Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
ChemMedChem ; : e202400269, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724444

RÉSUMÉ

Targeting the protein arginine methyltransferase 1 (PRMT1) has emerged as a promising therapeutic strategy in cancer treatment. The phase 1 clinical trial for GSK3368715, the first PRMT1 inhibitor to enter the clinic, was terminated early due to a lack of clinical efficacy, extensive treatment-emergent effects, and dose-limiting toxicities. The incidence of the latter two events may be associated with inhibition-driven pharmacology as a high and sustained concentration of inhibitor is required for therapeutic effect. The degradation of PRMT1 using a proteolysis targeting chimera (PROTAC) may be superior to inhibition as proceeds via event-driven pharmacology where a PROTAC acts catalytically at a low dose. PROTACs containing the same pharmacophore as GSK3368715, combined with a motif that recruits the VHL or CRBN E3-ligase, were synthesised. Suitable cell permeability and target engagement were shown for selected candidates by the detection of downstream effects of PRMT1 inhibition and by a NanoBRET assay for E3-ligase binding, however the candidates did not induce PRMT1 degradation. This paper is the first reported investigation of PRMT1 for targeted protein degradation and provides hypotheses and insights to assist the design of PROTACs for PRMT1 and other novel target proteins.

3.
Mol Cancer Res ; 21(6): 591-604, 2023 06 01.
Article de Anglais | MEDLINE | ID: mdl-36930833

RÉSUMÉ

Estrogen receptor alpha (ER/ESR1) mutations occur in 30% to 40% of endocrine resistant ER-positive (ER+) breast cancer. Forkhead box A1 (FOXA1) is a key pioneer factor mediating ER-chromatin interactions and endocrine response in ER+ breast cancer, but its role in ESR1-mutant breast cancer remains unclear. Our previous FOXA1 chromatin immunoprecipitation sequencing (ChIP-seq) identified a large portion of redistributed binding sites in T47D genome-edited Y537S and D538G ESR1-mutant cells. Here, we further integrated FOXA1 genomic binding profile with the isogenic ER cistrome, accessible genome, and transcriptome data of T47D cell model. FOXA1 redistribution was significantly associated with transcriptomic alterations caused by ESR1 mutations. Furthermore, in ESR1-mutant cells, FOXA1-binding sites less frequently overlapped with ER, and differential gene expression was less associated with the canonical FOXA1-ER axis. Motif analysis revealed a unique enrichment of retinoid X receptor (RXR) motifs in FOXA1-binding sites of ESR1-mutant cells. Consistently, ESR1-mutant cells were more sensitive to growth stimulation with the RXR agonist LG268. The mutant-specific response was dependent on two RXR isoforms, RXR-α and RXR-ß, with a stronger dependency on the latter. In addition, T3, the agonist of thyroid receptor (TR) also showed a similar growth-promoting effect in ESR1-mutant cells. Importantly, RXR antagonist HX531 blocked growth of ESR1-mutant cells and a patient-derived xenograft (PDX)-derived organoid with an ESR1 D538G mutation. Collectively, our data support the evidence for a stronger RXR response associated with FOXA1 reprograming in ESR1-mutant cells, suggesting development of therapeutic strategies targeting RXR pathways in breast tumors with ESR1 mutation. IMPLICATIONS: It provides comprehensive characterization of the role of FOXA1 in ESR1-mutant breast cancer and potential therapeutic strategy through blocking RXR activation.


Sujet(s)
Tumeurs du sein , Récepteur alpha des oestrogènes , Facteur nucléaire hépatocytaire HNF-3 alpha , Femelle , Humains , Tumeurs du sein/anatomopathologie , Chromatine , Récepteur alpha des oestrogènes/métabolisme , Facteur nucléaire hépatocytaire HNF-3 alpha/métabolisme , Mutation , Récepteurs X des rétinoïdes/génétique , Transcriptome
4.
Br J Pharmacol ; 180(2): 214-234, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36102035

RÉSUMÉ

BACKGROUND AND PURPOSE: Traumatic brain injury (TBI) remains a leading cause of mortality and morbidity in young adults. The role of iron in potentiating neurodegeneration following TBI has gained recent interest as iron deposition has been detected in the injured brain in the weeks to months post-TBI, in both the preclinical and clinical setting. A failure in iron homeostasis can lead to oxidative stress, inflammation and excitotoxicity; and whether this is a cause or consequence of the long-term effects of TBI remains unknown. EXPERIMENTAL APPROACH: We investigated the role of iron and the effect of therapeutic intervention using a brain-permeable iron chelator, deferiprone, in a controlled cortical impact mouse model of TBI. An extensive assessment of cognitive, motor and anxiety/depressive outcome measures were examined, and neuropathological and biochemical changes, over a 3-month period post-TBI. KEY RESULTS: Lesion volume was significantly reduced at 3 months, which was preceded by a reduction in astrogliosis, microglia/macrophages and preservation of neurons in the injured brain at 2 weeks and/or 1 month post-TBI in mice receiving oral deferiprone. Deferiprone treatment showed significant improvements in neurological severity scores, locomotor/gait performance and cognitive function, and attenuated anxiety-like symptoms post-TBI. Deferiprone reduced iron levels, lipid peroxidation/oxidative stress and altered expression of neurotrophins in the injured brain over this period. CONCLUSION AND IMPLICATIONS: Our findings support a detrimental role of iron in the injured brain and suggest that deferiprone (or similar iron chelators) may be promising therapeutic approaches to improve survival, functional outcomes and quality of life following TBI.


Sujet(s)
Lésions traumatiques de l'encéphale , Qualité de vie , Animaux , Souris , Défériprone/pharmacologie , Défériprone/usage thérapeutique , Souris de lignée C57BL , Lésions traumatiques de l'encéphale/traitement médicamenteux , Lésions traumatiques de l'encéphale/métabolisme , Fer
6.
Eur J Appl Physiol ; 121(12): 3421-3430, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34498135

RÉSUMÉ

INTRODUCTION: Maximal strength training (MST), performed with heavy loads (~ 90% of one repetition maximum; 1RM) and few repetitions, yields large improvements in efferent neural drive, skeletal muscle force production, and skeletal muscle efficiency. However, it is elusive whether neural adaptations following such high intensity strength training may be accompanied by alterations in energy-demanding muscular factors. METHODS: Sixteen healthy young males (24 ± 4 years) were randomized to MST 3 times per week for 8 weeks (n = 8), or a control group (CG; n = 8). Measurements included 1RM and rate of force development (RFD), and evoked potentials recordings (V-wave and H-reflex normalized to M-wave (M) in the soleus muscle) applied to assess efferent neural drive to maximally contracting skeletal muscle. Biopsies were obtained from vastus lateralis and analyzed by western blots and real-time PCR to investigate the relative protein expression and mRNA expression of Sarcoplasmic Reticulum Ca2+ ATPase (SERCA) 1 and SERCA2. RESULTS: Significant improvements in 1RM (17 ± 9%; p < 0.001) and early (0-100 ms), late (0-200 ms) and maximal RFD (31-53%; p < 0.01) were observed after MST, accompanied by increased maximal Vmax/Msup-ratio (9 ± 14%; p = 0.046), with no change in H-reflex to M-wave ratio. No changes were observed in the CG. No pre- to post-training differences were found in mRNA or protein expressions of SERCA1 and SERCA2 in either group. CONCLUSION: MST increased efferent neural drive to maximally contracting skeletal muscle, causing improved force production. No change was observed in SERCA expression, indicating that responses to high intensity strength training may predominantly be governed by neural adaptations.


Sujet(s)
Adaptation physiologique , Potentiels évoqués moteurs/physiologie , Contraction musculaire/physiologie , Muscles squelettiques/innervation , Muscles squelettiques/physiologie , Entraînement en résistance , Marqueurs biologiques/métabolisme , Humains , Mâle , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Jeune adulte
7.
Neurotherapeutics ; 18(2): 1081-1094, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33410108

RÉSUMÉ

The accumulation of neurofibrillary tangles (NFTs), which is composed of abnormally hyperphosphorylated tau aggregates, is the classic neuropathology associated with cognitive dysfunction in tauopathies such as Alzheimer's disease (AD). However, there is an emerging theory suggesting that dysregulation in cerebral iron may contribute to NFT formation. Iron is speculated to bind to tau and induce conformational changes of the protein, potentially leading to subsequent aggregation and cognitive decline. Deferiprone (DFP) is a clinically available iron chelator, which has demonstrated potential therapeutic advantages of chelating iron in neurodegenerative disorders, and is currently in clinical trials for AD. However, its effect on tau pathology remains unclear. Here, we report the effects of short-term DFP treatment (4 weeks, 100 mg/kg/daily, via oral gavage) in a mixed-gender cohort of the rTg(tauP301L)4510 mouse model of tauopathy. Our results revealed that DFP improved Y-maze and open field performance, accompanied by a 28% decrease in brain iron levels, measured by inductively coupled plasma mass spectrometry (ICP-MS) and reduced AT8-labeled p-tau within the hippocampus in transgenic tau mice. This data supports the notion that iron may play a neurotoxic role in tauopathies and may be a potential therapeutic target for this class of disorders that can be modulated by the clinically available metal chelator DFP.


Sujet(s)
Vieillissement/effets des médicaments et des substances chimiques , Vieillissement/anatomopathologie , Défériprone/usage thérapeutique , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Tauopathies/traitement médicamenteux , Tauopathies/anatomopathologie , Vieillissement/génétique , Animaux , Défériprone/pharmacologie , Femelle , Humains , Agents chélateurs du fer/pharmacologie , Mâle , Apprentissage du labyrinthe/physiologie , Souris , Souris transgéniques , Tauopathies/génétique , Résultat thérapeutique , Protéines tau/génétique
9.
J Alzheimers Dis ; 77(2): 753-771, 2020.
Article de Anglais | MEDLINE | ID: mdl-32741833

RÉSUMÉ

BACKGROUND: Abnormally hyperphosphorylated tau is a defining pathological feature of tauopathies, such as Alzheimer's disease (AD), and accumulating evidence suggests a role for iron in mediating tau pathology that may lead to cognitive decline in these conditions. The metal chelator deferiprone (DFP), which has a high affinity for iron, is currently in clinical trials for AD and Parkinson's disease. However, the effect of DFP on tau pathology remains underexplored. OBJECTIVE: We aimed to investigate the impact of chronic DFP treatment on tau pathology using a well-characterized mouse model of tauopathy (rTg(tauP301L)4510). METHODS: Animals were treated daily with DFP (100 mg/kg) via oral gavage for 16 weeks. After 14 weeks, mice were tested in the Y-maze, open field, Morris water maze, and rotorod. At the end of the study, brain tissue was collected to examine metal levels (using inductively coupled plasma-mass spectrometry) and for western blot analysis of DFP on tau and iron associated pathways. RESULTS: DFP significantly reduced anxiety-like behavior, and revealed a trend toward improved cognitive function. This was accompanied by a decrease in brain iron levels and sarkosyl-insoluble tau. Our data also showed downregulation of the tau kinases glycogen synthase kinase 3ß and cyclin dependent kinase-5 in DFP treated mice and an increase in the methylation of the catalytic subunit of protein phosphatase 2A. CONCLUSION: These data support the hypothesis that suggests that iron plays a neurotoxic role in tauopathies and may be a potential therapeutic target for this class of disorders.


Sujet(s)
Défériprone/usage thérapeutique , Modèles animaux de maladie humaine , Agents chélateurs du fer/usage thérapeutique , Phénotype , Tauopathies/traitement médicamenteux , Animaux , Défériprone/pharmacologie , Femelle , Fer/métabolisme , Agents chélateurs du fer/pharmacologie , Mâle , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Apprentissage du labyrinthe/physiologie , Souris , Souris transgéniques , Tauopathies/métabolisme
11.
Metallomics ; 12(2): 301-313, 2020 02 26.
Article de Anglais | MEDLINE | ID: mdl-31904058

RÉSUMÉ

A dysregulation in the homeostasis of metals such as copper, iron and zinc is speculated to be involved in the pathogenesis of tauopathies, which includes Alzheimer's disease (AD). In particular, there is a growing body of evidence to support a role for iron in facilitating the hyperphosphorylation and aggregation of the tau protein into neurofibrillary tangles (NFTs) - a primary neuropathological hallmark of tauopathies. Therefore, the aim of this study was to characterize the spatial and temporal brain metallomic profile in a mouse model of tauopathy (rTg(tauP301L)4510), so as to provide some insight into the potential interaction between tau pathology and iron. Using laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS), our results revealed an age-dependent increase in brain iron levels in both WT and rTg(tauP301L)4510 mice. In addition, size exclusion chromatography-ICP-MS (SEC-ICP-MS) revealed significant age-related changes in iron bound to metalloproteins such as ferritin. The outcomes from this study may provide valuable insight into the inter-relationship between iron and tau in ageing and neurodegeneration.


Sujet(s)
Encéphale/métabolisme , Cuivre/métabolisme , Fer/métabolisme , Métaux/métabolisme , Tauopathies/métabolisme , Zinc/métabolisme , Animaux , Chromatographie sur gel , Cuivre/analyse , Modèles animaux de maladie humaine , Fer/analyse , Métaux/analyse , Souris , Zinc/analyse , Protéines tau/métabolisme
12.
Nat Genet ; 52(2): 187-197, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31913353

RÉSUMÉ

Using genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) screens to understand endocrine drug resistance, we discovered ARID1A and other SWI/SNF complex components as the factors most critically required for response to two classes of estrogen receptor-alpha (ER) antagonists. In this context, SWI/SNF-specific gene deletion resulted in drug resistance. Unexpectedly, ARID1A was also the top candidate in regard to response to the bromodomain and extraterminal domain inhibitor JQ1, but in the opposite direction, with loss of ARID1A sensitizing breast cancer cells to bromodomain and extraterminal domain inhibition. We show that ARID1A is a repressor that binds chromatin at ER cis-regulatory elements. However, ARID1A elicits repressive activity in an enhancer-specific, but forkhead box A1-dependent and active, ER-independent manner. Deletion of ARID1A resulted in loss of histone deacetylase 1 binding, increased histone 4 lysine acetylation and subsequent BRD4-driven transcription and growth. ARID1A mutations are more frequent in treatment-resistant disease, and our findings provide mechanistic insight into this process while revealing rational treatment strategies for these patients.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Protéines du cycle cellulaire/métabolisme , Protéines de liaison à l'ADN/métabolisme , Histone Deacetylase 1/métabolisme , Facteurs de transcription/métabolisme , Acétylation , Animaux , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Protéines du cycle cellulaire/génétique , Prolifération cellulaire , Clustered regularly interspaced short palindromic repeats , Protéines de liaison à l'ADN/génétique , Résistance aux médicaments antinéoplasiques/génétique , Récepteur alpha des oestrogènes/génétique , Récepteur alpha des oestrogènes/métabolisme , Femelle , Régulation de l'expression des gènes tumoraux , Facteur nucléaire hépatocytaire HNF-3 alpha/génétique , Facteur nucléaire hépatocytaire HNF-3 alpha/métabolisme , Histone Deacetylase 1/génétique , Humains , Cellules MCF-7 , Souris de lignée NOD , Facteurs de transcription/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Front Mol Neurosci ; 11: 276, 2018.
Article de Anglais | MEDLINE | ID: mdl-30174587

RÉSUMÉ

There is an emerging link between the accumulation of iron in the brain and abnormal tau pathology in a number of neurodegenerative disorders, such as Alzheimer's disease (AD). Studies have demonstrated that iron can regulate tau phosphorylation by inducing the activity of multiple kinases that promote tau hyperphosphorylation and potentially also by impacting protein phosphatase 2A activity. Iron is also reported to induce the aggregation of hyperphosphorylated tau, possibly through a direct interaction via a putative iron binding motif in the tau protein, facilitating the formation of neurofibrillary tangles (NFTs). Furthermore, in human studies high levels of iron have been reported to co-localize with tau in NFT-bearing neurons. These data, together with our own work showing that tau has a role in mediating cellular iron efflux, provide evidence supporting a critical tau:iron interaction that may impact both the symptomatic presentation and the progression of disease. Importantly, this may also have relevance for therapeutic directions, and indeed, the use of iron chelators such as deferiprone and deferoxamine have been reported to alleviate the phenotypes, reduce phosphorylated tau levels and stabilize iron regulation in various animal models. As these compounds are also moving towards clinical translation, then it is imperative that we understand the intersection between iron and tau in neurodegeneration. In this article, we provide an overview of the key pathological and biochemical interactions between tau and iron. We also review the role of iron and tau in disease pathology and the potential of metal-based therapies for tauopathies.

14.
BMC Cancer ; 17(1): 68, 2017 01 21.
Article de Anglais | MEDLINE | ID: mdl-28109268

RÉSUMÉ

BACKGROUND: The peptide hormone gastrin exerts a growth-promoting effect in both normal and malignant gastrointestinal tissue. Gastrin mediates its effect via the cholecystokinin 2 receptor (CCKBR/CCK2R). Although a substantial part of the gastric adenocarcinomas express gastrin and CCKBR, the role of gastrin in tumor development is not completely understood. Autophagy has been implicated in mechanisms governing cytoprotection, tumor growth, and contributes to chemoresistance. This study explores the role of autophagy in response to gastrin in gastric adenocarcinoma cell lines. METHODS: Immunoblotting, survival assays and the xCELLigence system were used to study gastrin induced autophagy. Chemical inhibitors of autophagy were utilized to assess the role of this process in the regulation of cellular responses induced by gastrin. Further, knockdown studies using siRNA and immunoblotting were performed to explore the signaling pathways that activate autophagy in response to gastrin treatment. RESULTS: We demonstrate that gastrin increases the expression of the autophagy markers MAP1LC3B-II and SQSTM1 in gastric adenocarcinoma cells. Gastrin induces autophagy via activation of the STK11-PRKAA2-ULK1 and that this signaling pathway is involved in increased migration and cell survival. Furthermore, gastrin mediated increase in survival of cells treated with cisplatin is partially dependent on induced autophagy. CONCLUSION: This study reveals a novel role of gastrin in the regulation of autophagy. It also opens up new avenues in the treatment of gastric cancer by targeting CCKBR mediated signaling and/or autophagy in combination with conventional cytostatic drugs.


Sujet(s)
Adénocarcinome/génétique , Gastrines/métabolisme , Protéines associées aux microtubules/génétique , Séquestosome-1/génétique , Tumeurs de l'estomac/génétique , Adénocarcinome/métabolisme , Autophagie , Lignée cellulaire tumorale , Mouvement cellulaire , Survie cellulaire , Régulation de l'expression des gènes tumoraux , Humains , Récepteur de la cholécystokinine de type B/métabolisme , Transduction du signal , Tumeurs de l'estomac/métabolisme
15.
Peptides ; 79: 83-94, 2016 05.
Article de Anglais | MEDLINE | ID: mdl-27038741

RÉSUMÉ

The gastric hormone gastrin plays a role in organizing the gastric mucosa. Gastrin also regulates the expression of genes that have important actions in extracellular matrix modelling, including plasminogen activator inhibitor (PAI)-1 which is part of the urokinase plasminogen activator (uPA) system. The uPA system (including PAI-1) is associated with cancer progression, fibrosis and thrombosis. Its biological role in the stomach and molecular mechanisms of action are not well understood. The aim of this study was to examine the effect of PAI-1 on the trophic changes observed in gastric corpus mucosa in hypergastrinemia using PAI-1 and/or HK-ATPase beta subunit knockout (KO) mice. HK-ATPase beta subunit KO mice were used as a model of hypergastrinemia. In 12 month old female mice, intragastric acidity and plasma gastrin were measured. The stomachs were examined for macroscopic and histological changes. In mice null for both PAI-1 and HK-ATPase beta (double KO), there was exaggerated hypergastrinemia, increased stomach weight and corpus mucosal thickness, and more pronounced trophic and architectural changes in the corpus compared with HK-ATPase beta KO mice. Genome-wide microarray expression data for the gastric corpus mucosa showed a distinct gene expression profile for the HK-ATPase beta KO mice; moreover, enrichment analysis revealed changes in expression of genes regulating intracellular processes including cytoskeleton remodelling, cell adhesion, signal transduction and epithelial-to-mesenchymal transition (EMT). Genes differentially expressed in the double KO compared with HK-ATPase beta KO mice included the transcription factor Barx2 and the chromatin remodeler gene Tet2, which may be involved in both normal gastric physiology and development of gastric cancer. Based on the present data, we suggest that PAI-1 plays a role in maintaining gastric mucosal organization in hypergastrinemia.


Sujet(s)
Muqueuse gastrique/enzymologie , Gastrines/sang , Serpine E2/génétique , Animaux , Cellules entérochromaffines-like/enzymologie , Cellules entérochromaffines-like/anatomopathologie , Femelle , Muqueuse gastrique/anatomopathologie , H(+)-K(+)-Exchanging ATPase/génétique , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout , Antre pylorique/enzymologie , Antre pylorique/anatomopathologie , Serpine E2/métabolisme
16.
Malays J Med Sci ; 22(2): 69-72, 2015.
Article de Anglais | MEDLINE | ID: mdl-26023299

RÉSUMÉ

Kimura's disease is an uncommon entity that affects adults, with a predilection for the Asian population. This may rarely be encountered in children, and the knowledge of this fact is essential to rule out the remote possibility of Kimura's disease in children with a slow-growing painless mass in the head and neck region. In this case report, we document this disease in an 8-year-old boy with a slow-growing swelling in the right posterior auricular region.

17.
PLoS One ; 10(3): e0119857, 2015.
Article de Anglais | MEDLINE | ID: mdl-25769101

RÉSUMÉ

Alkylating agents are widely used chemotherapeutics in the treatment of many cancers, including leukemia, lymphoma, multiple myeloma, sarcoma, lung, breast and ovarian cancer. Melphalan is the most commonly used chemotherapeutic agent against multiple myeloma. However, despite a 70-80% initial response rate, virtually all patients eventually relapse due to the emergence of drug-resistant tumour cells. By using global proteomic and transcriptomic profiling on melphalan sensitive and resistant RPMI8226 cell lines followed by functional assays, we discovered changes in cellular processes and pathways not previously associated with melphalan resistance in multiple myeloma cells, including a metabolic switch conforming to the Warburg effect (aerobic glycolysis), and an elevated oxidative stress response mediated by VEGF/IL8-signaling. In addition, up-regulated aldo-keto reductase levels of the AKR1C family involved in prostaglandin synthesis contribute to the resistant phenotype. Finally, selected metabolic and oxidative stress response enzymes were targeted by inhibitors, several of which displayed a selective cytotoxicity against the melphalan-resistant cells and should be further explored to elucidate their potential to overcome melphalan resistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Melphalan/pharmacologie , Voies et réseaux métaboliques/génétique , Myélome multiple/traitement médicamenteux , Myélome multiple/génétique , Stress oxydatif/génétique , Transduction du signal/génétique , Antinéoplasiques alcoylants/pharmacologie , Lignée cellulaire tumorale , Humains , Interleukine-8/génétique , Voies et réseaux métaboliques/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Protéome/effets des médicaments et des substances chimiques , Protéome/génétique , Protéomique/méthodes , Transduction du signal/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques , Transcriptome/génétique , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/génétique , Facteur de croissance endothéliale vasculaire de type A/génétique
18.
PLoS One ; 9(11): e112485, 2014.
Article de Anglais | MEDLINE | ID: mdl-25384047

RÉSUMÉ

Salt-inducible kinase 1 (SIK1/Snf1lk) belongs to the AMP-activated protein kinase (AMPK) family of kinases, all of which play major roles in regulating metabolism and cell growth. Recent studies have shown that reduced levels of SIK1 are associated with poor outcome in cancers, and that this involves an invasive cellular phenotype with increased metastatic potential. However, the molecular mechanism(s) regulated by SIK1 in cancer cells is not well explored. The peptide hormone gastrin regulates cellular processes involved in oncogenesis, including proliferation, apoptosis, migration and invasion. The aim of this study was to examine the role of SIK1 in gastrin responsive adenocarcinoma cell lines AR42J, AGS-GR and MKN45. We show that gastrin, known to signal through the Gq/G11-coupled CCK2 receptor, induces SIK1 expression in adenocarcinoma cells, and that transcriptional activation of SIK1 is negatively regulated by the Inducible cAMP early repressor (ICER). We demonstrate that gastrin-mediated signalling induces phosphorylation of Liver Kinase 1B (LKB1) Ser-428 and SIK1 Thr-182. Ectopic expression of SIK1 increases gastrin-induced phosphorylation of histone deacetylase 4 (HDAC4) and enhances gastrin-induced transcription of c-fos and CRE-, SRE-, AP1- and NF-κB-driven luciferase reporter plasmids. We also show that gastrin induces phosphorylation and nuclear export of HDACs. Next we find that siRNA mediated knockdown of SIK1 increases migration of the gastric adenocarcinoma cell line AGS-GR. Evidence provided here demonstrates that SIK1 is regulated by gastrin and influences gastrin elicited signalling in gastric adenocarcinoma cells. The results from the present study are relevant for the understanding of molecular mechanisms involved in gastric adenocarcinomas.


Sujet(s)
Adénocarcinome/métabolisme , Gastrines/pharmacologie , Hormones/pharmacologie , Protein-Serine-Threonine Kinases/métabolisme , Tumeurs de l'estomac/métabolisme , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Modulateur de l'élément de réponse à l'AMP cyclique/métabolisme , Humains , Phosphorylation , Protein-Serine-Threonine Kinases/génétique , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie
19.
PLoS One ; 8(9): e76234, 2013.
Article de Anglais | MEDLINE | ID: mdl-24086717

RÉSUMÉ

The peptide hormone gastrin is known to play a role in differentiation, growth and apoptosis of cells in the gastric mucosa. In this study we demonstrate that gastrin induces Nuclear Receptor 4A2 (NR4A2) expression in the adenocarcinoma cell lines AR42J and AGS-GR, which both possess the gastrin/CCK2 receptor. In vivo, NR4A2 is strongly expressed in the gastrin responsive neuroendocrine ECL cells in normal mucosa, whereas gastric adenocarcinoma tissue reveals a more diffuse and variable expression in tumor cells. We show that NR4A2 is a primary early transient gastrin induced gene in adenocarcinoma cell lines, and that NR4A2 expression is negatively regulated by inducible cAMP early repressor (ICER) and zinc finger protein 36, C3H1 type-like 1 (Zfp36l1), suggesting that these gastrin regulated proteins exert a negative feedback control of NR4A2 activated responses. FRAP analyses indicate that gastrin also modifies the nucleus-cytosol shuttling of NR4A2, with more NR4A2 localized to cytoplasm upon gastrin treatment. Knock-down experiments with siRNA targeting NR4A2 increase migration of gastrin treated adenocarcinoma AGS-GR cells, while ectopically expressed NR4A2 increases apoptosis and hampers gastrin induced invasion, indicating a tumor suppressor function of NR4A2. Collectively, our results uncover a role of NR4A2 in gastric adenocarcinoma cells, and suggest that both the level and the localization of NR4A2 protein are of importance regarding the cellular responses of these cells.


Sujet(s)
Adénocarcinome/métabolisme , Gastrines/métabolisme , Régulation de l'expression des gènes tumoraux/physiologie , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Tumeurs de l'estomac/métabolisme , Transport nucléaire actif/physiologie , Technique de Western , Facteur BRF-1/métabolisme , Lignée cellulaire tumorale , Rétrocontrôle physiologique/physiologie , Cytométrie en flux , Redistribution de fluorescence après photoblanchiment , Analyse de profil d'expression de gènes , Techniques de knock-down de gènes , Humains , Immunohistochimie , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Petit ARN interférent/génétique , Réaction de polymérisation en chaine en temps réel
20.
Leuk Res ; 35(6): 821-30, 2011 Jun.
Article de Anglais | MEDLINE | ID: mdl-21216463

RÉSUMÉ

P276-00 is a novel cyclin-dependent kinase inhibitor especially potent for Cdk9-T1, Cdk4-D1 and Cdk1-B. Multiple myeloma (MM) is a B-cell malignancy characterized by the accumulation of malignant plasma cells. Treatment of MM cell lines with P276-00 resulted in apoptosis that correlated with transcription inhibition and a significant decline in Mcl-1 protein levels with the appearance of cleaved PARP in these cells. In vivo studies of P276-00 confirmed antitumor activity in RPMI-8226 xenograft. These results suggest that P276-00 causes multiple myeloma cell death by disrupting the balance between cell survival and apoptosis through inhibition of transcription and downregulation of Mcl-1.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Kinase-9 cycline-dépendante/antagonistes et inhibiteurs , Flavones/pharmacologie , Myélome multiple/traitement médicamenteux , Transcription génétique/effets des médicaments et des substances chimiques , Animaux , Protéines régulatrices de l'apoptose/métabolisme , Technique de Western , Cycle cellulaire/effets des médicaments et des substances chimiques , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Kinase-9 cycline-dépendante/génétique , Kinase-9 cycline-dépendante/métabolisme , Kinases cyclines-dépendantes/antagonistes et inhibiteurs , Kinases cyclines-dépendantes/génétique , Kinases cyclines-dépendantes/métabolisme , Relation dose-effet des médicaments , Humains , Souris , Souris SCID , Microscopie confocale , Myélome multiple/génétique , Myélome multiple/anatomopathologie , Protéine Mcl-1 , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Interférence par ARN , RNA polymerase II/métabolisme , RT-PCR , Facteurs temps , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE