Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 145
1.
Nat Commun ; 15(1): 4148, 2024 May 16.
Article En | MEDLINE | ID: mdl-38755149

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.


Alveolar Epithelial Cells , Cell Plasticity , Thyroid Nuclear Factor 1 , Animals , Mice , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/cytology , Thyroid Nuclear Factor 1/metabolism , Thyroid Nuclear Factor 1/genetics , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cell Differentiation , Epigenesis, Genetic , Mice, Inbred C57BL , Lung Injury/pathology , Lung Injury/metabolism , Lung Injury/genetics , Regeneration , Sendai virus/genetics , Sendai virus/physiology , Cell Proliferation , Mice, Knockout , Lung/metabolism
2.
PeerJ ; 12: e17338, 2024.
Article En | MEDLINE | ID: mdl-38708353

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.


Biomarkers, Tumor , Carcinoma, Squamous Cell , Lung Neoplasms , Thyroid Nuclear Factor 1 , Female , Humans , Male , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Nomograms , Prognosis , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics
3.
Diagn Cytopathol ; 52(7): E172-E175, 2024 Jul.
Article En | MEDLINE | ID: mdl-38650538

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).


Lung Neoplasms , Thyroid Nuclear Factor 1 , Humans , Male , Aged , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Thyroid Nuclear Factor 1/metabolism , Thyroid Nuclear Factor 1/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Endoscopic Ultrasound-Guided Fine Needle Aspiration , Transcription Factors/genetics , Transcription Factors/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , DNA-Binding Proteins
4.
Anticancer Res ; 44(5): 2159-2170, 2024 May.
Article En | MEDLINE | ID: mdl-38677751

BACKGROUND/AIM: Both mesonephric adenocarcinoma (MA) and mesonephric-like adenocarcinoma (MLA) express thyroid transcription factor 1 (TTF1). TTF1 is also considered a highly sensitive and specific diagnostic marker for primary lung adenocarcinoma (PLA). However, distinguishing PLA from pulmonary metastatic MA/MLA (PMM) based on the expression of TTF1 alone can be difficult. This study aimed to investigate the expression of TTF1 and paired box 8 (PAX8) and assess their value in distinguishing PMM from PLA. PATIENTS AND METHODS: We reviewed the electronic medical records and pathology slides of eight PMM cases. We conducted immunostaining for TTF1 and PAX8 in 6, 8, and 21 cases of primary MA/MLA, PMM, and PLA, respectively. RESULTS: Two patients with stage IB uterine MLA developed lung metastases at 5 and 57 months after hysterectomy. Solitary pulmonary nodules were suspected to be primary lung cancer in two patients. Compared to primary tumors, all matched PMMs exhibited reduced TTF1 immunoreactivity. In contrast, the majority of PLAs showed uniform and intense TTF1 expression. All except one PMM exhibited diffuse and strong PAX8 expression, while only one PLA showed focal and weak PAX8 expression. CONCLUSION: Immunostaining for TTF1 and PAX8 can help in distinguishing PMM from PLA in the diagnosis of pulmonary lesions detected in patients with a history of MA/MLA.


Adenocarcinoma of Lung , Adenocarcinoma , Biomarkers, Tumor , DNA-Binding Proteins , Immunohistochemistry , Lung Neoplasms , PAX8 Transcription Factor , Female , Humans , Male , Adenocarcinoma/metabolism , Adenocarcinoma/diagnosis , Adenocarcinoma/secondary , Adenocarcinoma/pathology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/secondary , Biomarkers, Tumor/metabolism , Diagnosis, Differential , Lung Neoplasms/metabolism , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Lung Neoplasms/secondary , PAX8 Transcription Factor/metabolism , Thyroid Nuclear Factor 1/metabolism , Transcription Factors/metabolism
5.
Thorac Cancer ; 15(12): 1029-1033, 2024 Apr.
Article En | MEDLINE | ID: mdl-38478955

Most lung carcinomas are subtyped by their morphologies; however, immunohistochemistry is usually performed when it is difficult to determine. The most reliable antibodies for distinguishing lung adenocarcinoma from squamous cell carcinoma are thyroid transcription factor-1 (TTF-1) and p40 (ΔNp63). In general, these markers are mutually exclusive in their expression of lung primary carcinoma; however, a few cases of non-small cell lung carcinoma (NSCLC) with coexpression of both markers have been reported. Examining a tissue microarray of 229 squamous cell carcinomas and 346 adenocarcinomas, we found one case of NSCLC with coexpression of TTF-1 and p40. Herein, we present a 71-year-old man, who had a mass lesion in the left lung apex. A transbronchial lung biopsy was performed, revealing NSCLC. He underwent left upper segmentectomy and lymph node dissection. Macroscopically, the mass showed a white-to-tan solid tumor on the cut surface. Microscopically, the tumor was composed of polygonal tumor cells which had round and vesicular nuclei with prominent nucleoli. They had an abundant amount of cytoplasm, which was slightly eosinophilic or amphophilic. Multinucleated cells with atypical nuclear features were observed to be scattered in some areas. Multifocal necrosis and hemorrhage were also noted. Distinct squamous features and obvious glandular features were absent. Immunohistochemically, the most tumor cells were coexpressed positive for both TTF-1 and p40. In our study, NSCLC with TTF-1 and p40 coexpression is rare; therefore, it is necessary to obtain further data and examine similar cases to establish more precise definitions and clinicopathological features.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Thyroid Nuclear Factor 1 , Transcription Factors , Tumor Suppressor Proteins , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Male , Aged , Thyroid Nuclear Factor 1/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Transcription Factors/metabolism , Biomarkers, Tumor/metabolism
6.
Endocrine ; 83(2): 519-526, 2024 Feb.
Article En | MEDLINE | ID: mdl-37775725

PURPOSE: Thyroid transcription factor-1 (TTF-1) assessed by immunohistochemistry (IHC) is a specific biomarker for lung adenocarcinoma, and is commonly used to confirm the pulmonary origin of neuroendocrine tumours (NET). The majority of the available data suggest that TTF-1 is favourable prognostic biomarker for lung adenocarcinomas, whereas its role is more conflicting for lung NET. The main aim of this multicenter retrospective study was to investigate the potentially relevant associations between TTF-1 biomarker and clinical and pathological features of the study population, as well as determine TTF-1 prognostic effect on the clinical outcome of the patients. METHODS: A multicentre retrospective study was conducted on 155 surgically-removed lung NET, with available IHC TTF-1 assessment. RESULTS: Median age was 59.5 years (range 13-86), 97 patients (62.6%) were females, 31 cases (20%) were atypical carcinoids, 4 (2.6%) had TNM stage IV. Mitotic count ≥2 per 10 high-power field was found in 35 (22.6%) subjects, whereas necrosis was detected in 20 patients (12.9%). TTF-1 was positive in 78 cases (50.3%). The median overall survival was 46.9 months (range 0.6-323) and the median progression-free survival was 39.1 months (range 0.6-323). Statistically significant associations were found between (1) TTF-1 positivity and female sex (p = 0.007); and among (2) TTF-1 positivity and the absence of necrosis (p = 0.018). CONCLUSIONS: This study highlights that TTF-1 positivity differs according to sex in lung NET, with a more common TTF-1 positive staining in female. Moreover, TTF-1 positivity correlated with the absence of necrosis. These data suggest that TTF-1 could potentially represent a gender-related biomarker for lung NET.


Adenocarcinoma of Lung , Carcinoma, Neuroendocrine , Lung Neoplasms , Neuroendocrine Tumors , Humans , Female , Adolescent , Young Adult , Adult , Middle Aged , Aged , Aged, 80 and over , Male , Neuroendocrine Tumors/metabolism , Retrospective Studies , Thyroid Gland/pathology , Biomarkers, Tumor/metabolism , Thyroid Nuclear Factor 1/metabolism , Lung/metabolism , Necrosis
7.
J Cell Physiol ; 238(12): 2867-2878, 2023 12.
Article En | MEDLINE | ID: mdl-37850660

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.


Fishes , Genes, Homeobox , Leptin , Animals , DNA-Binding Proteins/genetics , Fish Proteins/genetics , Fish Proteins/metabolism , Fishes/genetics , Fishes/metabolism , Leptin/genetics , Leptin/pharmacology , Promoter Regions, Genetic/genetics , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Thyroid Hormones , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism , Humans , HEK293 Cells
8.
Br J Cancer ; 128(10): 1862-1878, 2023 05.
Article En | MEDLINE | ID: mdl-36932191

BACKGROUND: One-third of cancers activate endogenous synthesis of serine/glycine, and can become addicted to this pathway to sustain proliferation and survival. Mechanisms driving this metabolic rewiring remain largely unknown. METHODS: NKX2-1 overexpressing and NKX2-1 knockdown/knockout T-cell leukaemia and lung cancer cell line models were established to study metabolic rewiring using ChIP-qPCR, immunoblotting, mass spectrometry, and proliferation and invasion assays. Findings and therapeutic relevance were validated in mouse models and confirmed in patient datasets. RESULTS: Exploring T-cell leukaemia, lung cancer and neuroendocrine prostate cancer patient datasets highlighted the transcription factor NKX2-1 as putative driver of serine/glycine metabolism. We demonstrate that transcription factor NKX2-1 binds and transcriptionally upregulates serine/glycine synthesis enzyme genes, enabling NKX2-1 expressing cells to proliferate and invade in serine/glycine-depleted conditions. NKX2-1 driven serine/glycine synthesis generates nucleotides and redox molecules, and is associated with an altered cellular lipidome and methylome. Accordingly, NKX2-1 tumour-bearing mice display enhanced tumour aggressiveness associated with systemic metabolic rewiring. Therapeutically, NKX2-1-expressing cancer cells are more sensitive to serine/glycine conversion inhibition by repurposed anti-depressant sertraline, and to etoposide chemotherapy. CONCLUSION: Collectively, we identify NKX2-1 as a novel transcriptional regulator of serine/glycine synthesis addiction across cancers, revealing a therapeutic vulnerability of NKX2-1-driven cancers. Transcription factor NKX2-1 fuels cancer cell proliferation and survival by hyperactivating serine/glycine synthesis, highlighting this pathway as a novel therapeutic target in NKX2-1-positive cancers.


Lung Neoplasms , Serine , Animals , Humans , Mice , Cell Line , Cell Line, Tumor , Glycine , Lung Neoplasms/pathology , Serine/metabolism , Thyroid Nuclear Factor 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Mol Metab ; 66: 101636, 2022 12.
Article En | MEDLINE | ID: mdl-36375792

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.


Leptin , Pro-Opiomelanocortin , Thyroid Nuclear Factor 1 , Animals , Mice , Hypothalamus/metabolism , Leptin/genetics , Leptin/metabolism , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism
10.
Development ; 149(20)2022 10 15.
Article En | MEDLINE | ID: mdl-36039869

Bud tip progenitors (BTPs) in the developing lung give rise to all epithelial cell types found in the airways and alveoli. This work aimed to develop an iPSC organoid model enriched with NKX2-1+ BTP-like cells. Building on previous studies, we optimized a directed differentiation paradigm to generate spheroids with more robust NKX2-1 expression. Spheroids were expanded into organoids that possessed NKX2-1+/CPM+ BTP-like cells, which increased in number over time. Single cell RNA-sequencing analysis revealed a high degree of transcriptional similarity between induced BTPs (iBTPs) and in vivo BTPs. Using FACS, iBTPs were purified and expanded as induced bud tip progenitor organoids (iBTOs), which maintained an enriched population of bud tip progenitors. When iBTOs were directed to differentiate into airway or alveolar cell types using well-established methods, they gave rise to organoids composed of organized airway or alveolar epithelium, respectively. Collectively, iBTOs are transcriptionally and functionally similar to in vivo BTPs, providing an important model for studying human lung development and differentiation.


Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Thyroid Nuclear Factor 1/metabolism , Alveolar Epithelial Cells , Cell Differentiation , Humans , Lung , Organoids
11.
Sci Rep ; 12(1): 2144, 2022 02 09.
Article En | MEDLINE | ID: mdl-35140269

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.


SOX9 Transcription Factor/metabolism , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/metabolism , Thyroid Gland/metabolism , Thyrotropin/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Cell Line , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Mice , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , Promoter Regions, Genetic , SOX9 Transcription Factor/genetics , Signal Transduction , Thyroid Gland/cytology , Thyroid Gland/embryology , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism , Thyrotropin/pharmacology , Transcription, Genetic , Transforming Growth Factor beta/pharmacology
12.
Acta Neurochir (Wien) ; 164(3): 737-742, 2022 03.
Article En | MEDLINE | ID: mdl-35029761

Posterior pituitary tumors (PPT) expressing thyroid transcription factor-1 (TTF-1) are extremely rare low-grade neoplasms. The recent discovery of BRAF mutations in these tumors offers a potential alternative treatment using targeted therapies. We present the case of a 57-year-old female with recurrent BRAFV600E-mutated TTF-1-positive PPT treated with a BRAF inhibitor monotherapy (dabrafenib) leading to tumor regression. After 18 months of uninterrupted treatment, ongoing radiological tumor regression was observed and the patient remained asymptomatic without any significant adverse event. BRAF inhibitor is potentially a valuable treatment option for recurrent TTF-1-positive PPT with BRAF mutation.


Pituitary Neoplasms , Proto-Oncogene Proteins B-raf/genetics , Female , Humans , Imidazoles/therapeutic use , Middle Aged , Mutation/genetics , Oximes/therapeutic use , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/genetics , Protein Kinase Inhibitors , Thyroid Nuclear Factor 1/metabolism
13.
Oncogene ; 41(2): 293-300, 2022 01.
Article En | MEDLINE | ID: mdl-34689179

The RAS→RAF→MEK→ERK pathway is hyperactivated in the majority of human lung adenocarcinoma (LUAD). However, the initial activating mutations induce homeostatic feedback mechanisms that limit ERK activity. How ERK activation reaches the tumor-promoting levels that overcome the feedback and drive malignant progression is unclear. We show here that the lung lineage transcription factor NKX2-1 suppresses ERK activity. In human tissue samples and cell lines, xenografts, and genetic mouse models, NKX2-1 induces the ERK phosphatase DUSP6, which inactivates ERK. In tumor cells from late-stage LUAD with silenced NKX2-1, re-introduction of NKX2-1 induces DUSP6 and inhibits tumor growth and metastasis. We show that DUSP6 is necessary for NKX2-1-mediated inhibition of tumor progression in vivo and that DUSP6 expression is sufficient to inhibit RAS-driven LUAD. Our results indicate that NKX2-1 silencing, and thereby DUSP6 downregulation, is a mechanism by which early LUAD can unleash ERK hyperactivation for tumor progression.


Dual Specificity Phosphatase 6/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lung Neoplasms/genetics , MAP Kinase Signaling System/genetics , Thyroid Nuclear Factor 1/metabolism , Animals , Disease Models, Animal , Disease Progression , Humans , Mice
14.
J Comp Neurol ; 530(5): 834-855, 2022 04.
Article En | MEDLINE | ID: mdl-34547112

The distribution patterns of a set of conserved brain developmental regulatory transcription factors were analyzed in the forebrain of the basal actinopterygian fish Acipenser ruthenus, consistent with the prosomeric model. In the telencephalon, the pallium was characterized by ventricular expression of Pax6. In the subpallium, the combined expression of Nkx2.1/Islet-1 (Isl1) allowed to propose ventral and dorsal areas, as the septo-pallidal (Nkx2.1/Isl1+) and striatal derivatives (Isl1+), respectively, and a dorsal portion of the striatal derivatives, ventricularly rich in Pax6 and devoid of Isl1 expression. Dispersed Orthopedia (Otp) cells were found in the supracommissural and posterior nuclei of the ventral telencephalon, related to the medial portion of the amygdaloid complex. The preoptic area was identified by the Nkx2.1/Isl1 expression. In the alar hypothalamus, an Otp-expressing territory, lacking Nkx2.1/Isl1, was identified as the paraventricular domain. The adjacent subparaventricular domain (Spa) was subdivided in a rostral territory expressing Nkx2.1 and an Isl1+ caudal one. In the basal hypothalamus, the tuberal region was defined by the Nkx2.1/Isl1 expression and a rostral Otp-expressing domain was identified. Moreover, the Otp/Nkx2.1 combination showed an additional zone lacking Isl1, tentatively identified as the mamillary area. In the diencephalon, both Pax6 and Isl1 defined the prethalamic domain, and within the basal prosomere 3, scattered Pax6- and Isl1-expressing cells were observed in the posterior tubercle. Finally, a small group of Pax6 cells was observed in the pretectal area. These results improve the understanding of the forebrain evolution and demonstrate that its basic bauplan is present very early in the vertebrate lineage.


Fish Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , PAX6 Transcription Factor/metabolism , Prosencephalon/metabolism , Thyroid Nuclear Factor 1/metabolism , Transcription Factors/metabolism , Animals , Fishes
15.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article En | MEDLINE | ID: mdl-34916298

The thyroid maintains systemic homeostasis by regulating serum thyroid hormone concentrations. Here we report the establishment of three-dimensional (3D) organoids from adult thyroid tissue representing murine and human thyroid follicular cells (TFCs). The TFC organoids (TFCOs) harbor the complete machinery of hormone production as visualized by the presence of colloid in the lumen and by the presence of essential transporters and enzymes in the polarized epithelial cells that surround a central lumen. Both the established murine as human thyroid organoids express canonical thyroid markers PAX8 and NKX2.1, while the thyroid hormone precursor thyroglobulin is expressed at comparable levels to tissue. Single-cell RNA sequencing and transmission electron microscopy confirm that TFCOs phenocopy primary thyroid tissue. Thyroid hormones are readily detectable in conditioned medium of human TFCOs. We show clinically relevant responses (increased proliferation and hormone secretion) of human TFCOs toward a panel of Graves' disease patient sera, demonstrating that organoids can model human autoimmune disease.


Gene Expression Regulation/physiology , Graves Disease/metabolism , Organoids/metabolism , Thyroid Epithelial Cells/physiology , Animals , Culture Media , Humans , Mice , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , Thyroglobulin/genetics , Thyroglobulin/metabolism , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism
16.
Elife ; 102021 12 01.
Article En | MEDLINE | ID: mdl-34851821

The septum is a ventral forebrain structure known to regulate innate behaviors. During embryonic development, septal neurons are produced in multiple proliferative areas from neural progenitors following transcriptional programs that are still largely unknown. Here, we use a combination of single-cell RNA sequencing, histology, and genetic models to address how septal neuron diversity is established during neurogenesis. We find that the transcriptional profiles of septal progenitors change along neurogenesis, coinciding with the generation of distinct neuron types. We characterize the septal eminence, an anatomically distinct and transient proliferative zone composed of progenitors with distinctive molecular profiles, proliferative capacity, and fate potential compared to the rostral septal progenitor zone. We show that Nkx2.1-expressing septal eminence progenitors give rise to neurons belonging to at least three morphological classes, born in temporal cohorts that are distributed across different septal nuclei in a sequential fountain-like pattern. Our study provides insight into the molecular programs that control the sequential production of different neuronal types in the septum, a structure with important roles in regulating mood and motivation.


Neurogenesis/genetics , Neurons/physiology , Septum of Brain/physiology , Thyroid Nuclear Factor 1/genetics , Transcription, Genetic , Animals , Female , Gene Expression Profiling , Male , Mice , Thyroid Nuclear Factor 1/metabolism
17.
Biomed Res Int ; 2021: 9957209, 2021.
Article En | MEDLINE | ID: mdl-34631891

Thyroid transcription factor-1 (TTF-1/NKx2.1) is a member of the NKx2 tissue-specific transcription factor family, which is expressed in thyroid follicle, parathyroid gland, alveolar epithelium, and diencephalon which originated from ectoderm, and participates in the differentiation, development, and functional maintenance of the above organs. Recent studies have shown that the abnormal expression of TTF-1 is closely related to the occurrence of a variety of human diseases and can be used as a potential new target for the diagnosis and treatment of related diseases. In this article, in order to strengthen the systematic understanding of TTF-1 and promote the progress of related research, we reviewed the structure, expression regulation, biological functions of TTF-1, and its role in the occurrence and development of human-related clinical diseases. Meanwhile, we prospect the future research direction of TTF-1, which might ultimately contribute to the understanding of the pathogenesis of related clinical diseases and the development of new prevention and treatment strategies.


Disease , Thyroid Nuclear Factor 1/chemistry , Thyroid Nuclear Factor 1/metabolism , Animals , Gene Expression Regulation , Humans , Models, Biological , Thyroid Nuclear Factor 1/genetics
18.
Oncoimmunology ; 10(1): 1957603, 2021.
Article En | MEDLINE | ID: mdl-34377595

Anti-PD1/PD-L1-directed immune checkpoint inhibitors are game changers in advanced non-small-cell lung cancer, but biomarkers are lacking. The aim of our study was to find clinically relevant biomarkers of the efficacy of ICI in non-squamous NSCLC. We conducted a retrospective study of patients receiving ICI for advanced non squamous NSCLC in two cohorts. For a subset of patients, RNAseq data were generated on tumor biopsy taken before ICI. The primary end point was progression-free survival under ICI. Secondary end point was overall survival from ICI initiation. In the cohort, we studied 231 patients. Clinico-pathological characteristics included KRAS mutant status (n = 88), TTF1-positive expression (n = 136), LIPI (Lung Immune Prognostic Index) score of 0 (n = 116). In our cohort, lack of TTF1 expression, LIPI score >0, line of treatment >1, and liver metastases were associated with poorer PFS. TTF1 and PD-L1 status could be used to stratify survival and improve the AUC for prediction of prognosis in comparison with the PD-L1 gold standard. Using an external cohort of 154 patients, we confirmed the independent prognostic role of TTF1. TTF1 expression and PD-L1 can be used to stratify risk and predict PFS and OS in patients treated with ICI for NS-NSCLC.


Adenocarcinoma of Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Thyroid Nuclear Factor 1/metabolism , Biomarkers/metabolism , Humans , Prognosis , Retrospective Studies
19.
Front Endocrinol (Lausanne) ; 12: 654569, 2021.
Article En | MEDLINE | ID: mdl-33959098

Thyroid differentiation of progenitor cells occurs during embryonic development and in the adult thyroid gland, and the molecular bases of these complex and finely regulated processes are becoming ever more clear. In this Review, we describe the most recent advances in the study of transcription factors, signaling molecules and regulatory pathways controlling thyroid differentiation and development in the mammalian embryo. We also discuss the maintenance of the adult differentiated phenotype to ensure the biosynthesis of thyroid hormones. We will focus on endoderm-derived thyroid epithelial cells, which are responsible for the formation of the thyroid follicle, the functional unit of the thyroid gland. The use of animal models and pluripotent stem cells has greatly aided in providing clues to the complicated puzzle of thyroid development and function in adults. The so-called thyroid transcription factors - Nkx2-1, Foxe1, Pax8 and Hhex - were the first pieces of the puzzle identified in mice. Other transcription factors, either acting upstream of or directly with the thyroid transcription factors, were subsequently identified to, almost, complete the puzzle. Among them, the transcription factors Glis3, Sox9 and the cofactor of the Hippo pathway Taz, have emerged as important players in thyroid differentiation and development. The involvement of signaling molecules increases the complexity of the puzzle. In this context, the importance of Bmps, Fgfs and Shh signaling at the onset of development, and of TSH, IGF1 and TGFß both at the end of terminal differentiation in embryos and in the adult thyroid, are well recognized. All of these aspects are covered herein. Thus, readers will be able to visualize the puzzle of thyroid differentiation with most - if not all - of the pieces in place.


Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Animals , Cell Differentiation , Endoderm/metabolism , Forkhead Transcription Factors/metabolism , Hippo Signaling Pathway , Homeodomain Proteins/metabolism , Humans , Mice , PAX8 Transcription Factor/metabolism , Signal Transduction , Stem Cells/metabolism , Thyroid Epithelial Cells/metabolism , Thyroid Gland/metabolism , Thyroid Nuclear Factor 1/metabolism , Trans-Activators/genetics
20.
Nat Commun ; 12(1): 2509, 2021 05 04.
Article En | MEDLINE | ID: mdl-33947861

Differential transcription of identical DNA sequences leads to distinct tissue lineages and then multiple cell types within a lineage, an epigenetic process central to progenitor and stem cell biology. The associated genome-wide changes, especially in native tissues, remain insufficiently understood, and are hereby addressed in the mouse lung, where the same lineage transcription factor NKX2-1 promotes the diametrically opposed alveolar type 1 (AT1) and AT2 cell fates. Here, we report that the cell-type-specific function of NKX2-1 is attributed to its differential chromatin binding that is acquired or retained during development in coordination with partner transcriptional factors. Loss of YAP/TAZ redirects NKX2-1 from its AT1-specific to AT2-specific binding sites, leading to transcriptionally exaggerated AT2 cells when deleted in progenitors or AT1-to-AT2 conversion when deleted after fate commitment. Nkx2-1 mutant AT1 and AT2 cells gain distinct chromatin accessible sites, including those specific to the opposite fate while adopting a gastrointestinal fate, suggesting an epigenetic plasticity unexpected from transcriptional changes. Our genomic analysis of single or purified cells, coupled with precision genetics, provides an epigenetic basis for alveolar cell fate and potential, and introduces an experimental benchmark for deciphering the in vivo function of lineage transcription factors.


Alveolar Epithelial Cells/metabolism , Cell Differentiation/genetics , Chromatin/metabolism , Epigenesis, Genetic , Histones/metabolism , Stem Cells/metabolism , Thyroid Nuclear Factor 1/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alveolar Epithelial Cells/cytology , Animals , Cell Lineage , Chromatin Immunoprecipitation , Chromatin Immunoprecipitation Sequencing , Gene Expression Regulation , Gene Knock-In Techniques , Lung/metabolism , Mice , Mutation , Protein Binding , RNA-Seq , Single-Cell Analysis , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
...