Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 168
Filtrer
1.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(3): 359-366, 2024 Mar 28.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38970509

RÉSUMÉ

OBJECTIVES: Adverse cardiovascular events are the leading cause of death in peritoneal dialysis patients. Identifying indicators that can predict adverse cardiovascular events in these patients is crucial for prognosis. This study aims to assess the value of dual-specificity phosphatase 6 (DUSP6) in peripheral blood mononuclear cells as a predictor of adverse cardiovascular events after peritoneal dialysis in diabetic nephropathy patients. METHODS: A total of 124 diabetic nephropathy patients underwent peritoneal dialysis treatment at the Department of Nephrology of the First Affiliated Hospital of Hebei North University from June to September 2022 were selected as study subjects. The levels of DUSP6 in peripheral blood mononuclear cells were determined using Western blotting. Patients were categorized into high-level and low-level DUSP6 groups based on the median DUSP6 level. Differences in body mass index, serum albumin, high-sensitivity C-reactive protein, and dialysis duration were compared between the 2 groups. Pearson, Spearman, and multiple linear regression analyses were performed to examine factors related to DUSP6. Patients were followed up to monitor the occurrence of adverse cardiovascular events, and risk factors for adverse cardiovascular events after peritoneal dialysis were analyzed using Kaplan-Meier and Cox regression. RESULTS: By the end of the follow-up, 33 (26.61%) patients had experienced at least one adverse cardiovascular event. The high-level DUSP6 group had higher body mass index, longer dialysis duration, and higher high-sensitivity C-reactive protein, but lower serum albumin levels compared to the low-level DUSP6 group (all P<0.05). DUSP6 was negatively correlated with serum albumin levels (r=-0.271, P=0.002) and positively correlated with dialysis duration (rs=0.406, P<0.001) and high-sensitivity C-reactive protein (rs=0.367, P<0.001). Multiple linear regression analysis revealed that dialysis duration and high-sensitivity C-reactive protein were independently correlated with DUSP6 levels (both P<0.05). The cumulative incidence of adverse cardiovascular events was higher in the high-level DUSP6 group than in the low-level DUSP6 group (46.67% vs 7.81%, P<0.001). Cox regression analysis indicated that low serum albumin levels (HR=0.836, 95% CI 0.778 to 0.899), high high-sensitivity C-reactive protein (HR=1.409, 95% CI 1.208 to 1.644), and high DUSP6 (HR=6.631, 95% CI 2.352 to 18.693) were independent risk factors for adverse cardiovascular events in peritoneal dialysis patients. CONCLUSIONS: Dialysis duration and high-sensitivity C-reactive protein are independently associated with DUSP6 levels in peripheral blood mononuclear cells of diabetic nephropathy patients undergoing peritoneal dialysis. High DUSP6 levels indicate a higher risk of adverse cardiovascular events.


Sujet(s)
Maladies cardiovasculaires , Néphropathies diabétiques , Dual Specificity Phosphatase 6 , Agranulocytes , Dialyse péritonéale , Humains , Dialyse péritonéale/effets indésirables , Maladies cardiovasculaires/étiologie , Néphropathies diabétiques/sang , Dual Specificity Phosphatase 6/génétique , Femelle , Mâle , Agranulocytes/métabolisme , Facteurs de risque , Protéine C-réactive/métabolisme , Adulte d'âge moyen , Pronostic , Sérumalbumine/métabolisme , Sérumalbumine/analyse
2.
EMBO Mol Med ; 16(7): 1603-1629, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38886591

RÉSUMÉ

Despite clinical benefits of tyrosine kinase inhibitors (TKIs) in cancer, most tumors can reactivate proliferation under TKI therapy. Here we present transcriptional profiling of HER2+ breast cancer cells transitioning from dormant drug tolerant cells to re-proliferating cells under continuous HER2 inhibitor (HER2i) therapy. Focusing on phosphatases, expression of dual-specificity phosphatase DUSP6 was found inhibited in dormant cells, but strongly induced upon regrowth. DUSP6 expression also selectively associated with poor patient survival in HER2+ breast cancers. DUSP6 overexpression conferred apoptosis resistance, whereas its pharmacological blockade prevented therapy tolerance development under HER2i therapy. DUSP6 targeting also synergized with clinically used HER2i combination therapies. Mechanistically DUSP6 is a positive regulator of HER3 expression, and its impact on HER2i tolerance was mediated by neuregulin-HER3 axis. In vivo, genetic targeting of DUSP6 reduced tumor growth in brain metastasis model, whereas its pharmacological targeting induced synthetic lethal therapeutic effect in combination with HER2i. Collectively this work demonstrates that DUSP6 drives escape from HER2i-induced dormancy, and that DUSP6 is a druggable target to overcome HER3-driven TKI resistance.


Sujet(s)
Tumeurs du sein , Dual Specificity Phosphatase 6 , Récepteur ErbB-2 , Récepteur ErbB-3 , Dual Specificity Phosphatase 6/métabolisme , Dual Specificity Phosphatase 6/génétique , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Femelle , Récepteur ErbB-2/métabolisme , Animaux , Récepteur ErbB-3/métabolisme , Récepteur ErbB-3/génétique , Récepteur ErbB-3/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Souris , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie
3.
J Mol Neurosci ; 74(3): 59, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890235

RÉSUMÉ

Binge drinking causes a range of problems especially damage to the nervous system, and the specific neural mechanism of brain loss and behavioral abnormalities caused by which is still unclear. Extracellular regulated protein kinases (ERK) maintain neuronal survival, growth, and regulation of synaptic plasticity by phosphorylating specific transcription factors to regulate expression of brain-derived neurotrophic factor (BDNF). Dual-specific phosphatase 1 (DUSP1) and DUSP6 dephosphorylate tyrosine and serine/threonine residues in ERK1/2 to inactivate them. To investigate the molecular mechanism by which alcohol affects memory and emotion, a chronic intermittent alcohol exposure (CIAE) model was established. The results demonstrated that mice in the CIAE group developed short-term recognition memory impairment and anxiety-like behavior; meanwhile, the expression of DUSP1 and DUSP66 in the mPFC was increased, while the levels of p-ERK and BDNF were decreased. Micro-injection of DUSP1/6 inhibitor BCI into the medial prefrontal cortex (mPFC) restored the dendritic morphology by reversing the activity of ERK-BDNF and ultimately improved cognitive and emotional impairment caused by CIAE. These findings indicate that CIAE inhibits ERK-BDNF by increasing DUSP1/6 in the mPFC that may be associated with cognitive and emotional deficits. Consequently, DUSP1 and DUSP6 appear to be potential targets for the treatment of alcoholic brain disorders.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Dual Specificity Phosphatase 1 , Éthanol , Souris de lignée C57BL , Cortex préfrontal , Animaux , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/génétique , Souris , Mâle , Dual Specificity Phosphatase 1/métabolisme , Dual Specificity Phosphatase 1/génétique , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Éthanol/toxicité , Éthanol/pharmacologie , Dual Specificity Phosphatase 6/métabolisme , Dual Specificity Phosphatase 6/génétique , Amino-acétonitrile/analogues et dérivés , Amino-acétonitrile/pharmacologie , Amino-acétonitrile/usage thérapeutique , Anxiété/traitement médicamenteux , Anxiété/étiologie , Système de signalisation des MAP kinases
4.
Biochem Pharmacol ; 224: 116247, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38697311

RÉSUMÉ

Current therapeutic options for renal cell carcinoma (RCC) are very limited, which is largely due to inadequate comprehension of molecular pathological mechanisms as well as RCC's resistance to chemotherapy. Dual-specificity phosphatase 6 (DUSP6) has been associated with numerous human diseases. However, its role in RCC is not well understood. Here, we show that diminished DUSP6 expression is linked to RCC progression and unfavorable prognosis. Mechanistically, DUSP6 serves as a tumor suppressor in RCC by intervening the TAF10 and BSCL2 via the ERK-AKT pathway. Further, DUSP6 is also transcriptionally regulated by HNF-4a. Moreover, docking experiments have indicated that DUSP6 expression is enhanced when bound by Calcium saccharate, which also inhibits RCC cell proliferation, metabolic rewiring, and sunitinib resistance. In conclusion, our study identifies Calcium saccharate as a prospective pharmacological therapeutic approach for RCC.


Sujet(s)
Antinéoplasiques , Néphrocarcinome , Dual Specificity Phosphatase 6 , Glycolyse , Tumeurs du rein , Protéines proto-oncogènes c-akt , Sunitinib , Humains , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/métabolisme , Néphrocarcinome/anatomopathologie , Sunitinib/pharmacologie , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/métabolisme , Tumeurs du rein/anatomopathologie , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/physiologie , Lignée cellulaire tumorale , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Dual Specificity Phosphatase 6/métabolisme , Dual Specificity Phosphatase 6/génétique , Antinéoplasiques/pharmacologie , Souris , Souris nude , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/physiologie , Mâle
5.
Anticancer Res ; 44(6): 2545-2554, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38821599

RÉSUMÉ

BACKGROUND/AIM: Epidermal growth factor receptor (EGFR) over-expression is commonly observed in advanced head and neck squamous cell carcinoma (HNSCC) and is correlated with poor patient outcomes. However, the role of dual-specificity phosphatase 6 (DUSP6) in EGFR-associated HNSCC progression remains poorly understood. This study aimed to investigate the correlation between DUSP6 expression and EGFR signaling in malignant HNSCC tissues. MATERIALS AND METHODS: Data mining and in vitro assays were employed to assess DUSP6 expression levels in HNSCC tissues compared to normal tissues. Additionally, the correlation between DUSP6 and EGFR expression was examined. Functional assays were conducted to investigate the modulation of DUSP6 expression by EGFR signaling and its involvement in EGF-induced cell migration and anoikis resistance. RESULTS: Our analysis revealed a significant elevation in DUSP6 expression in HNSCC tissues compared to normal tissues and a strong correlation between DUSP6 and EGFR expression. EGFR signaling modulated DUSP6 expression in a dose- and time-dependent manner, primarily through the extracellular signal-regulated kinase (ERK) pathway. Knockdown experiments demonstrated the functional role of DUSP6 in EGF-induced cell migration and anoikis resistance. CONCLUSION: The findings of this study elucidate the intricate signaling networks governing DUSP6 expression and its interplay with EGFR signaling in HNSCC. Moreover, the results provide insights into the potential role of DUSP6 as a therapeutic target and highlight the importance of personalized treatment strategies in HNSCC management.


Sujet(s)
Mouvement cellulaire , Dual Specificity Phosphatase 6 , Tumeurs de la tête et du cou , Carcinome épidermoïde de la tête et du cou , Humains , Anoïkis/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique , Carcinome épidermoïde/métabolisme , Lignée cellulaire tumorale , Évolution de la maladie , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/métabolisme , Transduction du signal , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme
6.
Biochem Biophys Res Commun ; 715: 150004, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38678784

RÉSUMÉ

Megakaryopoiesis and platelet production is a complex process that is underpotential regulation at multiple stages. Many long non-coding RNAs (lncRNAs) are distributed in hematopoietic stem cells and platelets. lncRNAs may play important roles as key epigenetic regulators in megakaryocyte differentiation and proplatelet formation. lncRNA NORAD can affect cell ploidy by sequestering PUMILIO proteins, although its direct effect on megakaryocyte differentiation and thrombopoiesis is still unknown. In this study, we demonstrate NORAD RNA is highly expressed in the cytoplasm during megakaryocyte differentiation. Interestingly, we identified for the first time that NORAD has a strong inhibitory effect on megakaryocyte differentiation and proplatelet formation from cultured megakaryocytes. DUSP6/ERK1/2 pathway is activated in response to NORAD knockdown during megakaryocytopoiesis, which is achieved by sequestering PUM2 proteins. Finally, compared with the wild-type control mice, NORAD knockout mice show a faster platelet recovery after severe thrombocytopenia induced by 6 Gy total body irradiation. These findings demonstrate lncRNA NORAD has a key role in regulating megakaryocyte differentiation and thrombopoiesis, which provides a promising molecular target for the treatment of platelet-related diseases such as severe thrombocytopenia.


Sujet(s)
Plaquettes , Différenciation cellulaire , Dual Specificity Phosphatase 6 , Mégacaryocytes , ARN long non codant , Thrombopoïèse , Animaux , Humains , Souris , Plaquettes/métabolisme , Différenciation cellulaire/génétique , Cellules cultivées , Dual Specificity Phosphatase 6/métabolisme , Dual Specificity Phosphatase 6/génétique , Système de signalisation des MAP kinases , Mégacaryocytes/métabolisme , Mégacaryocytes/cytologie , Souris de lignée C57BL , Souris knockout , ARN long non codant/génétique , ARN long non codant/métabolisme , Thrombopénie/génétique , Thrombopénie/métabolisme , Thrombopénie/anatomopathologie , Thrombopoïèse/génétique
7.
Chemosphere ; 344: 140358, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37797900

RÉSUMÉ

Particulate matter (PM), one of the most serious air contaminants, could easily pass through the airway and deposit at the deep alveoli. Thus, it might trigger respiratory diseases like inflammation, asthma and lung cancer on human. Long non-coding RNAs (lncRNAs) are considered as important regulator in promotion and progression of diverse cancers. However, the molecular mechanism of lncRNAs mediating PM-induced lung carcinogenesis remains unclear. In this study, we established a 16HBE malignant transformed cell induced by PM (Cells were treated with 20 µg/ml PM, which named PM-T cells) and explored the roles and mechanisms of lncRNAs in the malignant transformation induced by PM. Compared with 16HBE cells, various biological functions were changed in PM-T cells, such as cell proliferation, migration, cell cycle and apoptosis. LncRNA SPRY4-IT1 was significant down-regulated expression and associated with these biological effects. Our results showed that lncRNA SPRY4-IT1 overexpression reversed these functional changes mentioned above. The further studies indicated that lncRNA SPRY4-IT1 involved in PM-induced cell transformation by modulating Chk1 expression via negative regulation of DUSP6-ERK1/2. In conclusion, our studies suggested that lncRNA SPRY4-IT1 played the role as a tumor suppressor gene and might mediate 16HBE cells malignant transformation induced by PM through regulating DUSP6-ERK1/2-Chk1 signaling pathway.


Sujet(s)
ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Lignée cellulaire tumorale , Système de signalisation des MAP kinases , Régulation de l'expression des gènes tumoraux , Transduction du signal , Prolifération cellulaire/génétique , Mouvement cellulaire , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme
8.
Arch Esp Urol ; 76(6): 445-453, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-37681336

RÉSUMÉ

BACKGROUND: Nucleolar prominence is a biomarker of prostate cancer (CaP), and the nucleolar protein block of proliferation 1 (BOP1) participates in the development of CaP, which has great significance for CaP therapy. Thus, this study explored the mechanism of BOP1 in CaP development. METHODS: BOP1 expression levels in the tumor tissues of CaP patients and in PC3 tumor cells were determined. The viability, apoptosis rate of PC3 cells, and apoptosis-related proteins levels were determined to explore the effect of BOP1 on tumor-cell growth in vitro. BOP1 function in the metastasis of PC3 cells was further assessed by Transwell experiment. We also studied the influence of BOP1 on the expression of mitogen-activated protein kinase (MAPK) pathway-related proteins and investigated the regulatory effect of BOP1 on dual-specificity phosphatase 6 (DUSP6). RESULTS: BOP1 expression was upregulated in the tumor tissues and PC3 cells of CaP patients. BOP1 knockout reduced the activity of PC3 cells and induced apoptosis, significantly inhibiting the metastasis of PC3 cells. DUSP6 was overexpressed in tumor tissues and PC3 cells. BOP1 knockout inhibited DUSP6 expression and the MAPK pathway. DUSP6 overexpression reversed the inhibition of BOP1 siRNA (si-BOP1) on PC3 cells and the activated MAPK signaling pathway. CONCLUSIONS: This finding demonstrated that BOP1 promoted CaP progression by regulating the DUSP6/MAPK pathway.


Sujet(s)
Mitogen-Activated Protein Kinases , Tumeurs de la prostate , Mâle , Humains , Tumeurs de la prostate/génétique , Transduction du signal , Prolifération cellulaire , Dual Specificity Phosphatase 6/génétique , Protéines de liaison à l'ARN
9.
J Assist Reprod Genet ; 40(7): 1597-1610, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-37300650

RÉSUMÉ

PURPOSE: Dysregulated behaviors of trophoblast cells leading to defective placentation are considered the main cause of preeclampsia (PE). Abnormal miRNA expression profiles have been observed in PE placental tissue, indicating the significant role of miRNAs in PE development. This study aimed to investigate the expression of miR-101-5p in PE placental tissue and its biological functions. METHODS: The expression of miR-101-5p in placental tissue was detected by quantitative real-time PCR (qRT-PCR). The localization of miR-101-5p in term placental tissue and decidual tissue was determined by the fluorescence in situ hybridization (FISH)-immunofluorescence (IF) double labeling assay. The effect of miR-101-5p on the migration, invasion, proliferation, and apoptosis of the HTR8/SVneo trophoblast cells was investigated. Online databases combined with transcriptomics were used to identify potential target genes and related pathways of miR-101-5p. Finally, the interaction between miR-101-5p and the target gene was verified by qRT-PCT, WB, dual-luciferase reporter assay, and rescue experiments. RESULTS: The study found that miR-101-5p was upregulated in PE placental tissue compared to normal controls and was mainly located in various trophoblast cell subtypes in placental and decidual tissues. Overexpression of miR-101-5p impaired the migration and invasion of HTR8/SVneo cells. DUSP6 was identified as a potential downstream target of miR-101-5p. The expression of miR-101-5p was negatively correlated with DUSP6 expression in HTR8/SVneo cells, and miR-101-5p directly bound to the 3' UTR region of DUSP6. DUSP6 upregulation rescued the migratory and invasive abilities of HTR8/SVneo cells in the presence of miR-101-5p overexpression. Additionally, miR-101-5p downregulated DUSP6, resulting in enhanced ERK1/2 phosphorylation. CONCLUSION: This study revealed that miR-101-5p inhibits the migration and invasion of HTR8/SVneo cells by regulating the DUSP6-ERK1/2 axis, providing a new molecular mechanism for the pathogenesis of PE.


Sujet(s)
microARN , Pré-éclampsie , Humains , Grossesse , Femelle , Placenta/métabolisme , Trophoblastes/métabolisme , Pré-éclampsie/anatomopathologie , Hybridation fluorescente in situ , Système de signalisation des MAP kinases/génétique , Lignée cellulaire , microARN/génétique , microARN/métabolisme , Prolifération cellulaire/génétique , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme
10.
Sci Rep ; 13(1): 5683, 2023 04 07.
Article de Anglais | MEDLINE | ID: mdl-37029196

RÉSUMÉ

Cultured human pluripotent stem cells (hPSCs) grow as colonies that require breakdown into small clumps for further propagation. Although cell death mechanism by single-cell dissociation of hPSCs has been well defined, how hPSCs respond to the deadly stimulus and recover the original status remains unclear. Here we show that dissociation of hPSCs immediately activates ERK, which subsequently activates RSK and induces DUSP6, an ERK-specific phosphatase. Although the activation is transient, DUSP6 expression persists days after passaging. DUSP6 depletion using the CRISPR/Cas9 system reveals that DUSP6 suppresses the ERK activity over the long term. Elevated ERK activity by DUSP6 depletion increases both viability of hPSCs after single-cell dissociation and differentiation propensity towards mesoderm and endoderm lineages. These findings provide new insights into how hPSCs respond to dissociation in order to maintain pluripotency.


Sujet(s)
Cellules souches pluripotentes , Transduction du signal , Humains , Rétroaction , Différenciation cellulaire , Mort cellulaire , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme
11.
Int J Mol Sci ; 24(6)2023 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-36982663

RÉSUMÉ

We investigated the expression and biological function of retinoic acid inducible gene I (RIG-I) in esophageal squamous cell carcinoma (ESCC). Materials and methods: An immunohistochemical analysis was performed on 86 pairs of tumor tissue and adjacent normal tissue samples of patients with ESCC. We generated RIG-I-overexpressing ESCC cell lines KYSE70 and KYSE450, and RIG-I- knockdown cell lines KYSE150 and KYSE510. Cell viability, migration and invasion, radioresistance, DNA damage, and cell cycle were evaluated using CCK-8, wound-healing and transwell assay, colony formation, immunofluorescence, and flow cytometry and Western blotting, respectively. RNA sequencing was performed to determine the differential gene expression between controls and RIG-I knockdown. Tumor growth and radioresistance were assessed in nude mice using xenograft models. RIG-I expression was higher in ESCC tissues compared with that in matched non-tumor tissues. RIG-I overexpressing cells had a higher proliferation rate than RIG-I knockdown cells. Moreover, the knockdown of RIG-I slowed migration and invasion rates, whereas the overexpression of RIG-I accelerated migration and invasion rates. RIG-I overexpression induced radioresistance and G2/M phase arrest and reduced DNA damage after exposure to ionizing radiations compared with controls; however, it silenced the RIG-I enhanced radiosensitivity and DNA damage, and reduced the G2/M phase arrest. RNA sequencing revealed that the downstream genes DUSP6 and RIG-I had the same biological function; silencing DUSP6 can reduce the radioresistance caused by the overexpression of RIG-I. RIG-I knockdown depleted tumor growth in vivo, and radiation exposure effectively delayed the growth of xenograft tumors compared with the control group. RIG-I enhances the progression and radioresistance of ESCC; therefore, it may be a new potential target for ESCC-targeted therapy.


Sujet(s)
Carcinome épidermoïde , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Animaux , Humains , Souris , Carcinogenèse/génétique , Carcinome épidermoïde/génétique , Carcinome épidermoïde/radiothérapie , Carcinome épidermoïde/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Transformation cellulaire néoplasique/génétique , Dual Specificity Phosphatase 6/génétique , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/radiothérapie , Tumeurs de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/génétique , Régulation de l'expression des gènes tumoraux , Souris nude , Récepteurs à l'acide rétinoïque/métabolisme
12.
Cancer Lett ; 558: 216092, 2023 04 01.
Article de Anglais | MEDLINE | ID: mdl-36806557

RÉSUMÉ

Despite many advances in treatment over the past few years, the poor 5-year survival rate and high recurrence rate of gastric cancer (GC) remain unsatisfactory. As the most abundant epigenetic modification in the eukaryotic mRNA, N6-methyladenosine (m6A) methylation participates in tumor progression and tissue development. During tumor progression, DNA damage repair mechanisms can be reprogrammed to give new growth advantages on tumor clones whose genomic integrity is disturbed. Here we detected the elevated SUV39H2 expression in GC tissues and cell lines. Functionally, SUV39H2 promoted GC proliferation and inhibited apoptosis in vitro and in vivo. Mechanistically, METTL3-mediated m6A modification promotes mRNA stability of SUV39H2 in an IGF2BP2 dependent manner, resulting in upregulated mRNA expression of SUV39H2. As a histone methyltransferase, SUV39H2 was verified to increase the phosphorylation level of ATM through transcriptional repression of DUSP6, thereby promoting HRR and ultimately inhibiting GC chemosensitivity to cisplatin. Collectively, these results indicate the specific mechanism of m6A-modified SUV39H2 as a histone methyltransferase promoting HRR to inhibit the chemosensitivity of GC. SUV39H2 is expected to become a key target in the precision targeted therapy of GC.


Sujet(s)
Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/anatomopathologie , Répression épigénétique , Lignée cellulaire tumorale , Recombinaison homologue , Histone méthyltransférases/génétique , ARN messager , Methyltransferases/métabolisme , Protéines de liaison à l'ARN/génétique , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme , Histone-lysine N-methyltransferase/génétique
14.
Pathol Int ; 72(11): 529-540, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36161420

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) develops via dysplastic changes in the epithelia graded as low- and high-grade with accumulation of molecular alterations. Constitutive activation of mitogen-activated protein kinase (MAPK) contributed by attenuation of DUSP6 plays a key role in sustaining PDAC. Active MAPK induces various molecules that function as effectors to sustain PDAC. AURKA and SON are downstream effectors that contribute substantially to the proliferation and survival of PDAC cells and are potentially useful as therapeutic targets. Active MAPK also promote microRNAs that modulate the proliferation of PDAC cells and are useful as diagnostic markers. Familial pancreatic cancer kindreds in Japan show various germline mutations supposed to increase a pancreatic cancer risk. Intraductal papillary mucinous neoplasms (IPMNs) consist of dilated ducts lined by papillary neoplastic epithelia of various shapes and varying grades of atypia. Various papillae of IPMNs are classified into four subtypes that are associated with clinicopathological features, including patient prognosis. GNAS is a specific driver gene for the development of IPMN through gain-of-function mutations. Tracing of molecular alterations has elucidated the mechanism of progression of IPMN from dysplasia to carcinoma, as well as one type of papillae. Intraductal tubulopapillary neoplasms belong to a distinct class of pancreatic neoplasms.


Sujet(s)
Carcinogenèse , Carcinome du canal pancréatique , Tumeurs intracanalaires pancréatiques , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/anatomopathologie , Tumeurs intracanalaires pancréatiques/génétique , Tumeurs intracanalaires pancréatiques/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Dual Specificity Phosphatase 6/génétique , Aurora kinase A/génétique
15.
Acta Biochim Biophys Sin (Shanghai) ; 54(10): 1431-1440, 2022 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-36017891

RÉSUMÉ

ERK1/2 are essential proteins mediating mitogen-activated protein kinase signaling downstream of RAS in pancreatic adenocarcinoma (PDAC). Our previous study reveals that ARF6 plays a positive regulatory role in ERK1/2 pathway in a feedback loop manner. A significant part of the literature on ARF6 has emphasized its oncogenic effect as an essential downstream molecule of ERK1/2, and no research has been done on the regulation mechanisms of the feedback loop between ARF6 and the ERK1/2 signaling pathway. In the present study, we explore the gene network downstream of ARF6 and find that DUSP6 may be the critical signal molecule in the positive feedback loop between ARF6 and ERK1/2. Specifically, to elucidate the negative correlations between ARF6 and DUSP6 in pancreatic cancer, we examine their expressions in pancreatic cancer tissues by immunohistochemical staining. Then the impact of DUSP6 on the proliferation and apoptosis of PDAC cells are investigated by gain-of-function and loss-of-function approaches. Mechanism explorations uncover that ARF6 suppresses the expression of DUSP6, which is responsible for the dephosphorylation of ERK1/2. Altogether, these results indicate that DUSP6 plays a tumor-suppressive role and acts as an intermediate molecule between ARF6 and ERK1/2 in PDAC cells, thereby forming a positive feedback loop.


Sujet(s)
Adénocarcinome , Tumeurs du pancréas , Humains , Système de signalisation des MAP kinases , Rétroaction , Tumeurs du pancréas/génétique , Mitogen-Activated Protein Kinase 1/métabolisme , Transduction du signal , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme , Tumeurs du pancréas
16.
Dev Biol ; 488: 81-90, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35598626

RÉSUMÉ

Pre-placodal ectoderm (PPE), a horseshoe-shaped narrow region formed during early vertebrate development, gives rise to multiple types of sensory organs and ganglia. For PPE induction, a certain level of FGF signal activation is required. However, it is difficult to reproducibly induce the narrow region with variations in gene expression, including FGF, among individuals. An intracellular regulatory factor of FGF signaling, Dusp6, is expressed by FGF signal activation and inactivates a downstream regulator, ERK1/2, in adult tissues; however, its role in early development is not well known. Here, we reveal that Dusp6 is expressed in an FGF-dependent manner in Xenopus PPE. Gain- and loss-of-function experiments showed that Dusp6 is required for expression of a PPE gene, Six1, and patterning of adjacent regions, neural plate, and neural crest. To reveal the importance of Dusp6 in variable FGF production, we performed Dusp6 knockdown with FGF-bead implantation, which resulted in varying Six1 expression patterns. Taken together, these results suggest that Dusp6 is required for PPE formation and that it contributes to the robust patterning of PPE by mediating FGF signaling.


Sujet(s)
Ectoderme , Plaque neurale , Animaux , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme , Ectoderme/métabolisme , Régulation de l'expression des gènes au cours du développement/génétique , Protéines à homéodomaine/métabolisme , Humains , Crête neurale/métabolisme , Plaque neurale/métabolisme , Protéines de Xénope/génétique , Protéines de Xénope/métabolisme , Xenopus laevis/génétique , Xenopus laevis/métabolisme
17.
FEBS Open Bio ; 12(7): 1388-1405, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35478300

RÉSUMÉ

Neuroblastoma (NB) is a heterogeneous cancer of the sympathetic nervous system, which accounts for 7-10% of paediatric malignancies worldwide. Due to the lack of targetable molecular aberrations in NB, most treatment options remain relatively nonspecific. Here, we investigated the therapeutic potential of BCI, an inhibitor of DUSP1 and DUSP6, in cultured NB cells. BCI was cytotoxic in a range of NB cell lines and induced a short-lived activation of the AKT and stress-inducible MAP kinases, although ERK phosphorylation was unaffected. Furthermore, a phosphoproteomic screen identified significant upregulation of JNK signalling components and suppression in mTOR and R6K signalling. To assess the specificity of BCI, CRISPR-Cas9 was employed to introduce insertions and deletions in the DUSP1 and DUSP6 genes. Surprisingly, BCI remained fully cytotoxic in NB cells with complete loss of DUSP6 and partial depletion of DUSP1, suggesting that BCI exerts cytotoxicity in NB cells through a complex mechanism that is unrelated to these phosphatases. Overall, these data highlight the risk of using an inhibitor such as BCI as supposedly specific DUSP1/6, without understanding its full range of targets in cancer cells.


Sujet(s)
Antinéoplasiques , Dual Specificity Phosphatase 1 , Dual Specificity Phosphatase 6 , Neuroblastome , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Dual Specificity Phosphatase 1/génétique , Dual Specificity Phosphatase 6/génétique , Humains , Neuroblastome/traitement médicamenteux , Neuroblastome/génétique , Phosphorylation , Transduction du signal
18.
Oncogene ; 41(20): 2811-2823, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35418690

RÉSUMÉ

The cytoplasmic phosphatase DUSP6 and its nuclear counterpart DUSP5 are negative regulators of RAS/ERK signalling. Here we use deletion of either Dusp5 or Dusp6 to explore the roles of these phosphatases in a murine model of KRASG12D-driven pancreatic cancer. By 56-days, loss of either DUSP5 or DUSP6 causes a significant increase in KRASG12D-driven pancreatic hyperplasia. This is accompanied by increased pancreatic acinar to ductal metaplasia (ADM) and the development of pre-neoplastic pancreatic intraepithelial neoplasia (PanINs). In contrast, by 100-days, pancreatic hyperplasia is reversed with significant atrophy of pancreatic tissue and weight loss observed in animals lacking either DUSP5 or DUSP6. On further ageing, Dusp6-/- mice display accelerated development of metastatic pancreatic ductal adenocarcinoma (PDAC), while in Dusp5-/- animals, although PDAC development is increased this process is attenuated by atrophy of pancreatic acinar tissue and severe weight loss in some animals before cancer could progress. Our data suggest that despite a common target in the ERK MAP kinase, DUSP5 and DUSP6 play partially non-redundant roles in suppressing oncogenic KRASG12D signalling, thus retarding both tumour initiation and progression. Our data suggest that loss of either DUSP5 or DUSP6, as observed in certain human tumours, including the pancreas, could promote carcinogenesis.


Sujet(s)
Carcinome du canal pancréatique , Dual Specificity Phosphatase 6 , Dual-specificity phosphatases , Tumeurs du pancréas , Animaux , Atrophie/anatomopathologie , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Dual Specificity Phosphatase 6/génétique , Dual-specificity phosphatases/génétique , Hyperplasie , Souris , Pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes p21(ras)/génétique , Perte de poids , Tumeurs du pancréas
19.
J Invest Dermatol ; 142(9): 2499-2507.e6, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35189148

RÉSUMÉ

A subset of dual-specificity phosphatases is a major negative regulator of MAPKs, and their involvement in tumorigenesis remains controversial. Among them, DUSP4 is reported to preferentially dephosphorylate extracellular signal‒regulated kinase (ERK) 1/2 and c-Jun N-terminal kinase over p38. In this study, we aimed to identify a possible role of DUSP4 in melanoma genesis. An examination of large-scale public data on gene expression and dependency revealed a considerably high DUSP4 expression and dependency of the melanoma cell lines compared with those of other tumor cell lines, which was not apparent for the other 24 dual-specificity phosphatases genes encoded in the human genome. Using two melanoma lines, we confirmed that DUSP4 depletion impaired cell growth without notably inducing apoptosis. Interestingly, immunoblotting and kinase translocation reporter data revealed that DUSP4 depletion induces a decrease in ERK1/2 phosphorylation but barely affects c-Jun N-terminal kinase phosphorylation, suggesting that neither ERK nor c-Jun N-terminal kinase is a direct target of DUSP4 in our experimental setting. Notably, DUSP4 depletion led to an increase in DUSP6 level, possibly through a post-transcriptional process, and DUSP6 knockout almost eliminated the DUSP4-depletion effect on cell growth and ERK activity. Our findings suggest that DUSP4 plays a role in maintaining a high ERK1/2 activity by negatively regulating DUSP6 and thus contributes to the survival and growth of melanoma cells.


Sujet(s)
Dual Specificity Phosphatase 6 , Dual-specificity phosphatases , Système de signalisation des MAP kinases , Mélanome , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme , Dual-specificity phosphatases/génétique , Dual-specificity phosphatases/métabolisme , Humains , JNK Mitogen-Activated Protein Kinases/métabolisme , Mélanome/génétique , Mitogen-Activated Protein Kinase Phosphatases/génétique , Mitogen-Activated Protein Kinase Phosphatases/métabolisme , Phosphorylation , Régulation positive
20.
Int J Mol Med ; 49(3)2022 Mar.
Article de Anglais | MEDLINE | ID: mdl-35014672

RÉSUMÉ

Airway epithelial cell (AEC) dysfunction has been proven to be involved in the pathogenesis of asthma, which may be induced by the use of dexamethasone (Dex). The altered expression of microRNAs (miRNAs/miRs) has been found in asthma. However, the detailed mechanisms responsible for the effects of miR­375 on Dex­induced AEC dysfunction remain elusive. Thus, the present study aimed to elucidate these mechanisms. Following treatment with Dex for 0, 6, 12 and 24 h, AEC viability, migration, invasion and apoptosis were examined using Cell Counting Kit­8 (CCK­8), wound healing and Transwell assays, and flow cytometry, respectively. The expression levels of miR­375, dual specificity phosphatase 6 (DUSP6) and apoptosis­related proteins (Bcl­2, Bax, cleaved caspase­3) were measured using reverse transcription­quantitative polymerase chain reaction and western blot analysis. The target genes and potential binding sites of miR­375 and DUSP6 were predicted using TargetScan and confirmed using dual­luciferase reporter assay. The viability, migration, invasion and apoptosis of Dex­treated AECs were further assessed with or without miR­375 and DUSP6. In the AECs (9HTE cells), Dex treatment suppressed cell viability and miR­375 expression, whereas it promoted cell apoptosis and the expression of DUSP6, the target gene of miR­375. The overexpression of miR­375 reversed the effects of Dex treatment on miR­375 expression, cell viability, migration and invasion, and apoptosis­related protein expression; in turn, these effects were reversed by the overexpression of DUSP6, with the exception of miR­375 expression. On the whole, the present study demonstrates that the overexpression of miR­375 counteracts the effects of Dex treatment on AEC viability, migration, invasion and apoptosis by targeting DUSP6. Thus, it was suggested that the downregulated expression of miR­375 may be a therapeutic target for AEC dysfunction.


Sujet(s)
Dual Specificity Phosphatase 6 , microARN , Apoptose/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Dexaméthasone/pharmacologie , Dual Specificity Phosphatase 6/génétique , Dual Specificity Phosphatase 6/métabolisme , Cellules épithéliales/métabolisme , Régulation de l'expression des gènes tumoraux , Humains , microARN/génétique , microARN/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...