Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 699
Filter
Add more filters

Publication year range
1.
Cell ; 149(6): 1269-83, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682249

ABSTRACT

Cell-cycle arrest, apoptosis, and senescence are widely accepted as the major mechanisms by which p53 inhibits tumor formation. Nevertheless, it remains unclear whether they are the rate-limiting steps in tumor suppression. Here, we have generated mice bearing lysine to arginine mutations at one (p53(K117R)) or three (p53(3KR); K117R+K161R+K162R) of p53 acetylation sites. Although p53(K117R/K117R) cells are competent for p53-mediated cell-cycle arrest and senescence, but not apoptosis, all three of these processes are ablated in p53(3KR/3KR) cells. Surprisingly, unlike p53 null mice, which rapidly succumb to spontaneous thymic lymphomas, early-onset tumor formation does not occur in either p53(K117R/K117R) or p53(3KR/3KR) animals. Notably, p53(3KR) retains the ability to regulate energy metabolism and reactive oxygen species production. These findings underscore the crucial role of acetylation in differentially modulating p53 responses and suggest that unconventional activities of p53, such as metabolic regulation and antioxidant function, are critical for suppression of early-onset spontaneous tumorigenesis.


Subject(s)
Apoptosis , Cell Cycle Checkpoints , Cellular Senescence , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Fibroblasts/metabolism , Gene Knock-In Techniques , Humans , Lymphoma/metabolism , Mice , Molecular Sequence Data , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Sequence Alignment , Thymus Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics
2.
Cancer Sci ; 115(11): 3636-3647, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39166351

ABSTRACT

Thymic epithelial tumors (TETs) are rare tumors arising from the mediastinum. Among TETs, thymoma type B2, B3 and thymic carcinoma are highly malignant and often present invasion and dissemination. However, the biological characteristics of TETs have not been thoroughly studied, and their mechanisms of invasion and dissemination are largely unknown. α-Actinin 4 (ACTN4) is a member of actin-binding proteins and reportedly plays important roles in the progression of several cancers. In this study, we investigated the relationship between ACTN4 and characteristics of the malignant potential of TETs, such as invasion and dissemination. In vitro experiments using Ty-82 thymic carcinoma cells revealed that overexpression of ACTN4 enhanced the proliferative and invasive ability of Ty-82 cells; conversely, knockdown of ACTN4 attenuated the proliferative and invasive potential of Ty-82 cells. In western blotting (WB) experiments, ACTN4 induced the phosphorylation of extracellular signal-regulated kinase and glycogen synthase kinase 3ß to regulate the ß-catenin/Slug pathway. Furthermore, WB analysis of cancer tissue-origin spheroids from patients with TETs showed results similar to those for Ty-82 cells. In vivo experiments showed that the knockdown of ACTN4 significantly suppressed the dissemination of Ty-82 cells. A WB and immunohistochemistry staining comparison of primary and disseminated lesions of TETs using surgical specimens showed upregulated expression of ACTN4, ß-catenin, and Slug proteins in disseminated lesions. In summary, our study suggests ACTN4 is associated with malignant potential characteristics such as invasion and dissemination in TETs via the ß-catenin/Slug pathway.


Subject(s)
Actinin , Cell Proliferation , Signal Transduction , Snail Family Transcription Factors , Thymus Neoplasms , beta Catenin , Actinin/metabolism , Actinin/genetics , Humans , beta Catenin/metabolism , beta Catenin/genetics , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Thymus Neoplasms/genetics , Cell Line, Tumor , Animals , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Mice , Male , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/genetics , Female , Neoplasm Invasiveness , Middle Aged , Glycogen Synthase Kinase 3 beta/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Aged , Mice, Nude , Cell Movement/genetics
3.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34433672

ABSTRACT

Anti-PD-1 therapies can activate tumor-specific T cells to destroy tumors. However, whether and how T cells with different antigen specificity and affinity are differentially regulated by PD-1 remain vaguely understood. Upon antigen stimulation, a variety of genes is induced in T cells. Recently, we found that T cell receptor (TCR) signal strength required for the induction of genes varies across different genes and PD-1 preferentially inhibits the induction of genes that require stronger TCR signal. As each T cell has its own response characteristics, inducibility of genes likely differs across different T cells. Accordingly, the inhibitory effects of PD-1 are also expected to differ across different T cells. In the current study, we investigated whether and how factors that modulate T cell responsiveness to antigenic stimuli influence PD-1 function. By analyzing TCRs with different affinities to peptide-MHC complexes (pMHC) and pMHCs with different affinities to TCR, we demonstrated that PD-1 inhibits the expression of TCR-inducible genes efficiently when TCR:pMHC affinity is low. In contrast, affinities of peptides to MHC and MHC expression levels did not affect PD-1 sensitivity of TCR-inducible genes although they markedly altered the dose responsiveness of T cells by changing the efficiency of pMHC formation, suggesting that the strength of individual TCR signal is the key determinant of PD-1 sensitivity. Accordingly, we observed a preferential expansion of T cells with low-affinity to tumor-antigen in PD-1-deficient mice upon inoculation of tumor cells. These results demonstrate that PD-1 imposes qualitative control of T cell responses by preferentially suppressing low-affinity T cells.


Subject(s)
Antigens, Neoplasm/immunology , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/physiology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymoma/immunology , Thymus Neoplasms/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology
4.
EMBO J ; 38(14): e101564, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31304633

ABSTRACT

DOT1L methylates histone H3K79 and is aberrantly regulated in MLL-rearranged leukemia. Inhibitors have been developed to target DOT1L activity in leukemia, but cellular mechanisms that regulate DOT1L are still poorly understood. We have identified the histone deacetylase Rpd3 as a negative regulator of budding yeast Dot1. At its target genes, the transcriptional repressor Rpd3 restricts H3K79 methylation, explaining the absence of H3K79me3 at a subset of genes in the yeast genome. Similar to the crosstalk in yeast, inactivation of the murine Rpd3 homolog HDAC1 in thymocytes led to an increase in H3K79 methylation. Thymic lymphomas that arise upon genetic deletion of Hdac1 retained the increased H3K79 methylation and were sensitive to reduced DOT1L dosage. Furthermore, cell lines derived from Hdac1Δ/Δ thymic lymphomas were sensitive to a DOT1L inhibitor, which induced apoptosis. In summary, we identified an evolutionarily conserved crosstalk between HDAC1 and DOT1L with impact in murine thymic lymphoma development.


Subject(s)
Histone Deacetylase 1/genetics , Histone Deacetylase 2/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Lymphoma/metabolism , Thymus Neoplasms/metabolism , Acetylation , Animals , Cell Line, Tumor , Gene Deletion , Histone Deacetylases/genetics , Humans , Lymphoma/genetics , Methylation , Mice , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Thymus Neoplasms/genetics
5.
Histopathology ; 83(5): 798-809, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37565303

ABSTRACT

AIMS: Metaplastic thymoma is a rare thymic tumour characterized by Yes Associated Protein 1 (YAP1) and Mastermind Like Transcriptional Coactivator 2 (MAML2) gene fusions resulting from an intrachromosomal inversion of chromosome 11. Immunohistochemistry with an antibody directed against the C-terminus of YAP1 has shown loss of expression in YAP1-rearranged vascular neoplasms, poromas, and porocarcinomas. This study aimed to validate an anti-YAP1 C-terminal antibody as an ancillary immunohistochemical marker for the diagnosis of metaplastic thymoma. MATERIALS AND METHODS: Ten metaplastic thymomas were selected for the current study. Fluorescence in situ hybridization (FISH), next-generation sequencing (NGS), and reverse transcription-polymerase chain reaction (RT-PCR) analyses were performed to detect YAP1::MAML2 fusions. We then performed immunohistochemistry to detect YAP1 C-terminus expression in 10 metaplastic thymomas, 50 conventional thymomas (10 each of type A thymoma, type AB thymoma, type B1 thymoma, type B2 thymoma, and type B3 thymoma) and seven thymic carcinomas. RESULTS: All 10 cases showed narrow split signals with a distance of nearly two signal diameters and sometimes had false-negative results in YAP1 and MAML2 break-apart FISH (BA-FISH). Abnormal colocalized signals of the YAP1::MAML2 fusion were observed in all 10 cases using fusion FISH (F-FISH) assays. Eight of 10 cases with adequate nucleic acids were successfully sequenced and all showed YAP1::MAML2 fusions; in two cases the fusions were detected by both DNA and RNA sequencing and in six cases by RNA sequencing only. YAP1::MAML2 fusion transcripts were identified in four cases by RT-PCR. Metaplastic thymoma showed loss of YAP1 C-terminus expression in all 10 (100%) cases. All other thymic neoplasms showed retained YAP1 C-terminus expression. CONCLUSION: YAP1 C-terminus immunohistochemistry is a highly sensitive and specific ancillary marker that distinguishes metaplastic thymoma from its mimics. BA-FISH assays could not effectively detect YAP1::MAML2 fusions due to the proximity of the two genes. Loss of YAP1 C-terminus expression is a reliable surrogate for the detection of YAP1::MAML2 fusions in metaplastic thymoma.


Subject(s)
Thymoma , Thymus Neoplasms , Humans , Thymoma/diagnosis , Thymoma/genetics , Thymoma/metabolism , In Situ Hybridization, Fluorescence , Transcription Factors/genetics , Transcription Factors/metabolism , Thymus Neoplasms/diagnosis , Thymus Neoplasms/genetics , Thymus Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/genetics , Gene Rearrangement , Trans-Activators/genetics
6.
Neuroimmunomodulation ; 30(1): 69-80, 2023.
Article in English | MEDLINE | ID: mdl-36780882

ABSTRACT

INTRODUCTION: Dendritic cells (DCs) play critical roles in the pathogenesis of myasthenia gravis (MG), and a series of DC-based experimental strategies for MG have recently been developed. However, the definite roles of different DC subsets in the mechanism of MG have scarcely been covered by previous studies. The present study aimed to investigate the levels of three main DC subsets, plasmacytoid DCs (pDCs) (CD303 positive) and two distinct subsets of conventional DCs (cDCs), namely CD1c+ cDCs and CD141+ cDCs, in MG patients and analyze related clinical features. METHODS: From January 2016 to December 2020, 160 newly diagnosed MG patients and matched healthy controls (n = 160) were included in the study, and their clinical data were collected. The blood samples from MG patients before treatment and controls were collected for flow cytometry analysis. A total of 14 MG thymoma, 24 control thymoma, and 3 thymic cysts were used to immunostain the DC subsets. RESULTS: The flow cytometry analysis showed a significantly higher frequency of circulating pDCs, CD1c+ cDCs, and CD141+ cDCs in MG patients than in healthy controls (p < 0.001 for all). Patients with early-onset MG (<50 years old) had a lower frequency of circulating pDCs but a higher frequency of circulating CD1c+ cDCs than those with late-onset MG (≥50 years old) (p = 0.014 and p = 0.025, respectively). The frequency of circulating pDCs was positively associated with the clinical severity of late-onset MG patients (r = 0.613, p < 0.001). 64.3% (9/14) of MG thymoma is of type B2 under the World Health Organization classification, which is higher than that in control thymoma (33.3%, 8/24) (p = 0.019). For type B2 thymoma, there were significantly more pDCs but fewer CD1c+ cDCs in MG thymoma than in the controls. CONCLUSION: The distribution of aberrant pDCs, CD1c+ cDCs, and CD141+ cDCs in MG patients displayed age- and thymoma-related differences, which may contribute to the impaired immune tolerance and lead to the onset of MG.


Subject(s)
Myasthenia Gravis , Thymoma , Thymus Neoplasms , Humans , Middle Aged , Thymoma/metabolism , Immune Tolerance , Thymus Neoplasms/metabolism , Dendritic Cells/metabolism
7.
Cell ; 135(5): 907-18, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-19041753

ABSTRACT

Genomic instability and alterations in gene expression are hallmarks of eukaryotic aging. The yeast histone deacetylase Sir2 silences transcription and stabilizes repetitive DNA, but during aging or in response to a DNA break, the Sir complex relocalizes to sites of genomic instability, resulting in the desilencing of genes that cause sterility, a characteristic of yeast aging. Using embryonic stem cells, we show that mammalian Sir2, SIRT1, represses repetitive DNA and a functionally diverse set of genes across the mouse genome. In response to DNA damage, SIRT1 dissociates from these loci and relocalizes to DNA breaks to promote repair, resulting in transcriptional changes that parallel those in the aging mouse brain. Increased SIRT1 expression promotes survival in a mouse model of genomic instability and suppresses age-dependent transcriptional changes. Thus, DNA damage-induced redistribution of SIRT1 and other chromatin-modifying proteins may be a conserved mechanism of aging in eukaryotes.


Subject(s)
Aging/genetics , Chromatin/metabolism , Genomic Instability , Sirtuins/genetics , Animals , Brain/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair , Embryonic Stem Cells , Gene Knockout Techniques , Humans , Lymphoma/metabolism , Mice , Molecular Sequence Data , Oxidative Stress , Sirtuin 1 , Specific Pathogen-Free Organisms , Thymus Neoplasms/metabolism , Yeasts/cytology , Yeasts/metabolism
8.
Br J Cancer ; 126(5): 754-763, 2022 03.
Article in English | MEDLINE | ID: mdl-34876673

ABSTRACT

BACKGROUND: Thymic epithelial tumours (TETs) are rare tumours comprised of thymomas and thymic carcinoma. Novel therapies are needed, especially in thymic carcinoma where the 5-year survival rate hovers at 30%. Mesothelin (MSLN), a surface glycoprotein that is cleaved to produce mature MSLN (mMSLN) and megakaryocyte potentiating factor (MPF), is expressed in limited tissues. However, its expression is present in various cancers, including thymic carcinoma, where it is expressed in 79% of cases. METHODS: We utilised flow cytometry, in vitro cytotoxicity assays, and an in vivo xenograft model in order to demonstrate the ability of the MSLN targeting antibody-drug conjugate (ADC) anetumab ravtansine (ARav) in inhibiting the growth of thymic carcinoma. RESULTS: Thymoma and thymic carcinoma cell lines express MSLN, and anetumab, the antibody moiety of ARav, was capable of binding MSLN expressing thymic carcinoma cells and internalising. ARav was effective at inhibiting the growth of thymic carcinoma cells stably transfected with mMSLN in vitro. In vivo, 15 mg/kg ARav inhibited T1889 xenograft tumour growth, while combining 7.5 mg/kg ARav with 4 mg/kg cisplatin yielded an additive effect on inhibiting tumour growth. CONCLUSIONS: These data demonstrate that anetumab ravtansine inhibits the growth of MSLN positive thymic carcinoma cells in vitro and in vivo.


Subject(s)
Immunoconjugates/administration & dosage , Maytansine/analogs & derivatives , Mesothelin/genetics , Mesothelin/metabolism , Neoplasms, Glandular and Epithelial/drug therapy , Thymoma/drug therapy , Thymus Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Immunoconjugates/pharmacology , Maytansine/administration & dosage , Maytansine/pharmacology , Mice , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Thymoma/genetics , Thymoma/metabolism , Thymus Neoplasms/genetics , Thymus Neoplasms/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
9.
Mod Pathol ; 34(10): 1831-1838, 2021 10.
Article in English | MEDLINE | ID: mdl-34135467

ABSTRACT

The expression of immunohistochemical markers has been extensively investigated in thymomas to assist in the differential diagnosis. We have studied six select markers to determine their utility in the evaluation of these tumors. A series of 126 thymomas including 33 type A, 27 type AB, 20 type B1, 22 type B2, and 24 type B3, were examined utilizing a tissue microarray (TMA) technique with antibodies to e-cadherin, ß-catenin, PAX8, bcl-2, EMA, and MIB-1. Keratin AE1/AE3 and p63 were used for quality control. A significant finding was strong and consistent positivity for bcl-2 in type A (90%) and type AB (88.8%) thymoma, while 100% of B1, B2, and B3 were negative. The distribution of e-cadherin and ß-catenin was not useful for differential diagnosis. E-cadherin and ß-catenin were expressed in a high proportion of all the tumors (92-100%), except for B2 thymoma which showed only 45% expression. A significant increase in the expression of the MIB-1 proliferation marker (mean: 12.8% nuclear positivity) was also observed in B3 thymoma compared with the other histologic types. Statistical significance was confirmed using Kruskal's non-parameterized test for distribution. EMA was generally negative except for spindle cells in the fibrous septa in types A and AB thymoma. PAX8 showed less consistent nuclear staining than p63 and was only widely expressed in 55.7% of cases. Bcl-2 may serve as a useful marker to separate spindle cell thymomas (Type A and AB) from the other types, and the MIB1 proliferation index may be of use to differentiate type B2 from type B3 thymoma.


Subject(s)
Biomarkers, Tumor/metabolism , Thymoma/diagnosis , Thymus Neoplasms/diagnosis , beta Catenin/metabolism , Cadherins/metabolism , Diagnosis, Differential , E2F6 Transcription Factor/metabolism , Humans , Ki-67 Antigen/metabolism , PAX8 Transcription Factor/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology
10.
Pathol Int ; 71(7): 471-479, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33902161

ABSTRACT

Cancer testis antigens (CTAs) are detected in cancer cells but not in healthy normal tissues, with the exception of gametogenic tissues. However, to our knowledge, expression of the antigens in thymic epithelial tumors has not been examined yet. We examined the immunohistochemical expression of five CTAs (MAGE-A, NY-ESO-1, MAGE-C1, SAGE and GAGE7) in 192 cases of thymic epithelial tumor. The CTAs were variably expressed in the thymic epithelial tumors. Type B component of type AB thymomas, type B1/B2/B3 thymomas, and thymic carcinomas showed a generally positive correlation between the malignancy grades and positive expression rates in four CTAs other than MAGE-C1. In thymic squamous cell carcinomas (SqCCs), four antigens except for MAGE-C1 showed high expression rates ranging from 23.1% to 43.6%. In the prognostic analysis, a positive expression of SAGE (P = 0.0485) and GAGE7 (P = 0.0289) were associated with a shorter overall survival in type B2/B3 thymomas, respectively. In thymic SqCC, a positive MAGE-A expression was significantly associated with an increased level of programmed death ligand in tumor-infiltrating lymphocytes (P = 0.0181). We showed (i) a frequent CTA expression, (ii) a general correlation of CTA expression with tumor malignancy grades and (iii) a prognostic impact in some of the CTAs.


Subject(s)
Antigens, Neoplasm/metabolism , Neoplasms, Glandular and Epithelial , Thymus Neoplasms , Adult , Aged , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Prognosis , Testis/metabolism , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology
11.
Med Sci Monit ; 27: e929727, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34219124

ABSTRACT

BACKGROUND Thymoma is the most common tumor of the anterior mediastinum, and can be caused by infrequent malignancies arising from the epithelial cells of the thymus. Unfortunately, blood-based diagnostic markers are not currently available. High-throughput sequencing technologies, such as RNA-seq with next-generation sequencing, have facilitated the detection and characterization of both coding and non-coding RNAs (ncRNAs), which play significant roles in genomic regulation, transcriptional and post-transcriptional regulation, and imprinting and epigenetic modification. The knowledge about fusion genes and ncRNAs in thymomas is scarce. MATERIAL AND METHODS For this study, we gathered large-scale RNA-seq data belonging to samples from 25 thymomas and 25 healthy thymus specimens and analyzed them to identify fusion genes, lncRNAs, and miRNAs. RESULTS We found 21 fusion genes, including KMT2A-MAML2, HADHB-REEP1, COQ3-CGA, MCM4-SNTB1, and IFT140-ACTN4, as the most frequent and significant in thymomas. We also detected 65 differentially-expressed lncRNAs in thymomas, including AFAP1-AS1, LINC00324, ADAMTS9-AS1, VLDLR-AS1, LINC00968, and NEAT1, that have been validated with the TCGA database. Moreover, we identified 1695 miRNAs from small RNA-seq data that were overexpressed in thymomas. Our network analysis of the lncRNA-mRNA-miRNA regulation axes identified a cluster of miRNAs upregulated in thymomas, that can trigger the expression of target protein-coding genes, and lead to the disruption of several biological pathways, including the PI3K-Akt signaling pathway, FoxO signaling pathway, and HIF-1 signaling pathway. CONCLUSIONS Our results show that overexpression of this miRNA cluster activates PI3K-Akt, FoxO, HIF-1, and Rap-1 signaling pathways, suggesting pathway inhibitors may be therapeutic candidates against thymoma.


Subject(s)
RNA, Long Noncoding/metabolism , Thymoma/metabolism , Thymus Neoplasms/metabolism , Adult , Aged , Biomarkers, Tumor/metabolism , Female , Humans , Male , Middle Aged
12.
Surg Today ; 51(2): 322-326, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32535710

ABSTRACT

This study aimed to investigate the association between the volume-dependent parameters in 18F-fluorodeoxyglucose-positron emission tomography (18F-FDG PET/CT) and a recurrence of thymic carcinoma. A retrospective chart review was performed based on our multi-institutional database to identify patients undergoing PET prior to resection of thymic carcinoma or neuroendocrine carcinoma between 1991 and 2018. The PET parameters (metabolic tumor volume and total lesion glycolysis) were evaluated retrospectively. The relevant factors were extracted and a survival analysis was performed using the Kaplan-Meier method. Sixteen patients were thus deemed to be eligible for analysis. The median follow-up period following resection was 2.65 years (range: 0.96-0.68 years). The recurrence-free survival was significantly longer in patients with a metabolic tumor volume < = 22.755 cm3 and with total lesion glycolysis < = 105.4006 g/mL (p = 0.001 and 0.001, respectively, by a log-rank test). The metabolic tumor volume and total lesion glycolysis may, therefore, be predictive of the postoperative recurrence of thymic carcinoma.


Subject(s)
Positron Emission Tomography Computed Tomography , Thymoma/diagnostic imaging , Thymoma/surgery , Thymus Neoplasms/diagnostic imaging , Thymus Neoplasms/surgery , Disease-Free Survival , Fluorodeoxyglucose F18 , Follow-Up Studies , Glycolysis , Humans , Predictive Value of Tests , Radiopharmaceuticals , Recurrence , Retrospective Studies , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Time Factors
13.
J Autoimmun ; 106: 102337, 2020 01.
Article in English | MEDLINE | ID: mdl-31594658

ABSTRACT

Thymomas are associated with a very high risk of developing Myasthenia Gravis (MG). Our objectives were to identify histological and biological parameters to allow early diagnosis of thymoma patients susceptible to developing MG. We conducted a detailed retrospective analysis from a patient database, searching for differences between patients with thymoma-associated MG (MGT, n = 409) and thymoma without MG (TOMA, n = 111) in comparison with nonthymomatous MG patients (MG, n = 1246). We also performed multiplex and single molecule arrays to measure the serum levels of cytokines in these groups of patients and controls (n = 14-22). We identified a set of parameters associated with MG development in thymoma patients: 1) detection of anti-acetylcholine receptor (AChR) antibodies, 2) development of B1 or B2 thymoma subtypes, 3) presence of ectopic thymic germinal centers (GCs), 4) local invasiveness of thymoma, and 5) being a woman under 50 years old. Among these parameters, 58.8% of MGT patients displayed GCs with a positive correlation between the number of GCs and anti-AChR titers. By immunohistochemistry, we found thymic GCs in the adjacent tissues of thymomas encircled by high endothelial venules (HEVs) that could favor peripheral cell recruitment. We also clearly associated MG symptoms with higher IFN-γ, IL-1ß and sCD40L serum levels, specifically in MGT patients compared to TOMA patients. Altogether, these analyses allowed the clear identification of histological, in particular the presence of GCs, and biological parameters that would facilitate the evaluation of the probability of the MG outcome postoperatively in thymoma patients.


Subject(s)
Germinal Center/pathology , Myasthenia Gravis/etiology , Thymoma/complications , Thymus Neoplasms/complications , Adult , Autoantibodies/metabolism , CD40 Ligand/metabolism , Female , Germinal Center/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Male , Middle Aged , Myasthenia Gravis/metabolism , Receptors, Cholinergic/metabolism , Retrospective Studies , Risk Factors , Thymoma/metabolism , Thymus Neoplasms/metabolism
14.
Cytokine ; 133: 155149, 2020 09.
Article in English | MEDLINE | ID: mdl-32512341

ABSTRACT

OBJECTIVES: To understand underlying changes in pretreatment serum inflammatory markers associated with thymic epithelial tumors (TETs) development. METHODS: A retrospective analysis of 113 TETs patients who underwent 18-fluorine fluorodeoxyglucose (18F-FDG) positron emission tomography combined computed tomography (PET/CT) one to two weeks before tumor resection or biopsy was performed. Pretreatment serum neutrophil, monocyte, platelet, and lymphocyte counts, and fibrinogen and C-reaction protein (CRP) concentrations were measured one day before surgery or biopsy. Neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), and platelet-to-lymphocyte ratio (PLR) were calculated by dividing corresponding cells counts by lymphocyte counts, respectively. The maximum standard uptake value (SUVmax) of 18F-FDG of primary TETs was applied to reflect tumor glycolytic activity. The student's t-test, one-way ANOVA analysis, Chi-square test, receiver operating characteristic curve analysis, and Logistic regression analysis were used for statistical analysis. RESULTS: The serum NLR and MLR were significantly higher in TETs patients than in healthy volunteers (P both ≤ 0.001). High serum NLR and MLR were related to the thymic carcinomas (TCs) subtype, elevated Masaoka-Koga (M-K) tumor stage, and metastasis of TETs (P all < 0.005). High serum NLR and MLR were also associated with high SUVmax values of TETs (P all < 0.005), with increasingly differences between groups as the cut-off values defining low-SUVmax and high-SUVmax groups increased. With the medium cutoff of NLR, MLR, and SUVmax of 3.07, 0.25, and 8.00 respectively, the high NLR and MLR levels were significantly associated with high SUVmax level of TETs (P both < 0.005). Moreover, the incidences of co-high SUVmax/NLR and co-high SUVmax/MLR were higher in TETs patients older than 55 years, with TCs, in M-K stage IV, and with metastasis (P all < 0.05). Both the co-high SUVmax/NLR and co-high SUVmax/MLR increased the risk of TETs metastasis (P both < 0.001), while the co-high SUVmax/MLR was also an independent risk factor for TETs metastasis (odds ratio: 3.92, 95% confidence interval: 1.02-15.12, P = 0.047). CONCLUSION: Pretreatment serum NLR and MLR of TETs patients are two tumor-progression- and tumor-glycolysis-related inflammatory markers. Enhanced tumor glycolytic activity and associated systemic inflammatory reaction may play a synergistic role in TETs metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Inflammation/pathology , Lymphocytes/pathology , Monocytes/pathology , Neoplasms, Glandular and Epithelial/pathology , Neutrophils/pathology , Thymus Neoplasms/pathology , Adult , Aged , Blood Platelets/metabolism , Blood Platelets/pathology , C-Reactive Protein/metabolism , Case-Control Studies , Female , Humans , Inflammation/metabolism , Leukocyte Count/methods , Lymphocyte Count/methods , Lymphocytes/metabolism , Male , Middle Aged , Monocytes/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neutrophils/metabolism , Positron Emission Tomography Computed Tomography/methods , ROC Curve , Retrospective Studies , Thymus Neoplasms/metabolism , Young Adult
15.
Clin Exp Pharmacol Physiol ; 47(6): 989-996, 2020 06.
Article in English | MEDLINE | ID: mdl-32048308

ABSTRACT

Imbalance of T helper 17 (Th17)/regulatory T (Treg) cells is involved in the pathogenesis of myasthenia gravis with thymoma (MG-T). Long non-coding RNAs (lncRNAs) are implicated in the regulation of Th17/Treg balance. This study was designed to explore the role of XLOC_003810, a novel lncRNA, in regulating the Th17/Treg balance in MG-T. The thymic CD4+ T cells were isolated from control subjects and MG-T patients. The Th17/Treg balance was evaluated by determining proportions of Th17 and Treg cells and expression of Th17- and Treg- associated molecules. Lentivirus-mediated silencing and overexpression of XLOC_003810 in CD4+ T cells were performed. The results showed that XLOC_003810 expression was higher in MG-T thymic CD4+ T cells than that in the control group. Furthermore, the ratio of Th17/Treg cells, proportion of Th17 cells and levels of Th17-associated molecules were significantly increased, whereas the proportion of Treg cells and levels of Treg-associated molecules were decreased in MG-T thymic CD4+ T cells. Importantly, the Th17/Treg imbalance in MG-T thymic CD4+ T cells was aggravated by XLOC_003810 overexpression, whereas it was attenuated by XLOC_003810 silencing. Collectively, XLOC_003810 modulates thymic Th17/Treg balance in MG-T patients, providing the scientific basis for the clinical targeted therapy of MG-T.


Subject(s)
Myasthenia Gravis/metabolism , RNA, Long Noncoding/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Thymoma/metabolism , Thymus Gland/metabolism , Thymus Neoplasms/metabolism , Case-Control Studies , Cytokines/metabolism , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Myasthenia Gravis/genetics , Myasthenia Gravis/immunology , Phenotype , Prospective Studies , RNA, Long Noncoding/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Thymoma/genetics , Thymoma/immunology , Thymus Gland/immunology , Thymus Neoplasms/genetics , Thymus Neoplasms/immunology , Transforming Growth Factor beta1/metabolism
16.
Cancer Immunol Immunother ; 68(11): 1769-1778, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31616964

ABSTRACT

Various immune cells are recruited in the tumor microenvironment. It is well established that cellular immune responses, such as cytotoxic or suppressive activities, play an important role in regulating tumor growth and metastasis. However, the contribution of humoral immune responses against tumors is poorly understood. Fc receptors constitute critical elements for the up- or downregulation of immune responses through immune complexes. Here, we examined the potential role of the inhibitory Fc receptor, Fcγ receptor IIB (FcγRIIB), in tumor immunity using a mouse model. Our findings indicated that tumor-specific antibodies are induced in tumor-bearing mice and control tumor immunity. FcγRIIB deletion significantly improved both cellular and humoral immunity against tumors and delayed tumor growth. These findings indicated that spontaneous antibodies against tumors create a suppressive tumor microenvironment through FcγRIIB signaling, thus suggesting an attractive therapeutic target for cancer immunotherapy.


Subject(s)
Antibodies/immunology , Carcinoma, Lewis Lung/therapy , Immunotherapy , Receptors, IgG/physiology , Thymoma/therapy , Thymus Neoplasms/therapy , Tumor Microenvironment/immunology , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgG/immunology , Thymoma/immunology , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/immunology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Neuroimmunomodulation ; 26(5): 234-238, 2019.
Article in English | MEDLINE | ID: mdl-31661704

ABSTRACT

OBJECTIVE: To explore the diversity and clinical features of anti-glutamate decarboxylase (GAD) antibody-associated neurological diseases. METHODS: Clinical data of a series of 5 patients positive for anti-GAD antibodies were retrospectively analyzed. RESULTS: All 5 patients were female, with a median age of 41.5 years (range 19-60 years). Their neurological symptoms included stiff-person syndrome (SPS), encephalitis, myelitis, cramp, visual loss, and paresthesia. Three patients (60%) were diagnosed with tumors, 2 cases of thymic tumor and 1 of breast cancer. On immunohistochemistry for tumor pathology, expression of GAD65 was found only in 1 patient. Four patients (80%) had abnormal brain MRI findings. All patients received immunotherapy and improved significantly after treatment, but 4 (80%) then experienced a relapse. CONCLUSIONS: Neurological manifestations in anti-GAD-positive patients are diverse and include SPS, encephalitis, myelitis, cramp, visual loss, and paresthesia.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases of the Nervous System/physiopathology , Glutamate Decarboxylase/immunology , Paraneoplastic Syndromes, Nervous System/physiopathology , Adult , Autoimmune Diseases of the Nervous System/diagnostic imaging , Autoimmune Diseases of the Nervous System/immunology , Brain/diagnostic imaging , Breast Neoplasms/metabolism , Encephalitis/diagnostic imaging , Encephalitis/immunology , Encephalitis/physiopathology , Female , Glutamate Decarboxylase/metabolism , Humans , Magnetic Resonance Imaging , Middle Aged , Muscle Cramp/immunology , Muscle Cramp/physiopathology , Myelitis/immunology , Myelitis/physiopathology , Paraneoplastic Syndromes, Nervous System/diagnostic imaging , Paraneoplastic Syndromes, Nervous System/immunology , Paresthesia/immunology , Paresthesia/physiopathology , Recurrence , Retrospective Studies , Stiff-Person Syndrome/immunology , Stiff-Person Syndrome/physiopathology , Thymus Neoplasms/metabolism , Vision Disorders/immunology , Vision Disorders/physiopathology , Young Adult
18.
Cancer Sci ; 109(12): 3737-3750, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30302856

ABSTRACT

The antitumor activity of activated CD8+ T cells in the tumor microenvironment seems to be limited due to their being metabolically unfit. This metabolic unfitness is closely associated with T-cell exhaustion and impairment of memory formation, which are barriers to successful antitumor adoptive immunotherapy. We therefore assessed the role of glutamine metabolism in the antitumor activity of CD8+ T cells using a tumor-inoculated mouse model. The adoptive transfer of tumor-specific CD8+ T cells cultured under glutamine-restricted (dGln) conditions or CD8+ T cells treated with specific inhibitors of glutamine metabolism efficiently eliminated tumors and led to better survival of tumor-inoculated mice than with cells cultured under control (Ctrl) conditions. The decreased expression of PD-1 and increased Ki67 positivity among tumor-infiltrating CD8+ T cells cultured under dGln conditions suggested that the inhibition of glutamine metabolism prevents CD8+ T-cell exhaustion in vivo. Furthermore, the transferred CD8+ T cells cultured under dGln conditions expanded more efficiently against secondary OVA stimulation than did CD8+ T cells under Ctrl conditions. We found that the expression of a pro-survival factor and memory T cell-related transcription factors was significantly higher in CD8+ T cells cultured under dGln conditions than in those cultured under Ctrl conditions. Given these findings, our study uncovered an important role of glutamine metabolism in the antitumor activity of CD8+ T cells. The novel adoptive transfer of tumor-specific CD8+ T cells cultured in glutamine-restricted conditions may be a promising approach to improve the efficacy of cell-based adoptive immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Glutamine/deficiency , Thymoma/therapy , Thymus Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Culture Techniques , Cell Line, Tumor , Culture Media/chemistry , Humans , Immunotherapy, Adoptive/methods , Mice , Thymoma/immunology , Thymoma/metabolism , Thymus Neoplasms/immunology , Thymus Neoplasms/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
19.
Mod Pathol ; 31(12): 1801-1806, 2018 12.
Article in English | MEDLINE | ID: mdl-29973653

ABSTRACT

Poorly differentiated non-keratinizing squamous cell carcinoma of the thymus, also known as lymphoepithelioma-like carcinoma, is a rare primary malignant neoplasm of thymic origin. The mainstay of treatment for these tumors is surgical and they tend to respond poorly to chemotherapy. The checkpoint programmed cell death ligand-1 protein (PD-L1) bound to its receptor (PD-1) has been demonstrated to be an important therapeutic target for many different tumors. Expression of PD-L1/PD-1 in lymphoepithelioma-like carcinoma of the thymus may indicate that these tumors are potential targets for inhibitor therapy. Twenty-one cases of lymphoepithelioma-like carcinoma of the thymus were collected and reviewed. Tissue microarrays were created using triplicate 2 mm cores for each case. PD-L1/PD-1 staining pattern (neoplastic cells versus tumor infiltrating lymphocytes) was documented for each case. Out of 21 cases, 15 (71.4%) showed various degrees of membranous PD-L1 staining. Of the positive cases, 48% showed high expression of PD-L1 (>50% of tumor cells) and 24% showed low expression (<50%). PD-1 staining showed focal positivity in 12/20 (60%) cases among tumor infiltrating lymphocytes. PD-L1/PD-1 inhibitor therapy has been applied successfully in other solid malignant tumors with high expression of PD-L1/PD-1. The high level of PD-L1 expression in our cases indicates that PD-L1 may play a role in the pathogenesis of these tumors and that PD-L1/PD-1 blockade may be a viable therapeutic option for patients with lymphoepithelioma-like carcinoma of the thymus who have failed other first-line therapies.


Subject(s)
B7-H1 Antigen/biosynthesis , Biomarkers, Tumor/analysis , Programmed Cell Death 1 Receptor/biosynthesis , Squamous Cell Carcinoma of Head and Neck/metabolism , Thymus Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/analysis , Female , Humans , Male , Middle Aged , Programmed Cell Death 1 Receptor/analysis , Squamous Cell Carcinoma of Head and Neck/pathology , Thymus Neoplasms/pathology , Young Adult
20.
Proc Natl Acad Sci U S A ; 112(46): E6321-30, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578792

ABSTRACT

Centrosomes are microtubule-organizing centers that facilitate bipolar mitotic spindle assembly and chromosome segregation. Recognizing that centrosome amplification is a common feature of aneuploid cancer cells, we tested whether supernumerary centrosomes are sufficient to drive tumor development. To do this, we constructed and analyzed mice in which centrosome amplification can be induced by a Cre-recombinase-mediated increase in expression of Polo-like kinase 4 (Plk4). Elevated Plk4 in mouse fibroblasts produced supernumerary centrosomes and enhanced the expected mitotic errors, but proliferation continued only after inactivation of the p53 tumor suppressor. Increasing Plk4 levels in mice with functional p53 produced centrosome amplification in liver and skin, but this did not promote spontaneous tumor development in these tissues or enhance the growth of chemically induced skin tumors. In the absence of p53, Plk4 overexpression generated widespread centrosome amplification, but did not drive additional tumors or affect development of the fatal thymic lymphomas that arise in animals lacking p53. We conclude that, independent of p53 status, supernumerary centrosomes are not sufficient to drive tumor formation.


Subject(s)
Asymmetric Cell Division/physiology , Centrosome/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Thymus Neoplasms/genetics , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL