Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.744
Filtrer
1.
BMC Endocr Disord ; 24(1): 116, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39010034

RÉSUMÉ

BACKGROUND: Somatic mutations have been observed to induce aldosterone-producing adenomas (APAs). These may be accelerated during pregnancy. Somatic PRKACA mutations are common in cortisol-producing adenomas (CPAs). However, their role in APAs, particularly aldosterone- and cortisol-producing adenomas (A/CPAs), is not well understood. This study aims to investigate the association between PRKACA mutations and the accelerated development of A/CPAs during pregnancy. CASE PRESENTATION: A patient with primary aldosteronism (PA) associated with severe Cushing's syndrome (CS) underwent surgical resection of an adrenal tumor one year after delivery. Pathologic examination revealed an adrenocortical adenoma characterized primarily by zona glomerulosa hyperplasia. Somatic mutation analysis revealed the presence of the somatic PRKACA mutation, which was validated as a deleterious mutation by various computational databases. Immunohistochemical results showed positive staining for cytochrome P450 family 11 subfamily B member 1 (CYP11B1), cytochrome P450 family 11 subfamily B member 2 (CYP11B2), and luteinizing hormone/chorionic gonadotropin receptor (LHCGR). Our study included a review of 20 previously documented cases of aldosterone- and cortisol-producing adenomas (A/CPAs), two of which were concurrently positive for both CYP11B1 and CYP11B2, consistent with our findings. CONCLUSION: Somatic mutations in PRKACA may correlate with the upregulation of LHCGR, which synergistically drives the accelerated growth of co-secretion tumors during pregnancy, thereby exacerbating disease progression.


Sujet(s)
Tumeurs corticosurrénaliennes , Adénome corticosurrénalien , Aldostérone , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits , Hydrocortisone , Mutation , Complications tumorales de la grossesse , Humains , Femelle , Grossesse , Adulte , Hydrocortisone/métabolisme , Adénome corticosurrénalien/génétique , Adénome corticosurrénalien/anatomopathologie , Adénome corticosurrénalien/métabolisme , Adénome corticosurrénalien/chirurgie , Tumeurs corticosurrénaliennes/génétique , Tumeurs corticosurrénaliennes/anatomopathologie , Tumeurs corticosurrénaliennes/métabolisme , Aldostérone/métabolisme , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/génétique , Complications tumorales de la grossesse/génétique , Complications tumorales de la grossesse/anatomopathologie , Hyperaldostéronisme/génétique , Hyperaldostéronisme/anatomopathologie , Hyperaldostéronisme/chirurgie , Syndrome de Cushing/génétique , Syndrome de Cushing/anatomopathologie , Adénomes/génétique , Adénomes/anatomopathologie , Adénomes/métabolisme
2.
Mol Biol Rep ; 51(1): 764, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874740

RÉSUMÉ

BACKGROUND AND AIM: Colorectal cancer (CRC) originates from pre-existing polyps in the colon. The development of different subtypes of CRC is influenced by various genetic and epigenetic characteristics. CpG island methylator phenotype (CIMP) is found in about 15-20% of sporadic CRCs and is associated with hypermethylation of certain gene promoters. This study aims to find prognostic genes and compare their expression and methylation status as potential biomarkers in patients with serrated sessile adenomas/polyps (SSAP) and CRC, in order to evaluate which, one is a better predictor of disease. METHOD: This study employed a multi-phase approach to investigate genes associated with CRC and SSAP. Initially, two gene expression datasets were analyzed using R and Limma package to identify differentially expressed genes (DEGs). Venn diagram analysis further refined the selection, revealing four genes from the Weissenberg panel with significant changes. These genes, underwent thorough in silico evaluations. Once confirmed, they proceeded to wet lab experimentation, focusing on expression and methylation status. This comprehensive methodology ensured a robust examination of the genes involved in CRC and SSAP. RESULT: This study identified cancer-specific genes, with 8,351 and 1,769 genes specifically down-regulated in SSAP and CRC tissues, respectively. The down-regulated genes were associated with cell adhesion, negative regulation of cell proliferation, and drug response. Four highly downregulated genes in the Weissenberg panel, including CACNA1G, IGF2, MLH1, and SOCS1. In vitro analysis showed that they are hypermethylated in both SSAP and CRC samples while their expressions decreased only in CRC samples. CONCLUSION: This suggests that the decrease in gene expression could help determine whether a polyp will become cancerous. Using both methylation status and gene expression status of genes in the Weissenberg panel in prognostic tests may lead to better prognoses for patients.


Sujet(s)
Tumeurs colorectales , Ilots CpG , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Facteur de croissance IGF-II , Protéine-1 homologue de MutL , Protéine-1 suppressive de la signalisation des cytokines , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Protéine-1 suppressive de la signalisation des cytokines/génétique , Protéine-1 suppressive de la signalisation des cytokines/métabolisme , Méthylation de l'ADN/génétique , Facteur de croissance IGF-II/génétique , Facteur de croissance IGF-II/métabolisme , Régulation de l'expression des gènes tumoraux/génétique , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Ilots CpG/génétique , Femelle , Polypes coliques/génétique , Polypes coliques/métabolisme , Polypes coliques/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Mâle , Régulation négative/génétique , Simulation numérique , Adulte d'âge moyen , Adénomes/génétique , Adénomes/métabolisme , Adénomes/anatomopathologie , Régions promotrices (génétique)/génétique , Canaux calciques de type T/génétique , Canaux calciques de type T/métabolisme , Analyse de profil d'expression de gènes/méthodes , Sujet âgé , Pronostic
3.
Front Endocrinol (Lausanne) ; 15: 1258233, 2024.
Article de Anglais | MEDLINE | ID: mdl-38841301

RÉSUMÉ

Technetium-99m sestamibi single-photon emission computed tomography/computed tomography (99mTc-sestamibi SPECT/CT) is a mainstay of the pre-operative localization of parathyroid lesions. We report here the case of a 30 year-old woman with a fortuitously discovered 2 cm cervical mass for which a parathyroid origin was originally suspected due to its retro-thyroidal localization and a personal history of nephrolithiasis. Normal serum calcium and parathyroid hormone (PTH) levels excluded primary hyperparathyroidism, raising suspicion of a non-functional parathyroid adenoma, and SPECT/CT imaging showed that the mass was 99mTc-sestamibi-avid. Fine-needle aspiration (FNA) was performed; cytology was non-diagnostic but the needle washout was negative for thyroglobulin, calcitonin and PTH, arguing against a thyroidal or parathyroidal origin of the mass. Core needle biopsy revealed a schwannoma, ostensibly originating from the recurrent laryngeal nerve; upon surgical resection, it was finally found to arise from the esophageal submucosa. This case illustrates the fact that endocrinologists, radiologists, nuclear medicine, head and neck, and other specialists investigating patients with cervical masses should be aware that schwannomas need to be considered in the differential diagnosis of focal 99mTc-sestamibi uptake in the neck region.


Sujet(s)
Adénomes , Neurinome , Tumeurs de la parathyroïde , Technétium (99mTc) sestamibi , Humains , Femelle , Tumeurs de la parathyroïde/imagerie diagnostique , Tumeurs de la parathyroïde/anatomopathologie , Tumeurs de la parathyroïde/chirurgie , Tumeurs de la parathyroïde/diagnostic , Adulte , Neurinome/imagerie diagnostique , Neurinome/anatomopathologie , Neurinome/diagnostic , Diagnostic différentiel , Adénomes/imagerie diagnostique , Adénomes/diagnostic , Adénomes/anatomopathologie , Adénomes/métabolisme , Tumeurs de l'oesophage/imagerie diagnostique , Tumeurs de l'oesophage/diagnostic , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/chirurgie , Tomographie par émission monophotonique couplée à la tomodensitométrie , Radiopharmaceutiques
4.
Pathol Oncol Res ; 30: 1611734, 2024.
Article de Anglais | MEDLINE | ID: mdl-38873175

RÉSUMÉ

Background: Gastric epithelial neoplasm of the fundic-gland mucosa lineages (GEN-FGMLs) are rare forms of gastric tumors that encompass oxyntic gland adenoma (OGA), gastric adenocarcinoma of the fundic-gland type (GA-FG), and gastric adenocarcinoma of the fundic-gland mucosa type (GA-FGM). There is no consensus on the cause, classification, and clinicopathological features of GEN-FGMLs, and misdiagnosis is common because of similarities in symptoms. Methods: 37 cases diagnosed with GEN-FGMLs were included in this study. H&E-stained slides were reviewed and clinicopathological parameters were recorded. Immunohistochemical staining was conducted for MUC2, MUC5AC, MUC6, CD10, CD56, synaptophysin, chromograninA, p53, Ki67, pepsinogen-I, H+/K+-ATPase and Desmin. Results: The patients' ages ranged from 42 to 79 years, with a median age of 60. 17 were male and 20 were female. Morphologically, 19 OGAs, 16 GA-FGs, and two GA-FGMs were identified. Histopathological similarities exist between OGA, GA-FG, and GA-FGM. The tumors demonstrated well-formed glands, expanding with dense growth patterns comprising pale, blue-grey columnar cells with mild nuclear atypia. These cells resembled fundic gland cells. None of the OGA invaded the submucosal layer. The normal gastric pit epithelium covered the entire surface of the OGA and GA-FG, but the dysplasia pit epithelium covered the GA-FGM. Non-atrophic gastritis was observed in more than half of the background mucosa. All cases were diffusely positive for MUC6 and pepsinogen-I on immunohistochemistry. H+/K+-ATPase staining was negative or showed a scattered pattern in most cases. MUC5AC was expressed on the surface of GA-FGMs. p53 was focally expressed and the Ki67 index was low (1%-20%). Compared with OGA, GA-FG and GA-FGM were more prominent in the macroscopic view (p < 0.05) and had larger sizes (p < 0.0001). Additionally, GA-FG and GA-FGM exhibited higher Ki67 indices than OGA (p < 0.0001). Specimens with Ki-67 proliferation indices >2.5% and size >4.5 mm are more likely to be diagnosed with GA-FG and GA-FGM than OGA. Conclusion: GEN-FGMLs are group of well-differentiated gastric tumors with favourable biological behaviours, low cellular atypia, and low proliferation. Immunohistochemistry is critical for confirming diagnosis. Compared with OGA, GA-FG and GA-FGM have larger sizes and higher Ki67 proliferation indices, indicating that they play a critical role in the identification of GEN-FGML. Pathologists and endoscopists should be cautious to prevent misdiagnosis and overtreatment, especially in biopsy specimens.


Sujet(s)
Marqueurs biologiques tumoraux , Muqueuse gastrique , Antigène KI-67 , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Antigène KI-67/métabolisme , Muqueuse gastrique/anatomopathologie , Muqueuse gastrique/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/analyse , Adénocarcinome/anatomopathologie , Adénocarcinome/métabolisme , Fundus gastrique/anatomopathologie , Fundus gastrique/métabolisme , Adénomes/anatomopathologie , Adénomes/métabolisme , Pronostic
5.
Clin Transl Med ; 14(6): e1734, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38888967

RÉSUMÉ

BACKGROUND: Sporadic parathyroid adenoma (PA) is the most common cause of hyperparathyroidism, yet the mechanisms involved in its pathogenesis remain incompletely understood. METHODS: Surgically removed PA samples, along with normal parathyroid gland (PG) tissues that were incidentally dissected during total thyroidectomy, were analysed using single-cell RNA-sequencing with the 10× Genomics Chromium Droplet platform and Cell Ranger software. Gene set variation analysis was conducted to characterise hallmark pathway gene signatures, and single-cell regulatory network inference and clustering were utilised to analyse transcription factor regulons. Immunohistochemistry and immunofluorescence were performed to validate cellular components of PA tissues. siRNA knockdown and gene overexpression, alongside quantitative polymerase chain reaction, Western blotting and cell proliferation assays, were conducted for functional investigations. RESULTS: There was a pervasive increase in gene transcription in PA cells (PACs) compared with PG cells. This is associated with high expression of histone-lysine N-methyltransferase 2A (KMT2A). High KMT2A levels potentially contribute to promoting PAC proliferation through upregulation of the proto-oncogene CCND2, which is mediated by the transcription factors signal transducer and activator of transcription 3 (STAT3) and GATA binding protein 3 (GATA3). PA tissues are heavily infiltrated with myeloid cells, while fibroblasts, endothelial cells and macrophages in PA tissues are commonly enriched with proinflammatory gene signatures relative to their counterparts in PG tissues. CONCLUSIONS: We revealed the previously underappreciated involvement of the KMT2A‒STAT3/GATA3‒CCND2 axis and chronic inflammation in the pathogenesis of PA. These findings underscore the therapeutic promise of KMT2A inhibition and anti-inflammatory strategies, highlighting the need for future investigations to translate these molecular insights into practical applications. HIGHLIGHTS: Single-cell RNA-sequencing reveals a transcriptome catalogue comparing sporadic parathyroid adenomas (PAs) with normal parathyroid glands. PA cells show a pervasive increase in gene expression linked to KMT2A upregulation. KMT2A-mediated STAT3 and GATA3 upregulation is key to promoting PA cell proliferation via cyclin D2. PAs exhibit a proinflammatory microenvironment, suggesting a potential role of chronic inflammation in PA pathogenesis.


Sujet(s)
Adénomes , Histone-lysine N-methyltransferase , Inflammation , Tumeurs de la parathyroïde , Humains , Tumeurs de la parathyroïde/génétique , Tumeurs de la parathyroïde/métabolisme , Tumeurs de la parathyroïde/anatomopathologie , Adénomes/génétique , Adénomes/métabolisme , Adénomes/anatomopathologie , Inflammation/génétique , Inflammation/métabolisme , Histone-lysine N-methyltransferase/génétique , Histone-lysine N-methyltransferase/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/génétique , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Proto-oncogène Mas , Prolifération cellulaire/génétique
6.
Inn Med (Heidelb) ; 65(7): 672-680, 2024 Jul.
Article de Allemand | MEDLINE | ID: mdl-38869654

RÉSUMÉ

The widespread use of diagnostic imaging has led to an increase in the incidence of pituitary tumors. The majority of incidentalomas are hormone-inactive (HI) pituitary microadenomas. The most common clinically relevant pituitary adenomas are prolactin-secreting, followed by HI, and far less common are growth hormone (GH)-, adrenocorticotropic hormone (ACTH)- and thyroid-stimulating hormone (TSH)-secreting adenomas. Pituitary adenomas are usually benign, although aggressive growth and invasion occurs in individual cases. Very rarely, they give rise to metastases and are then termed pituitary carcinomas. All pituitary tumors require endocrine testing for pituitary hormone excess. In addition to the medical history and clinical examination, laboratory diagnostics are very important. Symptoms such as irregular menstruation, loss of libido or galactorrhea often lead to the timely diagnosis of prolactinomas, and hyperprolactinemia can easily confirm the diagnosis (considering the differential diagnoses). Diagnosis is more difficult for all other hormone-secreting pituitary adenomas (acromegaly, Cushing's disease, TSHoma), as the symptoms are often non-specific (i.e., headaches, weight gain, fatigue, joint pain). Furthermore, comorbidities such as hypertension, diabetes, and depression are such widespread diseases that pituitary adenomas are rarely considered as the underlying cause. Timely diagnosis and appropriate treatment have a significant impact on morbidity, mortality, and quality of life. Therefore, the role of primary care physicians is very important for achieving an early diagnosis. In addition, patients with pituitary adenomas should always be referred to endocrinologists to ensure optimal diagnosis as well as treatment.


Sujet(s)
Tumeurs de l'hypophyse , Humains , Tumeurs de l'hypophyse/diagnostic , Tumeurs de l'hypophyse/métabolisme , Tumeurs de l'hypophyse/anatomopathologie , Diagnostic différentiel , Adénomes/métabolisme , Adénomes/anatomopathologie , Adénomes/diagnostic , Prolactinome/diagnostic , Prolactinome/métabolisme , Prolactinome/anatomopathologie
7.
Front Endocrinol (Lausanne) ; 15: 1407615, 2024.
Article de Anglais | MEDLINE | ID: mdl-38919490

RÉSUMÉ

Acromegaly is a rare endocrine disorder caused by hypersecretion of growth hormone (GH) from a pituitary adenoma. Elevated GH levels stimulate excess production of insulin-like growth factor 1 (IGF-1) which leads to the insidious onset of clinical manifestations. The most common primary central nervous system (CNS) tumors, meningiomas originate from the arachnoid layer of the meninges and are typically benign and slow-growing. Meningiomas are over twice as common in women as in men, with age-adjusted incidence (per 100,000 individuals) of 10.66 and 4.75, respectively. Several reports describe co-occurrence of meningiomas and acromegaly. We aimed to determine whether patients with acromegaly are at elevated risk for meningioma. Investigation of the literature showed that co-occurrence of a pituitary adenoma and a meningioma is a rare phenomenon, and the majority of cases involve GH-secreting adenomas. To the best of our knowledge, a systematic review examining the association between meningiomas and elevated GH levels (due to GH-secreting adenomas in acromegaly or exposure to exogenous GH) has never been conducted. The nature of the observed coexistence between acromegaly and meningioma -whether it reflects causation or mere co-association -is unclear, as is the pathophysiologic etiology. Systematic review registration: https://www.crd.york.ac.uk/prospero/, identifier CRD42022376998.


Sujet(s)
Acromégalie , Tumeurs des méninges , Méningiome , Humains , Méningiome/complications , Méningiome/étiologie , Méningiome/anatomopathologie , Méningiome/épidémiologie , Acromégalie/complications , Tumeurs des méninges/complications , Tumeurs des méninges/épidémiologie , Tumeurs des méninges/anatomopathologie , Hormone de croissance humaine/métabolisme , Hormone de croissance humaine/sang , Facteurs de risque , Adénomes/complications , Adénomes/métabolisme , Adénomes/anatomopathologie , Adénomes/épidémiologie
8.
Oncol Res ; 32(6): 1079-1091, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827318

RÉSUMÉ

Approximately 30%-40% of growth hormone-secreting pituitary adenomas (GHPAs) harbor somatic activating mutations in GNAS (α subunit of stimulatory G protein). Mutations in GNAS are associated with clinical features of smaller and less invasive tumors. However, the role of GNAS mutations in the invasiveness of GHPAs is unclear. GNAS mutations were detected in GHPAs using a standard polymerase chain reaction (PCR) sequencing procedure. The expression of mutation-associated maternally expressed gene 3 (MEG3) was evaluated with RT-qPCR. MEG3 was manipulated in GH3 cells using a lentiviral expression system. Cell invasion ability was measured using a Transwell assay, and epithelial-mesenchymal transition (EMT)-associated proteins were quantified by immunofluorescence and western blotting. Finally, a tumor cell xenograft mouse model was used to verify the effect of MEG3 on tumor growth and invasiveness. The invasiveness of GHPAs was significantly decreased in mice with mutated GNAS compared with that in mice with wild-type GNAS. Consistently, the invasiveness of mutant GNAS-expressing GH3 cells decreased. MEG3 is uniquely expressed at high levels in GHPAs harboring mutated GNAS. Accordingly, MEG3 upregulation inhibited tumor cell invasion, and conversely, MEG3 downregulation increased tumor cell invasion. Mechanistically, GNAS mutations inhibit EMT in GHPAs. MEG3 in mutated GNAS cells prevented cell invasion through the inactivation of the Wnt/ß-catenin signaling pathway, which was further validated in vivo. Our data suggest that GNAS mutations may suppress cell invasion in GHPAs by regulating EMT through the activation of the MEG3/Wnt/ß-catenin signaling pathway.


Sujet(s)
Chromogranine , Transition épithélio-mésenchymateuse , Sous-unités alpha Gs des protéines G , Adénome hypophysaire à GH , Mutation , Invasion tumorale , ARN long non codant , Sous-unités alpha Gs des protéines G/génétique , Sous-unités alpha Gs des protéines G/métabolisme , Animaux , Humains , Adénome hypophysaire à GH/génétique , Adénome hypophysaire à GH/anatomopathologie , Adénome hypophysaire à GH/métabolisme , Souris , Chromogranine/génétique , Chromogranine/métabolisme , Transition épithélio-mésenchymateuse/génétique , ARN long non codant/génétique , Femelle , Mâle , Lignée cellulaire tumorale , Adénomes/génétique , Adénomes/anatomopathologie , Adénomes/métabolisme , Adulte d'âge moyen , Adulte , Prolifération cellulaire/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Voie de signalisation Wnt/génétique , Régulation de l'expression des gènes tumoraux
9.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38731846

RÉSUMÉ

Activated TGFß signaling in the tumor microenvironment, which occurs independently of epithelial cancer cells, has emerged as a key driver of tumor progression in late-stage colorectal cancer (CRC). This study aimed to elucidate the contribution of TGFß-activated stroma to serrated carcinogenesis, representing approximately 25% of CRCs and often characterized by oncogenic BRAF mutations. We used a transcriptional signature developed based on TGFß-responsive, stroma-specific genes to infer TGFß-dependent stromal activation and conducted in silico analyses in 3 single-cell RNA-seq datasets from a total of 39 CRC samples and 12 bulk transcriptomic datasets consisting of 2014 CRC and 416 precursor samples, of which 33 were serrated lesions. Single-cell analyses validated that the signature was expressed specifically by stromal cells, effectively excluding transcriptional signals derived from epithelial cells. We found that the signature was upregulated during malignant transformation and cancer progression, and it was particularly enriched in CRCs with mutant BRAF compared to wild-type counterparts. Furthermore, across four independent precursor datasets, serrated lesions exhibited significantly higher levels of TGFß-responsive stromal activation compared to conventional adenomas. This large-scale analysis suggests that TGFß-dependent stromal activation occurs early in serrated carcinogenesis. Our study provides novel insights into the molecular mechanisms underlying CRC development via the serrated pathway.


Sujet(s)
Tumeurs colorectales , Régulation de l'expression des gènes tumoraux , Cellules stromales , Facteur de croissance transformant bêta , Humains , Adénomes/génétique , Adénomes/anatomopathologie , Adénomes/métabolisme , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Analyse de profil d'expression de gènes , Mutation , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Transduction du signal , Analyse sur cellule unique , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie , Transcriptome , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Microenvironnement tumoral/génétique
10.
J Proteome Res ; 23(6): 2054-2066, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38775738

RÉSUMÉ

The metabolites and microbiota in tongue coating display distinct characteristics in certain digestive disorders, yet their relationship with colorectal cancer (CRC) remains unexplored. Here, we employed liquid chromatography coupled with tandem mass spectrometry to analyze the lipid composition of tongue coating using a nontargeted approach in 30 individuals with colorectal adenomas (CRA), 32 with CRC, and 30 healthy controls (HC). We identified 21 tongue coating lipids that effectively distinguished CRC from HC (AUC = 0.89), and 9 lipids that differentiated CRC from CRA (AUC = 0.9). Furthermore, we observed significant alterations in the tongue coating lipid composition in the CRC group compared to HC/CRA groups. As the adenoma-cancer sequence progressed, there was an increase in long-chain unsaturated triglycerides (TG) levels and a decrease in phosphatidylethanolamine plasmalogen (PE-P) levels. Furthermore, we noted a positive correlation between N-acyl ornithine (NAOrn), sphingomyelin (SM), and ceramide phosphoethanolamine (PE-Cer), potentially produced by members of the Bacteroidetes phylum. The levels of inflammatory lipid metabolite 12-HETE showed a decreasing trend with colorectal tumor progression, indicating the potential involvement of tongue coating microbiota and tumor immune regulation in early CRC development. Our findings highlight the potential utility of tongue coating lipid analysis as a noninvasive tool for CRC diagnosis.


Sujet(s)
Tumeurs colorectales , Lipidomique , Phosphatidyléthanolamine , Spectrométrie de masse en tandem , Langue , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/microbiologie , Lipidomique/méthodes , Mâle , Femelle , Langue/microbiologie , Langue/métabolisme , Langue/anatomopathologie , Langue/composition chimique , Adulte d'âge moyen , Spectrométrie de masse en tandem/méthodes , Phosphatidyléthanolamine/métabolisme , Phosphatidyléthanolamine/analyse , Sujet âgé , Chromatographie en phase liquide , Lipides/analyse , Lipides/composition chimique , Triglycéride/métabolisme , Triglycéride/analyse , Adénomes/métabolisme , Adénomes/microbiologie , Sphingomyéline/analyse , Sphingomyéline/métabolisme , Acide éicosatétraénoïque-5,8,10,14 hydroxy-12/métabolisme , Acide éicosatétraénoïque-5,8,10,14 hydroxy-12/composition chimique , Acétalphosphatides/analyse , Acétalphosphatides/métabolisme , Acétalphosphatides/composition chimique , Études cas-témoins , Éthanolamines/métabolisme , Éthanolamines/analyse , Éthanolamines/composition chimique , Céramides/métabolisme , Céramides/analyse , Adulte
11.
Clin Chim Acta ; 560: 119732, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38772522

RÉSUMÉ

AIM: Our study focuses on the microbial and metabolomic profile changes during the adenoma stage, as adenomas can be considered potential precursors to colorectal cancer through the adenoma-carcinoma sequence. Identifying possible intervention targets at this stage may aid in preventing the progression of colorectal adenoma (CRA) to malignant lesions. Furthermore, we evaluate the efficacy of combined microbial and metabolite biomarkers in detecting CRA. METHODS: Fecal metagenomic and serum metabolomic analyses were performed for the discovery of alterations of gut microbiome and metabolites in CRA patients (n = 26), Colorectal cancer (CRC) patients (n = 19), Familial Adenomatous Polyposis (FAP) patients (n = 10), and healthy controls (n = 20). Finally, analyzing the associations between gut microbes and metabolites was performed by a Receiver Operating Characteristic (ROC) curve. RESULTS: Our analysis present that CRA patients differ significantly in gut microflora and serum metabolites compared with healthy controls, especially for Lachnospiraceae and Parasutterella. Its main metabolite, butyric acid, concentrations were raised in CRA patients compared with the healthy controls, indicating its role as a promoter of colorectal tumorigenesis. α-Linolenic acid and lysophosphatidylcholine represented the other healthy metabolite for CRA. Combining five microbial and five metabolite biomarkers, we differentiated CRA from CRC with an Area Under the Curve (AUC) of 0.85 out of this performance vastly superior to the specificity recorded by traditional markers CEA and CA199 in such differentiation of these conditions. CONCLUSIONS: The study underlines significant microbial and metabolic alterations in CRA with a novel insight into screening and early intervention of its tumorigenesis.


Sujet(s)
Adénomes , Tumeurs colorectales , Dépistage précoce du cancer , Microbiome gastro-intestinal , Humains , Tumeurs colorectales/sang , Tumeurs colorectales/diagnostic , Tumeurs colorectales/métabolisme , Adénomes/métabolisme , Adénomes/sang , Adénomes/diagnostic , Mâle , Femelle , Adulte d'âge moyen , Adulte , Sujet âgé , Fèces/microbiologie , Fèces/composition chimique , Marqueurs biologiques tumoraux/sang , Carcinomes/métabolisme , Carcinomes/sang , Carcinomes/diagnostic
12.
Eur J Endocrinol ; 190(6): 458-466, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38771697

RÉSUMÉ

OBJECTIVE: The aim of this study is to compare the response to first-line medical treatment in treatment-naive acromegaly patients with pure growth hormone (GH)-secreting pituitary adenoma (GH-PA) and those with GH and prolactin cosecreting PA (GH&PRL-PA). DESIGN: This is a retrospective multicentric study of acromegaly patients followed from 2003 to 2023 in 33 tertiary Spanish hospitals with at least 6 months of first-line medical treatment. METHODS: Baseline characteristics, first-line medical treatment strategies, and outcomes were analyzed. We employed a multiple logistic regression full model to estimate the impact of some baseline characteristics on disease control after each treatment modality. RESULTS: Of the 144 patients included, 72.9% had a GH-PA, and 27.1% had a GH&PRL-PA. Patients with GH&PRL-PA were younger (43.9 ± 15.0 vs 51.9 ± 12.7 years, P < .01) and harboring more frequently macroadenomas (89.7% vs 72.1%, P = .03). First-generation somatostatin receptor ligand (fgSRL) as monotherapy was given to 106 (73.6%) and a combination treatment with fgSRL and cabergoline in the remaining 38 (26.4%). Patients with GH&PRL-PA received more frequently a combination therapy (56.4% vs 15.2%, P < .01). After 6 months of treatment, in the group of patients under fgSRL as monotherapy, those patients with GH&PRL-PA had worse control compared to GH-PAs (29.4% vs 55.1%, P = .04). However, these differences in the rate of disease control between both groups disappeared when both received combination treatment with fgSRL and cabergoline. CONCLUSION: In GH&PRL-PA, the biochemical control achieved with fgSRL as monotherapy is substantially worse than in patients harboring GH-PA, supporting the inclusion of cabergoline as first-line medical treatment in combination with fgSRLs in these subgroups of patients.


Sujet(s)
Acromégalie , Cabergoline , Prolactine , Humains , Acromégalie/traitement médicamenteux , Acromégalie/sang , Femelle , Mâle , Adulte d'âge moyen , Études rétrospectives , Adulte , Cabergoline/usage thérapeutique , Résultat thérapeutique , Prolactine/sang , Adénome hypophysaire à GH/traitement médicamenteux , Adénome hypophysaire à GH/sang , Adénome hypophysaire à GH/métabolisme , Hormone de croissance humaine , Adénomes/traitement médicamenteux , Adénomes/sang , Adénomes/métabolisme , Adénomes/complications , Sujet âgé , Association de médicaments , Somatostatine/analogues et dérivés , Somatostatine/usage thérapeutique , Tumeurs de l'hypophyse/traitement médicamenteux , Tumeurs de l'hypophyse/sang , Tumeurs de l'hypophyse/métabolisme , Tumeurs de l'hypophyse/complications , Espagne/épidémiologie
13.
Eur J Endocrinol ; 190(6): 421-433, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38701338

RÉSUMÉ

INTRODUCTION: Growth hormone (GH)-secreting pituitary tumors (GHomas) are the most common acromegaly cause. At diagnosis, most of them are macroadenomas, and up to 56% display cavernous sinus invasion. Biomarker assessment associated with tumor growth and invasion is important to optimize their management. OBJECTIVES: The study aims to identify clinical/hormonal/molecular biomarkers associated with tumor size and invasiveness in GHomas and to analyze the influence of pre-treatment with somatostatin analogs (SSAs) or dopamine agonists (DAs) in key molecular biomarker expression. METHODS: Clinical/analytical/radiological variables were evaluated in 192 patients from the REMAH study (ambispective multicenter post-surgery study of the Spanish Society of Endocrinology and Nutrition). The expression of somatostatin/ghrelin/dopamine system components and key pituitary/proliferation markers was evaluated in GHomas after the first surgery. Univariate/multivariate regression studies were performed to identify association between variables. RESULTS: Eighty percent of patients harbor macroadenomas (63.8% with extrasellar growth). Associations between larger and more invasive GHomas with younger age, visual abnormalities, higher IGF1 levels, extrasellar/suprasellar growth, and/or cavernous sinus invasion were found. Higher GH1 and lower PRL/POMC/CGA/AVPR1B/DRD2T/DRD2L expression levels (P < .05) were associated with tumor invasiveness. Least Absolute Shrinkage and Selection Operator's penalized regression identified combinations of clinical and molecular features with areas under the curve between 0.67 and 0.82. Pre-operative therapy with DA or SSAs did not alter the expression of any of the markers analyzed except for DRD1/AVPR1B (up-regulated with DA) and FSHB/CRHR1 (down-regulated with SSAs). CONCLUSIONS: A specific combination of clinical/analytical/molecular variables was found to be associated with tumor invasiveness and growth capacity in GHomas. Pre-treatment with first-line drugs for acromegaly did not significantly modify the expression of the most relevant biomarkers in our association model. These findings provide valuable insights for risk stratification and personalized management of GHomas.


Sujet(s)
Acromégalie , Adénomes , Adénome hypophysaire à GH , Invasion tumorale , Humains , Mâle , Femelle , Acromégalie/métabolisme , Adulte d'âge moyen , Adulte , Adénome hypophysaire à GH/anatomopathologie , Adénome hypophysaire à GH/métabolisme , Adénomes/métabolisme , Adénomes/anatomopathologie , Sujet âgé , Agonistes de la dopamine/usage thérapeutique , Marqueurs biologiques tumoraux/métabolisme , Somatostatine/analogues et dérivés , Somatostatine/usage thérapeutique , Hormone de croissance humaine/métabolisme
14.
Pathol Res Pract ; 258: 155332, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38696856

RÉSUMÉ

Necroptosis can either be the cause of tumorigenesis or it can impede its process. Recently, it has been proved that long non-coding RNAs (lncRNAs) have different crucial roles at cellular level, especially on cell death. Regarding the important role of necroptosis and lncRNAs in the pathogenesis of different cancers, especially pituitary adenomas (PAs), we assessed expression levels of two necroptosis related genes, namely TRADD and BIRC2, in addition to three related lncRNAs, namely FLVCR1-DT, MAGI2-AS3, and NEAT1 in PAs compared with adjacent normal tissues (ANTs). TRADD had no significant difference between two groups; however, BIRC2, FLVCR1-DT, MAGI2-AS3, and NEAT1 were upregulated in PAs compared to ANTs (Expression ratios [95% CI] = 2.3 [1.47-3.6], 2.13 [1.02-4.44], 3.01 [1.76-5.16] and 2.47 [1.37-4.45], respectively). When taking into account different types of PAs, significant upregulation of BIRC2, MAGI2-AS3 and NEAT1 was recorded in non-functioning PAs compared with corresponding ANTs (Expression ratios [95% CI] =1.9 [1.04-3.43], 2.69 [1.26-5.72] and 2.22 [0.98-5.01], respectively). Additionally, higher levels of BIRC2 were associated with higher flow of CSF (P value=0.048). Moreover, higher Knosp classified tumors had lower levels of BIRC2 (P value=0.001). Finally, lower levels of MAGI2-AS3 were associated with larger tumor size (P value=0.006). NEAT1 expression was correlated with FLVCR1-DT and TRADD. TRADD expression was correlated with FLVCR1-DT. Additional correlation was observed between expression of BIRC2 and MAGI2-AS3. In sum, this study provides evidence that dysregulated levels of studied genes could contribute to the pathogenesis of pituitary tumors.


Sujet(s)
Nécroptose , Tumeurs de l'hypophyse , ARN long non codant , Humains , ARN long non codant/génétique , Tumeurs de l'hypophyse/génétique , Tumeurs de l'hypophyse/anatomopathologie , Tumeurs de l'hypophyse/métabolisme , Mâle , Adulte d'âge moyen , Femelle , Adulte , Nécroptose/génétique , Sujet âgé , Régulation de l'expression des gènes tumoraux/génétique , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/métabolisme , Adénomes/génétique , Adénomes/anatomopathologie , Adénomes/métabolisme
15.
Tumour Biol ; 46(1): 1-11, 2024.
Article de Anglais | MEDLINE | ID: mdl-38728194

RÉSUMÉ

BACKGROUND: It is well established that most colorectal carcinomas arise from conventional adenomas through the adenoma-carcinoma sequence (ACS) model. mitogen-activated protein kinases (MAPKs) pathway has been reported as a crucial player in tumorigenesis. The MAPK signaling pathway is activated by different extracellular signals involving the "mitogen-activated/extracellular signal-regulated kinase 1 (MEK1)", and this induces the expression of genes involved in proliferation and cellular transformation. Diaphanous-related formin-3 (DIAPH3) acts as a potential metastasis regulator through inhibiting the cellular transition to amoeboid behavior in different cancer types. OBJECTIVE: The aim of the study was to investigate the pattern of immunohistochemical expression of MEK1 and DIAPH3 in colorectal adenoma (CRA) and corresponding colorectal carcinoma (CRC) specimens. METHODS: The immunohistochemical expression of DIAPH3 and MEK1 was examined in 43 cases of CRC and their associated adenomas using tissue microarray technique. RESULTS: MEK1 was overexpressed in 23 CRC cases (53.5%) and in 20 CRA cases (46.5%). DIAPH3 was overexpressed in 11 CRA cases (about 29%) which were significantly lower than CRC (22 cases; 58%) (P = 0.011). Both MEK1 and DIAPH3 overexpression were significantly correlated in CRC (P = 0.009) and CRA cases (P = 0.002). Tumors with MEK1 overexpression had a significantly higher tumor grade (P = 0.050) and perineural invasion (P = 0.017). CONCLUSIONS: Both MEK1 and DIAPH3 are overexpressed across colorectal ACS with strong correlation between them. This co- expression suggests a possible synergistic effect of MEK1 and DIAPH-3 in colorectal ACS. Further large-scale studies are required to investigate the potential functional aspects of MEK1 and DIAPH3 in ACS and their involvement in tumor initiation and the metastatic process.


Sujet(s)
Adénomes , Tumeurs colorectales , Formines , MAP Kinase Kinase 1 , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Formines/génétique , Formines/métabolisme , Adénomes/anatomopathologie , Adénomes/génétique , Adénomes/métabolisme , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , MAP Kinase Kinase 1/génétique , MAP Kinase Kinase 1/métabolisme , Adulte , Immunohistochimie , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Régulation de l'expression des gènes tumoraux , Carcinomes/anatomopathologie , Carcinomes/génétique , Carcinomes/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique
16.
CNS Neurosci Ther ; 30(5): e14749, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38739004

RÉSUMÉ

AIMS: A bone-invasive pituitary adenoma exhibits aggressive behavior, leading to a worse prognosis. We have found that TNF-α promotes bone invasion by facilitating the differentiation of osteoclasts, however, before bone-invasive pituitary adenoma invades bone tissue, it needs to penetrate the dura mater, and this mechanism is not yet clear. METHODS: We performed transcriptome microarrays on specimens of bone-invasive pituitary adenomas (BIPAs) and noninvasive pituitary adenomas (NIPAs) and conducted differential expressed gene analysis and enrichment analysis. We altered the expression of TNF-α through plasmids, then validated the effects of TNF-α on GH3 cells and verified the efficacy of the TNF-α inhibitor SPD304. Finally, the effects of TNF-α were validated in in vivo experiments. RESULTS: Pathway act work showed that the MAPK pathway was significantly implicated in the pathway network. The expression of TNF-α, MMP9, and p-p38 is higher in BIPAs than in NIPAs. Overexpression of TNF-α elevated the expression of MAPK pathway proteins and MMP9 in GH3 cells, as well as promoted proliferation, migration, and invasion of GH3 cells. Flow cytometry indicated that TNF-α overexpression increased the G2 phase ratio in GH3 cells and inhibited apoptosis. The expression of MMP9 was reduced after blocking the P38 MAPK pathway; overexpression of MMP9 promoted invasion of GH3 cells. In vivo experiments confirm that the TNF-α overexpression group has larger tumor volumes. SPD304 was able to suppress the effects caused by TNF-α overexpression. CONCLUSION: Bone-invasive pituitary adenoma secretes higher levels of TNF-α, which then acts on itself in an autocrine manner, activating the MAPK pathway and promoting the expression of MMP9, thereby accelerating the membrane invasion process. SPD304 significantly inhibits the effect of TNF-α and may be applied in the clinical treatment of bone-invasive pituitary adenoma.


Sujet(s)
Adénomes , Système de signalisation des MAP kinases , Matrix metalloproteinase 9 , Invasion tumorale , Tumeurs de l'hypophyse , Facteur de nécrose tumorale alpha , Facteur de nécrose tumorale alpha/métabolisme , Tumeurs de l'hypophyse/métabolisme , Tumeurs de l'hypophyse/anatomopathologie , Humains , Adénomes/anatomopathologie , Adénomes/métabolisme , Animaux , Matrix metalloproteinase 9/métabolisme , Système de signalisation des MAP kinases/physiologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mâle , Lignée cellulaire tumorale , Femelle , Souris , Souris nude , Communication autocrine/physiologie , Communication autocrine/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Adulte , Rats , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/physiologie , Transduction du signal/physiologie , Transduction du signal/effets des médicaments et des substances chimiques
18.
BMC Cancer ; 24(1): 611, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773399

RÉSUMÉ

RNA interactomes and their diversified functionalities have recently benefited from critical methodological advances leading to a paradigm shift from a conventional conception on the regulatory roles of RNA in pathogenesis. However, the dynamic RNA interactomes in adenoma-carcinoma sequence of human CRC remain unexplored. The coexistence of adenoma, cancer, and normal tissues in colorectal cancer (CRC) patients provides an appropriate model to address this issue. Here, we adopted an RNA in situ conformation sequencing technology for mapping RNA-RNA interactions in CRC patients. We observed large-scale paired RNA counts and identified some unique RNA complexes including multiple partners RNAs, single partner RNAs, non-overlapping single partner RNAs. We focused on the antisense RNA OIP5-AS1 and found that OIP5-AS1 could sponge different miRNA to regulate the production of metabolites including pyruvate, alanine and lactic acid. Our findings provide novel perspectives in CRC pathogenesis and suggest metabolic reprogramming of pyruvate for the early diagnosis and treatment of CRC.


Sujet(s)
Adénomes , Tumeurs colorectales , microARN , Acide pyruvique , ARN long non codant , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , microARN/génétique , microARN/métabolisme , Adénomes/génétique , Adénomes/métabolisme , Adénomes/anatomopathologie , Acide pyruvique/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Régulation de l'expression des gènes tumoraux ,
19.
Neoplasia ; 53: 101005, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38761506

RÉSUMÉ

Colorectal cancer (CRC) stands as a prevalent malignancy globally. A pivotal event in CRC pathogenesis involves the loss-of-function mutation in the APC gene, leading to the formation of benign polyps. Despite the well-established role of APC, the contribution of CUL4B to CRC initiation in the pre-tumorous stage remains poorly understood. In this investigation, we generated a murine model by crossing ApcMin/+ mice with Cul4bΔIEC mice to achieve specific deletion of Cul4b in the gut epithelium against an ApcMin/+ background. By employing histological methods, RNA-sequencing (RNA-seq), and flow cytometry, we assessed alterations and characterized the immune microenvironment. Our results unveiled that CUL4B deficiency in gut epithelium expedited ApcMin/+ adenoma formation. Notably, CUL4B in adenomas restrained the accumulation of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). In vivo inhibition of MDSCs significantly delayed the growth of CUL4B deleted ApcMin/+ adenomas. Furthermore, the addition of MDSCs to in vitro cultured ApcMin/+; Cul4bΔIEC adenoma organoids mitigated their alterations. Mechanistically, CUL4B directly interacted with the promoter of Csf3, the gene encoding granulocyte-colony stimulating factor (G-CSF) by coordinating with PRC2. Inhibiting CUL4B epigenetically activated the expression of G-CSF, promoting the recruitment of MDSCs. These findings offer novel insights into the tumor suppressor-like roles of CUL4B in regulating ApcMin/+ adenomas, suggesting a potential therapeutic strategy for CRC initiation and progression in the context of activated Wnt signaling.


Sujet(s)
Adénomes , Cullines , Modèles animaux de maladie humaine , Cellules myéloïdes suppressives , Animaux , Cullines/génétique , Cullines/métabolisme , Souris , Cellules myéloïdes suppressives/métabolisme , Cellules myéloïdes suppressives/anatomopathologie , Adénomes/anatomopathologie , Adénomes/génétique , Adénomes/métabolisme , Protéine de la polypose adénomateuse colique/génétique , Humains , Microenvironnement tumoral/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/étiologie , Délétion de gène , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme
20.
Discov Med ; 36(184): 971-980, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38798256

RÉSUMÉ

OBJECTIVE: Tissue inhibitors of matrix metalloproteinases (TIMPs) are prognostic markers in cancers. However, the role of TIMPs in DNA methylation during invasive pituitary adenoma (PA) remains unclear. The purpose of this study was to assess the effects of TIMP2 and TIMP3 promoter demethylation on the proliferation, migration, and invasion of invasive PA cells. METHODS: Methylation-specific polymerase chain reaction (PCR), quantitative PCR, and western blots were used to analyze the promoter methylation and expression of TIMP1-3. Cell counting kit-8 (CCK-8), wound healing, and transwell assays were carried out to determine the effects of TIMP2 and TIMP3 demethylation. RESULTS: TIMP1-3 showed downregulated expression in invasive PA tissues and cell lines (p < 0.05). The low expression of TIMP1-3 was due to promoter methylation of these genes (p < 0.05). The results showed that downregulation of TIMP2 and TIMP3 can promote cell proliferation, migration, and invasion (p < 0.05), whereas overexpression of TIMP2 and TIMP3 can inhibit cell proliferation, migration, and invasion (p < 0.05). After treatment with 5-azacytidine (5-AzaC), the cell activity decreased, the proliferation rate decreased, and the invasion ability weakened (p < 0.05). Treatment with 5-AzaC increased TIMP2 and TIMP3 expression and decreased DNA (cytosine-5-)-methyltransferase 1 (DNMT1), DNMT3a, and DNMT3b expression (p < 0.05). CONCLUSIONS: We showed that DNA methylation causes the silencing of TIMP2 and TIMP3 in invasive PA, it can also lead to malignant cell proliferation and cause pathological changes, whereas the use of 5-AzaC can inhibit the methylation process and can inhibit cell proliferation. Our results provide a novel method for clinical diagnosis and prevention of invasive PA.


Sujet(s)
Adénomes , Mouvement cellulaire , Prolifération cellulaire , Méthylation de l'ADN , Invasion tumorale , Tumeurs de l'hypophyse , Inhibiteur tissulaire de métalloprotéinase-2 , Inhibiteur tissulaire de métalloprotéinase-3 , Humains , Inhibiteur tissulaire de métalloprotéinase-3/génétique , Inhibiteur tissulaire de métalloprotéinase-3/métabolisme , Prolifération cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs de l'hypophyse/génétique , Tumeurs de l'hypophyse/anatomopathologie , Tumeurs de l'hypophyse/métabolisme , Mouvement cellulaire/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Inhibiteur tissulaire de métalloprotéinase-2/génétique , Inhibiteur tissulaire de métalloprotéinase-2/métabolisme , Adénomes/génétique , Adénomes/anatomopathologie , Adénomes/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Mâle , Femelle , Régions promotrices (génétique)/génétique , Adulte d'âge moyen , Adulte , Azacitidine/pharmacologie , DNA methyltransferase 3A/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE