Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.108
Filtrer
1.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38999946

RÉSUMÉ

The tumor cells reprogram their metabolism to cover their high bioenergetic demands for maintaining uncontrolled growth. This response can be mediated by cytokines such as IL-2, which binds to its receptor and activates the JAK/STAT pathway. Some reports show a correlation between the JAK/STAT pathway and cellular metabolism, since the constitutive activation of STAT proteins promotes glycolysis through the transcriptional activation of genes related to energetic metabolism. However, the role of STAT proteins in the metabolic switch induced by cytokines in cervical cancer remains poorly understood. In this study, we analyzed the effect of IL-2 on the metabolic switch and the role of STAT5 in this response. Our results show that IL-2 induces cervical cancer cell proliferation and the tyrosine phosphorylation of STAT5. Also, it induces an increase in lactate secretion and the ratio of NAD+/NADH, which suggest a metabolic reprogramming of their metabolism. When STAT5 was silenced, the lactate secretion and the NAD+/NADH ratio decreased. Also, the expression of HIF1α and GLUT1 decreased. These results indicate that STAT5 regulates IL-2-induced cell proliferation and the metabolic shift to aerobic glycolysis by regulating genes related to energy metabolism. Our results suggest that STAT proteins modulate the metabolic switch in cervical cancer cells to attend to their high demand of energy required for cell growth and proliferation.


Sujet(s)
Prolifération cellulaire , Interleukine-2 , Facteur de transcription STAT-5 , Tumeurs du col de l'utérus , Humains , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/génétique , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Interleukine-2/métabolisme , Interleukine-2/pharmacologie , Glycolyse/effets des médicaments et des substances chimiques , Métabolisme énergétique/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 1/génétique , NAD/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Acide lactique/métabolisme
2.
Cell Rep ; 43(7): 114446, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38996065

RÉSUMÉ

The gene encoding the NUAK family kinase 1 (NUAK1) is frequently amplified and its expression is upregulated, activating oncogenic signaling in various cancers. However, little is known about its role in gastric cancer (GC). We investigate the mechanistic links among NUAK1, Hedgehog signaling, and tumorigenesis in GC. NUAK1 overexpression is validated in local and public GC cohorts. Patient-derived xenograft and transgenic mouse models demonstrate that NUAK1 depletion or inhibition dramatically ameliorates gastric tumorigenesis. NUAK1 upregulates GLI1 expression by activating STAT5-mediated transcription and stabilizing GLI1 protein. NUAK1 depletion or inhibition impairs cancer cell expansion, tumor formation, and chemotherapy resistance in in vitro and in vivo models. Clinicopathological analysis confirms that upregulated NUAK1 expression correlates with poor prognosis and chemotherapy resistance in human GC. Our findings demonstrate that the signaling axis NUAK1/STAT5/GLI1 promotes cancer cell expansion and tumorigenesis and indicate that NUAK1 is an attractive therapeutic target and prognostic factor in GC.


Sujet(s)
Prolifération cellulaire , Résistance aux médicaments antinéoplasiques , Facteurs de transcription SOX-B1 , Facteur de transcription STAT-5 , Transduction du signal , Tumeurs de l'estomac , Protéine à doigt de zinc GLI1 , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/traitement médicamenteux , Humains , Protéine à doigt de zinc GLI1/métabolisme , Protéine à doigt de zinc GLI1/génétique , Résistance aux médicaments antinéoplasiques/génétique , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Animaux , Lignée cellulaire tumorale , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Facteurs de transcription SOX-B1/métabolisme , Facteurs de transcription SOX-B1/génétique , Régulation de l'expression des gènes tumoraux , Souris nude , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Mâle , Femelle , Carcinogenèse/anatomopathologie , Carcinogenèse/génétique
3.
Cell Rep Med ; 5(7): 101645, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39019012

RÉSUMÉ

Fms-like tyrosine kinase 3 (FLT3) mutations, present in over 30% of acute myeloid leukemia (AML) cases and dominated by FLT3-internal tandem duplication (FLT3-ITD), are associated with poor outcomes in patients with AML. While tyrosine kinase inhibitors (TKIs; e.g., gilteritinib) are effective, they face challenges such as drug resistance, relapse, and high costs. Here, we report that metformin, a cheap, safe, and widely used anti-diabetic agent, exhibits a striking synergistic effect with gilteritinib in treating FLT3-ITD AML. Metformin significantly sensitizes FLT3-ITD AML cells (including TKI-resistant ones) to gilteritinib. Metformin plus gilteritinib (low dose) dramatically suppresses leukemia progression and prolongs survival in FLT3-ITD AML mouse models. Mechanistically, the combinational treatment cooperatively suppresses polo-like kinase 1 (PLK1) expression and phosphorylation of FLT3/STAT5/ERK/mTOR. Clinical analysis also shows improved survival rates in patients with FLT3-ITD AML taking metformin. Thus, the metformin/gilteritinib combination represents a promising and cost-effective treatment for patients with FLT3-mutated AML, particularly for those with low income/affordability.


Sujet(s)
Dérivés de l'aniline , Protéines du cycle cellulaire , Synergie des médicaments , Leucémie aigüe myéloïde , Metformine , Mutation , , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Pyrazines , Transduction du signal , Tyrosine kinase-3 de type fms , Metformine/pharmacologie , Metformine/usage thérapeutique , Tyrosine kinase-3 de type fms/génétique , Tyrosine kinase-3 de type fms/métabolisme , Tyrosine kinase-3 de type fms/antagonistes et inhibiteurs , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/antagonistes et inhibiteurs , Humains , Animaux , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Pyrazines/pharmacologie , Pyrazines/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Souris , Mutation/génétique , Lignée cellulaire tumorale , Thiophènes/pharmacologie , Thiophènes/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Femelle , Tests d'activité antitumorale sur modèle de xénogreffe , Mâle , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Sérine-thréonine kinases TOR/métabolisme
4.
EMBO Rep ; 25(8): 3678-3706, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39080439

RÉSUMÉ

Adult neural stem cells (NSCs) in the hippocampal dentate gyrus continuously proliferate and generate new neurons throughout life. Although various functions of organelles are closely related to the regulation of adult neurogenesis, the role of endoplasmic reticulum (ER)-related molecules in this process remains largely unexplored. Here we show that Derlin-1, an ER-associated degradation component, spatiotemporally maintains adult hippocampal neurogenesis through a mechanism distinct from its established role as an ER quality controller. Derlin-1 deficiency in the mouse central nervous system leads to the ectopic localization of newborn neurons and impairs NSC transition from active to quiescent states, resulting in early depletion of hippocampal NSCs. As a result, Derlin-1-deficient mice exhibit phenotypes of increased seizure susceptibility and cognitive dysfunction. Reduced Stat5b expression is responsible for adult neurogenesis defects in Derlin-1-deficient NSCs. Inhibition of histone deacetylase activity effectively induces Stat5b expression and restores abnormal adult neurogenesis, resulting in improved seizure susceptibility and cognitive dysfunction in Derlin-1-deficient mice. Our findings indicate that the Derlin-1-Stat5b axis is indispensable for the homeostasis of adult hippocampal neurogenesis.


Sujet(s)
Hippocampe , Homéostasie , Protéines membranaires , Cellules souches neurales , Neurogenèse , Facteur de transcription STAT-5 , Animaux , Neurogenèse/génétique , Souris , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Hippocampe/métabolisme , Hippocampe/cytologie , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris knockout , Crises épileptiques/métabolisme , Crises épileptiques/génétique , Gyrus denté/métabolisme , Gyrus denté/cytologie , Transduction du signal , Prolifération cellulaire
5.
Hematology ; 29(1): 2375045, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39012197

RÉSUMÉ

OBJECTIVES: Constitutive activation of Janus kinase 2 (JAK2)/signal transducer and activator of transcription (STAT) signaling pathway is central to the pathogenesis of myeloproliferative neoplasms (MPNs). Long noncoding RNAs (lncRNAs) regulate diverse biological processes. However, the role of lncRNAs in MPN pathogenesis is not well studied. METHODS: The expression of lnc-AC004893 in MPN patients was measured by quantitative real-time PCR (qRT-PCR). Gene-specific short hairpin RNAs (shRNAs) were designed to inhibit the expression of lnc-AC004893, and western blot was performed to explore the role of lnc-AC004893 via regulating the JAK2/STAT5 signaling pathway. Furthermore, co-IP was performed to determine the binding ability of lnc-AC004893 and STAT5 protein. Finally, the BaF3-JAK2V617F-transplanted mouse model was used to assess the biological role of lnc-ac004893 in vivo. RESULTS: We report that lnc-AC004893, a poorly conserved pseudogene-209, is substantially upregulated in MPN cells compared with normal controls (NCs). Knockdown of lnc-AC004893 by specific shRNAs suppressed cell proliferation and decreased colony formation. Furthermore, the knockdown of lnc-AC004893 reduced the expression of p-STAT5 but not total STAT5 in HEL and murine IL-3-dependent Ba/F3 cells, which present constitutive and inducible activation of JAK2/STAT5 signaling. In addition, inhibition of murine lnc-ac004893 attenuated BaF3-JAK2V617F-transplanted phenotypes and extended the overall survival. Mechanistically, knockdown of lnc-AC004893 enhanced the binding ability of STAT5 and protein tyrosine phosphatase SHP1. Furthermore, knockdown of lnc-AC004893 decreased STAT5-lnc-AC004893 interaction but not SHP1-lnc-AC004893 interaction. CONCLUSION: Lnc-AC004893 regulates STAT5 phosphorylation by affecting the interaction of STAT5 and SHP1. Lnc-AC004893 might be a potential therapeutic target for MPN patients.


Sujet(s)
Syndromes myéloprolifératifs , ARN long non codant , Facteur de transcription STAT-5 , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , ARN long non codant/génétique , Humains , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/métabolisme , Souris , Animaux , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 6/génétique , Kinase Janus-2/métabolisme , Kinase Janus-2/génétique , Transduction du signal , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux
6.
Oncogene ; 43(30): 2338-2354, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38879589

RÉSUMÉ

N6-methyladenosine (m6A) is the predominant post-transcriptional RNA modification in eukaryotes and plays a pivotal regulatory role in various aspects of RNA fate determination, such as mRNA stability, alternative splicing, and translation. Dysregulation of the critical m6A methyltransferase METTL3 is implicated in tumorigenesis and development. Here, this work showed that METTL3 is upregulated in gastric cancer tissues and is associated with poor prognosis. METTL3 methylates the A2318 site within the coding sequence (CDS) region of STAT5A. IGF2BP2 recognizes and binds METTL3-mediated m6A modification of STAT5A through its GXXG motif in the KH3 and KH4 domains, leading to increased stability of STAT5A mRNA. In addition, both METTL3 and IGF2BP2 are positively correlated with STAT5A in human gastric cancer tissue samples. Helicobacter pylori infection increased the expression level of METTL3 in gastric cancer cells, thereby leading to the upregulation of STAT5A. Functional studies indicated that STAT5A overexpression markedly enhances the proliferation and migration of GC cells, whereas STAT5A knockdown has inhibitory effects. Further nude mouse experiments showed that STAT5A knockdown effectively inhibits the growth and metastasis of gastric cancer in vivo. Moreover, as a transcription factor, STAT5A represses KLF4 transcription by binding to its promoter region. The overexpression of KLF4 can counteract the oncogenic impact of STAT5A. Overall, this study highlights the crucial role of m6A in gastric cancer and provides potential therapeutic targets for gastric cancer.


Sujet(s)
Adénosine , Prolifération cellulaire , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Facteur-4 de type Kruppel , Facteurs de transcription Krüppel-like , Methyltransferases , Souris nude , Protéines de liaison à l'ARN , Facteur de transcription STAT-5 , Tumeurs de l'estomac , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Humains , Adénosine/analogues et dérivés , Adénosine/métabolisme , Methyltransferases/métabolisme , Methyltransferases/génétique , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Animaux , Facteurs de transcription Krüppel-like/génétique , Facteurs de transcription Krüppel-like/métabolisme , Souris , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Mâle , Femelle , Infections à Helicobacter/génétique , Infections à Helicobacter/métabolisme , Infections à Helicobacter/anatomopathologie , Mouvement cellulaire/génétique , Souris de lignée BALB C , Helicobacter pylori/génétique , Protéines suppresseurs de tumeurs
7.
J Vet Med Sci ; 86(7): 816-823, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38777776

RÉSUMÉ

Signal transducers and activators of transcription (STATs) are a family of transcription factors involved in various normal physiological cellular processes. Moreover, STATs have been recently identified as novel therapeutic targets for various human tumors. STAT3, STAT5a, and STAT6 have been suggested to be involved in tumorigenesis in human breast cancer. Owing to the similarity between feline mammary carcinomas (FMCs) and human breast cancers, these factors may play an important role in FMCs. However, studies on the expression of STATs in animal tumors are limited. Therefore, in this study, we aimed to characterize the expression of total STAT5 (tSTAT5) and phosphorylated STAT5 (pSTAT5) in FMCs, feline mammary adenomas, non-neoplastic proliferative mammary gland lesions, and normal feline mammary glands using immunohistochemistry. High expression of tSTAT5 was observed in the cytoplasm of all the samples assessed in this study. Moreover, high expression of tSTAT5 was observed in the nucleus; however, its levels varied depending on the lesion. The percentage of pSTAT5-nuclear positive cells varied among normal feline mammary glands (40.1 ± 25.1%), and non-neoplastic lesions, including mammary hyperplasia (43.2 ± 28.6%) and fibroadenomatous changes (18.0 ± 13.6%). Moreover, the percentage of pSTAT5-nuclear-positive cells in feline mammary adenomas was 24.5 ± 19.2%, which was significantly reduced in feline mammary carcinomas (2.4 ± 5.6%), regardless of histopathological subtype. This study suggests that decreased STAT5 activity may be involved in the development and malignant progression of feline mammary carcinomas.


Sujet(s)
Maladies des chats , Tumeurs mammaires de l'animal , Facteur de transcription STAT-5 , Animaux , Chats , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Femelle , Tumeurs mammaires de l'animal/métabolisme , Tumeurs mammaires de l'animal/anatomopathologie , Maladies des chats/métabolisme , Maladies des chats/anatomopathologie , Phosphorylation , Régulation de l'expression des gènes tumoraux , Immunohistochimie/médecine vétérinaire , Glandes mammaires animales/métabolisme , Glandes mammaires animales/anatomopathologie
8.
Cell Commun Signal ; 22(1): 254, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702781

RÉSUMÉ

IL-3/STAT5 signaling pathway is crucial for the development and activation of immune cells, contributing to the cellular response to infections and inflammatory stimuli. Dysregulation of the IL-3/STAT5 signaling have been associated with inflammatory and autoimmune diseases characterized by inflammatory cell infiltration and organ damage. IL-3 receptor α (IL-3Rα) specifically binds to IL-3 and initiates intracellular signaling, resulting in the phosphorylation of STAT5. However, the regulatory mechanisms of IL-3Rα remain unclear. Here, we identified the E3 ubiquitin ligase RNF128 as a negative regulator of IL-3/STAT5 signaling by targeting IL-3Rα for lysosomal degradation. RNF128 was shown to selectively bind to IL-3Rα, without interacting with the common beta chain IL-3Rß, which shares the subunit with GM-CSF. The deficiency of Rnf128 had no effect on GM-CSF-induced phosphorylation of Stat5, but it resulted in heightened Il-3-triggered activation of Stat5 and increased transcription of the Id1, Pim1, and Cd69 genes. Furthermore, we found that RNF128 promoted the K27-linked polyubiquitination of IL-3Rα in a ligase activity-dependent manner, ultimately facilitating its degradation through the lysosomal pathway. RNF128 inhibited the activation and chemotaxis of macrophages in response to LPS stimulation, thereby attenuating excessive inflammatory responses. Collectively, these results reveal that RNF128 negatively regulates the IL-3/STAT5 signaling pathway by facilitating K27-linked polyubiquitination of IL-3Rα. This study uncovers E3 ubiquitin ligase RNF128 as a novel regulator of the IL-3/STAT5 signaling pathway, providing potential molecular targets for the treatment of inflammatory diseases.


Sujet(s)
Interleukine-3 , Facteur de transcription STAT-5 , Transduction du signal , Ubiquitin-protein ligases , Ubiquitination , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Humains , Animaux , Interleukine-3/métabolisme , Souris , Lysosomes/métabolisme , Cellules HEK293 , Phosphorylation , Récepteurs à l'interleukine-3/métabolisme , Récepteurs à l'interleukine-3/génétique
9.
Endocrinology ; 165(7)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38728240

RÉSUMÉ

GH acts in numerous organs expressing the GH receptor (GHR), including the brain. However, the mechanisms behind the brain's permeability to GH and how this hormone accesses different brain regions remain unclear. It is well-known that an acute GH administration induces phosphorylation of the signal transducer and activator of transcription 5 (pSTAT5) in the mouse brain. Thus, the pattern of pSTAT5 immunoreactive cells was analyzed at different time points after IP or intracerebroventricular GH injections. After a systemic GH injection, the first cells expressing pSTAT5 were those near circumventricular organs, such as arcuate nucleus neurons adjacent to the median eminence. Both systemic and central GH injections induced a medial-to-lateral pattern of pSTAT5 immunoreactivity over time because GH-responsive cells were initially observed in periventricular areas and were progressively detected in lateral brain structures. Very few choroid plexus cells exhibited GH-induced pSTAT5. Additionally, Ghr mRNA was poorly expressed in the mouse choroid plexus. In contrast, some tanycytes lining the floor of the third ventricle expressed Ghr mRNA and exhibited GH-induced pSTAT5. The transport of radiolabeled GH into the hypothalamus did not differ between wild-type and dwarf Ghr knockout mice, indicating that GH transport into the mouse brain is GHR independent. Also, single-photon emission computed tomography confirmed that radiolabeled GH rapidly reaches the ventral part of the tuberal hypothalamus. In conclusion, our study provides novel and valuable information about the pattern and mechanisms behind GH transport into the mouse brain.


Sujet(s)
Encéphale , Hormone de croissance , Récepteur STH , Facteur de transcription STAT-5 , Animaux , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Encéphale/métabolisme , Hormone de croissance/métabolisme , Souris , Récepteur STH/métabolisme , Récepteur STH/génétique , Mâle , Souris knockout , Souris de lignée C57BL , Phosphorylation , Plexus choroïde/métabolisme , Hypothalamus/métabolisme , Injections ventriculaires
10.
J Exp Clin Cancer Res ; 43(1): 144, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38745318

RÉSUMÉ

BACKGROUND: Neuroendocrine prostate cancer (NEPC) is a lethal subset of prostate cancer which is characterized by neuroendocrine differentiation and loss of androgen receptor (AR) signaling. Growing evidence reveals that cell lineage plasticity is crucial in the failure of NEPC therapies. Although studies suggest the involvement of the neural transcription factor PAX6 in drug resistance, its specific role in NEPC remains unclear. METHODS: The expression of PAX6 in NEPC was identified via bioinformatics and immunohistochemistry. CCK8 assay, colony formation assay, tumorsphere formation assay and apoptosis assay were used to illustrate the key role of PAX6 in the progression of in vitro. ChIP and Dual-luciferase reporter assays were conducted to confirm the binding sequences of AR in the promoter region of PAX6, as well as the binding sequences of PAX6 in the promoter regions of STAT5A and MET. For in vivo validation, the xenograft model representing NEPC subtype underwent pathological analysis to verify the significant role of PAX6 in disease progression. Complementary diagnoses were established through public clinical datasets and transcriptome sequencing of specific cell lines. ATAC-seq was used to detect the chromatin accessibility of specific cell lines. RESULTS: PAX6 expression was significantly elevated in NEPC and negatively regulated by AR signaling. Activation of PAX6 in non-NEPC cells led to NE trans-differentiation, while knock-down of PAX6 in NEPC cells inhibited the development and progression of NEPC. Importantly, loss of AR resulted in an enhanced expression of PAX6, which reprogramed the lineage plasticity of prostate cancer cells to develop NE phenotypes through the MET/STAT5A signaling pathway. Through ATAC-seq, we found that a high expression level of PAX6 elicited enhanced chromatin accessibility, mainly through attenuation of H4K20me3, which typically causes chromatin silence in cancer cells. CONCLUSION: This study reveals a novel neural transcription factor PAX6 could drive NEPC progression and suggest that it might serve as a potential therapeutic target for the management of NEPC.


Sujet(s)
Chromatine , Facteur de transcription PAX6 , Tumeurs de la prostate , Facteur de transcription STAT-5 , Animaux , Humains , Mâle , Souris , Lignée cellulaire tumorale , Chromatine/métabolisme , Chromatine/génétique , Régulation de l'expression des gènes tumoraux , Facteur de transcription PAX6/métabolisme , Facteur de transcription PAX6/génétique , Phénotype , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Transduction du signal , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/métabolisme
11.
Int Immunopharmacol ; 133: 112166, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38678673

RÉSUMÉ

Dendritic cells (DCs) are specialized antigen-presenting cells that play an important role in inducing and maintaining immune tolerance. The altered distribution and/or function of DCs contributes to defective tolerance in autoimmune diseases such as type 1 diabetes (T1D). In human T1D and in NOD mouse models, DCs share some defects and are often described as less tolerogenic and excessively immunogenic. In the NOD mouse model, the autoimmune response is associated with a defect in the Stat5b signaling pathway. We have reported that expressing a constitutively active form of Stat5b in DCs of transgenic NOD mice (NOD.Stat5b-CA), re-established their tolerogenic function, restored autoimmune tolerance and conferred protection from diabetes. However, the role and molecular mechanisms of Stat5b signaling in regulating splenic conventional DCs tolerogenic signature remained unclear. In this study, we reported that, compared to immunogenic splenic DCs of NOD, splenic DCs of NOD.Stat5b-CA mice exhibited a tolerogenic profile marked by elevated PD-L1 and PD-L2 expression, reduced pro-inflammatory cytokine production, increased frequency of the cDC2 subset and decreased frequency of the cDC1 subset. This tolerogenic profile was associated with increased Ezh2 and IRF4 but decreased IRF8 expression. We also found an upregulation of PD-L1 in the cDC1 subset and high PD-L1 and PD-L2 expression in cDC2 of NOD.Stat5b-CA mice. Mechanistically, we demonstrated that Ezh2 plays an important role in the maintenance of high PD-L1 expression in cDC1 and cDC2 subsets and that Ezh2 inhibition resulted in PD-L1 but not PD-L2 downregulation which was more drastic in the cDC2 subset. Additionally, Ezh2 inhibition severely reduced the cDC2 subset and increased the cDC1 subset and Stat5b-CA.DC pro-inflammatory cytokine production. Together our data suggest that the Stat5b-Ezh2 axis is critical for the maintenance of tolerogenic high PD-L1-expressing cDC2 and autoimmune tolerance in NOD.Stat5b-CA mice.


Sujet(s)
Antigène CD274 , Cellules dendritiques , Diabète de type 1 , Protéine-2 homologue de l'activateur de Zeste , Facteur de transcription STAT-5 , Animaux , Femelle , Humains , Souris , Antigène CD274/métabolisme , Antigène CD274/génétique , Cellules cultivées , Cytokines/métabolisme , Cellules dendritiques/immunologie , Diabète de type 1/immunologie , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Tolérance immunitaire , Souris de lignée NOD , Souris transgéniques , Transduction du signal , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique
12.
Am J Hematol ; 99(6): 1108-1118, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38563187

RÉSUMÉ

We investigated using a custom NGS panel of 149 genes the mutational landscape of 64 consecutive adult patients with tyrosine kinase fusion-negative hypereosinophilia (HE)/hypereosinophilic syndrome (HES) harboring features suggestive of myeloid neoplasm. At least one mutation was reported in 50/64 (78%) patients (compared to 8/44 (18%) patients with idiopathic HE/HES/HEUS used as controls; p < .001). Thirty-five patients (54%) had at least one mutation involving the JAK-STAT pathway, including STAT5B (n = 18, among which the hotspot N642H, n = 13), JAK1 (indels in exon 13, n = 5; V658F/L, n = 2), and JAK2 (V617F, n = 6; indels in exon 13, n = 2). Other previously undescribed somatic mutations were also found in JAK2, JAK1, STAT5B, and STAT5A, including three patients who shared the same STAT5A V707fs mutation and features consistent with primary polycythemia. Nearly all JAK-STAT mutations were preceded by (or associated with) myelodysplasia-related gene mutations, especially in RNA-splicing genes or chromatin modifiers. In multivariate analysis, neurologic involvement (hazard ratio [HR] 4.95 [1.87-13.13]; p = .001), anemia (HR 5.50 [2.24-13.49]; p < .001), and the presence of a high-risk mutation (as per the molecular international prognosis scoring system: HR 6.87 [2.39-19.72]; p < .001) were independently associated with impaired overall survival. While corticosteroids were ineffective in all treated JAK-STAT-mutated patients, ruxolitinib showed positive hematological responses including in STAT5A-mutated patients. These findings emphasize the usefulness of NGS for the workup of tyrosine kinase fusion-negative HE/HES patients and support the use of JAK inhibitors in this setting. Updated classifications could consider patients with JAK-STAT mutations and eosinophilia as a new "gene mutated-entity" that could be differentiated from CEL, NOS, and idiopathic HES.


Sujet(s)
Syndrome hyperéosinophilique , Mutation , Facteur de transcription STAT-5 , Humains , Syndrome hyperéosinophilique/génétique , Syndrome hyperéosinophilique/traitement médicamenteux , Mâle , Femelle , Adulte d'âge moyen , Adulte , Sujet âgé , Facteur de transcription STAT-5/génétique , Kinase Janus-2/génétique , Transduction du signal , Janus kinase 1/génétique , Sujet âgé de 80 ans ou plus , Pyrimidines/usage thérapeutique , Jeune adulte
13.
J Clin Invest ; 134(8)2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38618957

RÉSUMÉ

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive immature T cell cancer. Mutations in IL7R have been analyzed genetically, but downstream effector functions such as STAT5A and STAT5B hyperactivation are poorly understood. Here, we studied the most frequent and clinically challenging STAT5BN642H driver in T cell development and immature T cell cancer onset and compared it with STAT5A hyperactive variants in transgenic mice. Enhanced STAT5 activity caused disrupted T cell development and promoted an early T cell progenitor-ALL phenotype, with upregulation of genes involved in T cell receptor (TCR) signaling, even in absence of surface TCR. Importantly, TCR pathway genes were overexpressed in human T-ALL and mature T cell cancers and activation of TCR pathway kinases was STAT5 dependent. We confirmed STAT5 binding to these genes using ChIP-Seq analysis in human T-ALL cells, which were sensitive to pharmacologic inhibition by dual STAT3/5 degraders or ZAP70 tyrosine kinase blockers in vitro and in vivo. We provide genetic and biochemical proof that STAT5A and STAT5B hyperactivation can initiate T-ALL through TCR pathway hijacking and suggest similar mechanisms for other T cell cancers. Thus, STAT5 or TCR component blockade are targeted therapy options, particularly in patients with chemoresistant clones carrying STAT5BN642H.


Sujet(s)
Leucémie-lymphome lymphoblastique à précurseurs T , Animaux , Humains , Souris , Souris transgéniques , Leucémie-lymphome lymphoblastique à précurseurs T/génétique , Protein-tyrosine kinases , Récepteurs aux antigènes des cellules T/génétique , Transduction du signal , Facteur de transcription STAT-5/génétique
14.
J Interferon Cytokine Res ; 44(4): 178-189, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38579140

RÉSUMÉ

Chronic myeloid leukemia (CML) is a clonal myeloproliferative hematological disease characterized by the chimeric breakpoint-cluster region/Abelson kinase1 (BCR::ABL1) oncoprotein; playing a pivotal role in CML molecular pathology, diagnosis, treatment, and possible resistance arising from the success and tolerance of tyrosine kinase inhibitor (TKI)-based therapy. The transcription factor STAT5 constitutive signaling, which is influenced by the cytokine signaling network, triggers BCR::ABL1-based CML pathogenesis and is also relevant to acquired TKI resistance. The unsuccessful therapeutic approaches targeting BCR::ABL1, in particular third-line therapy with ponatinib, still need to be further developed with alternative combination strategies to overcome drug resistance. As treatment with the STAT5 inhibitor pimozide in combination with ponatinib resulted in an efficient and synergistic therapeutic approach in TKI-resistant CML cells, this study focused on identifying the underlying amplification of ponatinib response mechanisms by determining different cytokine expression profiles in parental and ponatinib-resistant CML cells, in vitro. The results showed that expression of interleukin (IL) 1B, IL9, and IL12A-B was increased by 2-fold, while IL18 was downregulated by 2-fold in the ponatinib-resistant cells compared to sensitive ones. Importantly, ponatinib treatment upregulated the expression of 21 of the 23 interferon and IL genes in the ponatinib-resistant cells, while treatment with pimozide or a combination dose resulted in a reduction in the expression of 19 different cytokine genes, such as for example, inflammatory cytokines, IL1A-B and IL6 or cytokine genes associated with supporting tumor progression, leukemia stem cell growth or poor survival, such as IL3, IL8, IL9, IL10, IL12, or IL15. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis results showed that the genes were mainly enriched in the regulation of receptor signaling through the Janus kinase/signal transducer and activator of transcription pathway, cytokine-cytokine receptor interaction, and hematopoietic cell lineage. Protein-protein interaction analysis showed that IL2, IL6, IL15, IFNG, and others appeared in the top lists of pathways, indicating their high centrality and importance in the network. Therefore, pimozide could be a promising agent to support TKI therapies in ponatinib resistance. This research would help to clarify the role of cytokines in ponatinib resistance and advance the development of new therapeutics to utilize the STAT5 inhibitor pimozide in combination with TKIs.


Sujet(s)
Imidazoles , Leucémie myéloïde chronique BCR-ABL positive , Pimozide , Pyridazines , Humains , Pimozide/pharmacologie , Pimozide/usage thérapeutique , Cytokines/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Protéines de fusion bcr-abl/génétique , Protéines de fusion bcr-abl/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Facteur de transcription STAT-5/génétique , Facteur de transcription STAT-5/métabolisme , Interleukine-15/métabolisme , Interleukine-15/usage thérapeutique , Interleukine-6/métabolisme , Interleukine-9/métabolisme , Interleukine-9/usage thérapeutique , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie
16.
Blood ; 143(24): 2474-2489, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38498036

RÉSUMÉ

ABSTRACT: Patients with T- and natural killer (NK)-cell neoplasms frequently have somatic STAT5B gain-of-function mutations. The most frequent STAT5B mutation is STAT5BN642H, which is known to drive murine T-cell leukemia, although its role in NK-cell malignancies is unclear. Introduction of the STAT5BN642H mutation into human NK-cell lines enhances their potential to induce leukemia in mice. We have generated a mouse model that enables tissue-specific expression of STAT5BN642H and have selectively expressed the mutated STAT5B in hematopoietic cells (N642Hvav/+) or exclusively in NK cells (N642HNK/NK). All N642Hvav/+ mice rapidly develop an aggressive T/NKT-cell leukemia, whereas N642HNK/NK mice display an indolent NK-large granular lymphocytic leukemia (NK-LGLL) that progresses to an aggressive leukemia with age. Samples from patients with NK-cell leukemia have a distinctive transcriptional signature driven by mutant STAT5B, which overlaps with that of murine leukemic N642HNK/NK NK cells. To our knowledge, we have generated the first reliable STAT5BN642H-driven preclinical mouse model that displays an indolent NK-LGLL progressing to aggressive NK-cell leukemia. This novel in vivo tool will enable us to explore the transition from an indolent to an aggressive disease and will thus permit the study of prevention and treatment options for NK-cell malignancies.


Sujet(s)
Cellules tueuses naturelles , Leucémie à grands lymphocytes granuleux , Facteur de transcription STAT-5 , Animaux , Facteur de transcription STAT-5/génétique , Facteur de transcription STAT-5/métabolisme , Souris , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/anatomopathologie , Humains , Leucémie à grands lymphocytes granuleux/génétique , Leucémie à grands lymphocytes granuleux/anatomopathologie , Modèles animaux de maladie humaine , Lignage cellulaire/génétique , Mutation , Souris transgéniques
17.
J Cell Mol Med ; 28(3): e18114, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38323741

RÉSUMÉ

Patients with Philadelphia chromosome-like acute lymphoblastic leukaemia (Ph-like ALL) often face a grim prognosis, with PDGFRB gene fusions being commonly detected in this subgroup. Our study has unveiled a newfound fusion gene, TERF2::PDGFRB, and we have found that patients carrying this fusion gene exhibit sensitivity to dasatinib. Ba/F3 cells harbouring the TERF2::PDGFRB fusion display IL-3-independent cell proliferation through activation of the p-PDGFRB and p-STAT5 signalling pathways. These cells exhibit reduced apoptosis and demonstrate sensitivity to imatinib in vitro. When transfused into mice, Ba/F3 cells with the TERF2::PDGFRB fusion gene induce tumorigenesis and a shortened lifespan in cell-derived graft models, but this outcome can be improved with imatinib treatment. In summary, we have identified the novel TERF2::PDGFRB fusion gene, which exhibits oncogenic potential both in vitro and in vivo, making it a potential therapeutic target for tyrosine kinase inhibitors (TKIs).


Sujet(s)
Protéines de fusion oncogènes , Leucémie-lymphome lymphoblastique à précurseurs B et T , Récepteur au PDGF bêta , Protéine-2 de liaison aux répétitions télomériques , Animaux , Humains , Souris , Carcinogenèse , Transformation cellulaire néoplasique , Mésilate d'imatinib , Inhibiteurs de protéines kinases/pharmacologie , Récepteur au PDGF bêta/génétique , Transduction du signal , Facteur de transcription STAT-5/génétique , Protéine-2 de liaison aux répétitions télomériques/génétique , Protéines de fusion oncogènes/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique
18.
Cell Death Dis ; 15(2): 128, 2024 02 10.
Article de Anglais | MEDLINE | ID: mdl-38341429

RÉSUMÉ

Previous study showed that higher expression of prolactin (PRL) was found in CRPC samples compared with hormone-naive prostate cancer (HNPC) and benign prostatic hyperplasia (BPH) samples. We further investigate the function of PRL in prostate cancer (PCa) and explored its downstream effects. We found heterogeneous expression of the PRLR in clinical prostate samples. The VCaP and 22Rv1 cells exhibited PRLR expression. Among the downstream proteins, STAT5B was the dominant subtype in clinical samples and cell lines. Human recombinant PRL stimulation of PCa cells with PRLR expression resulted in increased phosphorylation of STAT5B(pSTAT5B) and progression of PCa in vitro and in vivo, and STAT5B knockdown can suppress the malignant behavior of PCa. To understand the mechanism further, we performed Bioinformatic analysis, ChIP qPCR, and luciferase reporter gene assay. The results revealed that ARRB2 was the transcription target gene of STAT5B, and higher expression of ARRB2 was related to higher aggression and poorer prognosis of PCa. Additionally, Gene set enrichment analysis indicated that higher expression of ARRB2 was significantly enriched in the MAPK signaling pathway. Immunohistochemistry (IHC) demonstrated elevated pSTAT5B, ARRB2, and pERK1/2 expression levels in CRPC tissues compared to HNPC and BPH. Mechanically, ARRB2 enhanced the activation of the MAPK pathway by binding to ERK1/2, thereby promoting the phosphorylation of ERK1/2 (pERK1/2). In conclusion, our study demonstrated that PRL stimulation can promote the progression of PCa through STAT5B/ARRB2 pathway and activation of MAPK signaling, which can be suppressed by intervention targeting STAT5B. Blockade of the STAT5B can be a potential therapeutic target for PCa.


Sujet(s)
Hyperplasie de la prostate , Tumeurs prostatiques résistantes à la castration , Tumeurs de la prostate , Mâle , Humains , Prolactine/génétique , Prolactine/métabolisme , Hyperplasie de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Récepteur prolactine/métabolisme , Phosphorylation , Lignée cellulaire tumorale , Facteur de transcription STAT-5/génétique , Facteur de transcription STAT-5/métabolisme , bêta-Arrestine 2/métabolisme
19.
Growth Horm IGF Res ; 74: 101572, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38281404

RÉSUMÉ

OBJECTIVE: GATA2 is a key transcription factor involved in the differentiation and determination of thyrotrophs and gonadotrophs in pituitary and hematopoietic development. However, studies on the upstream ligands of the GATA2 signal transduction pathway have been limited. To identify upstream ligands, we examined growth hormone (GH) as a plausible stimulator. DESIGN: We evaluated GH-induced GATA2 expression in murine TtT/GF thyrotrophic pituitary tumor cells and its direct impact on the GHR/JAK/STAT5 pathway using a combination of a reporter assay, real-time quantitative polymerase chain reaction, and western blotting. RESULTS: GATA2 expression increased with activated STAT5B in a dose-dependent manner and was inhibited by a STAT5 specific inhibitor. Moreover, we found functional STAT5B binding site consensus sequences at -359 bp in the GATA2 promoter region. CONCLUSION: These findings suggest that GH directly stimulates GATA2 via the GHR/JAK/STAT pathway and participates in various developmental phenomena mediated by GATA2.


Sujet(s)
Hormone de croissance , Hormone de croissance humaine , Souris , Animaux , Hormone de croissance/métabolisme , Facteur de transcription STAT-5/génétique , Facteur de transcription STAT-5/métabolisme , Janus kinases/métabolisme , Transduction du signal , Facteurs de transcription STAT/métabolisme , Hormone de croissance humaine/métabolisme , Protéines de lait
20.
Gene ; 900: 148131, 2024 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-38216003

RÉSUMÉ

Precursor B cell acute lymphoblastic leukemia (Pre-B-ALL) arises from developing B cells and frequently involves mutations in genes encoding transcription factors. In this study, we investigated the function of mutations in the transcription factor IKZF3 (Aiolos), R137* and H195Y, discovered in a mouse model of pre-B-ALL. R137* IKZF3 mutation resulted in a truncated protein, while electrophoretic mobility shift assay showed that H195Y IKZF3 mutation resulted in a protein with altered DNA binding. 38B9 pre-B cell lines were generated expressing WT and H195Y IKZF3 proteins. Anti-IKZF3 ChIP-seq showed that H195Y IKZF3 interacted with a larger number of sites that were different than WT IKZF3. Treatment with interleukin-7 induced changes in gene expression in 38B9 cells expressing WT IKZF3, but did not induce any changes in gene expression in cells expressing H195Y IKZF3. Anti-STAT5 ChIP-seq showed that expression of H195Y IKZF3 resulted in redistribution of STAT5 binding sites in the genome. H195Y IKZF3 binding sites overlapped with a subset of STAT5 binding sites, including in the promoter of the Cish gene. These findings suggest that H195Y mutation of IKZF3 results in altered DNA binding specificity and altered binding of STAT5 to target genes.


Sujet(s)
Leucémie B , Leucémie-lymphome lymphoblastique à précurseurs B , Animaux , Souris , Sites de fixation , ADN , Expression des gènes , Protéines de lait/génétique , Mutation , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Facteur de transcription STAT-5/génétique , Facteur de transcription STAT-5/métabolisme , Transactivateurs/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE