in NKTCL patients treated with the steroid-dexamethasone, methotrexate, ifosfamide, l-asparaginase, and etoposide regimen. Altogether, our findings show that PTPRK underexpression leads to STAT3 activation and contributes to NKTCL pathogenesis.


Subject(s)
Lymphoma, Extranodal NK-T-Cell/metabolism , Nose Neoplasms/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , STAT3 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Apoptosis , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , DNA Methylation , DNA Mutational Analysis , Down-Regulation , Female , Gene Deletion , Gene Knockdown Techniques , Humans , Lymphoma, Extranodal NK-T-Cell/genetics , Lymphoma, Extranodal NK-T-Cell/pathology , Male , Middle Aged , Neoplasm Invasiveness , Nose Neoplasms/genetics , Nose Neoplasms/pathology , Phosphorylation , Prognosis , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , STAT3 Transcription Factor/chemistry , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
8.
Proc Natl Acad Sci U S A ; 110(4): 1398-403, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23292937

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common form of lymphoma in adults. The disease exhibits a striking heterogeneity in gene expression profiles and clinical outcomes, but its genetic causes remain to be fully defined. Through whole genome and exome sequencing, we characterized the genetic diversity of DLBCL. In all, we sequenced 73 DLBCL primary tumors (34 with matched normal DNA). Separately, we sequenced the exomes of 21 DLBCL cell lines. We identified 322 DLBCL cancer genes that were recurrently mutated in primary DLBCLs. We identified recurrent mutations implicating a number of known and not previously identified genes and pathways in DLBCL including those related to chromatin modification (ARID1A and MEF2B), NF-κB (CARD11 and TNFAIP3), PI3 kinase (PIK3CD, PIK3R1, and MTOR), B-cell lineage (IRF8, POU2F2, and GNA13), and WNT signaling (WIF1). We also experimentally validated a mutation in PIK3CD, a gene not previously implicated in lymphomas. The patterns of mutation demonstrated a classic long tail distribution with substantial variation of mutated genes from patient to patient and also between published studies. Thus, our study reveals the tremendous genetic heterogeneity that underlies lymphomas and highlights the need for personalized medicine approaches to treating these patients.


Subject(s)
Genetic Heterogeneity , Lymphoma, Large B-Cell, Diffuse/genetics , Adult , Base Sequence , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , DNA Mutational Analysis , DNA, Neoplasm/genetics , Exome , Gene Expression , Genetic Variation , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Models, Molecular , Molecular Sequence Data , Molecular Targeted Therapy , Mutation , Oncogenes , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Protein Conformation , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Sequence Homology, Nucleic Acid , Signal Transduction/genetics
9.
Nat Genet ; 44(12): 1321-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143597

ABSTRACT

Burkitt lymphoma is characterized by deregulation of MYC, but the contribution of other genetic mutations to the disease is largely unknown. Here, we describe the first completely sequenced genome from a Burkitt lymphoma tumor and germline DNA from the same affected individual. We further sequenced the exomes of 59 Burkitt lymphoma tumors and compared them to sequenced exomes from 94 diffuse large B-cell lymphoma (DLBCL) tumors. We identified 70 genes that were recurrently mutated in Burkitt lymphomas, including ID3, GNA13, RET, PIK3R1 and the SWI/SNF genes ARID1A and SMARCA4. Our data implicate a number of genes in cancer for the first time, including CCT6B, SALL3, FTCD and PC. ID3 mutations occurred in 34% of Burkitt lymphomas and not in DLBCLs. We show experimentally that ID3 mutations promote cell cycle progression and proliferation. Our work thus elucidates commonly occurring gene-coding mutations in Burkitt lymphoma and implicates ID3 as a new tumor suppressor gene.


Subject(s)
Burkitt Lymphoma/genetics , Mutation , Ammonia-Lyases/genetics , Base Sequence , Cell Line, Tumor , Chaperonin Containing TCP-1/genetics , DNA Helicases/genetics , DNA-Binding Proteins , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Genes, myc/genetics , Genome, Human , Glutamate Formimidoyltransferase/genetics , Homeodomain Proteins/genetics , Humans , Inhibitor of Differentiation Proteins/genetics , Intracellular Signaling Peptides and Proteins , Lymphoma, Large B-Cell, Diffuse/genetics , Membrane Proteins/genetics , Molecular Sequence Data , Multifunctional Enzymes , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-ret/genetics , Sequence Analysis, DNA , Transcription Factors/genetics , Translocation, Genetic
10.
J Transl Med ; 10: 209, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-23050586

ABSTRACT

BACKGROUND: Inactivaion of tumor suppressor genes (TSGs) by promoter CpG methylation frequently occurs in tumorigenesis, even in the early stages, contributing to the initiation and progression of human cancers. Deleted in lung and esophageal cancer 1 (DLEC1), located at the 3p22-21.3 TSG cluster, has been identified frequently silenced by promoter CpG methylation in multiple carcinomas, however, no study has been performed for lymphomas yet. METHODS: We examined the expression of DLEC1 by semi-quantitative reverse transcription (RT)-PCR, and evaluated the promoter methylation of DLEC1 by methylation-specific PCR (MSP) and bisulfite genomic sequencing (BGS) in common lymphoma cell lines and tumors. RESULTS: Here we report that DLEC1 is readily expressed in normal lymphoid tissues including lymph nodes and PBMCs, but reduced or silenced in 70% (16/23) of non-Hodgkin and Hodgkin lymphoma cell lines, including 2/6 diffuse large B-cell (DLBCL), 1/2 peripheral T cell lymphomas, 5/5 Burkitt, 6/7 Hodgkin and 2/3 nasal killer (NK)/T-cell lymphoma cell lines. Promoter CpG methylation was frequently detected in 80% (20/25) of lymphoma cell lines and correlated with DLEC1 downregulation/silencing. Pharmacologic demethylation reversed DLEC1 expression in lymphoma cell lines along with concomitant promoter demethylation. DLEC1 methylation was also frequently detected in 32 out of 58 (55%) different types of lymphoma tissues, but not in normal lymph nodes. Furthermore, DLEC1 was specifically methylated in the sera of 3/13 (23%) Hodgkin lymphoma patients. CONCLUSIONS: Thus, methylation-mediated silencing of DLEC1 plays an important role in multiple lymphomagenesis, and may serve as a non-invasive tumor marker for lymphoma diagnosis.


Subject(s)
Chromosomes, Human, Pair 3 , Epigenesis, Genetic , Gene Silencing , Hodgkin Disease/genetics , Lymphoma, Non-Hodgkin/genetics , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics , CpG Islands , DNA Methylation , Humans , Reverse Transcriptase Polymerase Chain Reaction
11.
Am J Pathol ; 180(6): 2375-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22538089

ABSTRACT

IL-10-producing CD1d(hi)CD5(+) B cells, also known as B10 cells, have been shown to possess a regulatory function in the inhibition of immune responses, but whether and how B10 cells suppress the development of autoimmune arthritis remain largely unclear. In this study, we detected significantly decreased numbers of IL-10-producing B cells, but increased IL-17-producing CD4(+) T (Th17) cells in both spleen and draining lymph nodes of mice during the acute stage of collagen-induced arthritis (CIA) when compared with adjuvant-treated control mice. On adoptive transfer of in vitro expanded B10 cells, collagen-immunized mice showed a marked delay of arthritis onset with reduced severity of both clinical symptoms and joint damage, accompanied by a substantial reduction in the number of Th17 cells. To determine whether B10 cells directly inhibit the generation of Th17 cells in culture, naive CD4(+) T cells labeled with carboxyfluorescein succinimidyl ester (CFSE) were co-cultured with B10 cells. These B10 cells suppressed Th17 cell differentiation via the reduction of STAT3 phosphorylation and retinoid-related orphan receptor γt (RORγt) expression. Moreover, Th17 cells showed significantly decreased proliferation when co-cultured with B10 cells. Although adoptive transfer of Th17 cells triggered the development of collagen-induced arthritis in IL-17(-/-)DBA/1J mice, co-transfer of B10 cells with Th17 cells profoundly delayed the onset of arthritis. Thus, our findings suggest a novel regulatory role of B10 cells in arthritic progression via the suppression of Th17 cell generation.


Subject(s)
Arthritis, Experimental/prevention & control , B-Lymphocyte Subsets/immunology , Interleukin-10/biosynthesis , Th17 Cells/immunology , Adoptive Transfer/methods , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocyte Subsets/transplantation , Cell Differentiation/immunology , Cells, Cultured , Interleukin-17/biosynthesis , Interleukin-17/deficiency , Lymph Nodes/immunology , Lymphocyte Transfusion/methods , Mice , Mice, Inbred DBA , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phosphorylation/immunology , STAT3 Transcription Factor/metabolism , Spleen/immunology
12.
Med Oncol ; 29(5): 3009-16, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22528514

ABSTRACT

To examine the correlation of survivin (both total and nuclear survivin) with clinicopathological parameters of esophageal squamous cell carcinoma (ESCC) patients. Tumors and non-tumor tissues near the proximal resection margins were resected from ESCC patients undergone esophagectomy. Quantitative polymerase chain reaction (qPCR) was performed to detect survivin mRNA expression level in the 10 paired tumor and adjacent non-tumor tissues. To confirm with the clinical situation, survivin mRNA and protein expression were measured by qPCR and immunoblot, respectively, in 5 ESCC cell lines and a non-neoplastic esophageal epithelial cell line. Immunohistochemistry was employed to reveal the cellular localization of survivin in tumor tissues isolated from the 64 ESCC patients undergone surgery alone. Up-regulation of survivin mRNA and protein was found in 5 ESCC lines (HKESC-1, HKESC-2, HKESC-3, HKESC-4, and SLMT-1) when compared to a non-neoplastic esophageal epithelial cell line NE-1. In particular, HKESC-3, HKESC-4, and SLMT-1 cells demonstrated ~50-fold increase in survivin mRNA. High level of survivin mRNA in tumor tissues when compared to non-tumor tissues was found in 70 % (7 of 10) of clinical cases. The increase in expression ranged from ~twofold to ~16-fold. Immunohistochemistry results showed that survivin was found at the cell nuclei in all specimens examined. Nuclear expression of survivin was inversely associated with the likelihood of developing nodal metastasis (p = 0.021) and significantly associated with early-stage ESCC (p = 0.039). Nuclear survivin could serve as a marker for indicating disease status in ESCC patients.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/metabolism , Cell Nucleus/metabolism , Esophageal Neoplasms/metabolism , Inhibitor of Apoptosis Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Female , Humans , Immunoblotting , Immunohistochemistry , Inhibitor of Apoptosis Proteins/analysis , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Survivin
13.
Cancer Sci ; 103(6): 1145-54, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22364398

ABSTRACT

Emerging evidence has indicated a role of the bone morphogenetic proteins (BMP) in the pathogenesis of certain cancers. The signaling of BMP family members is tightly regulated by their antagonists, including noggin and SOST, which are, in turn, positively regulated by BMP, thereby forming a negative feedback loop. Consequently, the expression of these antagonists should be taken into account in studies on the prognostic significance of BMP. In the present paper, we correlated protein and mRNA expression levels of BMP6, noggin and SOST, alone or in combination, with patient survival in various types of cancer. We found that BMP6 alone was not significantly correlated with esophageal squamous cell carcinoma patient survival. Instead, a high level of inhibitor of differentiation 1, a downstream factor of BMP6, was associated with shorter survival in patients whose tumors stained strongly for BMP6. Knockdown of noggin in esophageal cancer cell line EC109, which expresses BMP6 strongly and SOST weakly, enhanced the non-adherent growth of the cells. Noggin and SOST expression levels, when analyzed alone, were not significantly correlated with patient survival. However, high BMP6 activity, defined by strong BMP6 expression coupled with weak noggin or SOST expression, was significantly associated with shorter survival in esophageal squamous cell carcinoma patients. We further confirmed that BMP6 activity could be used as a prognostic indicator in prostate, bladder and colorectal cancers, using publicly available data on BMP6, noggin and SOST mRNA expression and patient survival. Our results strongly suggest that BMP6, noggin and SOST could be used in combination as a prognostic indicator in cancer progression.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Esophageal Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Proteins/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carrier Proteins/genetics , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Disease Progression , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Female , Genetic Markers/genetics , Humans , Inhibitor of Differentiation Protein 1/metabolism , Male , Middle Aged , Prostatic Neoplasms/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Signal Transduction , Urinary Bladder Neoplasms/metabolism
14.
World J Surg ; 36(1): 90-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21976009

ABSTRACT

UNLABELLED: Esophageal cancer is a deadly cancer with esophageal squamous cell carcinoma (ESCC) as the major type. Until now there has been a lack of reliable prognostic markers for this malignancy. This study aims to investigate the clinical correlation between Forkhead box M1 (FoxM1)and patients' parameters in ESCC. METHODS: Immunohistochemistry was performed to investigate the expression and localization of FoxM1 in 64ESCC tissues and 10 nontumor esophageal tissues randomly selected from 64 patients before these data were used for clinical correlations. RESULTS: Cytoplasmic and nuclear expressions of FoxM1 were found in 63 and 16 of the 64 ESCC tissues, respectively.Low cytoplasmic expression of FoxM1 was correlated with early pathological stage in ESCC (P = 0.018),while patients with nuclear FoxM1 were younger in age than those without nuclear expression (P\0.001).Upregulation of FoxM1 mRNA was found in five ESCCcell lines (HKESC-1, HKESC-2, HKESC-3, HKESC-4,and SLMT-1) when compared to non-neoplastic esophageal squamous cell line NE-1 using quantitative polymerase chain reaction (qPCR). Except for HKESC-3, all studied ESCC cell lines demonstrated a high expression of FoxM1 protein using immunoblot. A high mRNA level of FoxM1 was observed in all of the ESCC tissues examined when compared to their adjacent nontumor tissues using qPCR. CONCLUSION: Cytoplasmic FoxM1 was correlated with pathological stage and might be a biomarker for advanced ESCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/surgery , Case-Control Studies , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Neoplasms/surgery , Esophagectomy , Female , Forkhead Box Protein M1 , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
15.
Med Oncol ; 29(3): 1699-706, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22086736

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is the most common type of esophageal cancer. RON is a transmembrane receptor overexpressed in various cancers; however, the clinical significance of its phosphorylated form (pRON) is not fully deciphered. This report is the first to investigate the expression and clinical significance of pRON in human ESCC. Quantitative polymerase chain reaction revealed an up-regulation of RON mRNA in 70% (7/10) of ESCC tissues when compared to the adjacent nontumor tissues. An overexpression of pRON protein was found in most of the ESCC cell lines studied (4/5) when compared to two non-neoplastic esophageal epithelial cells using immunoblot. In 64 ESCC tissues, pRON was localized at the cell membrane, cytoplasm and nucleus in 15 (23.4%), 63 (98.4%) and 61 (95.3%) cases using immunohistochemistry. Patients having high expression of cytoplasmic pRON significantly associated with shorter median survival when compared to those with low expression (25.41 months vs. 14.43 months), suggesting cytoplasmic pRON as a potential marker for poor prognosis in ESCC patients.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Humans , Immunoblotting , Immunohistochemistry , Kaplan-Meier Estimate , Phosphorylation , Prognosis , Receptor Protein-Tyrosine Kinases/analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Int J Cancer ; 130(1): 83-95, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21387288

ABSTRACT

Suppressive effects of DUSP6 in tumorigenesis and EMT-associated properties were observed. Dual-specificity phosphatase (DUSP6) is a MAP kinase phosphatase (MKP) negatively regulating the activity of ERK, one of the major molecular switches in the MAPK signaling cascade propagating the signaling responses during malignancies. The impact of DUSP6 in EMT and its contribution to tumor dissemination has not yet been characterized. Due to differences in tumor microenvironments affecting cell signaling during cancer progression, DUSP6 may play varying roles in tumor development. We sought to examine the potential role of DUSP6-mediated tumorigenesis and EMT-associated properties in two aerodigestive tract cancers, namely, esophageal squamous cell carcinoma (ESCC) and nasopharyngeal carcinoma (NPC). Significant loss of DUSP6 was observed in 100% of 11 ESCC cell lines and 71% of seven NPC cell lines. DUSP6 expression was down-regulated in 40% of 30 ESCC tumor tissues and 75% of 20 NPC tumor tissues compared to their respective normal counterparts. Suppressive effects of DUSP6 in tumor formation and cancer cell mobility are seen in in vivo tumorigenicity assay and in vitro colony formation, three-dimensional Matrigel culture, cell migration and invasion chamber tests. Notably, overexpression of DUSP6 impairs EMT-associated properties. Furthermore, tissue microarray analysis reveals a clinical association of DUSP6 expression with better patient survival. Taken together, our study provides a novel insight into understanding the functional impact of DUSP6 in tumorigenesis and metastasis of ESCC and NPC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Movement , Dual Specificity Phosphatase 6/metabolism , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/pathology , Nasopharyngeal Neoplasms/pathology , Animals , Blotting, Western , Carcinoma , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , DNA Methylation , Dual Specificity Phosphatase 6/genetics , Epigenomics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Neoplasm Invasiveness , Neoplasm Staging , Phenotype , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Rate , Tissue Array Analysis
17.
Cancer Lett ; 316(1): 39-45, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22154084

ABSTRACT

The group 2 LIM domain protein, Cysteine-rich intestinal protein 2 (CRIP2) was found to play an important role in esophageal squamous cell carcinoma (ESCC) tumorigenesis. Subcellular fractionation studies show that CRIP2 is expressed in the nucleus. Real-time quantitative PCR shows CRIP2 expression is down-regulated in ESCC tissues and cell lines. Functional studies reveal that CRIP2 reduces colony formation, growth, and invasion abilities. Furthermore, over-expression of CRIP2 induces apoptosis through induction of active caspases 3 and 9 proteins. In conclusion, this study shows CRIP2 plays an important role in the development of ESCC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/genetics , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Growth Processes/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Down-Regulation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Real-Time Polymerase Chain Reaction/methods , Up-Regulation
18.
Proc Natl Acad Sci U S A ; 108(50): 20119-24, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22143801

ABSTRACT

Natural killer cell lymphoma (NKCL) constitutes a rare and aggressive form of non-Hodgkin lymphoma, and there is little insight into its pathogenesis. Here we show that PRDM1 is a tumor suppressor gene in NKCLs that is inactivated by a combination of monoallelic deletion and promoter CpG island hypermethylation. We observed monoallelic deletion of PRDM1 loci in 8 of 18 (44%) NKCL cases. The other allele showed significant promoter methylation in 12 of 17 (71%) cases. In support of its role as a tumor suppressor gene, the reconstitution of PRDM1 in PRDM1-null NK cell lines led to G2/M cell cycle arrest, increased apoptosis, and a strong negative selection pressure with progressive elimination of PRDM1-expressing cells, which was enhanced when IL-2 concentration is limiting. We observed a progressive increase in PRDM1 expression--in particular, PRDM1α--in normal NK cells in response to IL-2 and in normal NK cells activated with an engineered NK cell target, K562-Cl9-mb21, suggesting its role in NK cell homeostasis. In support of this role, knockdown of PRDM1 by shRNA in normal NK cells resulted in the positive selection of these cells. We identified MYC and 4-1BBL as targets of PRDM1 in NK cells. Disruption of homeostatic control by PRDM1 may be an important pathogenetic mechanism for NKCL.


Subject(s)
Killer Cells, Natural/pathology , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Apoptosis/drug effects , Biopsy , Cell Division/drug effects , Cell Division/genetics , Culture Media/pharmacology , DNA Copy Number Variations/drug effects , DNA Copy Number Variations/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , DNA Mutational Analysis , G2 Phase/drug effects , G2 Phase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Interleukin-2/metabolism , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Promoter Regions, Genetic/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Time Factors , Transduction, Genetic , Tumor Suppressor Proteins/metabolism
19.
PLoS One ; 6(11): e27346, 2011.
Article in English | MEDLINE | ID: mdl-22087297

ABSTRACT

BACKGROUND: PRDM (PRDI-BF1 and RIZ domain containing) proteins are zinc finger proteins involved in multiple cellular regulations by acting as epigenetic modifiers. We studied a recently identified PRDM member PRDM5 for its epigenetic abnormality and tumor suppressive functions in multiple tumorigeneses. METHODOLOGY/PRINCIPAL FINDINGS: Semi-quantitative RT-PCR showed that PRDM5 was broadly expressed in human normal tissues, but frequently silenced or downregulated in multiple carcinoma cell lines due to promoter CpG methylation, including 80% (4/5) nasopharyngeal, 44% (8/18) esophageal, 76% (13/17) gastric, 50% (2/4) cervical, and 25% (3/12) hepatocellular carcinoma cell lines, but not in any immortalized normal epithelial cell lines. PRDM5 expression could be restored by 5-aza-2'-deoxycytidine demethylation treatment in silenced cell lines. PRDM5 methylation was frequently detected by methylation-specific PCR (MSP) in multiple primary tumors, including 93% (43/46) nasopharyngeal, 58% (25/43) esophageal, 88% (37/42) gastric and 63% (29/46) hepatocellular tumors. PRDM5 was further found a stress-responsive gene, but its response was impaired when the promoter was methylated. Ectopic PRDM5 expression significantly inhibited tumor cell clonogenicity, accompanied by the inhibition of TCF/ß-catenin-dependent transcription and downregulation of CDK4, TWIST1 and MDM2 oncogenes, while knocking down of PRDM5 expression lead to increased cell proliferation. ChIP assay showed that PRDM5 bound to its target gene promoters and suppressed their transcription. An inverse correlation between the expression of PRDM5 and activated ß-catenin was also observed in cell lines. CONCLUSIONS/SIGNIFICANCE: PRDM5 functions as a tumor suppressor at least partially through antagonizing aberrant WNT/ß-catenin signaling and oncogene expression. Frequent epigenetic silencing of PRDM5 is involved in multiple tumorigeneses, which could serve as a tumor biomarker.


Subject(s)
DNA-Binding Proteins/physiology , Epigenesis, Genetic , Gene Silencing , Neoplasms/genetics , Transcription Factors/physiology , Wnt Signaling Pathway , Humans , Tumor Suppressor Proteins/metabolism , beta Catenin/metabolism
20.
Am J Pathol ; 179(2): 992-1003, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21689625

ABSTRACT

The transcription factors Pea3, Erm, and Er81 can promote cancer initiation and progression in various types of solid tumors. However, their role in esophageal squamous cell carcinoma (ESCC) has not been elucidated. In this study, we found that the expression levels of Pea3 and Erm, but not that of Er81, were significantly higher in ESCC compared with nontumor esophageal epithelium. A high level of Pea3 expression was significantly correlated with a shorter overall survival in a cohort of 81 patients with ESCC and the subgroup with N1 stage tumor (Wilcoxon-Gehan test, P = 0.016 and P = 0.001, respectively). Pea3 was overexpressed in seven ESCC cell lines compared with two immortalized esophageal cell lines. Pea3 knockdown reduced cell proliferation and suppressed nonadherent growth, migration, and invasion in ESCC cells in vitro. In addition, Pea3 knockdown in ESCC cells resulted in a down-regulation of phospho-Akt and matrix metalloproteinase 13, whereas a significant positive correlation in the expression levels was observed between Pea3 and phospho-Akt (r = 0.281, P < 0.013) and between Pea3 and matrix metalloproteinase 13 in the human specimens (r = 0.462, P < 0.001). Moreover, Pea3 modulated the sensitivity of EC109 cells to doxorubicin, probably via reduced activity of the phosphatidylinositol 3-kinase-Akt-mammalian target of Rapamycin complex 1 pathway on Pea3 knockdown. In conclusion, our results suggest that Pea3 plays an important role in the progression of ESCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Transcription Factors/physiology , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...