Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1390140, 2024.
Article in English | MEDLINE | ID: mdl-38828408

ABSTRACT

Objective: The aim of this study was to identify potential causal cytokines in thymic malignancies and benign tumors from the FinnGen database using Mendelian randomization (MR). Methods: In this study, data from genome-wide association studies (GWAS) of 91 cytokines were used as exposure factors, and those of thymic malignant tumors and thymic benign tumors were the outcome variables. Two methods were used to determine the causal relationship between exposure factors and outcome variables: inverse variance weighting (IVW) and MR-Egger regression. Sensitivity analysis was performed using three methods, namely, the heterogeneity test, the pleiotropy test, and the leave-one-out test. Results: There was a causal relationship between the expression of fibroblast growth factor 5, which is a risk factor for thymic malignant tumors, and thymic malignant tumors. C-C motif chemokine 19 expression, T-cell surface glycoprotein CD5 levels, and interleukin-12 subunit beta levels were causally related to thymic malignant tumors and were protective. Adenosine deaminase levels, interleukin-10 receptor subunit beta expression, tumor necrosis factor (TNF)-related apoptosis-inducing ligand levels, and TNF-related activation-induced cytokine levels showed a causal relationship with thymic benign tumors, which are its risk factors. Caspase 8 levels, C-C motif chemokine 28 levels, interleukin-12 subunit beta levels, latency-associated peptide transforming growth factor beta 1 levels, and programmed cell death 1 ligand 1 expression showed a causal relationship with thymic benign tumors, which are protective factors. Sensitivity analysis showed no heterogeneity. Conclusion: Cytokines showed a causal relationship with benign and malignant thymic tumors. Interleukin-12 subunit beta is a common cytokine that affects malignant and benign thymic tumors.


Subject(s)
Cytokines , Genome-Wide Association Study , Mendelian Randomization Analysis , Proteomics , Thymus Neoplasms , Humans , Cytokines/metabolism , Cytokines/genetics , Thymus Neoplasms/genetics , Proteomics/methods , Biomarkers, Tumor/genetics , Risk Factors
2.
J Neuroinflammation ; 21(1): 126, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734662

ABSTRACT

Myasthenia gravis (MG) is an immune-mediated disease frequently associated with thymic changes. Increased T helper 17 (Th17) cell activity and dysfunctional regulatory T (Treg) cells have been demonstrated in subgroups of MG. On the other hand, hypoxia-inducible factor 1 (HIF-1) has been shown to regulate the Th17/Treg balance by inducing Th17 differentiation while attenuating Treg development. To identify the underlying mechanisms of different thymic pathologies in MG development, we evaluated thymic samples from thymoma-associated myasthenia gravis (TAMG), MG with hyperplasia (TFH-MG) and thymoma without MG (TOMA) patients. Differential gene expression analysis revealed that TAMG and TFH-MG cells are associated with different functional pathways. A higher RORC/FOXP3 ratio provided evidence for Th17/Treg imbalance in TAMG potentially related to increased HIF1A. The hypoxic microenvironment in thymoma may be a driver of TAMG by increasing HIF1A. These findings may lead to new therapeutic approaches targeting HIF1A in the development of TAMG.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Myasthenia Gravis , Th17 Cells , Thymoma , Female , Humans , Male , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myasthenia Gravis/genetics , Myasthenia Gravis/immunology , Myasthenia Gravis/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/metabolism , Th17 Cells/immunology , Thymoma/complications , Thymoma/genetics , Thymoma/immunology , Thymus Gland/pathology , Thymus Neoplasms/complications , Thymus Neoplasms/genetics
3.
Clin Immunol ; 263: 110233, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697554

ABSTRACT

Ataxia-telangiectasia (A-T) is a rare disorder caused by genetic defects of A-T mutated (ATM) kinase, a key regulator of stress response, and characterized by neurodegeneration, immunodeficiency, and high incidence of cancer. Here we investigated NK cells in a mouse model of A-T (Atm-/-) showing that they are strongly impaired at killing tumor cells due to a block of early signaling events. On the other hand, in Atm-/- littermates with thymic lymphoma NK cell cytotoxicity is enhanced as compared with ATM-proficient mice, possibly via tumor-produced TNF-α. Results also suggest that expansion of exhausted NKG2D+ NK cells in Atm-/- mice is driven by low-level expression of stress-inducible NKG2D ligands, whereas development of thymoma expressing the high-affinity MULT1 ligand is associated with NKG2D down-regulation on NK cells. These results expand our understanding of immunodeficiency in A-T and encourage exploring NK cell biology in A-T patients in the attempt to identify cancer predictive biomarkers and novel therapeutic targets.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily K , Animals , Killer Cells, Natural/immunology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Mice , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/immunology , Mice, Knockout , Mice, Inbred C57BL , Thymoma/immunology , Thymoma/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/immunology , Cytotoxicity, Immunologic , Thymus Neoplasms/immunology , Thymus Neoplasms/genetics , Signal Transduction , Membrane Proteins , Histocompatibility Antigens Class I
4.
Respir Investig ; 62(4): 517-519, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38631274

ABSTRACT

Microsatellite instability (MSI) is a valuable biomarker for immune checkpoint inhibitors. We report the first case of MSI-high thymoma successfully treated with pembrolizumab. This patient had pleural dissemination and was treated with two cytotoxic chemotherapy regimens including carboplatin and paclitaxel combination therapy and pemetrexed, which did not have the desired effect. Because MSI status was high by using the surgical specimen, pembrolizumab was administered as 3rd line chemotherapy. After three courses, the pleural lesions dramatically shrunk, which confirmed a partial response. Although MSI-high thymoma is rare, our results suggest the necessity to evaluate MSI status in patients with thymoma.


Subject(s)
Antibodies, Monoclonal, Humanized , Microsatellite Instability , Thymoma , Thymus Neoplasms , Humans , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Thymoma/drug therapy , Thymoma/genetics , Thymus Neoplasms/drug therapy , Thymus Neoplasms/genetics , Thymus Neoplasms/pathology , Treatment Outcome , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Male , Carboplatin/administration & dosage , Middle Aged , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Female
5.
Cancer Sci ; 115(6): 1808-1819, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38572512

ABSTRACT

Rev1 has two important functions in the translesion synthesis pathway, including dCMP transferase activity, and acts as a scaffolding protein for other polymerases involved in translesion synthesis. However, the role of Rev1 in mutagenesis and tumorigenesis in vivo remains unclear. We previously generated Rev1-overexpressing (Rev1-Tg) mice and reported that they exhibited a significantly increased incidence of intestinal adenoma and thymic lymphoma (TL) after N-methyl-N-nitrosourea (MNU) treatment. In this study, we investigated mutagenesis of MNU-induced TL tumorigenesis in wild-type (WT) and Rev1-Tg mice using diverse approaches, including whole-exome sequencing (WES). In Rev1-Tg TLs, the mutation frequency was higher than that in WT TL in most cases. However, no difference in the number of nonsynonymous mutations in the Catalogue of Somatic Mutations in Cancer (COSMIC) genes was observed, and mutations involved in Notch1 and MAPK signaling were similarly detected in both TLs. Mutational signature analysis of WT and Rev1-Tg TLs revealed cosine similarity with COSMIC mutational SBS5 (aging-related) and SBS11 (alkylation-related). Interestingly, the total number of mutations, but not the genotypes of WT and Rev1-Tg, was positively correlated with the relative contribution of SBS5 in individual TLs, suggesting that genetic instability could be accelerated in Rev1-Tg TLs. Finally, we demonstrated that preleukemic cells could be detected earlier in Rev1-Tg mice than in WT mice, following MNU treatment. In conclusion, Rev1 overexpression accelerates mutagenesis and increases the incidence of MNU-induced TL by shortening the latency period, which may be associated with more frequent DNA damage-induced genetic instability.


Subject(s)
DNA-Directed DNA Polymerase , Methylnitrosourea , Mutagenesis , Nucleotidyltransferases , Thymus Neoplasms , Animals , Mice , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Exome Sequencing , Lymphoma/genetics , Lymphoma/chemically induced , Lymphoma/pathology , Methylnitrosourea/toxicity , Mice, Transgenic , Mutation , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Thymus Neoplasms/genetics , Thymus Neoplasms/chemically induced , Thymus Neoplasms/pathology
6.
Int J Mol Sci ; 25(3)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38338833

ABSTRACT

Thymic epithelial tumors (TETs) are characterized by their extreme rarity and variable clinical presentation, with the inadequacy of the use of histological classification alone to distinguish biologically indolent from aggressive cases. The utilization of Next Generation Sequencing (NGS) to unravel the intricate genetic landscape of TETs could offer us a comprehensive understanding that is crucial for precise diagnoses, prognoses, and potential therapeutic strategies. Despite the low tumor mutational burden of TETS, NGS allows for exploration of specific genetic signatures contributing to TET onset and progression. Thymomas exhibit a limited mutational load, with prevalent GTF2I and HRAS mutations. On the other hand, thymic carcinomas (TCs) exhibit an elevated mutational burden, marked by frequent mutations in TP53 and genes associated with epigenetic regulation. Moreover, signaling pathway analyses highlight dysregulation in crucial cellular functions and pathways. Targeted therapies, and ongoing clinical trials show promising results, addressing challenges rooted in the scarcity of actionable mutations and limited genomic understanding. International collaborations and data-sharing initiatives are crucial for breakthroughs in TETs research.


Subject(s)
Neoplasms, Glandular and Epithelial , Thymoma , Thymus Neoplasms , Humans , Epigenesis, Genetic , Thymus Neoplasms/genetics , Thymus Neoplasms/pathology , Thymoma/genetics , Thymoma/pathology , Neoplasms, Glandular and Epithelial/genetics
8.
Chinese Journal of Pathology ; (12): 1237-1243, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1012399

ABSTRACT

Objective: To investigate the clinicopathological features, and molecular genetic alterations of metaplastic thymoma (MT). Methods: A total of ten MT cases, diagnosed from 2011 to 2021, were selected from the Department of Pathology of Jinling Hospital, Nanjing University Medical School, Nanjing, China for clinicopathological and immunohistochemical (IHC) examination and clinical follow-up. Fluorescence in situ hybridization (FISH), next-generation sequencing (NGS), and YAP1 C-terminus (YAP1-CT) IHC were performed to detect YAP1::MAML2 fusions. Results: There were four males and six females, ranging in age from 29 to 60 years (mean 50 years, median 54 years). Microscopically, all tumors showed a typical biphasic morphology consisting of epithelial components and gradually or abruptly transitioning spindle cell components. The two components were present in varying proportions in different cases. Immunophenotypically, the epithelial cells were diffusely positive for CKpan, CK5/6 and p63. The spindle cells were diffusely positive for vimentin and focally positive for EMA. TdT was negative in the background lymphocytes. Ki-67 proliferation index was less than 5%. YAP1 and MAML2 break-apart FISH analyses showed that all ten cases had narrow split signals with a distance of nearly 2 signal diameters and may be considered false-negative. Using YAP1::MAML2 fusion FISH assays, abnormal fusion signals were observed in all the ten cases. NGS demonstrated YAP1::MAML2 fusions in all eight cases with adequate nucleic acids; in two cases the fusions were detected by DNA sequencing and in eight cases by RNA sequencing. All ten cases of MT demonstrated loss of YAP1 C-terminal expression in epithelioid cells. Conclusions: MT is a rare and low-grade thymic tumor characterized by a biphasic pattern and YAP1::MAML2 fusions. Break-apart FISH assays may sometimes show false-negative results due to the proximity of YAP1 and MAML2, while YAP1 C-terminal IHC is a highly sensitive and specific marker for MT. Loss of YAP1 C-terminal expression can also be used to screen YAP1::MAML2 fusions for possible MT cases.


Subject(s)
Male , Female , Humans , Adult , Middle Aged , Thymoma/genetics , In Situ Hybridization, Fluorescence , Transcription Factors/genetics , Mutation , Thymus Neoplasms/genetics
9.
Medicina (B.Aires) ; 68(1): 43-47, ene.-feb. 2008. ilus, tab
Article in Spanish | LILACS | ID: lil-633513

ABSTRACT

El mediastino anterior es un sitio frecuente de localización de tumores ricos en elementos linfoides. La identificación correcta de cada entidad es de importancia en el tratamiento de los pacientes. En ocasiones puede plantearse el diagnóstico diferencial entre timoma y linfoma linfoblástico con fenotipo de precursor T (LLB-T). La Citometría de Flujo (CF) es una técnica complementaria útil para estos tumores de la cual se obtiene información cualitativa y cuantitativa. Revisamos 38 tumores mediastinales que tenían estudio de CF. Además comparamos los resultados de CF de timomas y tejido tímico normal con 42 casos de LLB-T de otras localizaciones anatómicas. De los 38 tumores mediastinales 6 eran lesiones benignas, 9 linfomas difusos de células grandes con fenotipo B (LDCG-B), 10 linfomas de Hodgkin (LH), 11 timomas y 2 LLB-T. En 24 casos la CF aportó información positiva, definiendo el inmunofenotipo de las células linfoides neoplásicas, o los linfocitos característicos que acompañan a los timomas. La CF en los 10 casos de LH y en 4 lesiones benignas permitió descartar otros tipos de linfoma (LDCG-B, LLB-T, etc.). Las marcaciones para CD3, CD4 y CD8 fueron las más útiles para el diagnóstico diferencial entre timomas y LLB-T. En conclusión, la CF es una técnica complementaria de utilidad que aporta información en lesiones mediastinales de manera rápida, requiriendo cantidades pequeñas de material, tanto para el diagnóstico inicial como para el monitoreo de estas enfermedades.


The anterior mediastinum is a common site of tumors with abundant lymphoid elements. Flow cytometry is a useful complementary technique to analyze this type of tumors, which provides qualitative and quantitative information. A differential diagnosis can be sometimes made between thymoma and precursor T-lymphoblastic lymphoma (T-LBL). Correct identification is of utmost importance for patient treatment. A total of 38 mediastinal tumors were analyzed, and samples were separated for flow cytometry. Flow cytometry data from thymomas and normal thymic tissue were compared with 42 cases of T-LBL from other anatomical locations. Among 38 mediastinal tumors, we found 6 benign lesions, 9 diffuse large B-cell lymphomas (DLBCL), 10 Hodgkin lymphomas (HL), 11 thymomas and 2 T-LL. Flow cytometry provided positive information in 24 cases, and defined lymphoid neoplastic cells immunophenotype or the typical lymphocytes accompanying thymomas. Flow cytometry helped differentiate 10 cases of HL and 4 benign lesions from other lymphomas (DLBCL, T-LBL, etc.). CD3, CD4 and CD8 expressions were most useful for the differential diagnosis of thymomas and T-LL. To conclude, flow cytometry is a valid complementary technique, which promptly provides information on mediastinal lesions, requiring small quantities of tissue for both early diagnosis and follow up of these diseases.


Subject(s)
Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Young Adult , Antigens, CD/analysis , Biomarkers, Tumor , Flow Cytometry , Mediastinal Neoplasms/pathology , Thymoma/pathology , Thymus Neoplasms/pathology , /analysis , /analysis , /analysis , Hyperplasia , Mediastinal Neoplasms/genetics , Mediastinal Neoplasms/immunology , Phenotype , Retrospective Studies , T-Lymphocytes/immunology , Thymoma/genetics , Thymoma/immunology , Thymus Neoplasms/genetics , Thymus Neoplasms/immunology
10.
Article in English | WPRIM (Western Pacific) | ID: wpr-205422

ABSTRACT

Germ-line mutations in BRCA2 predispose to early-onset cancer. Homozygous mutant mouse, which has Brca2 truncated in exon 11 exhibit paradoxic occurrence of growth retardation and development of thymic lymphomas. However, due to its large embryonic lethality, cohort studies on the thymic lymphomas were not feasible. With the aid of Cre-loxP system, we demonstrate here that thymus-specific disruption of Brca2 allele without crossing it to p53-mutant background leads to the development of thymic lymphomas. Varying from 16 weeks to 66 weeks after birth, 25% of mice disrupted of Brca2 in the thymus died of thymic lymphomas, whereas previous report did not observe lymphomagenesis using similar Cre-loxP system. Future analysis of thymic lymphomas from these mice presented here will provide information on the cooperative mutations that are required for the BRCA2-associated pathogenesis of cancer.


Subject(s)
Animals , Mice , BRCA2 Protein/deficiency , CD4-CD8 Ratio , Cell Separation , Flow Cytometry , Integrases/genetics , Lymphoma/genetics , Mice, Knockout , Organ Specificity , Sequence Deletion , T-Lymphocytes/enzymology , Thymus Gland/immunology , Thymus Neoplasms/genetics , Tumor Suppressor Protein p53/deficiency
11.
Clin. transl. oncol. (Print) ; 8(5): 349-353, mayo 2006. graf
Article in En | IBECS | ID: ibc-047682

ABSTRACT

Background. It is well documented that over-expressionof the c-myc proto-oncogene occurs in thevast majority of mouse thymic lymphomas inducedby gamma-irradiation, evidencing the importance of thisgene in T-cell lymphomagenesis. However, it remainsunknown whether elevated levels of c-mycexpression are driven by extra c-myc copy numbers.Materials and methods. Here we use a quantitativetest on the basis of real-time PCR to determine thecellular copy number of c-myc in a set of 14 g-radiation-induced thymic lymphomas obtained from(C57BL/6J x BALB/cJ) F1 hybrid mice with increasedmRNA c-myc expression.Results. Since 5 out of 14 (35.7%) cases had no extracopy numbers of c-myc, gene amplification was obviouslynot the cause of c-myc over-expression inthese tumours. In the remaining 9 tumours, c-mycover-expression was also accompanied with extraDNA copy numbers. Therefore, c-myc amplificationmight be a consequence of the genomic instabilitysubsequent to the up-regulation of c-myc. However,linear regression analysis showed a lack of correlationbetween increasing DNA copy numbers andmRNA over expression of c-myc in these tumours (r= 0.029, p = 0.94).Conclusion. De-regulation of c-myc does not necessarilyimply amplification of this gene in these tumours.This report is, to our knowledge, the firstone comparing c-myc amplification with expressionin lymphomas of the T-cell lineage


No disponible


Subject(s)
Rats , Animals , Neoplasms, Radiation-Induced , Thymus Neoplasms/genetics , Genetic Markers , Genes, myc , RNA, Messenger/analysis , Polymerase Chain Reaction/methods , Gamma Rays
12.
Article in English | WPRIM (Western Pacific) | ID: wpr-171355

ABSTRACT

Breast cancer susceptibility gene, BRCA2, is a tumor suppressor and individuals who inherit one defected copy of BRCA2 allele experience early onset breast cancer or ovarian cancer accompanied by the loss of the wild type allele. Mouse model for Brca2 mutation shows growth retardation and paradoxical occurrence of thymic lymphomas. Thymic lymphomas from Brca2-mutant mice harbor mutations in p53, Bub1, and BubR1, which function as mitotic checkpoint proteins. Therefore, interplay between Brca2 and mitotic checkpoint has been suggested in the maintenance of genetic fidelity, although it has not been assessed whether the unique mutations in Bub1 and BubR1 found in Brca2-mutant mice are responsible for the abolishment of mitotic checkpoint function. This report demonstrates that Bub1 and BubR1 mutant proteins from Brca2(-/-)thymic lymphomas have defects in the phosphorylation and kinetochore localization after spindle damage. Thus, the mutations of Bub1 and BubR1 found in Brca2- mutant mice indeed are responsible for the chromosome instability in Brca2-mutated tumors.


Subject(s)
Animals , Mice , BRCA2 Protein/genetics , Cell Cycle Proteins , Cell Transformation, Neoplastic/metabolism , Mitosis , Mutation/genetics , Phosphorylation , Protein Kinases/metabolism , Protein Transport , T-Lymphocytes/metabolism , Thymus Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL