Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 38.894
Filtrer
1.
J Gene Med ; 26(7): e3709, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38949077

RÉSUMÉ

BACKGROUND: Colorectal cancer is the third most common malignancy worldwide and is one of the leading causes of cancer-related mortality. Ubiquitin-specific peptidase 18 (USP18) protein has been reported to exert different tumor-related effects in distinct tumor types. Here, we initially investigated the expression and signaling pathways of USP18 in colon adenocarcinoma (COAD). METHODS: A quantitative real-time PCR was conducted to evaluate the mRNA level of USP18 in cultured cells. Immunohistochemical staining was used to explore the protein expression of USP18 in clinical COAD samples. Specific knockdown was achieved by transient transfection of small interfering RNAs into SW480 and HT29 cells using Lipo3000. Cell conting kit-8 assay, transwell assay and matrigel-transwell assays were conducted to evaluate proliferation, migration and invasion capacities, respectively. Western blotting was performed to analyze downstream signaling pathways. A chi-squared test and univariate and multivariate analyses were used to evaluate the clinical data. Xenografts from mice model were assessed to validate the in vitro findings. RESULTS: Higher USP18 level was identified in COAD tissues and was positively correlated with advanced tumor stage. High USP18 protein expression indicated poorer prognosis of COAD patients. Silencing USP18 suppressed COAD cell proliferation and invasion via destabilizing extracellular signal-regulated kinase (ERK) protein and suppressing ERK downstream pathways. Simultaneously silencing interferon-stimulated gene 15 (ISG15) with USP18 can partially rescue the tumor cell viability, indicating its involvement in USP18 signaling. The oncogenic effects of USP18 were also confirmed in mice models. CONCLUSIONS: USP18 plays oncogenic effects in colon adenocarcinoma via ISG15-ERK pathways. High USP18 expression indicates poor clinical outcomes for colon adenocarcinoma patients.


Sujet(s)
Adénocarcinome , Mouvement cellulaire , Prolifération cellulaire , Tumeurs du côlon , Régulation de l'expression des gènes tumoraux , Transduction du signal , Ubiquitin thiolesterase , Humains , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Animaux , Souris , Adénocarcinome/génétique , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Tumeurs du côlon/génétique , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Mâle , Mouvement cellulaire/génétique , Femelle , Lignée cellulaire tumorale , Évolution de la maladie , Adulte d'âge moyen , Pronostic , Système de signalisation des MAP kinases , Extracellular Signal-Regulated MAP Kinases/métabolisme , Cellules HT29 , Souris nude
2.
Oncotarget ; 15: 424-438, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953895

RÉSUMÉ

Single-agent TAS102 (trifluridine/tipiracil) and regorafenib are FDA-approved treatments for metastatic colorectal cancer (mCRC). We previously reported that regorafenib combined with a fluoropyrimidine can delay disease progression in clinical case reports of multidrug-resistant mCRC patients. We hypothesized that the combination of TAS102 and regorafenib may be active in CRC and other gastrointestinal (GI) cancers and may in the future provide a treatment option for patients with advanced GI cancer. We investigated the therapeutic effect of TAS102 in combination with regorafenib in preclinical studies employing cell culture, colonosphere assays that enrich for cancer stem cells, and in vivo. TAS102 in combination with regorafenib has synergistic activity against multiple GI cancers in vitro including colorectal and gastric cancer, but not liver cancer cells. TAS102 inhibits colonosphere formation and this effect is potentiated by regorafenib. In vivo anti-tumor effects of TAS102 plus regorafenib appear to be due to anti-proliferative effects, necrosis and angiogenesis inhibition. Growth inhibition by TAS102 plus regorafenib occurs in xenografted tumors regardless of p53, KRAS or BRAF mutations, although more potent tumor suppression was observed with wild-type p53. Regorafenib significantly inhibits TAS102-induced angiogenesis and microvessel density in xenografted tumors, as well inhibits TAS102-induced ERK1/2 activation regardless of RAS or BRAF status in vivo. TAS102 plus regorafenib is a synergistic drug combination in preclinical models of GI cancer, with regorafenib suppressing TAS102-induced increase in microvessel density and p-ERK as contributing mechanisms. The TAS102 plus regorafenib drug combination may be further tested in gastric and other GI cancers.


Sujet(s)
Association médicamenteuse , Synergie des médicaments , Tumeurs gastro-intestinales , Mutation , Cellules souches tumorales , Néovascularisation pathologique , Phénylurées , Protéines proto-oncogènes B-raf , Protéines proto-oncogènes p21(ras) , Pyridines , Pyrrolidines , Facteur de transcription STAT-3 , Thymine , Trifluorothymidine , Uracile , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Trifluorothymidine/pharmacologie , Phénylurées/pharmacologie , Animaux , Pyridines/pharmacologie , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Tumeurs gastro-intestinales/traitement médicamenteux , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/anatomopathologie , Tumeurs gastro-intestinales/métabolisme , Uracile/pharmacologie , Uracile/analogues et dérivés , Souris , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Thymine/pharmacologie , Lignée cellulaire tumorale , Pyrrolidines/pharmacologie , Pyrrolidines/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques ,
3.
Proc Natl Acad Sci U S A ; 121(28): e2402407121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38959045

RÉSUMÉ

Trade-offs between evolutionary gain and loss are prevalent in nature, yet their genetic basis is not well resolved. The evolution of insect resistance to insecticide is often associated with strong fitness costs; however, how the fitness trade-offs operates remains poorly understood. Here, we show that the mitogen-activated protein kinase (MAPK) pathway and its upstream and downstream actors underlie the fitness trade-offs associated with insecticide resistance in the whitefly Bemisia tabaci. Specifically, we find a key cytochrome P450 gene CYP6CM1, that confers neonicotinoids resistance to in B. tabaci, is regulated by the MAPKs p38 and ERK through their activation of the transcription factor cAMP-response element binding protein. However, phosphorylation of p38 and ERK also leads to the activation of the transcription repressor Cap "n" collar isoform C (CncC) that negatively regulates exuperantia (Ex), vasa (Va), and benign gonial cell neoplasm (Bg), key genes involved in oogenesis, leading to abnormal ovary growth and a reduction in female fecundity. We further demonstrate that the transmembrane G protein-coupled receptor (GPCR) neuropeptide FF receptor 2 (NPFF2) triggers the p38 and ERK pathways via phosphorylation. Additionally, a positive feedback loop between p38 and NPFF2 leads to the continuous activation of the MAPK pathways, thereby constitutively promoting neonicotinoids resistance but with a significant reproductive cost. Collectively, these findings provide fundamental insights into the role of cis-trans regulatory networks incurred by GPCR-MAPK signaling pathways in evolutionary trade-offs and applied knowledge that can inform the development of strategies for the sustainable pest control.


Sujet(s)
Hemiptera , Protéines d'insecte , Résistance aux insecticides , Système de signalisation des MAP kinases , Récepteurs couplés aux protéines G , Animaux , Hemiptera/génétique , Hemiptera/métabolisme , Résistance aux insecticides/génétique , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Protéines d'insecte/métabolisme , Protéines d'insecte/génétique , Femelle , Insecticides/pharmacologie , Cytochrome P-450 enzyme system/métabolisme , Cytochrome P-450 enzyme system/génétique
4.
FASEB J ; 38(13): e23769, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38958951

RÉSUMÉ

Renal ischemia-reperfusion injury (IRI) is an integral process in renal transplantation, which results in compromised graft survival. Macrophages play an important role in both the early inflammatory period and late fibrotic period in response to IRI. In this study, we investigated whether scutellarin (SCU) could protect against renal IRI by regulating macrophage polarization. Mice were given SCU (5-50 mg/kg) by gavage 1 h earlier, followed by a unilateral renal IRI. Renal function and pathological injury were assessed 24 h after reperfusion. The results showed that administration of 50 mg/kg SCU significantly improved renal function and renal pathology in IRI mice. In addition, SCU alleviated IRI-induced apoptosis. Meanwhile, it reduced macrophage infiltration and inhibited pro-inflammatory macrophage polarization. Moreover, in RAW 264.7 cells and primary bone marrow-derived macrophages (BMDMs) exposed to SCU, we found that 150 µM SCU inhibited these cells to polarize to an inflammatory phenotype induced by lipopolysaccharide (LPS) and interferon-γ (IFN-γ). However, SCU has no influence on anti-inflammatory macrophage polarization in vivo and in vitro induced by in interleukin-4 (IL-4). Finally, we explored the effect of SCU on the activation of the mitogen-activated protein kinase (MAPK) pathway both in vivo and in vitro. We found that SCU suppressed the activation of the MAPK pathway, including the extracellular signal-regulated kinase (ERK), Jun N-terminal kinase (JNK), and p38. Our results demonstrated that SCU protects the kidney against IRI by inhibiting macrophage infiltration and polarization toward pro-inflammatory phenotype via the MAPK pathway, suggesting that SCU may be therapeutically important in treatment of IRI.


Sujet(s)
Apigénine , Glucuronates , Système de signalisation des MAP kinases , Macrophages , Souris de lignée C57BL , Lésion d'ischémie-reperfusion , Animaux , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Souris , Apigénine/pharmacologie , Glucuronates/pharmacologie , Glucuronates/usage thérapeutique , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Cellules RAW 264.7 , Mâle , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/prévention et contrôle , Inflammation/anatomopathologie
5.
Cancer Discov ; 14(7): 1143-1144, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38946324

RÉSUMÉ

In this issue, Ryan and colleagues describe the preclinical development of a pan-RAF:MEK molecular glue with superior efficacy, brain penetrance, and tolerability in xenograft models of Ras/Raf/MAPK pathway-driven tumors. See related article by Ryan et al., p. 1190 (1).


Sujet(s)
Inhibiteurs de protéines kinases , Humains , Animaux , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Kinases raf/métabolisme , Kinases raf/génétique , Souris , Système de signalisation des MAP kinases , Mitogen-Activated Protein Kinase Kinases/métabolisme , Tumeurs/génétique , Tumeurs/métabolisme
6.
Proc Natl Acad Sci U S A ; 121(28): e2404887121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968100

RÉSUMÉ

The timing of seed germination is controlled by the combination of internal dormancy and external factors. Temperature is a major environmental factor for seed germination. The permissive temperature range for germination is narrow in dormant seeds and expands during after-ripening (AR) (dormancy release). Quantitative trait loci analyses of preharvest sprouting in cereals have revealed that MKK3, a mitogen-activated protein kinase (MAPK) cascade protein, is a negative regulator of grain dormancy. Here, we show that the MAPKKK19/20-MKK3-MPK1/2/7/14 cascade modulates the germination temperature range in Arabidopsis seeds by elevating the germinability of the seeds at sub- and supraoptimal temperatures. The expression of MAPKKK19 and MAPKKK20 is induced around optimal temperature for germination in after-ripened seeds but repressed in dormant seeds. MPK7 activation depends on the expression levels of MAPKKK19/20, with expression occurring under conditions permissive for germination. Abscisic acid (ABA) and gibberellin (GA) are two major phytohormones which are involved in germination control. Activation of the MKK3 cascade represses ABA biosynthesis enzyme gene expression and induces expression of ABA catabolic enzyme and GA biosynthesis enzyme genes, resulting in expansion of the germinable temperature range. Our data demonstrate that the MKK3 cascade integrates temperature and AR signals to phytohormone metabolism and seed germination.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Régulation de l'expression des gènes végétaux , Germination , Graines , Température , Germination/physiologie , Germination/génétique , Arabidopsis/génétique , Arabidopsis/métabolisme , Arabidopsis/croissance et développement , Graines/croissance et développement , Graines/métabolisme , Graines/génétique , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/génétique , MAP Kinase Kinase 3/métabolisme , MAP Kinase Kinase 3/génétique , Système de signalisation des MAP kinases/physiologie , Dormance des plantes/génétique , Dormance des plantes/physiologie , Transduction du signal , Mitogen-Activated Protein Kinases/métabolisme , Mitogen-Activated Protein Kinases/génétique
7.
FASEB J ; 38(13): e23757, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38965999

RÉSUMÉ

Hepatic stellate cells (HSCs) are responsible for liver fibrosis accompanied by its activation into myofibroblasts and the abundant production of extracellular matrix. However, the HSC contribution to progression of liver inflammation has been less known. We aimed to elucidate the mechanism in HSCs underlying the inflammatory response and the function of tumor necrosis factor α-related protein A20 (TNFAIP3). We established A20 conditional knockout (KO) mice crossing Twist2-Cre and A20 floxed mice. Using these mice, the effect of A20 was analyzed in mouse liver and HSCs. The human HSC line LX-2 was also used to examine the role and underlying molecular mechanism of A20. In this KO model, A20 was deficient in >80% of HSCs. Spontaneous inflammation with mild fibrosis was found in the liver of the mouse model without any exogenous agents, suggesting that A20 in HSCs suppresses chronic hepatitis. Comprehensive RNA sequence analysis revealed that A20-deficient HSCs exhibited an inflammatory phenotype and abnormally expressed chemokines. A20 suppressed JNK pathway activation in HSCs. Loss of A20 function in LX-2 cells also induced excessive chemokine expression, mimicking A20-deficient HSCs. A20 overexpression suppressed chemokine expression in LX-2. In addition, we identified DCLK1 in the genes regulated by A20. DCLK1 activated the JNK pathway and upregulates chemokine expression. DCLK1 inhibition significantly decreased chemokine induction by A20-silencing, suggesting that A20 controlled chemokine expression in HSCs via the DCLK1-JNK pathway. In conclusion, A20 suppresses chemokine induction dependent on the DCLK1-JNK signaling pathway. These findings demonstrate the therapeutic potential of A20 and the DCLK1-JNK pathway for the regulation of inflammation in chronic hepatitis.


Sujet(s)
Chimiokines , Cellules étoilées du foie , Système de signalisation des MAP kinases , Souris knockout , Protein-Serine-Threonine Kinases , Protéine-3 induite par le facteur de nécrose tumorale alpha , Animaux , Cellules étoilées du foie/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/génétique , Souris , Humains , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Chimiokines/métabolisme , Chimiokines/génétique , Hépatite chronique/métabolisme , Hépatite chronique/anatomopathologie , Hépatite chronique/génétique , Kinases de type doublecortine , Souris de lignée C57BL , Lignée cellulaire , Mâle
8.
Int J Med Mushrooms ; 26(8): 41-57, 2024.
Article de Anglais | MEDLINE | ID: mdl-38967210

RÉSUMÉ

Bone metastasis in metastatic breast cancer commonly results in osteolytic lesions due to osteoclast activity, promoting bone destruction and tumor progression. The bioactive fungal isolates, 4-acetyl-antroquinonol B (4-AAQB) and erinacine A, have diverse pharmacological and biological activities. However, their effects on breast cancer bone metastasis treatment remain unclear. Our study aimed to examine the impact of 4-AAQB or erinacine A on breast cancer metastases in bone. The effects of 4-AAQB and erinacine A on breast cancer-induced osteoclastogenesis, breast cancer migration, production of prometastatic cytokine (TGF-ß) and marker (MMP-9), as well as potential MAPK signaling transductions were assessed. The results revealed that 4-AAQB and erinacine A effectively suppressed breast cancer-induced osteoclastogenesis and migration, and reduced TGF-ß and MMP-9 production via Erk or JNK signaling transductions, specifically in breast cancer cells or in breast cancer cells-induced osteoclasts. Based on these findings, either 4-AAQB or erinacine A showed promise in preventing breast cancer metastases in bone.


Sujet(s)
Tumeurs du sein , Matrix metalloproteinase 9 , Ostéoclastes , Ostéogenèse , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Humains , Femelle , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 9/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Animaux , Facteur de croissance transformant bêta/métabolisme , Tumeurs osseuses/secondaire , Tumeurs osseuses/traitement médicamenteux , Souris , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Cyclohexanones , 4-Butyrolactone/analogues et dérivés
9.
Curr Pharm Des ; 30(17): 1377, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38975683

RÉSUMÉ

A typographical error appeared in the title of the article "Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophora Decoction", published in Current Pharmaceutical Design, 2022; 28(42): 3456-3468 [1]. Details of the error and a correction are provided below. Original: Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophora Decoction Corrected: Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophorae Decoction We regret the error and apologize to readers. The original article can be found online at: https://www.eurekaselect.com/article/127740.


Sujet(s)
Colite , Sulfate dextran , Protéines du choc thermique HSP90 , Animaux , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/métabolisme , Souris , Colite/traitement médicamenteux , Colite/induit chimiquement , Sophora/composition chimique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
10.
Commun Biol ; 7(1): 807, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961177

RÉSUMÉ

Glaucoma is the leading cause of irreversible blindness and is characterized by progressive retinal ganglion cell (RGC) loss and retinal nerve fiber layer thinning. Currently, no existing treatment is effective for the preservation of RGCs. MicroRNA-22-3p (miR22) and small extracellular vesicles derived from mesenchymal stem cells (MSC-sEVs) have neuroprotective effects. In this study, we apply miR22-overexpressing MSC-sEVs in an N-methyl-D-aspartic acid (NMDA)-induced RGC injury model to assess their short-term therapeutic effects and explore the underlying mechanisms. We find that mice in the miR22-sEVs-treated group have thicker retinas, fewer apoptotic cells, more reserved RGCs, better retinal function, and lower expression levels of Bax and caspase-3. MiR22-sEVs treatment promotes viability, inhibits apoptosis and inhibits Bax and caspase-3 expression in RGC-5 cells. MiR22 targets mitogen-activated protein kinase kinase kinase 12 to inhibit apoptosis by regulating the mitogen-activated protein kinase (MAPK) signaling pathway. Collectively, our results suggest that miR22-sEVs ameliorate NMDA-induced RGC injury through the inhibition of MAPK signaling pathway-mediated apoptosis, providing a potential therapy for glaucoma and other diseases that involve RGC damage.


Sujet(s)
Vésicules extracellulaires , Système de signalisation des MAP kinases , Cellules souches mésenchymateuses , microARN , Cellules ganglionnaires rétiniennes , Cellules ganglionnaires rétiniennes/métabolisme , microARN/génétique , microARN/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Vésicules extracellulaires/métabolisme , Souris , Apoptose , Souris de lignée C57BL , Glaucome/génétique , Glaucome/métabolisme , Glaucome/anatomopathologie , Glaucome/thérapie , Mâle
11.
J Biochem Mol Toxicol ; 38(7): e23762, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967723

RÉSUMÉ

Given the malignancy of gastric cancer, developing highly effective and low-toxic targeted drugs is essential to prolong patient survival and improve patient outcomes. In this study, we conducted structural optimizations based on the benzimidazole scaffold. Notably, compound 8 f presented the most potent antiproliferative activity in MGC803 cells and induced cell cycle arrest at the G0/G1 phase. Further mechanistic studies demonstrated that compound 8 f caused the apoptosis of MGC803 cells by elevating intracellular reactive oxygen species (ROS) levels and activating the mitogen-activated protein kinase (MAPK) signaling pathway, accompanied by corresponding markers change. In vivo investigations additionally validated the inhibitory effect of compound 8 f on tumor growth in xenograft models bearing MGC803 cells without obvious toxicity. Our studies suggest that compound 8 f holds promise as a potential and safe lead compound for developing anti-gastric cancer agents.


Sujet(s)
Antinéoplasiques , Benzimidazoles , Système de signalisation des MAP kinases , Espèces réactives de l'oxygène , Tumeurs de l'estomac , Benzimidazoles/pharmacologie , Benzimidazoles/composition chimique , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Humains , Espèces réactives de l'oxygène/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Lignée cellulaire tumorale , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Animaux , Souris , Tests d'activité antitumorale sur modèle de xénogreffe , Apoptose/effets des médicaments et des substances chimiques , Souris nude
12.
Oncol Res ; 32(7): 1197-1207, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948022

RÉSUMÉ

Breast cancer, a predominant global health issue, requires ongoing exploration of new therapeutic strategies. Palbociclib (PAL), a well-known cyclin-dependent kinase (CDK) inhibitor, plays a critical role in breast cancer treatment. While its efficacy is recognized, the interplay between PAL and cellular autophagy, particularly in the context of the RAF/MEK/ERK signaling pathway, remains insufficiently explored. This study investigates PAL's inhibitory effects on breast cancer using both in vitro (MCF7 and MDA-MB-468 cells) and in vivo (tumor-bearing nude mice) models. Aimed at elucidating the impact of PAL on autophagic processes and exploring the potential of combining it with trametinib (TRA), an MEK inhibitor, our research seeks to address the challenge of PAL-induced drug resistance. Our findings reveal that PAL significantly decreases the viability of MCF7 and MDA-MB-468 cells and reduces tumor size in mice while showing minimal cytotoxicity in MCF10A cells. However, PAL also induces protective autophagy, potentially leading to drug resistance via the RAF/MEK/ERK pathway activation. Introducing TRA effectively neutralized this autophagy, enhancing PAL's anti-tumor efficacy. A combination of PAL and TRA synergistically reduced cell viability and proliferation, and in vivo studies showed notable tumor size reduction. In conclusion, the PAL and TRA combination emerges as a promising strategy for overcoming PAL-induced resistance, offering a new horizon in breast cancer treatment.


Sujet(s)
Autophagie , Tumeurs du sein , Pipérazines , Pyridines , Pyridones , Pyrimidinones , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Autophagie/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Femelle , Pyrimidinones/pharmacologie , Pyrimidinones/usage thérapeutique , Souris , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Souris nude , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules MCF-7
13.
Mar Drugs ; 22(6)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38921597

RÉSUMÉ

Cadmium (Cd) is a toxic heavy metal that causes nephrosis, including acute kidney injury. To prevent and treat acute kidney injury (AKI) following Cd exposure, a tripeptide, Ser-Arg-Pro (SRP), from Sipunculus nudus L. was employed, and its potential efficacy in AKI was assessed. Oral administration of SRP significantly alleviated Cd-induced kidney damage, leading to improved renal function and the attenuation of structural abnormalities. A network pharmacology analysis revealed the potential of SRP in renal protection by targeting various pathways, including mitogen-activated protein kinase (MAPK) signaling, inflammatory response, and apoptosis pathways. Mechanistic studies indicated that SRP achieves renal protection by inhibiting the activation of MAPK pathways (phosphorylation of p38, p56, ERK, and JNK) in the oxidative stress cascade, suppressing inflammatory responses (iNOS, Arg1, Cox2, TNF-α, IL-1ß, and IL-6), and restoring altered apoptosis factors (caspase-9, caspase-3, Bax, and Bcl-2). Hence, SRP has the potential to be used as a therapeutic agent for the treatment of Cd-induced nephrotoxicity.


Sujet(s)
Atteinte rénale aigüe , Apoptose , Cadmium , Oligopeptides , Stress oxydatif , Animaux , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/induit chimiquement , Apoptose/effets des médicaments et des substances chimiques , Souris , Cadmium/toxicité , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Oligopeptides/pharmacologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Inflammation/traitement médicamenteux , Modèles animaux de maladie humaine , Pharmacologie des réseaux
14.
Biomolecules ; 14(6)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38927035

RÉSUMÉ

Lysophosphatidic acid (LPA) is a well-documented pro-oncogenic factor in different cancers, but relatively little is known on its biological activity in neuroblastoma. The LPA effects and the participation of the tyrosine kinase receptor anaplastic lymphoma kinase (ALK) in LPA mitogenic signaling were studied in human neuroblastoma cell lines. We used light microscopy and [3H]-thymidine incorporation to determine cell proliferation, Western blot to study intracellular signaling, and pharmacological and molecular tools to examine the role of ALK. We found that LPA stimulated the growth of human neuroblastoma cells, as indicated by the enhanced cell number, clonogenic activity, and DNA synthesis. These effects were curtailed by the selective ALK inhibitors NPV-TAE684 and alectinib. In a panel of human neuroblastoma cell lines harboring different ALK genomic status, the ALK inhibitors suppressed LPA-induced phosphorylation of extracellular signal-regulated kinases 1/2 (ERK1/2), which are major regulators of cell proliferation. ALK depletion by siRNA treatment attenuated LPA-induced ERK1/2 activation. LPA enhanced ALK phosphorylation and potentiated ALK activation by the ALK ligand FAM150B. LPA enhanced the inhibitory phosphorylation of the tumor suppressor FoxO3a, and this response was impaired by the ALK inhibitors. These results indicate that LPA stimulates mitogenesis of human neuroblastoma cells through a crosstalk with ALK.


Sujet(s)
Kinase du lymphome anaplasique , Prolifération cellulaire , Lysophospholipides , Neuroblastome , Transduction du signal , Humains , Lysophospholipides/métabolisme , Lysophospholipides/pharmacologie , Kinase du lymphome anaplasique/métabolisme , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Transduction du signal/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Pipéridines/pharmacologie , Carbazoles/pharmacologie , Protéine O3 à motif en tête de fourche/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Mitogen-Activated Protein Kinase 3/métabolisme , Mitogen-Activated Protein Kinase 3/génétique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
15.
Biomolecules ; 14(6)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38927100

RÉSUMÉ

Glaesserella parasuis (G. parasuis) causes serious inflammation and meningitis in piglets. Quercetin has anti-inflammatory and anti-bacterial activities; however, whether quercetin can alleviate brain inflammation and provide protective effects during G. parasuis infection has not been studied. Here, we established a mouse model of G. parasuis infection in vivo and in vitro to investigate transcriptome changes in the mouse cerebrum and determine the protective effects of quercetin on brain inflammation and blood-brain barrier (BBB) integrity during G. parasuis infection. The results showed that G. parasuis induced brain inflammation, destroyed BBB integrity, and suppressed PI3K/Akt/Erk signaling-pathway activation in mice. Quercetin decreased the expression of inflammatory cytokines (Il-18, Il-6, Il-8, and Tnf-α) and BBB-permeability marker genes (Mmp9, Vegf, Ang-2, and Et-1), increased the expression of angiogenetic genes (Sema4D and PlexinB1), reduced G. parasuis-induced tight junction disruption, and reactivated G. parasuis-induced suppression of the PI3K/Akt/Erk signaling pathway in vitro. Thus, we concluded that quercetin may protect BBB integrity via the PI3K/Akt/Erk signaling pathway during G. parasuis infection. This was the first attempt to explore the protective effects of quercetin on brain inflammation and BBB integrity in a G. parasuis-infected mouse model. Our findings indicated that quercetin is a promising natural agent for the prevention and treatment of G. parasuis infection.


Sujet(s)
Barrière hémato-encéphalique , Modèles animaux de maladie humaine , Système de signalisation des MAP kinases , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Quercétine , Animaux , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Quercétine/pharmacologie , Souris , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Méningite/microbiologie , Méningite/traitement médicamenteux , Méningite/métabolisme , Infections à Haemophilus/traitement médicamenteux , Infections à Haemophilus/microbiologie , Transduction du signal/effets des médicaments et des substances chimiques , Haemophilus parasuis/effets des médicaments et des substances chimiques , Haemophilus parasuis/pathogénicité , Cytokines/métabolisme , Suidae
16.
Int J Mol Sci ; 25(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38928105

RÉSUMÉ

Alcohol use disorder (AUD) is a chronic neurobehavioral condition characterized by a cycle of tolerance development, increased consumption, and reinstated craving and seeking behaviors during withdrawal. Understanding the intricate mechanisms of AUD necessitates reliable animal models reflecting its key features. Caenorhabditis elegans (C. elegans), with its conserved nervous system and genetic tractability, has emerged as a valuable model organism to study AUD. Here, we employ an ethanol vapor exposure model in Caenorhabditis elegans, recapitulating AUD features while maintaining high-throughput scalability. We demonstrate that ethanol vapor exposure induces intoxication-like behaviors, acute tolerance, and ethanol preference, akin to mammalian AUD traits. Leveraging this model, we elucidate the conserved role of c-jun N-terminal kinase (JNK) signaling in mediating acute ethanol tolerance. Mutants lacking JNK signaling components exhibit impaired tolerance development, highlighting JNK's positive regulation. Furthermore, we detect ethanol-induced JNK activation in C. elegans. Our findings underscore the utility of C. elegans with ethanol vapor exposure for studying AUD and offer novel insights into the molecular mechanisms underlying acute ethanol tolerance through JNK signaling.


Sujet(s)
Protéines de Caenorhabditis elegans , Caenorhabditis elegans , Tolérance aux médicaments , Éthanol , Système de signalisation des MAP kinases , Animaux , Caenorhabditis elegans/génétique , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , JNK Mitogen-Activated Protein Kinases/métabolisme , Alcoolisme/métabolisme , Alcoolisme/génétique , Modèles animaux de maladie humaine
17.
Mol Biol Rep ; 51(1): 774, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38904794

RÉSUMÉ

BACKGROUND: Olive is an evergreen tree of Oleaceae Olea with numerous bioactive components. While the anti-inflammatory properties of olive oil and the derivatives are well-documented, there remains a dearth of in-depth researches on the immunosuppressive effects of olive fruit water extract. This study aimed to elucidate the dose-effect relationship and underlying molecular mechanisms of olive fruit extract in mediating anti-inflammatory responses. METHODS AND RESULTS: The impacts of olive fruit extract on the release of nitric oxide (NO), tumor necrosis factor (TNF-α), interleukins-6 (IL-6) and reactive oxygen species (ROS) were assessed in RAW264.7 cells induced by lipopolysaccharide (LPS). For deeper understanding, the expression of genes encoding inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), TNF-α and IL-6 was quantitatively tested. Additionally, the expression patterns of MAPK and NF-κB pathways were further observed to analyze the action mechanisms. Results suggested that olive fruit extract (200, 500, 1000 µg/mL) markedly exhibited a dose-dependent reduction in the generation of NO, TNF-α, IL-6 and ROS, as well as the expression of correlative genes studied. The activation of ERK, JNK, p38, IκB-α and p65 were all suppressed when p65 nuclear translocation was further restricted by olive fruit extract in NF-κB and MAPK signal pathways. CONCLUSIONS: Olive fruit extract targeted imposing restrictions on the signal transduction of key proteins in NF-κB and MAPK pathways, and thereby lowered the level of inflammatory mediators, which put an enormous hindrance to inflammatory development. Accordingly, it is reasonable to consider olive fruit as a potent ingredient in immunomodulatory products.


Sujet(s)
Anti-inflammatoires , Fruit , Lipopolysaccharides , Facteur de transcription NF-kappa B , Monoxyde d'azote , Olea , Extraits de plantes , Espèces réactives de l'oxygène , Transduction du signal , Animaux , Olea/composition chimique , Souris , Cellules RAW 264.7 , Extraits de plantes/pharmacologie , Anti-inflammatoires/pharmacologie , Lipopolysaccharides/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Fruit/composition chimique , Espèces réactives de l'oxygène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Monoxyde d'azote/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Interleukine-6/métabolisme , Interleukine-6/génétique , Nitric oxide synthase type II/métabolisme , Nitric oxide synthase type II/génétique , Cyclooxygenase 2/métabolisme , Cyclooxygenase 2/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinases/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme
18.
J Cell Biol ; 223(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-38916917

RÉSUMÉ

Context-dependent physiological remodeling of the extracellular matrix (ECM) is essential for development and organ homeostasis. On the other hand, consumption of high-caloric diet leverages ECM remodeling to create pathological conditions that impede the functionality of different organs, including the heart. However, the mechanistic basis of high caloric diet-induced ECM remodeling has yet to be elucidated. Employing in vivo molecular genetic analyses in Drosophila, we demonstrate that high dietary sugar triggers ROS-independent activation of JNK signaling to promote fatty acid oxidation (FAO) in the pericardial cells (nephrocytes). An elevated level of FAO, in turn, induces histone acetylation-dependent transcriptional upregulation of the cytokine Unpaired 3 (Upd3). Release of pericardial Upd3 augments fat body-specific expression of the cardiac ECM protein Pericardin, leading to progressive cardiac fibrosis. Importantly, this pathway is quite distinct from the ROS-Ask1-JNK/p38 axis that regulates Upd3 expression under normal physiological conditions. Our results unravel an unknown physiological role of FAO in cytokine-dependent ECM remodeling, bearing implications in diabetic fibrosis.


Sujet(s)
Protéines de Drosophila , Matrice extracellulaire , Acides gras , Oxydoréduction , Animaux , Matrice extracellulaire/métabolisme , Acides gras/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Myocarde/métabolisme , Myocarde/anatomopathologie , Cytokines/métabolisme , Cytokines/génétique , Drosophila melanogaster/métabolisme , Système de signalisation des MAP kinases , Espèces réactives de l'oxygène/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Fibrose/métabolisme , Péricarde/métabolisme , Péricarde/anatomopathologie
19.
Biochim Biophys Acta Gen Subj ; 1868(8): 130651, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38825256

RÉSUMÉ

Cannabidiol (CBD) has antioxidant and anti-inflammatory activities. However, the anti-tumor effect of CBD on hepatocellular carcinoma (HCC) remains unclear. Here, we investigated whether CBD displays anti-tumorigenic effects in HCC cells and whether it could reduce tumorigenesis and metastases in vivo. First, this study treated HCC cells with different concentrations of CBD, followed by analyzing the changes in the proliferative, apoptotic, migratory and invasive abilities. The effects of CBD on the growth and metastasis of HCC cells in vivo were verified by tumorigenesis and metastasis assays. Subsequently, the target genes of CBD were predicted through the SwissTarget website and the genes differentially expressed in cells after CBD treatment were analyzed by microarray for intersection. The enrichment of the pathways after CBD treatment was analyzed by KEGG enrichment analysis, followed by western blot validation. Finally, rescue assays were used to validate the functions of genes as well as pathways in the growth and metastasis of HCC cells. A significant weakening of the ability of HCC cells to grow and metastasize in vitro and in vivo was observed upon CBD treatment. Mechanistically, CBD reduced GRP55 expression in HCC cells, along with increased TP53 expression and blocked MAPK signaling activation. In CBD-treated cells, the anti-tumor of HCC cells was restored after overexpression of GRP55 or deletion of TP53. CBD inhibits the MAPK signaling activation and increases the TP53 expression by downregulating GRP55 in HCC cells, thereby suppressing the growth and metastasis of HCC cells.


Sujet(s)
Cannabidiol , Carcinome hépatocellulaire , Tumeurs du foie , Récepteurs de cannabinoïdes , Protéine p53 suppresseur de tumeur , Cannabidiol/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Récepteurs de cannabinoïdes/métabolisme , Récepteurs de cannabinoïdes/génétique , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Phénotype , Souris nude
20.
Drug Des Devel Ther ; 18: 2301-2315, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911032

RÉSUMÉ

Background: Nepetoidin B (NB) has been reported to possess anti-inflammatory, antibacterial, and antioxidant properties. However, its effects on liver ischemia/reperfusion (I/R) injury remain unclear. Methods: In this study, a mouse liver I/R injury model and a mouse AML12 cell hypoxia reoxygenation (H/R) injury model were used to investigate the potential role of NB. Serum transaminase levels, liver necrotic area, cell viability, oxidative stress, inflammatory response, and apoptosis were evaluated to assess the effects of NB on liver I/R and cell H/R injury. Quantitative polymerase chain reaction (qPCR) and Western blotting were used to measure mRNA and protein expression levels, respectively. Molecular docking was used to predict the binding capacity of NB and mitogen-activated protein kinase phosphatase 5 (MKP5). Results: The results showed that NB significantly reduced serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, liver necrosis, oxidative stress, reactive oxygen species (ROS) content, inflammatory cytokine content and expression, inflammatory cell infiltration, and apoptosis after liver I/R and AML12 cells H/R injury. Additionally, NB inhibited the JUN protein amino-terminal kinase (JNK)/P38 pathway. Molecular docking results showed good binding between NB and MKP5 proteins, and Western blotting results showed that NB increased the protein expression of MKP5. MKP5 knockout (KO) significantly diminished the protective effects of NB against liver injury and its inhibitory effects on the JNK/P38 pathway. Conclusion: NB exerts hepatoprotective effects against liver I/R injury by regulating the MKP5-mediated P38/JNK signaling pathway.


Sujet(s)
Souris de lignée C57BL , Lésion d'ischémie-reperfusion , p38 Mitogen-Activated Protein Kinases , Animaux , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Souris , Mâle , p38 Mitogen-Activated Protein Kinases/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Simulation de docking moléculaire , Dual-specificity phosphatases/métabolisme , Dual-specificity phosphatases/antagonistes et inhibiteurs , Relation dose-effet des médicaments , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Relation structure-activité , Modèles animaux de maladie humaine , Structure moléculaire , Stress oxydatif/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...