Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.707
Filtrer
1.
Cells ; 13(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273030

RÉSUMÉ

The prolactin (PRL) hormone is a major regulator of mammary gland development and lactation. However, it remains unclear whether and how PRL contributes to mammary epithelial cell proliferation and secretion. The Boer and Macheng black crossbred goats are superior in reproduction, meat, and milk, and are popular in Hubei province. To elucidate the mechanisms of PRL on mammary growth and lactation, to improve the local goat economic trade, we have performed studies on these crossbred goats during pregnancy and early lactation, and in goat mammary epithelial cells (GMECs). Here, we first found that the amino acid transporters of SNAT1 and SNAT2 expression in vivo and in vitro were closely associated with PRL levels, the proliferation and secretion of GMECs; knockdown and over-expression of SNAT1/2 demonstrated that PRL modulated the proliferation and lactation of GMECs through regulating SNAT1/2 expression. Transcriptome sequencing and qPCR assays demonstrated the effect of PRL on the transcriptional regulation of SNAT1 and SNAT2 in GMECs. Dual-luciferase reporter gene assays further verified that the binding of the potential PRL response element in the SNAT1/2 promoter regions activated SNAT1/2 transcription after PRL stimulation. Additionally, silencing of either PRLR or STAT5 nearly abolished PRL-stimulated SNAT1/2 promoter activity, suggesting PRLR-STAT5 signaling is involved in the regulation of PRL on the transcriptional activation of SNAT1/2. These results illustrated that PRL modulates the proliferation and secretion of GMECs via PRLR-STAT5-mediated regulation of the SNAT1/2 pathway. This study provides new insights into how PRL affects ruminant mammary development and lactation through regulation of amino acid transporters.


Sujet(s)
Prolifération cellulaire , Cellules épithéliales , Capra , Lactation , Glandes mammaires animales , Prolactine , Animaux , Prolactine/métabolisme , Femelle , Cellules épithéliales/métabolisme , Glandes mammaires animales/métabolisme , Glandes mammaires animales/cytologie , Glandes mammaires animales/croissance et développement , Facteur de transcription STAT-5/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Grossesse , Régions promotrices (génétique)/génétique
2.
Theranostics ; 14(14): 5698-5724, 2024.
Article de Anglais | MEDLINE | ID: mdl-39310105

RÉSUMÉ

Background: Glioblastoma (GBM) is characterized by abundant neovascularization as an essential hallmark. Vasculogenic mimicry (VM) is a predominant pattern of GBM neovascularization. However, the biological functions of circRNAs prompting VM formation in GBM remains unclarified. Methods: The circular RNA circCMTM3 was identified through high-throughput sequencing and bioinformatics analysis. The expression of circCMTM3 in exosomes in glioma tissues and cells was verified via RT-qPCR and FISH. In vitro and in vivo assays, such as EdU, MTS, Transwell, and tube formation assays were performed to investigate functional roles of circCMTM3. Meanwhile, in situ tumorigenesis assay were implemented to explore the influences of circCMTM3 on the GBM progression. Additionally, RNA pull-down, RIP, ChIP, and dual-luciferase reporter gene assays were executed to confirm the underlying regulation mechanism of circCMTM3. Results: CircCMTM3, as a novel circular RNA, was packaged into exosomes derived from glioblastoma stem cells (GSCs), which facilitates the phenotypic transition of differentiated glioma cells (DGCs) to VM. Mechanistically, exosomal circCMTM3 is internalized by DGCs and disrupt the ubiquitination degradation of STAT5A and STAT5B by E3 ubiquitin ligase CNOT4. Additionally, through molecular scaffold function of circCMTM3, STAT5A is activated and triggers transcriptional regulation of target genes including the pro-vasculogenic factor CHI3L2 and the RNA-binding protein SRSF1. Subsequently, circCMTM3/STAT5A/SRSF1 positive feedback loop sustainably enhances VM formation and accelerates tumor progression in GBM. Conclusion: Exosomal circCMTM3 possessing growth factor-mimetic property activates the JAK2/STAT5A pathway via non-canonical manner, and promotes VM formation in GBM. The molecular communications between GSCs and DGCs offers a therapeutic strategy for targeting the neovascularization of GBM.


Sujet(s)
Tumeurs du cerveau , Exosomes , Glioblastome , Cellules souches tumorales , Néovascularisation pathologique , ARN circulaire , Facteur de transcription STAT-5 , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/génétique , ARN circulaire/métabolisme , ARN circulaire/génétique , Humains , Facteur de transcription STAT-5/métabolisme , Exosomes/métabolisme , Néovascularisation pathologique/métabolisme , Lignée cellulaire tumorale , Animaux , Cellules souches tumorales/métabolisme , Souris , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Phosphorylation , Souris nude , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Protéines suppresseurs de tumeurs
3.
J Agric Food Chem ; 72(37): 20331-20342, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39253853

RÉSUMÉ

Cytochrome P450 monooxygenases in insects have been verified to implicated in insecticide and phytochemical detoxification metabolism. However, the regulation of P450s, which are modulated by signal-regulated transcription factors (TFs), is less well studied in insects. Here, we found that the Malpighian tubule specific P450 gene SlCYP9A75b in Spodoptera litura is induced by xenobiotics. The transgenic Drosophila bioassay and RNAi results indicated that this P450 gene contributes to α-cypermethrin, cyantraniliprole, and nicotine tolerance. In addition, functional analysis revealed that the MAPKs p38, PI3K/Akt, and JAK-STAT activate the transcription factor fushi tarazu factor 1 (FTZ-F1) to regulate CYP9A75b expression. These findings provide mechanistic insights into the contributions of CYP9A genes to xenobiotic detoxification and support the possible involvement of different signaling pathways and TFs in tolerance to xenobiotics in insects.


Sujet(s)
Cytochrome P-450 enzyme system , Protéines d'insecte , Protéines proto-oncogènes c-akt , Transduction du signal , Spodoptera , Xénobiotique , Animaux , Spodoptera/génétique , Spodoptera/effets des médicaments et des substances chimiques , Spodoptera/métabolisme , Protéines d'insecte/génétique , Protéines d'insecte/métabolisme , Xénobiotique/métabolisme , Xénobiotique/pharmacologie , Cytochrome P-450 enzyme system/génétique , Cytochrome P-450 enzyme system/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Insecticides/pharmacologie , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , p38 Mitogen-Activated Protein Kinases/génétique
4.
Cancer Lett ; 603: 217215, 2024 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-39218290

RÉSUMÉ

Enhanced expression of methyltransferase-like 3 (METTL3) promotes the m6A modification of specific mRNAs, contributing to breast tumorigenesis. While the mRNA substrates targeted by METTL3 are well characterized, the factors dictating the selection of these specific mRNA remain elusive. This study aimed to examine the regulatory role of the transcription factor STAT5B in METTL3-induced m6A modification. METTL3 specifically interacts with STAT5B in response to mitogenic stimulation by epidermal growth factor (EGF). Chromatin immunoprecipitation and CRISPR/Cas9 mutagenesis showed that STAT5B recruits METTL3 to gene promoters like CCND1, where METTL3 interacts with RPB1, dependent on CDK9-mediated RPB1 (Ser2) phosphorylation during transcription elongation. Inhibition and depletion of either STAT5B or CDK9 prevented the EGF-induced m6A modification of CCND1. The translation efficiency of CCND1 was increased following m6A modification, thereby increasing cell proliferation. STAT5B facilitated METTL3-induced tumor formation by increasing CCND1 expression in an orthotopic mouse model. In clinical context, a positive correlation was observed between p-STAT5B and METTL3 expression in high-grade breast tumors. This study elucidates a novel mechanism that underlies the specificity of m6A modification in breast cancer cells, thereby underscoring its potential therapeutic value.


Sujet(s)
Tumeurs du sein , Cycline D1 , Methyltransferases , ARN messager , Facteur de transcription STAT-5 , Humains , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Femelle , Methyltransferases/génétique , Methyltransferases/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Animaux , ARN messager/génétique , ARN messager/métabolisme , Cycline D1/génétique , Cycline D1/métabolisme , Souris , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire , Kinase-9 cycline-dépendante/métabolisme , Kinase-9 cycline-dépendante/génétique , Carcinogenèse/génétique , Lignée cellulaire tumorale , Adénosine/analogues et dérivés , Adénosine/métabolisme , Phosphorylation , Régions promotrices (génétique) , Facteur de croissance épidermique/métabolisme , Facteur de croissance épidermique/génétique
5.
Prostaglandins Other Lipid Mediat ; 174: 106880, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39121944

RÉSUMÉ

Previous studies have shown prostaglandin E2 (PGE2) produced a marked increase in calcitonin secretion in human C-cells derived from medullary thyroid carcinoma. However, it's unclear whether PGE2 can increase the growth of C cells. In this study, we use TT cells as a C cell model to investigate the effect of PGE2 on the growth of C cells. The results revealed that both PGE2 and arachidonic acid (AA) significantly increased the count of TT cells, whereas indomethacin and Dup697 reduced this count. Notably, an increase in the level of AA was associated with an increase in the number of proliferating TT cells, indicating a dose-response relationship. PGE2 and its receptor agonists (sulprostone and butaprost) enhanced the proliferation of TT cells. By contrast, 17-phenyl-trinor-PGE2 exerted no significant effect on TT cell proliferation, whereas L161982 suppressed it. The positive effect of AA on TT cell proliferation was inhibited by indomethacin, NS398, Dup697 (complete inhibition), and SC560. Both PGE2 and AA increased the level of p-STAT5a. The positive effect of AA on p-STAT5a was completely inhibited by Dup697 but not indomethacin, NS398, or SC560. Treatment with indomethacin or Dup697 alone reduced the level of STAT5a in TT cells. AA increased the level of STAT5a, but this effect was inhibited by indomethacin, NS398, and Dup697. Overall, this study confirms the effect of PGE2 on the proliferation of TT cells. This effect is likely mediated through EP2, EP3, and EP4 receptors and associated with an increase in p-STAT5a level within TT cells.


Sujet(s)
Acide arachidonique , Prolifération cellulaire , Survie cellulaire , Dinoprostone , Indométacine , Dinoprostone/pharmacologie , Dinoprostone/métabolisme , Dinoprostone/analogues et dérivés , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Indométacine/pharmacologie , Acide arachidonique/pharmacologie , Lignée cellulaire tumorale , Division cellulaire/effets des médicaments et des substances chimiques , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/métabolisme , Facteur de transcription STAT-5/métabolisme , Alprostadil/pharmacologie , Alprostadil/analogues et dérivés
7.
J Transl Med ; 22(1): 788, 2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-39183280

RÉSUMÉ

Vascular dementia (VaD) is a prevalent form of dementia resulting from chronic cerebral hypoperfusion (CCH). However, the pathogenic mechanisms of VaD and corresponding therapeutic strategies are not well understood. Sirtuin 6 (SIRT6) has been implicated in various biological processes, including cellular metabolism, DNA repair, redox homeostasis, and aging. Nevertheless, its functional relevance in VaD remains unexplored. In this study, we utilized a bilateral common carotid artery stenosis (BCAS) mouse model of VaD to investigate the role of SIRT6. We detected a significant decrease in neuronal SIRT6 protein expression following CCH. Intriguingly, neuron-specific ablation of Sirt6 in mice exacerbated neuronal damage and cognitive deficits after CCH. Conversely, treatment with MDL-800, an agonist of SIRT6, effectively mitigated neuronal loss and facilitated neurological recovery. Mechanistically, SIRT6 inhibited excessive mitochondrial fission by suppressing the CCH-induced STAT5-PGAM5-Drp1 signaling cascade. Additionally, the gene expression of monocyte SIRT6 in patients with asymptomatic carotid stenosis showed a correlation with cognitive outcomes, suggesting translational implications in human subjects. Our findings provide the first evidence that SIRT6 prevents cognitive impairment induced by CCH, and mechanistically, this protection is achieved through the remodeling of mitochondrial dynamics in a STAT5-PGAM5-Drp1-dependent manner.


Sujet(s)
Dysfonctionnement cognitif , Dynamines , Dynamique mitochondriale , Facteur de transcription STAT-5 , Sirtuines , Sujet âgé , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Encéphalopathie ischémique/complications , Encéphalopathie ischémique/anatomopathologie , Encéphalopathie ischémique/métabolisme , Sténose carotidienne/complications , Sténose carotidienne/métabolisme , Maladie chronique , Dysfonctionnement cognitif/anatomopathologie , Dynamines/métabolisme , Dynamines/génétique , Souris de lignée C57BL , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Sirtuines/métabolisme , Sirtuines/génétique , Facteur de transcription STAT-5/métabolisme
8.
Nat Commun ; 15(1): 7372, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39191751

RÉSUMÉ

Cytokine-mediated STAT5 protein activation is vital for lymphocyte development and function. In vitro tyrosine phosphorylation of a C-terminal tyrosine is critical for activation of STAT5A and STAT5B; however, the importance of STAT5 tyrosine phosphorylation in vivo has not been assessed. Here we generate Stat5a and Stat5b tyrosine-to-phenylalanine mutant knockin mice and find they have greatly reduced CD8+ T-cell numbers and profoundly diminished IL-2-induced proliferation of these cells, and this correlates with reduced induction of Myc, pRB, a range of cyclins and CDKs, and a partial G1→S phase-transition block. These mutant CD8+ T cells also exhibit decreased IL-2-mediated activation of pERK and pAKT, which we attribute in part to diminished expression of IL-2Rß and IL-2Rγ. Our findings thus demonstrate that tyrosine phosphorylation of both STAT5A and STAT5B is essential for maximal IL-2 signaling. Moreover, our transcriptomic and proteomic analyses elucidate the molecular basis of the IL-2-induced proliferation of CD8+ T cells.


Sujet(s)
Lymphocytes T CD8+ , Prolifération cellulaire , Interleukine-2 , Facteur de transcription STAT-5 , Transduction du signal , Tyrosine , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Animaux , Interleukine-2/métabolisme , Phosphorylation , Tyrosine/métabolisme , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/immunologie , Souris , Sous-unité bêta du récepteur à l'interleukine-2/métabolisme , Sous-unité bêta du récepteur à l'interleukine-2/génétique , Sous-unité gamma commune aux récepteurs des interleukines/génétique , Sous-unité gamma commune aux récepteurs des interleukines/métabolisme , Souris de lignée C57BL , Techniques de knock-in de gènes , Activation des lymphocytes
9.
J Immunol ; 213(7): 952-964, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39140896

RÉSUMÉ

IL-7 and IL-2 are evolutionarily related cytokines that play critical roles in the development and expansion of immune cells. Although both IL-7R and IL-2R activate similar signaling molecules, whether their signals have specific or overlapping functions during lymphocyte differentiation remains unclear. To address this question, we generated IL-7R α-chain (IL-7Rα)/IL-2R ß-chain (IL-24ß) (72R) knock-in mice expressing a chimeric receptor consisting of the extracellular domain of IL-7Rα and the intracellular domain of IL-2Rß under the control of the endogenous IL-7Rα promoter. Notably, this 72R receptor induced higher levels of STAT5 and Akt phosphorylation in T cells. In the periphery of 72R mice, the number of T cells, B cells, and type 2 innate lymphoid cells (ILC2s) was increased, whereas early T cell progenitors and double-negative 2 thymocytes were reduced in the thymus. In addition, cell proliferation and Notch signaling were impaired in the early thymocytes of 72R mice, leading to their differentiation into thymic B cells. Interestingly, ILC2s were increased in the thymus of 72R mice. Early T cell progenitors from 72R mice, but not from wild-type mice, differentiated into NK cells and ILC2-like cells when cocultured with a thymic stromal cell line. Thus, this study indicates that the chimeric 72R receptor transduces more robust signals than the authentic IL-7Rα, thereby inducing the alternative differentiation of T cell progenitors into other cell lineages. This suggests that cytokine receptors may provide instructive signals for cell fate decisions.


Sujet(s)
Lymphocytes B , Différenciation cellulaire , Sous-unité bêta du récepteur à l'interleukine-2 , Récepteurs à l'interleukine-7 , Transduction du signal , Animaux , Souris , Différenciation cellulaire/immunologie , Récepteurs à l'interleukine-7/génétique , Récepteurs à l'interleukine-7/immunologie , Récepteurs à l'interleukine-7/métabolisme , Sous-unité bêta du récepteur à l'interleukine-2/génétique , Sous-unité bêta du récepteur à l'interleukine-2/immunologie , Transduction du signal/immunologie , Lymphocytes B/immunologie , Immunité innée , Souris de lignée C57BL , Techniques de knock-in de gènes , Protéines de fusion recombinantes/génétique , Facteur de transcription STAT-5/métabolisme
10.
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
11.
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
12.
Insect Biochem Mol Biol ; 173: 104164, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39068995

RÉSUMÉ

Janus kinase (JAK) and signal transducer and activator of transcription (STAT) signaling affect social aggregation, mood and psychiatric disorders, nociceptive and depressive behaviors. Olfactory dysfunction is one of the distinct symptoms of these behaviors, but function and mechanism of JAK and STAT in modulating olfaction remain largely unknown. Migratory locusts show olfactory preference for their own volatiles. We thus use this animal model to explore functions and mechanisms of JAK and STAT5B in mediating olfaction response to their own volatiles. Tissue distribution study shows that JAK and STAT5B express in antennae and brains, especially in antennal lobes and mushroom bodies in locust brains, and knockdown of these two genes by RNA interference (RNAi) in antennae and brains results in the loss of olfactory preference for locust volatiles, including chemical odorants indole and ß-ionone. RNA-seq analysis reveals that JAK and STAT5B RNAi knockdown downregulates a functional class of transcripts in nucleoprotein complex, including heterogeneous nuclear ribonucleoprotein C (hnRNPC) and small nuclear ribonucleoprotein polypeptide F (SNRPF). HnRNPC and SNRPF mRNAs and proteins are also expressed in antennae and brains, and RNAi knockdown of these two genes reduces the percentage of locusts preferring volatiles, including chemical odorants indole and ß-ionone. Furthermore, RNAi knockdown of dopamine receptor 1 (DopR1) results in the decrease of JAK mRNA level in antennae, and JAK/STAT5B, hnRNPC and SNRPF are required for dopamine receptor 1 (DopR1) to modulate olfactory preference for their own volatiles. This study confirms that JAK/STAT5B signaling modulates olfaction by affecting expression levels of hnRNPC and SNRPF, and this pathway is also required for DopR1 to modulate olfactory preference for their own volatiles. These findings highlight novel roles of JAK and STAT5B in modulating olfactory preference. This study provides novel insights into functional links among JAK/STAT5B signaling, RNA binding proteins and DopR1 underlying the modulation of olfactory behaviors.


Sujet(s)
Protéines d'insecte , Janus kinases , Animaux , Janus kinases/métabolisme , Janus kinases/génétique , Protéines d'insecte/métabolisme , Protéines d'insecte/génétique , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Odorat , Antennes des arthropodes/métabolisme , Locusta Migratoria/métabolisme , Locusta Migratoria/génétique , Encéphale/métabolisme , Transduction du signal , Interférence par ARN
13.
Int Immunopharmacol ; 139: 112681, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39068758

RÉSUMÉ

Lipid droplet (LD) accumulation is one of the features in various tumors, whereas the significance of LD accumulation in pancreatic cancer progression remains unclear under chemotherapeutic condition. Since chemoresistance towards gemcitabine (GEM) is an obstacle for clinical therapy of pancreatic cancer, we sought to investigate the contribution of LD accumulation to GEM resistance. Herein, triacsin C (an inhibitor of LD production) dampened the proliferation, migration, and invasion of pancreatic cancer cells. The inhibition of LD accumulation induced by triacsin C or silencing of perilipin 2 (a marker of LD) sensitized cells to GEM treatment. Next, 75 paraffin-embedded samples and 5 pairs of frozen samples from pancreatic cancer patients were obtained for the detection of lysophosphatidylcholine acyltransferase 2 (LPCAT2; a LD-located enzyme contributing phosphatidylcholine synthesis) expression. The results revealed that LPCAT2 was upregulated in pancreatic cancer tissues, and its expression was correlated with clinical parameters and the basal LD content of cancer cell lines. Loss of LPCAT2 repressed the LD accumulation, GEM resistance, and cell motility. The enhancement of chemotherapy sensitivity was further confirmed in a xenograft model of mice in vivo. The carcinogenesis role of LPCAT2 was at least partly mediated by the LD accumulation. Then, signal transducer and activator of transcription 5B (STAT5B) activated the transcription of LPCAT2. Both LPCAT2 downregulation and triacsin C reversed the STAT5B-induced potentiation of malignant phenotypes in pancreatic cancer cells. In conclusion, LPCAT2-mediated lipid droplet production supported pancreatic cancer chemoresistance and cell motility, which was triggered by STAT5B.


Sujet(s)
1-Acylglycerophosphocholine acyltransferase , Mouvement cellulaire , Désoxycytidine , Résistance aux médicaments antinéoplasiques , , Gouttelettes lipidiques , Tumeurs du pancréas , Humains , Mouvement cellulaire/effets des médicaments et des substances chimiques , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Animaux , 1-Acylglycerophosphocholine acyltransferase/métabolisme , 1-Acylglycerophosphocholine acyltransferase/génétique , Lignée cellulaire tumorale , Gouttelettes lipidiques/métabolisme , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique , Souris , Mâle , Femelle , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Facteur de transcription STAT-5/métabolisme , Adulte d'âge moyen , Souris de lignée BALB C , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques
14.
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
15.
Front Immunol ; 15: 1401949, 2024.
Article de Anglais | MEDLINE | ID: mdl-39076972

RÉSUMÉ

Introduction: Ubiquitin-specific proteases (USPs), a large subset of more than 50 deubiquitinase proteins, have recently emerged as promising targets in cancer. However, their role in immune cell regulation, particularly in T cell activation, differentiation, and effector functions, remains largely unexplored. Methods: We utilized a USP28 knockout mouse line to study the effect of USP28 on T cell activation and function, and its role in intestinal inflammation using the dextran sulfate sodium (DSS)-induced colitis model and a series of in vitro assays. Results: Our results show that USP28 exerts protective effects in acute intestinal inflammation. Mechanistically, USP28 knockout mice (USP28-/-) exhibited an increase in total T cells mainly due to an increased CD8+ T cell content. Additionally, USP28 deficiency resulted in early defects in T cell activation and functional changes. Specifically, we observed a reduced expression of IL17 and an increase in inducible regulatory T (iTreg) suppressive functions. Importantly, activated T cells lacking USP28 showed increased STAT5 phosphorylation. Consistent with these findings, these mice exhibited increased susceptibility to acute DSS-induced intestinal inflammation, accompanied by elevated IL22 cytokine levels. Conclusions: Our findings demonstrate that USP28 is essential for T cell functionality and protects mice from acute DSS-induced colitis by regulating STAT5 signaling and IL22 production. As a T cell regulator, USP28 plays a crucial role in immune responses and intestinal health.


Sujet(s)
Colite , , Interleukines , Facteur de transcription STAT-5 , Ubiquitin thiolesterase , Animaux , Souris , Colite/induit chimiquement , Colite/immunologie , Colite/métabolisme , Sulfate dextran , Modèles animaux de maladie humaine , Inflammation/immunologie , Inflammation/métabolisme , Interleukines/métabolisme , Interleukines/génétique , Intestins/immunologie , Intestins/anatomopathologie , Activation des lymphocytes/immunologie , Souris de lignée C57BL , Souris knockout , Phosphorylation , Facteur de transcription STAT-5/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/déficit
16.
EMBO Mol Med ; 16(8): 1886-1900, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39009886

RÉSUMÉ

Despite the re-emergence of the pioneering "Coley's toxin" concept in anti-cancer immune therapies highlighted by check-point inhibitors and CAR-T approaches, fundamental mechanisms responsible for the immune-enhancing efficacy of low-dose "Coley's toxin" remain poorly understood. This study examines the novel reprogramming of immune-enhancing neutrophils by super-low dose endotoxin conducive for anti-cancer therapies. Through integrated analyses including scRNAseq and functional characterizations, we examined the efficacy of reprogrammed neutrophils in treating experimental cancer. We observed that neutrophils trained by super-low dose endotoxin adopt a potent immune-enhancing phenotype characterized by CD177loCD11bloCD80hiCD40hiDectin2hi. Both murine and human neutrophils trained by super-low dose endotoxin exhibit relieved suppression of adaptive T cells as compared to un-trained neutrophils. Functionally, neutrophils trained by super-low dose endotoxin can potently reduce tumor burden when transfused into recipient tumor-bearing mice. Mechanistically, Super-low dose endotoxin enables the generation of immune-enhancing neutrophils through activating STAT5 and reducing innate suppressor IRAK-M. Together, our data clarify the long-held mystery of "Coley's toxin" in rejuvenating anti-tumor immune defense, and provide a proof-of-concept in developing innate neutrophil-based anti-tumor therapeutics.


Sujet(s)
Endotoxines , Granulocytes neutrophiles , Granulocytes neutrophiles/immunologie , Animaux , Souris , Humains , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Souris de lignée C57BL , Facteur de transcription STAT-5/métabolisme , Lignée cellulaire tumorale
17.
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
18.
Int J Biol Macromol ; 276(Pt 1): 133834, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39002899

RÉSUMÉ

IL-2 regulates the immune response by interacting with different IL-2 receptor (IL-2R) subunits. High dose of IL-2 binds to IL-2Rßγc heterodimer, which induce various side effects while activating immune function. Disrupting IL-2 and IL-2R interactions can block IL-2 mediated immune response. Here, we used a computational approach to de novo design mini-binder proteins against IL-2R ß chain (IL-2Rß) to block IL-2 signaling. The hydrophobic region where IL-2 binds to IL-2Rß was selected and the promising binding mode was broadly explored. Three mini-binders with amino acid numbers ranging from 55 to 65 were obtained and binder 1 showed the best effects in inhibiting CTLL-2 cells proliferation and STAT5 phosphorylation. Molecular dynamics simulation showed that the binding of binder 1 to IL-2Rß was stable; the free energy of binder1/IL-2Rß complex was lower, indicating that the affinity of binder 1 to IL-2Rß was higher than that of IL-2. Free energy decomposition suggested that the ARG35 and ARG131 of IL-2Rß might be the key to improve the affinity of binder. Our efforts provided new insights in developing of IL-2R blocker, offering a potential strategy for ameliorating the side effects of IL-2 treatment.


Sujet(s)
Sous-unité bêta du récepteur à l'interleukine-2 , Interleukine-2 , Simulation de dynamique moléculaire , Liaison aux protéines , Sous-unité bêta du récepteur à l'interleukine-2/métabolisme , Sous-unité bêta du récepteur à l'interleukine-2/composition chimique , Interleukine-2/métabolisme , Interleukine-2/composition chimique , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Facteur de transcription STAT-5/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Animaux , Simulation de docking moléculaire
19.
Mol Biol Rep ; 51(1): 832, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39037638

RÉSUMÉ

BACKGROUND: JAK/STAT signaling plays an important role in regulating cell proliferation. Reducing proliferation and inducing cell death with gene-specific inhibitors such as ruxolitinib, Receptor tyrosine kinases (RTK) inhibitor targeting JAK1/2, are therapeutic approaches. The use of nanoparticles can reduce the toxicity and side effects of drugs, as they act directly on cancer cells and can selectively increase drug accumulation in tumor cells. Poly-ɛ-caprolactone (PCL) is a polymer that is frequently used in drug development. In this study, Rux-PCL-NPs were synthesized to increase the effectiveness of ruxolitinib. In addition, this study aimed to determine the effect of Rux-PCL-NPs on JAK/STAT signaling and apoptotic cell death. METHODS AND RESULTS: Rux-PCL-NPs were synthesized by nanoprecipitation. The Rux-PCL-NPs had a spherical and mean particle size of 219 ± 88.66 nm and a zeta potential of 0.471 ± 0.453 mV. In vitro cytotoxicity and antiproliferative effects were determined by MTT and soft agar colony formation assays, respectively. The effects of ruxolitinib, PCL-NPs, and Rux-PCL-NPs on apoptosis and the JAK/STAT pathway in cells were examined by western blot analysis. PCL-NPs did not have a toxic effect on the cells. The IC50 value of Rux-PCL-NPs was decreased 50-fold compared to that of ruxolitinib. Rux-PCL-NPs promoted cell death by downregulating JAK2 and STAT5, thereby inhibiting the JAK/STAT pathway. CONCLUSIONS: Our results revealed that Rux-PCL-NPs, which increased the efficacy of ruxolitinib, regulated apoptosis and the JAK2/STAT5 pathway.


Sujet(s)
Apoptose , Tumeurs du sein , Prolifération cellulaire , Kinase Janus-2 , Nanoparticules , Nitriles , Polyesters , Pyrazoles , Pyrimidines , Facteur de transcription STAT-5 , Transduction du signal , Nitriles/pharmacologie , Humains , Pyrazoles/pharmacologie , Pyrimidines/pharmacologie , Kinase Janus-2/métabolisme , Facteur de transcription STAT-5/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Polyesters/composition chimique , Nanoparticules/composition chimique , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Protéines proto-oncogènes c-bcl-2/métabolisme
20.
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 , Protéines membranaires , Cellules souches neurales , Neurogenèse , Facteur de transcription STAT-5 , Animaux , Souris , Prolifération cellulaire , Gyrus denté/métabolisme , Gyrus denté/cytologie , Hippocampe/métabolisme , Hippocampe/cytologie , Homéostasie , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris knockout , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Crises épileptiques/métabolisme , Crises épileptiques/génétique , Transduction du signal , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE