Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 138
Filtrer
1.
Int J Mol Sci ; 25(12)2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38928294

RÉSUMÉ

It is known that V-set and immunoglobulin domain containing 1 (VSIG1) is a cell-cell adhesion molecule that can serve as an indicator of better survival in patients with gastric cancer. Its interaction with cytoplasmic thyroid transcription factor 1 (TTF-1) has been hypothesized to characterize gastric-type HCC, but its clinical importance is far from understood. As VSIG1 has also been supposed to be involved in the epithelial-mesenchymal transition (EMT) phenomenon, we checked for the first time in the literature the supposed interaction between VSIG1, TTF-1, and Vimentin (VIM) in HCCs. Immunohistochemical (IHC) stains were performed on 217 paraffin-embedded tissue samples that included tumor cells and normal hepatocytes, which served as positive internal controls. VSIG1 positivity was seen in 113 cases (52.07%). In 71 out of 217 HCCs (32.71%), simultaneous positivity for VSIG1 and TTF-1 was seen, being more specific for G1/G2 carcinomas with a trabecular architecture and a longer OS (p = 0.004). A negative association with VIM was revealed (p < 0.0001). Scirrhous-type HCC proved negative for all three examined markers. The present paper validates the hypothesis of the existence of a gastric-type HCC, which shows a glandular-like architecture and is characterized by double positivity for VSIG1 and TTF-1, vimentin negativity, and a significant OS.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Vimentine , Humains , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Mâle , Femelle , Adulte d'âge moyen , Vimentine/métabolisme , Sujet âgé , Adulte , Marqueurs biologiques tumoraux/métabolisme , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux , Sujet âgé de 80 ans ou plus , Facteur-1 de transcription de la thyroïde/métabolisme , Facteur-1 de transcription de la thyroïde/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Immunohistochimie
2.
PeerJ ; 12: e17338, 2024.
Article de Anglais | MEDLINE | ID: mdl-38708353

RÉSUMÉ

Background: This study was performed to determine the biological processes in which NKX2-1 is involved and thus its role in the development of lung squamous cell carcinoma (LUSC) toward improving the prognosis and treatment of LUSC. Methods: Raw RNA sequencing (RNA-seq) data of LUSC from The Cancer Genome Atlas (TCGA) were used in bioinformatics analysis to characterize NKX2-1 expression levels in tumor and normal tissues. Survival analysis of Kaplan-Meier curve, the time-dependent receiver operating characteristic (ROC) curve, and a nomogram were used to analyze the prognosis value of NKX2-1 for LUSC in terms of overall survival (OS) and progression-free survival (PFS). Then, differentially expressed genes (DEGs) were identified, and Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), and Gene Set Enrichment Analysis (GSEA) were used to clarify the biological mechanisms potentially involved in the development of LUSC. Moreover, the correlation between the NKX2-1 expression level and tumor mutation burden (TMB), tumor microenvironment (TME), and immune cell infiltration revealed that NKX2-1 participates in the development of LUSC. Finally, we studied the effects of NKX2-1 on drug therapy. To validate the protein and gene expression levels of NKX2-1 in LUSC, we employed immunohistochemistry(IHC) datasets, The Gene Expression Omnibus (GEO) database, and qRT-PCR analysis. Results: NKX2-1 expression levels were significantly lower in LUSC than in normal lung tissue. It significantly differed in gender, stage and N classification. The survival analysis revealed that high expression of NKX2-1 had shorter OS and PFS in LUSC. The multivariate Cox regression hazard model showed the NKX2-1 expression as an independent prognostic factor. Then, the nomogram predicted LUSC prognosis. There are 51 upregulated DEGs and 49 downregulated DEGs in the NKX2-1 high-level groups. GO, KEGG and GSEA analysis revealed that DEGs were enriched in cell cycle and DNA replication.The TME results show that NKX2-1 expression was positively associated with mast cells resting, neutrophils, monocytes, T cells CD4 memory resting, and M2 macrophages but negatively associated with M1 macrophages. The TMB correlated negatively with NKX2-1 expression. The pharmacotherapy had great sensitivity in the NKX2-1 low-level group, the immunotherapy is no significant difference in the NKX2-1 low-level and high-level groups. The analysis of GEO data demonstrated concurrence with TCGA results. IHC revealed NKX2-1 protein expression in tumor tissues of both LUAD and LUSC. Meanwhile qRT-PCR analysis indicated a significantly lower NKX2-1 expression level in LUSC compared to LUAD. These qRT-PCR findings were consistent with co-expression analysis of NKX2-1. Conclusion: We conclude that NKX2-1 is a potential biomarker for prognosis and treatment LUSC. A new insights of NKX2-1 in LUSC is still needed further research.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome épidermoïde , Tumeurs du poumon , Facteur-1 de transcription de la thyroïde , Femelle , Humains , Mâle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Carcinome épidermoïde/génétique , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/anatomopathologie , Régulation de l'expression des gènes tumoraux , Estimation de Kaplan-Meier , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Nomogrammes , Pronostic , Facteur-1 de transcription de la thyroïde/génétique , Facteur-1 de transcription de la thyroïde/métabolisme , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique
3.
Nat Commun ; 15(1): 4148, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755149

RÉSUMÉ

Cell plasticity theoretically extends to all possible cell types, but naturally decreases as cells differentiate, whereas injury-repair re-engages the developmental plasticity. Here we show that the lung alveolar type 2 (AT2)-specific transcription factor (TF), CEBPA, restricts AT2 cell plasticity in the mouse lung. AT2 cells undergo transcriptional and epigenetic maturation postnatally. Without CEBPA, both neonatal and mature AT2 cells reduce the AT2 program, but only the former reactivate the SOX9 progenitor program. Sendai virus infection bestows mature AT2 cells with neonatal plasticity where Cebpa mutant, but not wild type, AT2 cells express SOX9, as well as more readily proliferate and form KRT8/CLDN4+ transitional cells. CEBPA promotes the AT2 program by recruiting the lung lineage TF NKX2-1. The temporal change in CEBPA-dependent plasticity reflects AT2 cell developmental history. The ontogeny of AT2 cell plasticity and its transcriptional and epigenetic mechanisms have implications in lung regeneration and cancer.


Sujet(s)
Pneumocytes , Plasticité cellulaire , Facteur-1 de transcription de la thyroïde , Animaux , Souris , Pneumocytes/métabolisme , Pneumocytes/cytologie , Facteur-1 de transcription de la thyroïde/métabolisme , Facteur-1 de transcription de la thyroïde/génétique , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Protéines liant les séquences stimulatrices de type CCAAT/génétique , Différenciation cellulaire , Épigenèse génétique , Souris de lignée C57BL , Lésion pulmonaire/anatomopathologie , Lésion pulmonaire/métabolisme , Lésion pulmonaire/génétique , Régénération , Virus Sendai/génétique , Virus Sendai/physiologie , Prolifération cellulaire , Souris knockout , Poumon/métabolisme
4.
Diagn Cytopathol ; 52(7): E172-E175, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38650538

RÉSUMÉ

We present a case report of a 76-year-old male with a histologically confirmed KRAS mutated, thyroid transcription factor 1 (TTF1) positive, grade 1, mucinous adenocarcinoma with cytologically difficult to interpret lymph node metastasis showing loss of TTF1 expression and overlapping features with goblet cell hyperplasia. The case highlights the importance of molecular testing in aiding diagnosis and guiding treatment of non-small cell lung carcinomas (NSCLC).


Sujet(s)
Tumeurs du poumon , Facteur-1 de transcription de la thyroïde , Humains , Mâle , Sujet âgé , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Facteur-1 de transcription de la thyroïde/métabolisme , Facteur-1 de transcription de la thyroïde/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Cytoponction sous échoendoscopie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Protéines de liaison à l'ADN
5.
Eur Respir J ; 63(5)2024 May.
Article de Anglais | MEDLINE | ID: mdl-38575158

RÉSUMÉ

BACKGROUND: Several rare surfactant-related gene (SRG) variants associated with interstitial lung disease are suspected to be associated with lung cancer, but data are missing. We aimed to study the epidemiology and phenotype of lung cancer in an international cohort of SRG variant carriers. METHODS: We conducted a cross-sectional study of all adults with SRG variants in the OrphaLung network and compared lung cancer risk with telomere-related gene (TRG) variant carriers. RESULTS: We identified 99 SRG adult variant carriers (SFTPA1 (n=18), SFTPA2 (n=31), SFTPC (n=24), ABCA3 (n=14) and NKX2-1 (n=12)), including 20 (20.2%) with lung cancer (SFTPA1 (n=7), SFTPA2 (n=8), SFTPC (n=3), NKX2-1 (n=2) and ABCA3 (n=0)). Among SRG variant carriers, the odds of lung cancer was associated with age (OR 1.04, 95% CI 1.01-1.08), smoking (OR 20.7, 95% CI 6.60-76.2) and SFTPA1/SFTPA2 variants (OR 3.97, 95% CI 1.39-13.2). Adenocarcinoma was the only histological type reported, with programmed death ligand-1 expression ≥1% in tumour cells in three samples. Cancer staging was localised (I/II) in eight (40%) individuals, locally advanced (III) in two (10%) and metastatic (IV) in 10 (50%). We found no somatic variant eligible for targeted therapy. Seven cancers were surgically removed, 10 received systemic therapy, and three received the best supportive care according to their stage and performance status. The median overall survival was 24 months, with stage I/II cancers showing better survival. We identified 233 TRG variant carriers. The comparative risk (subdistribution hazard ratio) for lung cancer in SRG patients versus TRG patients was 18.1 (95% CI 7.1-44.7). CONCLUSIONS: The high risk of lung cancer among SRG variant carriers suggests specific screening and diagnostic and therapeutic challenges. The benefit of regular computed tomography scan follow-up should be evaluated.


Sujet(s)
Tumeurs du poumon , Protéine A associée au surfactant pulmonaire , Protéine C associée au surfactant pulmonaire , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Études transversales , Protéine C associée au surfactant pulmonaire/génétique , Protéine A associée au surfactant pulmonaire/génétique , Adulte , Facteur-1 de transcription de la thyroïde/génétique , Transporteurs ABC/génétique , Facteurs de risque , Prédisposition génétique à une maladie , Pneumopathies interstitielles/génétique , Hétérozygote , Protéines associées au surfactant pulmonaire/génétique
7.
Eur J Cancer ; 197: 113474, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38100920

RÉSUMÉ

OBJECTIVES: Thyroid transcription factor 1 (TTF-1) is a well-established independent prognostic factor in lung adenocarcinoma (LUAD), irrespective of stage. This study aims to determine if TTF-1's prognostic impact is solely based on histomorphological differentiation (tumor grading) or if it independently relates to a biologically more aggressive phenotype. We analyzed a large bi-centric LUAD cohort to accurately assess TTF-1's prognostic value in relation to tumor grade. PATIENTS AND METHODS: We studied 447 patients with resected LUAD from major German lung cancer centers (Berlin and Cologne), correlating TTF-1 status and grading with clinical, pathologic, and molecular data, alongside patient outcomes. TTF-1's impact was evaluated through univariate and multivariate Cox regression. Causal graph analysis was used to identify and account for potential confounders, improving the statistical estimation of TTF-1's predictive power for clinical outcomes. RESULTS: Univariate analysis revealed TTF-1 positivity associated with significantly longer disease-free survival (DFS) (median log HR -0.83; p = 0.018). Higher tumor grade showed a non-significant association with shorter DFS (median log HR 0.30; p = 0,62 for G1 to G2 and 0.68; p = 0,34 for G2 to G3). In multivariate analysis, TTF-1 positivity resulted in a significantly longer DFS (median log HR -0.65; p = 0.05) independent of all other parameters, including grading. Adjusting for potential confounders as indicated by the causal graph confirmed the superiority of TTF-1 over tumor grading in prognostics power. CONCLUSIONS: TTF-1 status predicts relapse and survival in LUAD independently of tumor grading. The prognostic power of tumor grading is limited to TTF-1-positive patients, and the effect size of TTF-1 surpasses that of tumor grading. We recommend including TTF1 status as a prognostic factor in the diagnostic guidelines of LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du poumon , Humains , Facteur-1 de transcription de la thyroïde/génétique , Grading des tumeurs , Stadification tumorale , Récidive tumorale locale/anatomopathologie , Adénocarcinome pulmonaire/anatomopathologie , Tumeurs du poumon/anatomopathologie , Pronostic
8.
J Cell Physiol ; 238(12): 2867-2878, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37850660

RÉSUMÉ

The melanocortin 4 receptor (MC4R) is a G protein-coupled transporter that mediates the regulation of thyroid hormones and leptin on energy balance and food intake. However, the mechanisms of transcriptional regulation of Mc4r by thyroid hormone and leptin in fish have been rarely reported. The messenger RNA expression of Mc4r gene was significantly higher in brain than those in other tissues of mandarin fish. We analyzed the structure and function of a 2029 bp sequence of Mc4r promoter. Meanwhile, overexpression of NKX2.1 and incubation with leptin significantly increased Mc4r promoter activity, but triiodothyronine showed the opposite effect. In addition, mutations in the NKX2.1 binding site abolished not only the activation of Mc4r promoter activity by leptin but also the inhibitory effect of thyroid hormones on Mc4r promoter activity. In summary, these results suggested that thyroid hormones and leptin might regulate the transcriptional expression of Mc4r through NKX2.1.


Sujet(s)
Poissons , Gènes homéotiques , Leptine , Animaux , Protéines de liaison à l'ADN/génétique , Protéines de poisson/génétique , Protéines de poisson/métabolisme , Poissons/génétique , Poissons/métabolisme , Leptine/génétique , Leptine/pharmacologie , Régions promotrices (génétique)/génétique , Récepteur de la mélanocortine de type 4/génétique , Récepteur de la mélanocortine de type 4/métabolisme , Hormones thyroïdiennes , Facteur-1 de transcription de la thyroïde/génétique , Facteur-1 de transcription de la thyroïde/métabolisme , Humains , Cellules HEK293
9.
Lung Cancer ; 176: 121-131, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36634573

RÉSUMÉ

TTF-1-expressing non-small cell lung cancer (NSCLC) is one of the most prevalent lung cancer types worldwide. However, theparadoxical activity of TTF-1 in both lung carcinogenesis and tumor suppression is believed to be context-dependentwhich calls for a deeper understanding about the pathological expression of TTF-1. In addition, the expression circuitry of TTF-1-target genes in NSCLC has not been well examined which necessitates to revisit the involvement of TTF-1- in a multitude of oncologic pathways. We used RNA-seq and clinical data of patients from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx), including ChIP-seq data from different NSCLC cell lines, and mapped the proteome of NSCLC tumor. Our analysis showed significant variability in TTF-1 expression among NSCLC,and further clarified that this variability is orchestrated at the transcriptional level. We also found that high TTF-1 expression could negatively influence the survival outcomes of stage 1 LUAD which may be attributed to growth factor receptor-driven activation of mitogenic and angiogenic pathways. Mechanistically, TTF-1 may also control the genes associated with pathways involved in acquired TKI drug resistance or response to immune checkpoint inhibitors. Lastly, proteome-based biomarker discovery in stage 1 LUAD showed that TTF-1 positivity is potentially associated with the upregulation of several oncogenes which includes interferon proteins, MUC1, STAT3, and EIF2AK2. Collectively, this study highlights the potential involvement of TTF-1 in cell proliferation, immune evasion, and angiogenesis in early-stage NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Facteur-1 de transcription de la thyroïde , Humains , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/anatomopathologie , Protéines nucléaires/génétique , Protéome , Facteur-1 de transcription de la thyroïde/génétique
10.
Gastric Cancer ; 26(1): 44-54, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36094595

RÉSUMÉ

BACKGROUND: Gastric adenocarcinoma of fundic-gland type (GA-FG) is a gastric malignancy with little relation to Helicobacter pylori. Clinical characteristics of GA-FG have been established, but molecular mechanisms leading to tumorigenesis have not yet been elucidated. METHODS: We subjected three GA-FG tumors-normal mucosa pairs to microarray analysis. Network analysis was performed for the top 30 up-regulated gene transcripts, followed by immunohistochemical staining to confirm the gene expression analysis results. AGS and NUGC4 cells were transfected with the gene-encoding NK2 homeobox 1/thyroid transcription factor 1 (NKX2-1/TTF-1) to evaluate transcriptional changes in its target genes. RESULTS: Comprehensive gene expression analysis identified 1410 up-regulated and 1395 down-regulated gene probes with ≥ two-fold difference in expression. Among the top 30 up-regulated genes in GA-FG, we identified transcription factor NKX2-1/TTF-1, a master regulator of lung/thyroid differentiation, together with surfactant protein B (SFTPB), SFTPC, and secretoglobin family 3A member 2(SCGB3A2), which are regulated by NKX2-1/TTF-1. Immunohistochemical analysis of 16 GA-FG specimens demonstrated significantly higher NKX2-1/TTF-1 and SFTPB levels, as compared to that in adjacent normal mucosa (P < 0.05), while SCGB3A2 levels did not differ (P = 0.341). Transduction of NKX2-1/TTF-1 into AGS and NUGC4 cells induced transactivation of SFTPB and SFTPC, indicating that NKX2-1/TTF-1 can function as normally in gastric cells as it can in the lung cells. CONCLUSIONS: Our first transcriptome analysis of GA-FG indicates significant expression of NKX2-1/TTF1 in GA-FG. Immunohistochemistry and cell biology show ectopic expression and normal transactivation ability of NKX2-1/TTF-1, suggesting that it plays an essential role in GA-FG development.


Sujet(s)
Adénocarcinome , Tumeurs de l'estomac , Humains , Facteur-1 de transcription de la thyroïde/génétique , Gènes homéotiques , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Analyse de profil d'expression de gènes
11.
Cells ; 11(23)2022 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-36497134

RÉSUMÉ

A developmental niche vacancy in host embryos is necessary for stem cell complementation-based organ regeneration (SCOG). Thyroid transcription factor 1 (TTF-1) is a tissue-specific transcription factor that regulates the embryonic development and differentiation of the thyroid and, more importantly, lungs; thus, it has been considered as a master gene to knockout in order to develop a lung vacancy host. TTF-1 knockout mice were originally produced by inserting a stop codon in Exon 3 of the gene (E3stop) through embryonic stem cell-based homologous recombination. The main problems of utilizing E3stop host embryos for lung SCOG are that these animals all have a tracheoesophageal fistula (TEF), which cannot be corrected by donor stem cells, and most of them have monolateral sac-like lungs. To improve the mouse model towards achieving SCOG-based lung generation, in this project, we used the CRISPR/Cas9 tool to remove Exon 2 of the gene by zygote microinjection and successfully produced TTF-1 knockout (E2del) mice. Similar to E3stop, E2del mice are birth-lethal due to retarded lung development with sac-like lungs and only a rudimentary bronchial tree, increased basal cells but without alveolar type II cells and blood vessels, and abnormal thyroid development. Unlike E3stop, 57% of the E2del embryos presented type I tracheal agenesis (TA, a kind of human congenital malformation) with a shortened trachea and clear separations of the trachea and esophagus, while the remaining 43% had TEF. Furthermore, all the E2del mice had bilateral sac-like lungs. Both TA and bilateral sac-like lungs are preferred in SCOG. Our work presents a new strategy for producing SCOG host embryos that may be useful for lung regeneration.


Sujet(s)
Systèmes CRISPR-Cas , Facteur-1 de transcription de la thyroïde , Fistule trachéo-oesophagienne , Animaux , Femelle , Humains , Souris , Grossesse , Systèmes CRISPR-Cas/génétique , Modèles animaux de maladie humaine , Poumon , Souris knockout , Facteur-1 de transcription de la thyroïde/génétique
12.
J Zhejiang Univ Sci B ; 23(11): 915-930, 2022 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-36379611

RÉSUMÉ

Targeted gene therapy has become a promising approach for lung cancer treatment. In our previous work, we reported that the targeted expression of microRNA-7 (miR-7) operated by thyroid transcription factor-1 (TTF-1) promoter inhibited the growth of human lung cancer cells in vitro and in vivo; however, the intervention efficiency needed to be further improved. In this study, we identified the core promoter of TTF-1 (from -1299 bp to -871 bp) by 5' deletion assay and screened out the putative transcription factors nuclear factor-1 (NF-1) and activator protein-1 (AP-1). Further analysis revealed that the expression level of NF-1, but not AP-1, was positively connected with the activation of TTF-1 core promoter in human non-small-cell lung cancer (NSCLC) cells. Moreover, the silencing of NF-1 could reduce the expression level of miR-7 operated by TTF-1 core promoter. Of note, we optimized four distinct sequences to form additional NF-1-binding sites (TGGCA) in the sequence of TTF-1 core promoter (termed as optTTF-1 promoter), and verified the binding efficiency of NF-1 on the optTTF-1 promoter by electrophoretic mobility shift assay (EMSA). As expected, the optTTF-1 promoter could more effectively drive miR-7 expression and inhibit the growth of human NSCLC cells in vitro, accompanied by a reduced transduction of NADH dehydrogenase (ubiquinone) 1α subcomplex 4 (NDUFA4)/protein kinase B (Akt) pathway. Consistently, optTTF-1 promoter-driven miR-7 expression could also effectively abrogate the growth and metastasis of tumor cells in a murine xenograft model of human NSCLC. Finally, no significant changes were detected in the biological indicators or the histology of some important tissues and organs, including heart, liver, and spleen. On the whole, our study revealed that the optimized TTF-1 promoter could more effectively operate miR-7 to influence the growth of human NSCLC cells, providing a new basis for the development of microRNA-based targeting gene therapy against clinical lung cancer.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , microARN , Animaux , Humains , Souris , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/thérapie , Tumeurs du poumon/génétique , Tumeurs du poumon/thérapie , Tumeurs du poumon/métabolisme , microARN/génétique , microARN/métabolisme , Protéines nucléaires/métabolisme , Glande thyroide/métabolisme , Glande thyroide/anatomopathologie , Facteur-1 de transcription de la thyroïde/génétique , Facteurs de transcription/métabolisme
13.
Mol Metab ; 66: 101636, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36375792

RÉSUMÉ

OBJECTIVE: Thyroid transcription factor-1 (TTF-1), a homeodomain-containing transcription factor, is predominantly expressed in discrete areas of the hypothalamus, which acts as the central unit for the regulation of whole-body energy homeostasis. Current study designed to identify the roles of TTF-1 on the responsiveness of the hypothalamic circuit activity to circulating leptin and the development of obesity linked to the insensitivity of leptin. METHODS: We generated conditional knock-out mice by crossing TTF-1flox/flox mice with leptin receptor (ObRb)Cre or proopiomelanocortin (POMC)Cre transgenic mice to interrogate the contributions of TTF-1 in leptin signaling and activity. Changes of food intake, body weight and energy expenditure were evaluated in standard or high fat diet-treated transgenic mice by using an indirect calorimetry instrument. Molecular mechanism was elucidated with immunohistochemistry, immunoblotting, quantitative PCR, and promoter assays. RESULTS: The selective deletion of TTF-1 gene expression in cells expressing the ObRb or POMC enhanced the anorexigenic effects of leptin as well as the leptin-induced phosphorylation of STAT3. We further determined that TTF-1 inhibited the transcriptional activity of the ObRb gene. In line with these findings, the selective deletion of the TTF-1 gene in ObRb-positive cells led to protective effects against diet-induced obesity via the amelioration of leptin resistance. CONCLUSIONS: Collectively, these results suggest that hypothalamic TTF-1 participates in the development of obesity as a molecular component involved in the regulation of cellular leptin signaling and activity. Thus, TTF-1 may represent a therapeutic target for the treatment, prevention, and control of obesity.


Sujet(s)
Leptine , Pro-opiomélanocortine , Facteur-1 de transcription de la thyroïde , Animaux , Souris , Hypothalamus/métabolisme , Leptine/génétique , Leptine/métabolisme , Souris knockout , Obésité/génétique , Obésité/métabolisme , Pro-opiomélanocortine/métabolisme , Facteur-1 de transcription de la thyroïde/génétique , Facteur-1 de transcription de la thyroïde/métabolisme
14.
Physiol Int ; 109(2): 261-277, 2022 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-35895569

RÉSUMÉ

Congenital hypothyroidism (CH) occurs with a relatively alarming prevalence in infants, and if not diagnosed and treated in time, it can have devastating consequences for the development of the nervous system. CH is associated with genetic changes in several genes that encode transcription factors responsible for thyroid development, including mutations in the NK2 homeobox 1 (NKX2.1) gene, which encodes the thyroid transcription factor-1 (TTF-1). Although CH is frequently observed in pediatric populations, there is still a limited understanding of the genetic factors and molecular mechanisms contributing to this disease. The sequence of the NKX2.1 gene was investigated in 75 pediatric patients with CH by polymerase chain reaction (PCR), single-stranded conformation polymorphism (SSCP), and direct DNA sequencing. Four missense heterozygous variations were identified in exon 3 of the NKX2.1 gene, including three novel missense variations, namely c.708A>G, p.Gln202Arg; c.713T>G, p.Tyr204Asp; c.833T>G, p.Tyr244Asp, and a previously reported variant rs781133468 (c.772C>G, p.His223Gln). Importantly, these variations occur in highly conserved residues of the TTF-1 DNA-binding domain and were predicted by bioinformatics analysis to alter the protein structure, with a probable alteration in the protein function. These results indicate that nucleotide changes in the NKX2.1 gene may contribute to CH pathogenesis.


Sujet(s)
Hypothyroïdie congénitale , Facteur-1 de transcription de la thyroïde , Enfant , Biologie informatique , Hypothyroïdie congénitale/génétique , Humains , Nourrisson , Iran , Mutation , Facteur-1 de transcription de la thyroïde/génétique , Facteurs de transcription/génétique
15.
Am J Respir Crit Care Med ; 206(12): 1480-1494, 2022 12 15.
Article de Anglais | MEDLINE | ID: mdl-35848993

RÉSUMÉ

Rationale: The current molecular classification of small-cell lung cancer (SCLC) on the basis of the expression of four lineage transcription factors still leaves its major subtype SCLC-A as a heterogeneous group, necessitating more precise characterization of lineage subclasses. Objectives: To refine the current SCLC classification with epigenomic profiles and to identify features of the redefined SCLC subtypes. Methods: We performed unsupervised clustering of epigenomic profiles on 25 SCLC cell lines. Functional significance of NKX2-1 (NK2 homeobox 1) was evaluated by cell growth, apoptosis, and xenograft using clustered regularly interspaced short palindromic repeats-Cas9 (CRISPR-associated protein 9)-mediated deletion. NKX2-1-specific cistromic profiles were determined using chromatin immunoprecipitation followed by sequencing, and its functional transcriptional partners were determined using coimmunoprecipitation followed by mass spectrometry. Rb1flox/flox; Trp53flox/flox and Rb1flox/flox; Trp53flox/flox; Nkx2-1flox/flox mouse models were engineered to explore the function of Nkx2-1 in SCLC tumorigenesis. Epigenomic landscapes of six human SCLC specimens and 20 tumors from two mouse models were characterized. Measurements and Main Results: We identified two epigenomic subclusters of the major SCLC-A subtype: SCLC-Aα and SCLC-Aσ. SCLC-Aα was characterized by the presence of a super-enhancer at the NKX2-1 locus, which was observed in human SCLC specimens and a murine SCLC model. We found that NKX2-1, a dual lung and neural lineage factor, is uniquely relevant in SCLC-Aα. In addition, we found that maintenance of this neural identity in SCLC-Aα is mediated by collaborative transcriptional activity with another neuronal transcriptional factor, SOX1 (SRY-box transcription factor 1). Conclusions: We comprehensively describe additional epigenomic heterogeneity of the major SCLC-A subtype and define the SCLC-Aα subtype by the core regulatory circuitry of NKX2-1 and SOX1 super-enhancers and their functional collaborations to maintain neuronal linage state.


Sujet(s)
Tumeurs du poumon , Facteurs de transcription SOX-B1 , Carcinome pulmonaire à petites cellules , Facteur-1 de transcription de la thyroïde , Animaux , Humains , Souris , Transformation cellulaire néoplasique , Poumon , Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/anatomopathologie , Facteurs de transcription SOX-B1/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Facteur-1 de transcription de la thyroïde/génétique
16.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(3): 396-400, 2022 Mar 28.
Article de Anglais, Chinois | MEDLINE | ID: mdl-35545334

RÉSUMÉ

Brain-lung-thyroid syndrome is a rare autosomal dominant disorder. More than 100 cases have been reported worldwide, but few cases have been reported in China. In December 2018, a boy with brain-lung-thyroid syndrome, aged 3 years and 10 months, was admitted to Xiangya Hospital of Central South University due to repeated cough for more than 3 years. In infancy of the boy, psychomotor retardation, repeated cough, and hypothyroidism were found. Gene detection showed that there was c.927delc heterozygous variation in NKX2-1 gene (NM-001079668: exon3: c.927delC). The variation of this gene locus has not been reported in relevant literature so far, which indicates a new mutation. According to the above clinical manifestations and examination results, the boy was diagnosed as brain-lung-thyroid syndrome, which mainly characterized by nervous system disorders, accompanied by respiratory manifestations and hypothyroidism. The boy was treated with oral dopasehydrazine to relieve tremor and levothyroxine sodium tablets to relieve hypothyroidism. Anti-infection, atomization, rehabilitation training and other symptomatic supporting treatment were also administered. The boy's language and movement have improved, the thyroid hormone level is normal, and there are still repeated respiratory tract infections.


Sujet(s)
Hypothyroïdie congénitale , Toux , Athétose/génétique , Chorée , Hypothyroïdie congénitale/diagnostic , Hypothyroïdie congénitale/génétique , Humains , Mâle , Syndrome de détresse respiratoire du nouveau-né , Facteur-1 de transcription de la thyroïde/génétique
17.
Neuroradiology ; 64(5): 1033-1035, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-35199208

RÉSUMÉ

The thyroid transcription factor 1 (TITF-1) gene plays an important role in the development of the ventral forebrain, thyroid and lungs. Mutations of this gene are known to cause benign hereditary chorea (BHC) and can cause the full spectrum of abnormalities seen in the brain-thyroid-lung syndrome. Abnormalities of the ventral forebrain on imaging have been variably documented in the literature. Multiple previous reports describe a cystic pituitary mass, as well as duplication of the pituitary stalk and communication between an intrasellar cyst and the third ventricle. The initial MRI performed in our case was interpreted as an intrasellar cyst, but the high-resolution MRI performed later was able to resolve this as a persisting embryonal infundibular recess (PEIR), rather than the cystic pituitary mass which has previously been described. This case illustrates the role of the TITF-1 gene in the development of the pituitary and hypothalamus.


Sujet(s)
Chorée , Kystes , Troisième ventricule , Chorée/génétique , Humains , Mutation , Hypophyse/imagerie diagnostique , Facteur-1 de transcription de la thyroïde/génétique
18.
Sci Rep ; 12(1): 2144, 2022 02 09.
Article de Anglais | MEDLINE | ID: mdl-35140269

RÉSUMÉ

While the signaling pathways and transcription factors involved in the differentiation of thyroid follicular cells, both in embryonic and adult life, are increasingly well understood, the underlying mechanisms and potential crosstalk between the thyroid transcription factors Nkx2.1, Foxe1 and Pax8 and inductive signals remain unclear. Here, we focused on the transcription factor Sox9, which is expressed in Nkx2.1-positive embryonic thyroid precursor cells and is maintained from embryonic development to adulthood, but its function and control are unknown. We show that two of the main signals regulating thyroid differentiation, TSH and TGFß, modulate Sox9 expression. Specifically, TSH stimulates the cAMP/PKA pathway to transcriptionally upregulate Sox9 mRNA and protein expression, a mechanism that is mediated by the binding of CREB to a CRE site within the Sox9 promoter. Contrastingly, TGFß signals through Smad proteins to inhibit TSH-induced Sox9 transcription. Our data also reveal that Sox9 transcription is regulated by the thyroid transcription factors, particularly Pax8. Interestingly, Sox9 significantly increased the transcriptional activation of Pax8 and Foxe1 promoters and, consequently, their expression, but had no effect on Nkx2.1. Our study establishes the involvement of Sox9 in thyroid follicular cell differentiation and broadens our understanding of transcription factor regulation of thyroid function.


Sujet(s)
Facteur de transcription SOX-9/métabolisme , Cellules épithéliales thyroïdiennes/cytologie , Cellules épithéliales thyroïdiennes/métabolisme , Glande thyroide/métabolisme , Thyréostimuline/métabolisme , Facteurs de transcription/métabolisme , Facteur de croissance transformant bêta/métabolisme , Animaux , Différenciation cellulaire , Lignée cellulaire , AMP cyclique/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Régulation de l'expression des gènes , Souris , Facteur de transcription PAX-8/génétique , Facteur de transcription PAX-8/métabolisme , Régions promotrices (génétique) , Facteur de transcription SOX-9/génétique , Transduction du signal , Glande thyroide/cytologie , Glande thyroide/embryologie , Facteur-1 de transcription de la thyroïde/génétique , Facteur-1 de transcription de la thyroïde/métabolisme , Thyréostimuline/pharmacologie , Transcription génétique , Facteur de croissance transformant bêta/pharmacologie
19.
J Pediatr Endocrinol Metab ; 35(3): 411-415, 2022 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-34710315

RÉSUMÉ

OBJECTIVES: Benign Hereditary Chorea (BHC) (MIM 118700) is a rare childhood-onset movements disorder characterized by non-progressive chorea. It is usually caused by variants in the thyroid transcription factor 1 (TITF-1/NKX2-1) gene and it is associated with thyroid dysfunction and pulmonary symptoms in the brain-lung-thyroid syndrome. CASE PRESENTATION: We reported the clinical case of a toddler presenting with neurological symptoms (hypotonia, delayed motor milestones, and axial dystonia) and subclinical hypothyroidism in which we found a 'de novo' variant in the NKX2-1 gene. CONCLUSIONS: The peculiarity of our case is that the mild alteration of thyroid-stimulating hormone (TSH) levels, hypotonia, and delayed motor milestones were associated with growth hormone deficiency.


Sujet(s)
Chorée , Hypothyroïdie congénitale , Enfant , Chorée/complications , Chorée/génétique , Hypothyroïdie congénitale/génétique , Hormone de croissance/génétique , Humains , Mutation , Protéines nucléaires/génétique , Facteur-1 de transcription de la thyroïde/génétique , Facteurs de transcription/génétique
20.
J Natl Cancer Inst ; 114(2): 290-301, 2022 02 07.
Article de Anglais | MEDLINE | ID: mdl-34524427

RÉSUMÉ

BACKGROUND: Approximately 20% of lung adenocarcinoma (LUAD) is negative for the lineage-specific oncogene Thyroid transcription factor 1 (TTF-1) and exhibits worse clinical outcome with a low frequency of actionable genomic alterations. To identify molecular features associated with TTF-1-negative LUAD, we compared the transcriptomic and proteomic profiles of LUAD cell lines. SRGN , a chondroitin sulfate proteoglycan Serglycin, was identified as a markedly overexpressed gene in TTF-1-negative LUAD. We therefore investigated the roles and regulation of SRGN in TTF-1-negative LUAD. METHODS: Proteomic and metabolomic analyses of 41 LUAD cell lines were done using mass spectrometry. The function of SRGN was investigated in 3 TTF-1-negative and 4 TTF-1-positive LUAD cell lines and in a syngeneic mouse model (n = 5 to 8 mice per group). Expression of SRGN was evaluated in 94 and 105 surgically resected LUAD tumor specimens using immunohistochemistry. All statistical tests were 2-sided. RESULTS: SRGN was markedly overexpressed at mRNA and protein levels in TTF-1-negative LUAD cell lines (P < .001 for both mRNA and protein levels). Expression of SRGN in LUAD tumor tissue was associated with poor outcome (hazard ratio = 4.22, 95% confidence interval = 1.12 to 15.86, likelihood ratio test, P = .03), and with higher expression of Programmed cell death 1 ligand 1 (PD-L1) in tumor cells and higher infiltration of Programmed cell death protein 1-positive lymphocytes. SRGN regulated expression of PD-L1 as well as proinflammatory cytokines, including Interleukin-6, Interleukin-8, and C-X-C motif chemokine 1 in LUAD cell lines; increased migratory and invasive properties of LUAD cells and fibroblasts; and enhanced angiogenesis. SRGN was induced by DNA demethylation resulting from Nicotinamide N-methyltransferase-mediated impairment of methionine metabolism. CONCLUSIONS: Our findings suggest that SRGN plays a pivotal role in tumor-stromal interaction and reprogramming into an aggressive and immunosuppressive tumor microenvironment in TTF-1-negative LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Protéines de liaison à l'ADN , Tumeurs du poumon , Protéoglycanes , Facteurs de transcription , Protéines du transport vésiculaire , Adénocarcinome pulmonaire/génétique , Animaux , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Souris , Phénotype , Protéoglycanes/métabolisme , Protéomique , Facteur-1 de transcription de la thyroïde/génétique , Microenvironnement tumoral , Protéines du transport vésiculaire/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...