Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.661
Filtrer
1.
J Hematol Oncol ; 17(1): 78, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39218945

RÉSUMÉ

BACKGROUND: Ferroptosis, characterized by iron-dependent lipid peroxidation, emerges as a promising avenue for hepatocellular carcinoma (HCC) intervention due to its tumor susceptibility. RNA N6-methyladenosine (m6A) modification has been involved in several types of regulated cell death. However, the roles and molecular mechanisms of m6A-related regulators in HCC cell ferroptosis remain unclear. METHODS: By examining a series of m6A modification enzymes upon ferroptosis induction or inhibition, we identified METTL16 as a novel ferroptotic repressor in HCC cells. The roles of METTL16 on ferroptosis and HCC development were investigated in multiple cell lines, human HCC organoids, subcutaneous xenografts and MYC/Trp53-/- HCC model in hepatocyte-specific Mettl16 knockout and overexpression mice. The underlying mechanism was elucidated with MeRIP/RIP-qPCR, luciferase assay, Co-IP assay and Mass Spectrometry. The clinical significance and relevance were evaluated in human samples. RESULTS: High METTL16 expression confers ferroptosis resistance in HCC cells and mouse models, and promotes cell viability and tumor progression. Mechanistically, METTL16 collaborates with IGF2BP2 to modulate SENP3 mRNA stability in an m6A-dependent manner, and the latter impedes the proteasome-mediated ubiquitination degradation of Lactotransferrin (LTF) via de-SUMOylation. Elevated LTF expression facilitates the chelation of free iron and reduces liable iron pool level. SENP3 and LTF are implicated in METTL16-mediated HCC progression and anti-ferroptotic effects both in vivo and in vitro. Clinically, METTL16 and SENP3 expression were positively correlated, and high METTL16 and SENP3 expression predicts poor prognosis in human HCC samples. CONCLUSIONS: Our study reveals a new METTL16-SENP3-LTF signaling axis regulating ferroptosis and driving HCC development. Targeting this axis is a promising strategy for sensitizing ferroptosis and against HCC.


Sujet(s)
Carcinome hépatocellulaire , Ferroptose , Tumeurs du foie , Methyltransferases , Protéines de liaison à l'ARN , Animaux , Humains , Souris , Carcinogenèse/métabolisme , Carcinogenèse/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Cysteine endopeptidases , Ferroptose/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Methyltransferases/métabolisme , Methyltransferases/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique
2.
Cell Death Dis ; 15(9): 642, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39227585

RÉSUMÉ

Paired immunoglobin-like type 2 receptor beta (PILRB) mainly plays a crucial role in regulating innate immunity, but whether PILRB is involved in cancer is poorly understood. Here, we report that PILRB potentiates the PI3K/AKT pathway to drive gastric tumorigenesis by binding and stabilizing IRS4, which could hyperactivate the PI3K/AKT pathway. Firstly, the levels of PILRB are upregulated in human gastric cancer (GC) specimens and associated with poor prognosis in patients with GC. In addition, our data show that PILRB promotes cell proliferation, colony formation, cell migration and invasion in GC cells in vitro and in vivo. Mechanistically, PILRB recruits the deubiquitination enzymes OTUB1 to IRS4 and relieves K48-linked ubiquitination of IRS4, protecting IRS4 protein from proteasomal-mediated degradation and subsequent activation of the PI3K/AKT pathway. Importantly, the levels of PILRB are positively correlated with IRS4 in GC specimens. Meanwhile, we also found that PILRB reprogrammed cholesterol metabolism by altering ABCA1 and SCARB1 expression levels, and PILRB-expression confers GC cell resistance to statin treatment. Taken together, our findings illustrate that the oncogenic role of PILRB in gastric tumorigenesis, providing new insights into the regulation of PI3K/AKT signaling in GC and establishing PILRB as a biomarker for simvastatin therapy resistance in GC.


Sujet(s)
Carcinogenèse , Cholestérol , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Animaux , Cholestérol/métabolisme , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Carcinogenèse/génétique , Lignée cellulaire tumorale , Souris , Souris nude , Prolifération cellulaire , Métastase tumorale , Mouvement cellulaire , Mâle , Souris de lignée BALB C
3.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39125864

RÉSUMÉ

The potential role of the transient receptor potential Vanilloid 1 (TRPV1) non-selective cation channel in gastric carcinogenesis remains unclear. The main objective of this study was to evaluate TRPV1 expression in gastric cancer (GC) and precursor lesions compared with controls. Patient inclusion was based on a retrospective review of pathology records. Patients were subdivided into five groups: Helicobacter pylori (H. pylori)-associated gastritis with gastric intestinal metaplasia (GIM) (n = 12), chronic atrophic gastritis (CAG) with GIM (n = 13), H. pylori-associated gastritis without GIM (n = 19), GC (n = 6) and controls (n = 5). TRPV1 expression was determined with immunohistochemistry and was significantly higher in patients with H. pylori-associated gastritis compared with controls (p = 0.002). TRPV1 expression was even higher in the presence of GIM compared with patients without GIM and controls (p < 0.001). There was a complete loss of TRPV1 expression in patients with GC. TRPV1 expression seems to contribute to gastric-mucosal inflammation and precursors of GC, which significantly increases in cancer precursor lesions but is completely lost in GC. These findings suggest TRPV1 expression to be a potential marker for precancerous conditions and a target for individualized treatment. Longitudinal studies are necessary to further address the role of TRPV1 in gastric carcinogenesis.


Sujet(s)
Infections à Helicobacter , Tumeurs de l'estomac , Canaux cationiques TRPV , Humains , Canaux cationiques TRPV/métabolisme , Canaux cationiques TRPV/génétique , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Infections à Helicobacter/métabolisme , Infections à Helicobacter/complications , Infections à Helicobacter/anatomopathologie , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Études rétrospectives , États précancéreux/métabolisme , États précancéreux/anatomopathologie , Helicobacter pylori/pathogénicité , Métaplasie/métabolisme , Métaplasie/anatomopathologie , Gastrite/métabolisme , Gastrite/anatomopathologie , Gastrite/microbiologie , Adulte , Immunohistochimie , Muqueuse gastrique/métabolisme , Muqueuse gastrique/anatomopathologie , Gastrite atrophique/métabolisme , Gastrite atrophique/anatomopathologie
4.
Cancer Treat Res ; 191: 33-55, 2024.
Article de Anglais | MEDLINE | ID: mdl-39133403

RÉSUMÉ

Several types of environmental, chemical and metabolic carcinogens exist both exogenously and endogenously. Humans are daily exposed to aforementioned carcinogens through various sources such as through water, air and food or through metabolic and inflammatory products. This chapter will summarize the links between exogenous and endogenous carcinogen exposure and their metabolism with the cancer pathogenesis and associated risks. This chapter will also cover the carcinogens acquired through lifestyle factors like tobacco use and occupational exposures to different chemicals like asbestos, arsenic, chloroform, vinyl chloride, etc. Moreover, environmental carcinogens such as radiation, sunlight, diet, smoke, etc. will also be discussed in this chapter. Furthermore, there are certain carcinogens that require bio-activation and various human enzymes that play a vital role in the metabolic carcinogenesis will also be recapitulated. Necessary preventive measures against carcinogenic exposure from the exogenous environment are significant to be taken into account to reduce the risks associated with the carcinogens.


Sujet(s)
Carcinogenèse , Cancérogènes , Humains , Carcinogenèse/métabolisme , Cancérogènes/toxicité , Tumeurs/étiologie , Exposition environnementale/effets indésirables , Animaux , Exposition professionnelle/effets indésirables
6.
J Cell Mol Med ; 28(16): e70021, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39153212

RÉSUMÉ

Accumulating evidence has demonstrated that F-box protein 22 (FBXO22) participates in tumour development and progression in various types of human malignancies. However, the functions and detailed molecular mechanisms of FBXO22 in osteosarcoma tumorigenesis and progression remain elusive. In this study, we aimed to determine the effects of FBXO22 on the cell proliferation, migration and invasion of osteosarcoma cells using cell counting kit-8 and Matrigel Transwell approaches. Moreover, we explored the molecular mechanisms by which FBXO22 mediated oncogenesis and progression in osteosarcoma via Western blotting, immunoprecipitation and ubiquitination. We found that FBXO22 depletion inhibited the proliferation, migration and invasion of osteosarcoma cells, whereas FBXO22 overexpression increased the proliferation and motility of osteosarcoma cells. Mechanistically, FBXO22 promoted the ubiquitination and degradation of FoxO1 in osteosarcoma cells. FBXO22 depletion reduced cell proliferation and motility via regulation of FoxO1. Taken together, our findings provide new insight into FBXO22-induced osteosarcoma tumorigenesis. The inhibition of FBXO22 could be a promising strategy for the treatment of osteosarcoma.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Protéines F-box , Protéine O1 à motif en tête de fourche , Régulation de l'expression des gènes tumoraux , Ostéosarcome , Ubiquitination , Ostéosarcome/métabolisme , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Humains , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Protéines F-box/métabolisme , Protéines F-box/génétique , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Protéolyse , Évolution de la maladie , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Invasion tumorale , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Récepteurs cytoplasmiques et nucléaires
7.
Mol Med ; 30(1): 114, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107723

RÉSUMÉ

Glucose transporter 5 (GLUT5) overexpression has gained increasing attention due to its profound implications for tumorigenesis. This manuscript provides a comprehensive overview of the key findings and implications associated with GLUT5 overexpression in cancer. GLUT5 has been found to be upregulated in various cancer types, leading to alterations in fructose metabolism and enhanced glycolysis, even in the presence of oxygen, a hallmark of cancer cells. This metabolic shift provides cancer cells with an alternative energy source and contributes to their uncontrolled growth and survival. Beyond its metabolic roles, recent research has unveiled additional aspects of GLUT5 in cancer biology. GLUT5 overexpression appears to play a critical role in immune evasion mechanisms, which further worsens tumor progression and complicates therapeutic interventions. This dual role of GLUT5 in both metabolic reprogramming and immune modulation highlights its significance as a potential diagnostic marker and therapeutic target. Understanding the molecular mechanisms driving GLUT5 overexpression is crucial for developing targeted therapeutic strategies that can disrupt the unique vulnerabilities of GLUT5-overexpressing cancer cells. This review emphasizes the complexities surrounding GLUT5's involvement in cancer and underscores the pressing need for continued research to unlock its potential as a diagnostic biomarker and therapeutic target, ultimately improving cancer management and patient outcomes.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Transporteur de glucose de type 5 , Tumeurs , Humains , Tumeurs/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie , Transporteur de glucose de type 5/métabolisme , Transporteur de glucose de type 5/génétique , Animaux , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Glycolyse , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique
8.
Life Sci ; 355: 122974, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39147318

RÉSUMÉ

BACKGROUND: Basic helix-loop-helix ARNT like 2 (ARNTL2) is a transcription factor that controls the circadian rhythm. Amounts of studies have demonstrated the carcinogenic function of ARNTL2 in human malignant tumors albeit the underlying mechanisms remain poorly understood. We aimed to study the significance of ARNTL2 in bladder cancer (BLCA). METHODS: Immunohistochemical staining, immunoblotting and the database from TCGA were used to analyze the clinical relevance of ARNTL2, enolase 1 (ENO1) and solute carrier family 31 member 1 (SLC31A1) in BLCA. The function of ARNTL2 was explored by cell proliferation assay, apoptosis, colony formation and xenografted tumorigenesis. The molecular mechanisms of ARNTL2-driving BLCA development were investigated by RT-qPCR, immunoblotting and luciferase assays. Glycolysis was checked by measuring glucose consumption and lactate production. ENO1 activity was assessed by using indicated assay kit. RESULTS: Overexpression of ARNTL2 facilitates the proliferation and tumorigenesis of BLCA cells through suppression of apoptosis and enhancement of glycolysis. Up-regulation of SLC31A1, ENO1, and enhancement of SLC31A1-mediated ENO1 activity were critical for ARNTL2-triggered glycolysis and malignant growth in BLCA cells. ARNTL2 was positively correlated with SLC31A1 and ENO1 in BLCA patients. High expression of ARNTL2, SLC31A1 or ENO1 predicted the poor prognosis of BLCA patients. Depletion of SLC31A1 and inhibition of glycolysis completely blunted the growth ability of BLCA cells. CONCLUSION: In summary, ARNTL2 facilitates the progression of BLCA via activating ENO1-mediated glycolysis in a SLC31A1-independent and -dependent manner. Inhibiting SLC31A1 and glycolysis may be an aspirational approach for the treatment of BLCA patients with overexpression of ARNTL2.


Sujet(s)
Facteurs de transcription ARNTL , Prolifération cellulaire , Protéines de liaison à l'ADN , Glycolyse , Enolase , Protéines suppresseurs de tumeurs , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/génétique , Humains , Enolase/métabolisme , Enolase/génétique , Animaux , Souris , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Facteurs de transcription ARNTL/métabolisme , Facteurs de transcription ARNTL/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Lignée cellulaire tumorale , Évolution de la maladie , Souris nude , Apoptose , Femelle , Régulation de l'expression des gènes tumoraux , Mâle , Souris de lignée BALB C , Carcinogenèse/métabolisme , Carcinogenèse/génétique , Marqueurs biologiques tumoraux
9.
J Exp Clin Cancer Res ; 43(1): 227, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39148124

RÉSUMÉ

BACKGROUND: The failure of proper recognition of the intricate nature of pathophysiology in colorectal cancer (CRC) has a substantial effect on the progress of developing novel medications and targeted therapy approaches. Imbalances in the processes of lipid oxidation and biosynthesis of fatty acids are significant risk factors for the development of CRC. Therapeutic intervention that specifically targets the peroxisome proliferator-activated receptor gamma (PPARγ) and its downstream response element, in response to lipid metabolism, has been found to promote the growth of tumors and has shown significant clinical advantages in cancer patients. METHODS: Clinical CRC samples and extensive in vitro and in vivo experiments were carried out to determine the role of ZDHHC6 and its downstream targets via a series of biochemical assays, molecular analysis approaches and lipid metabolomics assay, etc. RESULTS: To study the effect of ZDHHC6 on the progression of CRC and identify whether ZDHHC6 is a palmitoyltransferase that regulates fatty acid synthesis, which directly palmitoylates and stabilizes PPARγ, and this stabilization in turn activates the ACLY transcription-related metabolic pathway. In this study, we demonstrate that PPARγ undergoes palmitoylation in its DNA binding domain (DBD) section. This lipid-related modification enhances the stability of PPARγ protein by preventing its destabilization. As a result, palmitoylated PPARγ inhibits its degradation induced by the lysosome and facilitates its translocation into the nucleus. In addition, we have identified zinc finger-aspartate-histidine-cysteine 6 (ZDHHC6) as a crucial controller of fatty acid biosynthesis. ZDHHC6 directly interacts with and adds palmitoyl groups to stabilize PPARγ at the Cys-313 site within the DBD domain of PPARγ. Consequently, this palmitoylation leads to an increase in the expression of ATP citrate lyase (ACLY). Furthermore, our findings reveals that ZDHHC6 actively stimulates the production of fatty acids and plays a role in the development of colorectal cancer. However, we have observed a significant reduction in the cancer-causing effects when the expression of ZDHHC6 is inhibited in in vivo trials. Significantly, in CRC, there is a strong positive correlation between the high expression of ZDHHC6 and the expression of PPARγ. Moreover, this high expression of ZDHHC6 is connected with the severity of CRC and is indicative of a poor prognosis. CONCLUSIONS: We have discovered a mechanism in which lipid biosynthesis is controlled by ZDHHC6 and includes the signaling of PPARγ-ACLY in the advancement of CRC. This finding provides a justification for targeting lipid synthesis by blocking ZDHHC6 as a potential therapeutic approach.


Sujet(s)
Acyltransferases , Tumeurs du côlon , , Récepteur PPAR gamma , Animaux , Femelle , Humains , Mâle , Souris , Acyltransferases/métabolisme , Acyltransferases/génétique , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Lignée cellulaire tumorale , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/génétique , Métabolisme lipidique/génétique , Lipidomique/méthodes , /génétique , Récepteur PPAR gamma/métabolisme
10.
Nat Commun ; 15(1): 6915, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39134530

RÉSUMÉ

Protein post-translational modifications (PTMs) are crucial for cancer cells to adapt to hypoxia; however, the functional significance of lysine crotonylation (Kcr) in hypoxia remains unclear. Herein we report a quantitative proteomics analysis of global crotonylome under normoxia and hypoxia, and demonstrate 128 Kcr site alterations across 101 proteins in MDA-MB231 cells. Specifically, we observe a significant decrease in K131cr, K156cr and K220cr of phosphoglycerate kinase 1 (PGK1) upon hypoxia. Enoyl-CoA hydratase 1 (ECHS1) is upregulated and interacts with PGK1, leading to the downregulation of PGK1 Kcr under hypoxia. Abolishment of PGK1 Kcr promotes glycolysis and suppresses mitochondrial pyruvate metabolism by activating pyruvate dehydrogenase kinase 1 (PDHK1). A low PGK1 K131cr level is correlated with malignancy and poor prognosis of breast cancer. Our findings show that PGK1 Kcr is a signal in coordinating glycolysis and the tricarboxylic acid (TCA) cycle and may serve as a diagnostic indicator for breast cancer.


Sujet(s)
Tumeurs du sein , Cycle citrique , Glycolyse , Phosphoglycerate kinase , Phosphoglycerate kinase/métabolisme , Phosphoglycerate kinase/génétique , Humains , Glycolyse/génétique , Lignée cellulaire tumorale , Femelle , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lysine/métabolisme , Maturation post-traductionnelle des protéines , Animaux , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Régulation négative , Souris , Protéomique/méthodes , Souris nude , Régulation de l'expression des gènes tumoraux , Mitochondries/métabolisme , Hypoxie cellulaire , Pyruvate dehydrogenase acetyl-transferring kinase/métabolisme , Pyruvate dehydrogenase acetyl-transferring kinase/génétique
11.
Cells ; 13(16)2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39195234

RÉSUMÉ

Colorectal cancer remains a major global health concern. Colonoscopy, the gold-standard colorectal cancer diagnostic, relies on the visual detection of lesions and necessitates invasive biopsies for confirmation. Alternative diagnostic methods, based on nanomedicine, can facilitate early detection of malignancies. Here, we examine the uptake of surface-enhanced Raman scattering nanoparticles (SERS NPs) as a marker for intestinal tumor detection and imaging using an established Drosophila melanogaster model for gut disease. Young and old Oregon-R and w1118 flies were orally administered SERS NPs and scanned without and upon gut lumen clearance to assess nanoparticle retention as a function of aging. Neither young nor old flies showed significant NP retention in their body after gut lumen clearance. Moreover, tumorigenic flies of the esg-Gal4/UAS-RasV12 genotype were tested for SERS NP retention 2, 4 and 6 days after RasV12 oncogene induction in their midgut progenitor cells. Tumorigenic flies showed a statistically significant NP retention signal at 2 days, well before midgut epithelium impairment. The signal was then visualized in scans of dissected guts revealing areas of NP uptake in the posterior midgut region of high stem cell activity.


Sujet(s)
Vieillissement , Drosophila melanogaster , Nanoparticules , Analyse spectrale Raman , Animaux , Analyse spectrale Raman/méthodes , Drosophila melanogaster/métabolisme , Vieillissement/métabolisme , Nanoparticules/composition chimique , Carcinogenèse/anatomopathologie , Carcinogenèse/métabolisme
12.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119810, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39128596

RÉSUMÉ

Lung cancer is the leading cause of cancer-related deaths worldwide, with non-small cell lung cancer (NSCLC) constituting approximately 84 % of all lung cancer cases. The role of inflammation in the initiation and progression of NSCLC tumors has been the focus of extensive research. Among the various inflammatory mediators, prostaglandin E2 (PGE2) plays a pivotal role in promoting the aggressiveness of epithelial tumors through multiple mechanisms, including the stimulation of growth, evasion of apoptosis, invasion, and induction of angiogenesis. The Extracellular signal-Regulated Kinase 5 (ERK5), the last discovered member among conventional mitogen-activated protein kinases (MAPK), is implicated in cancer-associated inflammation. In this study, we explored whether ERK5 is involved in the process of tumorigenesis induced by PGE2. Using A549 and PC9 NSCLC cell lines, we found that PGE2 triggers the activation of ERK5 via the EP1 receptor. Moreover, both genetic and pharmacological inhibition of ERK5 reduced PGE2-induced proliferation, migration, invasion and stemness of A549 and PC9 cells, indicating that ERK5 plays a critical role in PGE2-induced tumorigenesis. In summary, our study underscores the pivotal role of the PGE2/EP1/ERK5 axis in driving the malignancy of NSCLC cells in vitro. Targeting this axis holds promise as a potential avenue for developing novel therapeutic strategies aimed at controlling the advancement of NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Mouvement cellulaire , Prolifération cellulaire , Dinoprostone , Tumeurs du poumon , Mitogen-Activated Protein Kinase 7 , Humains , Dinoprostone/métabolisme , Dinoprostone/pharmacologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Mitogen-Activated Protein Kinase 7/métabolisme , Mitogen-Activated Protein Kinase 7/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules A549 , Lignée cellulaire tumorale , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Phénotype
13.
J Biochem Mol Toxicol ; 38(9): e23815, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39171650

RÉSUMÉ

Drug transporters play a pivotal role in modulating drug disposition and are subject to alterations under inflammatory conditions. This study aimed to elucidate the intricate expression patterns of drug transporters during both acute and chronic inflammation, which are closely linked to malignant transformation. To investigate acute inflammation, we employed an in vitro model by subjecting Caco-2 cells to various inflammatory stimuli (IL-1ß, TNF-α, or LPS) individually or in combination. The successful induction of inflammation was confirmed by robust increases in IL-6 and NO production. Notably, inflamed Caco-2 cells exhibited significantly diminished levels of ABCB1 and ABCG2, while the expression of ABCC2 was upregulated. For chronic inflammation induction in vivo, we employed the well-established AOM/DSS mouse model known for its association with colitis-driven tumorigenesis. Persistent inflammation was effectively monitored throughout the experiment via elevated IL-6 and NO levels. The sequential stages of tumorigenesis were confirmed through Ki-67 immunohistochemistry. Intriguingly, we observed gradual alterations in the expression patterns of the studied drug transporters during stepwise induction, with ABCB1, ABCG2, and ABCC1 showing downregulation and ABCC2 exhibiting upregulation. Immunohistochemistry further revealed dynamic changes in the expression of ABCB1 and ABCC2 during the induction cycles, closely paralleling the gradual increase in Ki-67 expression observed during the development of precancerous lesions. Collectively, our findings underscore the significant impact of inflammation on drug transporter expression, potentially influencing the process of malignant transformation of the colon.


Sujet(s)
Oxyde de diméthyl-diazène , Tumeurs du côlon , Inflammation , Protéine-2 associée à la multirésistance aux médicaments , Humains , Tumeurs du côlon/métabolisme , Tumeurs du côlon/induit chimiquement , Tumeurs du côlon/anatomopathologie , Animaux , Cellules Caco-2 , Souris , Oxyde de diméthyl-diazène/toxicité , Inflammation/métabolisme , Inflammation/induit chimiquement , Inflammation/anatomopathologie , Carcinogenèse/métabolisme , Carcinogenèse/induit chimiquement , Protéines associées à la multirésistance aux médicaments/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Sous-famille B de transporteurs à cassette liant l'ATP/génétique , Protéines tumorales/métabolisme , Protéines tumorales/biosynthèse , Interleukine-6/métabolisme , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/induit chimiquement , Mâle
14.
Int J Mol Sci ; 25(16)2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39201498

RÉSUMÉ

Keratinocytes are major cellular components of the skin and are strongly involved in its homeostasis. Oncogenic events, starting mainly from excessive sun exposure, lead to the dysregulation of their proliferation and differentiation programs and promote the initiation and progression of non-melanoma skin cancers (NMSCs). Primary melanomas, which originate from melanocytes, initiate and develop in close interaction with keratinocytes, whose role in melanoma initiation, progression, and immune escape is currently being explored. Recent studies highlighted, in particular, unexpected modes of communication between melanocytic cells and keratinocytes, which may be of interest as sources of new biomarkers in melanomagenesis or potential therapeutic targets. This review aims at reporting the various contributions of keratinocytes in skin basal cell carcinoma (BCC), cutaneous squamous cell carcinoma (cSCC), and melanoma, with a greater focus on the latter in order to highlight some recent breakthrough findings. The readers are referred to recent reviews when contextual information is needed.


Sujet(s)
Carcinome basocellulaire , Carcinome épidermoïde , Évolution de la maladie , Kératinocytes , Tumeurs cutanées , Humains , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/métabolisme , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Carcinome basocellulaire/anatomopathologie , Carcinome basocellulaire/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme , Mélanome/métabolisme , Mélanome/anatomopathologie , Animaux , Carcinogenèse/anatomopathologie , Carcinogenèse/métabolisme
15.
Stem Cell Res Ther ; 15(1): 256, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39135143

RÉSUMÉ

BACKGROUND: One of major challenges in breast tumor therapy is the existence of breast cancer stem cells (BCSCs). BCSCs are a small subpopulation of tumor cells that exhibit characteristics of stem cells. BCSCs are responsible for progression, recurrence, chemoresistance and metastasis of breast cancer. Ca2+ signalling plays an important role in diverse processes in cancer development. However, the role of Ca2+ signalling in BCSCs is still poorly understood. METHODS: A highly effective 3D soft fibrin gel system was used to enrich BCSC-like cells from ER+ breast cancer lines MCF7 and MDA-MB-415. We then investigated the role of two Ca2+-permeable ion channels Orai1 and Orai3 in the growth and stemness of BCSC-like cells in vitro, and tumorigenicity in female NOD/SCID mice in vivo. RESULTS: Orai1 RNA silencing and pharmacological inhibition reduced the growth of BCSC-like cells in tumor spheroids, decreased the expression levels of BCSC markers, and reduced the growth of tumor xenografts in NOD/SCID mice. Orai3 RNA silencing also had similar inhibitory effect on the growth and stemness of BCSC-like cells in vitro, and tumor xenograft growth in vivo. Mechanistically, Orai1 and SPCA2 mediate store-operated Ca2+ entry. Knockdown of Orai1 or SPCA2 inhibited glycolysis pathway, whereas knockdown of Orai3 or STIM1 had no effect on glycolysis. CONCLUSION: We found that Orai1 interacts with SPCA2 to mediate store-independent Ca2+ entry, subsequently promoting the growth and tumorigenicity of BCSC-like cells via glycolysis pathway. In contrast, Orai3 and STIM1 mediate store-operated Ca2+ entry, promoting the growth and tumorigenicity of BCSC-like cells via a glycolysis-independent pathway. Together, our study uncovered a well-orchestrated mechanism through which two Ca2+ entry pathways act through distinct signalling axes to finely control the growth and tumorigenicity of BCSCs.


Sujet(s)
Tumeurs du sein , Canaux calciques , Souris de lignée NOD , Souris SCID , Cellules souches tumorales , Protéine ORAI1 , Protéine ORAI1/métabolisme , Protéine ORAI1/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Humains , Animaux , Femelle , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Souris , Canaux calciques/métabolisme , Canaux calciques/génétique , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Lignée cellulaire tumorale , Transduction du signal , Signalisation calcique , Cellules MCF-7
16.
J Extracell Vesicles ; 13(8): e12482, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39105261

RÉSUMÉ

It is known that small extracellular vesicles (sEVs) are released from cancer cells and contribute to cancer progression via crosstalk with recipient cells. We have previously reported that sEVs expressing the αVß3 integrin, a protein upregulated in aggressive neuroendocrine prostate cancer (NEPrCa), contribute to neuroendocrine differentiation (NED) in recipient cells. Here, we examine the impact of αVß3 expression on sEV protein content, density and function. sEVs used in this study were isolated by iodixanol density gradients and characterized by nanoparticle tracking analysis, immunoblotting and single vesicle analysis. Our proteomic profile of sEVs containing αVß3 shows downregulation of typical effectors involved in apoptosis and necrosis and an upregulation of tumour cell survival factors compared to control sEVs. We also show that the expression of αVß3 in sEVs causes a distinct reposition of EV markers (Alix, CD81, CD9) to a low-density sEV subpopulation. This low-density reposition is independent of extracellular matrix (ECM) protein interactions with sEVs. This sEV subset contains αVß3 and an αVß3 downstream effector, NgR2, a novel marker for NEPrCa. We show that sEVs containing αVß3 are loaded with higher amounts of NgR2 as compared to sEVs that do not express αVß3. Mechanistically, we demonstrate that sEVs containing NgR2 do not affect the sEV marker profile, but when injected in vivo intratumorally, they promote tumour growth and induce NED. We show that sEVs expressing NgR2 increase the activation of focal adhesion kinase (FAK), a known promoter of cancer cell proliferation, in recipient cells. We also show that NgR2 mimics the effect of sEVs containing αVß3 since it displays increased growth of NgR2 transfectants in vivo, as compared to control cells. Overall, our results describe the changes that occur in cargo, density and functions of cancer cell-derived sEVs containing the αVß3 integrin and its effector, NgR2, without affecting the sEV tetraspanin profiles.


Sujet(s)
Vésicules extracellulaires , Intégrine alphaVbêta3 , Tumeurs de la prostate , Mâle , Intégrine alphaVbêta3/métabolisme , Humains , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Vésicules extracellulaires/métabolisme , Animaux , Lignée cellulaire tumorale , Souris , Carcinogenèse/métabolisme
17.
Int J Mol Med ; 54(4)2024 10.
Article de Anglais | MEDLINE | ID: mdl-39092569

RÉSUMÉ

Non­SMC condensin I complex subunit D2 (NCAPD2) is a newly identified oncogene; however, the specific biological function and molecular mechanism of NCAPD2 in liver cancer progression remain unknown. In the present study, the aberrant expression of NCAPD2 in liver cancer was investigated using public tumor databases, including TNMplot, The Cancer Genome Atlas and the International Cancer Genome Consortium based on bioinformatics analyses, and it was validated using a clinical cohort. It was revealed that NCAPD2 was significantly upregulated in liver cancer tissues compared with in control liver tissues, and NCAPD2 served as an independent prognostic factor and predicted poor prognosis in liver cancer. In addition, the expression of NCAPD2 was positively correlated with the percentage of Ki67+ cells. Finally, single­cell sequencing data, gene­set enrichment analyses and in vitro investigations, including cell proliferation assay, Transwell assay, wound healing assay, cell cycle experiments, cell apoptosis assay and western blotting, were carried out in human liver cancer cell lines to assess the biological mechanisms of NCAPD2 in patients with liver cancer. The results revealed that the upregulation of NCAPD2 enhanced tumor cell proliferation, invasion and cell cycle progression at the G2/M­phase transition, and inhibited apoptosis in liver cancer cells. Furthermore, NCAPD2 overexpression was closely associated with the phosphatidylinositol 3­kinase (PI3K)­Akt­mammalian target of rapamycin (mTOR)/c­Myc signaling pathway and epithelial­mesenchymal transition (EMT) progression in HepG2 and Huh7 cells. In addition, upregulated NCAPD2 was shown to have adverse effects on overall survival and disease­specific survival in liver cancer. In conclusion, the overexpression of NCAPD2 was shown to lead to cell cycle progression at the G2/M­phase transition, activation of the PI3K­Akt­mTOR/c­Myc signaling pathway and EMT progression in human liver cancer cells.


Sujet(s)
Prolifération cellulaire , Tumeurs du foie , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Humains , Sérine-thréonine kinases TOR/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Transduction du signal/génétique , Phosphatidylinositol 3-kinases/métabolisme , Mâle , Femelle , Prolifération cellulaire/génétique , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Carcinogenèse/métabolisme , Adulte d'âge moyen , Régulation de l'expression des gènes tumoraux , Évolution de la maladie , Lignée cellulaire tumorale , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Transition épithélio-mésenchymateuse/génétique , Apoptose/génétique , Mouvement cellulaire/génétique , Pronostic
18.
Nature ; 633(8028): 189-197, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39143206

RÉSUMÉ

Fasting is associated with a range of health benefits1-6. How fasting signals elicit changes in the proteome to establish metabolic programmes remains poorly understood. Here we show that hepatocytes selectively remodel the translatome while global translation is paradoxically downregulated during fasting7,8. We discover that phosphorylation of eukaryotic translation initiation factor 4E (P-eIF4E) is induced during fasting. We show that P-eIF4E is responsible for controlling the translation of genes involved in lipid catabolism and the production of ketone bodies. Inhibiting P-eIF4E impairs ketogenesis in response to fasting and a ketogenic diet. P-eIF4E regulates those messenger RNAs through a specific translation regulatory element within their 5' untranslated regions (5' UTRs). Our findings reveal a new signalling property of fatty acids, which are elevated during fasting. We found that fatty acids bind and induce AMP-activated protein kinase (AMPK) kinase activity that in turn enhances the phosphorylation of MAP kinase-interacting protein kinase (MNK), the kinase that phosphorylates eIF4E. The AMPK-MNK-eIF4E axis controls ketogenesis, revealing a new lipid-mediated kinase signalling pathway that links ketogenesis to translation control. Certain types of cancer use ketone bodies as an energy source9,10 that may rely on P-eIF4E. Our findings reveal that on a ketogenic diet, treatment with eFT508 (also known as tomivosertib; a P-eIF4E inhibitor) restrains pancreatic tumour growth. Thus, our findings unveil a new fatty acid-induced signalling pathway that activates selective translation, which underlies ketogenesis and provides a tailored diet intervention therapy for cancer.


Sujet(s)
AMP-Activated Protein Kinases , Carcinogenèse , Facteur-4E d'initiation eucaryote , Jeûne , Biosynthèse des protéines , Animaux , Souris , Facteur-4E d'initiation eucaryote/métabolisme , Phosphorylation , Mâle , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Humains , AMP-Activated Protein Kinases/métabolisme , Acides gras/métabolisme , Corps cétoniques/métabolisme , Régime cétogène , Hépatocytes/métabolisme , Transduction du signal , Métabolisme lipidique , Femelle
19.
Cells ; 13(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38994962

RÉSUMÉ

Muscle-enriched A-type lamin-interacting protein (MLIP) is an emerging protein involved in cellular homeostasis and stress adaptation. Eukaryotic cells regulate various cellular processes, including metabolism, DNA repair, and cell cycle progression, to maintain cellular homeostasis. Disruptions in this homeostasis can lead to diseases such as cancer, characterized by uncontrolled cell growth and division. This review aims to explore for the first time the unique role MLIP may play in cancer development and progression, given its interactions with the PI3K/Akt/mTOR pathway, p53, MAPK9, and FOXO transcription factors, all critical regulators of cellular homeostasis and tumor suppression. We discuss the current understanding of MLIP's involvement in pro-survival pathways and its potential implications in cancer cells' metabolic remodeling and dysregulated homeostasis. Additionally, we examine the potential of MLIP as a novel therapeutic target for cancer treatment. This review aims to shed light on MLIP's potential impact on cancer biology and contribute to developing innovative therapeutic strategies.


Sujet(s)
Tumeurs , Transduction du signal , Humains , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Tumeurs/génétique , Animaux , Carcinogenèse/anatomopathologie , Carcinogenèse/métabolisme , Carcinogenèse/génétique
20.
FASEB J ; 38(13): e23809, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38967126

RÉSUMÉ

The neurofibromatosis type 2 (NF2) gene, known for encoding the tumor suppressor protein Merlin, is central to the study of tumorigenesis and associated cellular processes. This review comprehensively examines the multifaceted role of NF2/Merlin, detailing its structural characteristics, functional diversity, and involvement in various signaling pathways such as Wnt/ß-catenin, Hippo, TGF-ß, RTKs, mTOR, Notch, and Hedgehog. These pathways are crucial for cellular growth, proliferation, and differentiation. NF2 mutations are specifically linked to the development of schwannomas, meningiomas, and ependymomas, although the precise mechanisms of tumor formation in these specific cell types remain unclear. Additionally, the review explores Merlin's role in embryogenesis, highlighting the severe developmental defects and embryonic lethality caused by NF2 deficiency. The potential therapeutic strategies targeting these genetic aberrations are also discussed, emphasizing inhibitors of mTOR, HDAC, and VEGF as promising avenues for treatment. This synthesis of current knowledge underscores the necessity for ongoing research to elucidate the detailed mechanisms of NF2/Merlin and develop effective therapeutic strategies, ultimately aiming to improve the prognosis and quality of life for individuals with NF2 mutations.


Sujet(s)
Carcinogenèse , Neurofibromine-2 , Humains , Neurofibromine-2/génétique , Neurofibromine-2/métabolisme , Animaux , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Neurofibromatose de type 2/génétique , Neurofibromatose de type 2/métabolisme , Neurofibromatose de type 2/anatomopathologie , Transduction du signal , Mutation
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE