Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 19.918
Filtrer
1.
Pathol Oncol Res ; 30: 1611747, 2024.
Article de Anglais | MEDLINE | ID: mdl-39035053

RÉSUMÉ

Signaling pathways of Retinoblastoma (Rb) protein, Akt-kinase, and Erk-kinase (extracellular signal-regulated kinase) have an important role in the pathogenesis of acute myeloid leukemia. Constitutive activation of these proteins by phosphorylation contributes to cell survival by regulation of cell cycle, proliferation and proapoptotic signaling processes. According to previous data phosphorylated forms of these proteins represent a worse outcome for cancer patients. We investigated the presence of phosphorylated Rb (P-Rb), Akt (P-Akt) and Erk (P-Erk) proteins by Western blot technique using phospho-specific antibodies in bone marrow or peripheral blood samples of 69 AML patients, 36 patients with myelodysplastic syndrome (MDS) and 10 healthy volunteers. Expression level of PTEN (Phosphatase and tensin homolog) and PHLPP (PH domain and leucine-rich repeat Protein Phosphatase) phosphatases, the negative regulators of Akt kinase pathway were also examined. We tested the effect of these proteins on survival and on the correlation with known prognostic features in AML. We found 46.3% of AML patients had detectable P-Rb, 34.7% had P-Akt and 28.9% had P-Erk protein. 66.1% of patients expressing PTEN, 38.9% PHLPP, 37.2% both PTEN and PHLPP and 32.2% neither PTEN nor PHLPP phosphatases. Compared to nucleophosmin mutation (NPMc) negative samples P-Erk was significantly less in nucleophosmin mutated patients, P-Rb was significantly less in patients' group with more than 30 G/L peripheral leukocyte count by diagnosis. PHLPP was significantly present in FAB type M5. The expression of P-Rb represented significant better overall survival (OS), while P-Akt represented significantly worse event-free survival (EFS) in unfavorable cytogenetics patients. The presence of both PHLPP and PTEN phosphatases contributes to better OS and EFS, although the differences were not statistically significant. We confirmed significant positive correlation between P-Akt and PHLPP. Assessing the phosphorylation of Rb, Akt and Erk may define a subgroup of AML patients who would benefit especially from new targeted treatment options complemented the standard chemotherapy, and it may contribute to monitoring remission, relapse or progression of AML.


Sujet(s)
Leucémie aigüe myéloïde , Nucléophosmine , Phosphohydrolase PTEN , Protéines proto-oncogènes c-akt , Transduction du signal , Humains , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Pronostic , Femelle , Mâle , Phosphorylation , Adulte d'âge moyen , Sujet âgé , Adulte , Phosphohydrolase PTEN/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Sujet âgé de 80 ans ou plus , Phosphoprotein Phosphatases/métabolisme , Phosphoprotein Phosphatases/génétique , Jeune adulte , Taux de survie , Syndromes myélodysplasiques/métabolisme , Syndromes myélodysplasiques/anatomopathologie , Adolescent , Extracellular Signal-Regulated MAP Kinases/métabolisme , Protéines nucléaires
2.
Exp Dermatol ; 33(7): e15128, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38973249

RÉSUMÉ

Dry skin is common to many pruritic diseases and is difficult to improve with oral traditional antihistamines. Recently, increasing evidence indicated that histamine H4 receptor (H4R) plays an important role in the occurrence and development of pruritus. Extracellular signal-regulated kinase (ERK) phosphorylation activation in the spinal cord mediates histamine-induced acute and choric itch. However, whether the histamine H4 receptor regulates ERK activation in the dry skin itch remains unclear. In the study, we explore the role of the histamine H4 receptor and p-ERK in the spinal cord in a dry skin mouse model induced by acetone-ether-water (AEW). q-PCR, Western blot, pharmacology and immunofluorescence  were applied in the study. We established a dry skin itch model by repeated application of AEW on the nape of neck in mice. The AEW mice showed typically dry skin histological change and persistent spontaneous scratching behaviour. Histamine H4 receptor, instead of histamine H1 receptor, mediated spontaneous scratching behaviour in AEW mice. Moreover, c-Fos and p-ERK expression in the spinal cord neurons were increased and co-labelled with GRPR-positive neurons in AEW mice. Furthermore, H4R agonist 4-methyhistamine dihydrochloride (4-MH)induced itch. Both 4-MH-induced itch and the spontaneous itch in AEW mice were blocked by p-ERK inhibitor U0126. Finally, intrathecal H4R receptor antagonist JNJ7777120 inhibited spinal p-ERK expression in AEW mice. Our results indicated that spinal H4R mediates itch via ERK activation in the AEW-induced dry skin mice.


Sujet(s)
Acétone , Extracellular Signal-Regulated MAP Kinases , Prurit , Récepteur histaminergique H4 , Moelle spinale , Animaux , Prurit/induit chimiquement , Prurit/métabolisme , Récepteur histaminergique H4/métabolisme , Souris , Moelle spinale/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Mâle , Acétone/pharmacologie , Eau , Oxyde de diéthyle , Modèles animaux de maladie humaine , Phosphorylation , Indoles/pharmacologie , Butadiènes/pharmacologie , Pipérazines/pharmacologie , Nitriles/pharmacologie , Peau/métabolisme , Maladie chronique , Méthylhistamines , Protéines proto-oncogènes c-fos/métabolisme , Souris de lignée C57BL
3.
Cell Commun Signal ; 22(1): 356, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982464

RÉSUMÉ

BACKGROUND: Stem cell-derived extracellular vesicles (EVs) are an emerging class of therapeutics with excellent biocompatibility, bioactivity and pro-regenerative capacity. One of the potential targets for EV-based medicines are cardiovascular diseases (CVD). In this work we used EVs derived from human induced pluripotent stem cells (hiPSCs; hiPS-EVs) cultured under different oxygen concentrations (21, 5 and 3% O2) to dissect the molecular mechanisms responsible for cardioprotection. METHODS: EVs were isolated by ultrafiltration combined with size exclusion chromatography (UF + SEC), followed by characterization by nanoparticle tracking analysis, atomic force microscopy (AFM) and Western blot methods. Liquid chromatography and tandem mass spectrometry coupled with bioinformatic analyses were used to identify differentially enriched proteins in various oxygen conditions. We directly compared the cardioprotective effects of these EVs in an oxygen-glucose deprivation/reoxygenation (OGD/R) model of cardiomyocyte (CM) injury. Using advanced molecular biology, fluorescence microscopy, atomic force spectroscopy and bioinformatics techniques, we investigated intracellular signaling pathways involved in the regulation of cell survival, apoptosis and antioxidant response. The direct effect of EVs on NRF2-regulated signaling was evaluated in CMs following NRF2 inhibition with ML385. RESULTS: We demonstrate that hiPS-EVs derived from physiological hypoxia at 5% O2 (EV-H5) exert enhanced cytoprotective function towards damaged CMs compared to EVs derived from other tested oxygen conditions (normoxia; EV-N and hypoxia 3% O2; EV-H3). This resulted from higher phosphorylation rates of Akt kinase in the recipient cells after transfer, modulation of AMPK activity and reduced apoptosis. Furthermore, we provide direct evidence for improved calcium signaling and sustained contractility in CMs treated with EV-H5 using AFM measurements. Mechanistically, our mass spectrometry and bioinformatics analyses revealed differentially enriched proteins in EV-H5 associated with the antioxidant pathway regulated by NRF2. In this regard, EV-H5 increased the nuclear translocation of NRF2 protein and enhanced its transcription in CMs upon OGD/R. In contrast, inhibition of NRF2 with ML385 abolished the protective effect of EVs on CMs. CONCLUSIONS: In this work, we demonstrate a superior cardioprotective function of EV-H5 compared to EV-N and EV-H3. Such EVs were most effective in restoring redox balance in stressed CMs, preserving their contractile function and preventing cell death. Our data support the potential use of hiPS-EVs derived from physiological hypoxia, as cell-free therapeutics with regenerative properties for the treatment of cardiac diseases.


Sujet(s)
Antioxydants , Vésicules extracellulaires , Cellules souches pluripotentes induites , Myocytes cardiaques , Facteur-2 apparenté à NF-E2 , Protéines proto-oncogènes c-akt , Transduction du signal , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Vésicules extracellulaires/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Humains , Protéines proto-oncogènes c-akt/métabolisme , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Transduction du signal/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Animaux
4.
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
5.
Pharmazie ; 79(3): 67-71, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38872272

RÉSUMÉ

We examined the mechanism by which 24(R)-ethyllophenol (MAB28) isolated from the branches of Morus alba caused neurite outgrowth in rat pheochromocytoma cells (PC12). MAB28 significantly promoted neurite outgrowth to a similar degree as the positive control, nerve growth factor (NGF). After incubation with MAB28 in PC12 cells, phosphorylation of extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, and cyclic AMP response element-binding protein was detected, but the time course of phosphorylation was different from that induced by NGF. The expression of chloride intracellular channel protein 3 (CLIC3) was significantly decreased by MAB28. 5-Nitro-2-(3-phenylpropylamino)-benzoic acid (NPPB), an outward rectifying chloride channel inhibitor, significantly promoted neurite outgrowth in PC12 cells. These data suggested that MAB28 could induce neurite outgrowth by downregulating CLIC3 expression.


Sujet(s)
Morus , Neurites , Animaux , Cellules PC12 , Rats , Morus/composition chimique , Neurites/effets des médicaments et des substances chimiques , Excroissance neuronale/effets des médicaments et des substances chimiques , Facteur de croissance nerveuse/pharmacologie , Phosphorylation , p38 Mitogen-Activated Protein Kinases/métabolisme , Nitro-benzoates/pharmacologie , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Phénols/pharmacologie , Technique de Western , Extracellular Signal-Regulated MAP Kinases/métabolisme , Canaux chlorure
6.
FASEB J ; 38(11): e23731, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38855909

RÉSUMÉ

Ca2+ permeation through TRPV4 in fibroblasts is associated with pathological matrix degradation. In human gingival fibroblasts, IL-1ß binding to its signaling receptor (IL-1R1) induces activation of extracellular regulated kinase (ERK) and MMP1 expression, processes that require Ca2+ flux across the plasma membrane. It is not known how IL-1R1, which does not conduct Ca2+, generates Ca2+ signals in response to IL-1. We examined whether TRPV4 mediates the Ca2+ fluxes required for ERK signaling in IL-1 stimulated gingival fibroblasts. TRPV4 was immunostained in fibroblasts of human gingival connective tissue and in focal adhesions of cultured mouse gingival fibroblasts. Human gingival fibroblasts treated with IL-1ß showed no change of TRPV4 expression but there was increased MMP1 expression. In mouse, gingival fibroblasts expressing TRPV4, IL-1 strongly increased [Ca2+]i. Pre-incubation of cells with IL-1 Receptor Antagonist blocked Ca2+ entry induced by IL-1 or the TRPV4 agonist GSK101. Knockout of TRPV4 or expression of a non-Ca2+-conducting TRPV4 pore-mutant or pre-incubation with the TRPV4 inhibitor RN1734, blocked IL-1-induced Ca2+ transients and expression of the mouse interstitial collagenase, MMP13. Treatment of mouse gingival fibroblasts with GSK101 phenocopied Ca2+ and ERK responses induced by IL-1; these responses were absent in TRPV4-null cells or cells expressing a non-conducting TRPV4 pore-mutant. Immunostained IL-1R1 localized with TRPV4 in adhesions within cell extensions. While TRPV4 immunoprecipitates analyzed by mass spectrometry showed no association with IL-1R1, TRPV4 associated with Src-related proteins and Src co-immunoprecipitated with TRPV4. Src inhibition reduced IL-1-induced Ca2+ responses. The functional linkage of TRPV4 with IL-1R1 expands its repertoire of innate immune signaling processes by mediating IL-1-driven Ca2+ responses that drive matrix remodeling in fibroblasts. Thus, inhibiting TRPV4 activity may provide a new pharmacological approach for blunting matrix degradation in inflammatory diseases.


Sujet(s)
Signalisation calcique , Fibroblastes , Gencive , Canaux cationiques TRPV , Canaux cationiques TRPV/métabolisme , Canaux cationiques TRPV/génétique , Animaux , Humains , Souris , Fibroblastes/métabolisme , Gencive/métabolisme , Gencive/cytologie , Calcium/métabolisme , Système de signalisation des MAP kinases , Cellules cultivées , Extracellular Signal-Regulated MAP Kinases/métabolisme , Interleukine-1/métabolisme , Interleukine-1/pharmacologie , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 1/génétique , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie
7.
NPJ Syst Biol Appl ; 10(1): 65, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38834572

RÉSUMÉ

Understanding the dynamics of intracellular signaling pathways, such as ERK1/2 (ERK) and Akt1/2 (Akt), in the context of cell fate decisions is important for advancing our knowledge of cellular processes and diseases, particularly cancer. While previous studies have established associations between ERK and Akt activities and proliferative cell fate, the heterogeneity of single-cell responses adds complexity to this understanding. This study employed a data-driven approach to address this challenge, developing machine learning models trained on a dataset of growth factor-induced ERK and Akt activity time courses in single cells, to predict cell division events. The most predictive models were developed by applying discrete wavelet transforms (DWTs) to extract low-frequency features from the time courses, followed by using Ensemble Integration, a data integration and predictive modeling framework. The results demonstrated that these models effectively predicted cell division events in MCF10A cells (F-measure=0.524, AUC=0.726). ERK dynamics were found to be more predictive than Akt, but the combination of both measurements further enhanced predictive performance. The ERK model`s performance also generalized to predicting division events in RPE cells, indicating the potential applicability of these models and our data-driven methodology for predicting cell division across different biological contexts. Interpretation of these models suggested that ERK dynamics throughout the cell cycle, rather than immediately after growth factor stimulation, were associated with the likelihood of cell division. Overall, this work contributes insights into the predictive power of intra-cellular signaling dynamics for cell fate decisions, and highlights the potential of machine learning approaches in unraveling complex cellular behaviors.


Sujet(s)
Division cellulaire , Protéines proto-oncogènes c-akt , Protéines proto-oncogènes c-akt/métabolisme , Humains , Division cellulaire/physiologie , Apprentissage machine , Transduction du signal/physiologie , Modèles biologiques , Processus stochastiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Système de signalisation des MAP kinases/physiologie , Prolifération cellulaire/physiologie
8.
Nutrients ; 16(12)2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38931171

RÉSUMÉ

Taurine, a non-proteogenic amino acid and commonly used nutritional supplement, can protect various tissues from degeneration associated with the action of the DNA-damaging chemotherapeutic agent cisplatin. Whether and how taurine protects human ovarian cancer (OC) cells from DNA damage caused by cisplatin is not well understood. We found that OC ascites-derived cells contained significantly more intracellular taurine than cell culture-modeled OC. In culture, elevation of intracellular taurine concentration to OC ascites-cell-associated levels suppressed proliferation of various OC cell lines and patient-derived organoids, reduced glycolysis, and induced cell protection from cisplatin. Taurine cell protection was associated with decreased DNA damage in response to cisplatin. A combination of RNA sequencing, reverse-phase protein arrays, live-cell microscopy, flow cytometry, and biochemical validation experiments provided evidence for taurine-mediated induction of mutant or wild-type p53 binding to DNA, activation of p53 effectors involved in negative regulation of the cell cycle (p21), and glycolysis (TIGAR). Paradoxically, taurine's suppression of cell proliferation was associated with activation of pro-mitogenic signal transduction including ERK, mTOR, and increased mRNA expression of major DNA damage-sensing molecules such as DNAPK, ATM and ATR. While inhibition of ERK or p53 did not interfere with taurine's ability to protect cells from cisplatin, suppression of mTOR with Torin2, a clinically relevant inhibitor that also targets DNAPK and ATM/ATR, broke taurine's cell protection. Our studies implicate that elevation of intracellular taurine could suppress cell growth and metabolism, and activate cell protective mechanisms involving mTOR and DNA damage-sensing signal transducti.


Sujet(s)
Cisplatine , Altération de l'ADN , Tumeurs de l'ovaire , Sérine-thréonine kinases TOR , Taurine , Protéine p53 suppresseur de tumeur , Taurine/pharmacologie , Humains , Sérine-thréonine kinases TOR/métabolisme , Femelle , Tumeurs de l'ovaire/métabolisme , Altération de l'ADN/effets des médicaments et des substances chimiques , Cisplatine/pharmacologie , Protéine p53 suppresseur de tumeur/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Glycolyse/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Antinéoplasiques/pharmacologie
9.
J Mol Cell Cardiol ; 193: 100-112, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38851627

RÉSUMÉ

Nicotine, a key constituent of tobacco/electronic cigarettes causes cardiovascular injury and mortality. Nicotine is known to induce oxidative stress and mitochondrial dysfunction in cardiomyocytes leading to cell death. However, the underlying mechanisms remain unclear. Pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) is a member of metal-dependent protein phosphatase (PPM) family and is known to dephosphorylate several AGC family kinases and thereby regulate a diverse set of cellular functions including cell growth, survival, and death. Our lab has previously demonstrated that PHLPP1 removal reduced cardiomyocyte death and cardiac dysfunction following injury. Here, we present a novel finding that nicotine exposure significantly increased PHLPP1 protein expression in the adolescent rodent heart. Building upon our in vivo finding, we determined the mechanism of PHLPP1 expression in cardiomyocytes. Nicotine significantly increased PHLPP1 protein expression without altering PHLPP2 in cardiomyocytes. In cardiomyocytes, nicotine significantly increased NADPH oxidase 4 (NOX4), which coincided with increased reactive oxygen species (ROS) and increased cardiomyocyte apoptosis which were dependent on PHLPP1 expression. PHLPP1 expression was both necessary and sufficient for nicotine induced mitochondrial dysfunction. Mechanistically, nicotine activated extracellular signal-regulated protein kinases (ERK1/2) and subsequent eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) to increase PHLPP1 protein expression. Inhibition of protein synthesis with cycloheximide (CHX) and 4EGI-1 abolished nicotine induced PHLPP1 protein expression. Moreover, inhibition of ERK1/2 activity by U0126 significantly blocked nicotine induced PHLPP1 expression. Overall, this study reveals a novel mechanism by which nicotine regulates PHLPP1 expression through ERK-4E-BP1 signaling axis to drive cardiomyocyte injury.


Sujet(s)
Myocytes cardiaques , Nicotine , Stress oxydatif , Phosphoprotein Phosphatases , Animaux , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Phosphoprotein Phosphatases/métabolisme , Phosphoprotein Phosphatases/génétique , Nicotine/pharmacologie , Nicotine/effets indésirables , Stress oxydatif/effets des médicaments et des substances chimiques , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , NADPH Oxidase 4/métabolisme , NADPH Oxidase 4/génétique , Rat Sprague-Dawley , Souris , Extracellular Signal-Regulated MAP Kinases/métabolisme , Mâle
10.
Anticancer Res ; 44(7): 2861-2870, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38925807

RÉSUMÉ

BACKGROUND/AIM: Glutamine metabolism is crucial in cell proliferation, aging, and apoptosis across various cancer types. Existing research indicates that Sirtuin 4 (SIRT4), primarily located in mitochondria, modulates this process. This study aimed to clarify the regulatory relationship between SIRT4 and glutamine metabolism in cervical cancer. MATERIALS AND METHODS: SIRT4 mRNA levels and their clinical correlation to cervical cancer were analyzed using the UALCAN database. Immunohistochemistry (IHC) was performed to assess SIRT4 protein expression in tissue samples from cervical cancer patients. Transient transfection was employed to create Hela and Siha cell lines with overexpressed SIRT4, mitogen-activated extracellular signal-regulated kinase (MEK), and glutaminase 1 (GLS1). The impact on cellular functions was studied using MTT, soft agar, transwell, and western blotting assays. Glutamate and ATP levels were also measured to evaluate metabolic changes. RESULTS: Low levels of SIRT4 mRNA in cervical cancer tissues correlated with tumor metastasis and poor survival rates. Overexpression of SIRT4 led to suppressed cell proliferation, colony growth, and motility, along with significant down-regulation of GLS expression, a key contributor to glutamine metabolism. Additionally, SIRT4 overexpression resulted in the inactivation of the MEK/ERK/c-myc signaling pathway, while overexpression of MEK reversed these effects. Notably, the inhibitory effects of SIRT4 on cell proliferation, colony formation, migration, and invasion in Hela and Siha cells were significantly attenuated following GLS1 overexpression. CONCLUSION: SIRT4 acts as an anti-cancer agent in cervical cancer by inhibiting glutamine metabolism through the MEK/ERK/c-myc signaling pathway, providing a novel sight for cervical cancer therapy.


Sujet(s)
Prolifération cellulaire , Glutamine , Protéines proto-oncogènes c-myc , Sirtuines , Tumeurs du col de l'utérus , Humains , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/génétique , Femelle , Glutamine/métabolisme , Sirtuines/métabolisme , Sirtuines/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Cellules HeLa , Glutaminase/métabolisme , Glutaminase/antagonistes et inhibiteurs , Glutaminase/génétique , Système de signalisation des MAP kinases , Lignée cellulaire tumorale , Mouvement cellulaire , Régulation de l'expression des gènes tumoraux , Extracellular Signal-Regulated MAP Kinases/métabolisme , Apoptose , Protéines mitochondriales
11.
Discov Med ; 36(185): 1298-1305, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38926116

RÉSUMÉ

BACKGROUND: Oral squamous cell carcinoma (OSCC) is associated with high recurrence and poor prognosis. Baicalin has multiple pharmacological effects, including anti-inflammatory and anti-proliferative activities. Here, we examine the effect of baicalein on OSCC metastasis and its potential mechanism of action. METHODS: SCC-4 and CAL-27 cells were treated with different concentrations of baicalein. The proliferation of OSCC cells was evaluated by Methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. As for migration and metastasis, baicalein-treated OSCC cells were used for wound healing assay and Transwell assay. The levels of epithelial-mesenchymal transition-related proteins (E-cadherin, N-cadherin, vimentin) and extracellular regulated protein kinases (ERK)/ETS Transcription Factor ELK1 (ELK-1)/Snail signaling pathway-related proteins in baicalein-treated OSCC cells were evaluated by western blotting. RESULTS: The rates of cell proliferation and migration, along with the metastatic potential, of baicalein-treated cells were significantly lower than those of the control (p < 0.05), and the effects were concentration-dependent. Furthermore, compared to the control, baicalein significantly decreased the levels of N-cadherin and vimentin in SCC-4 and CAL-27 cells, and increased the E-cadherin level (p < 0.05). Mechanistically, baicalein downregulated the levels of p-ERK1/2, phospho-ETS Transcription Factor ELK1 (p-ELK-1), and Snail (p < 0.05). Finally, the ERK/ELK-1/Snail pathway inhibitor (U0126) promoted the effect of baicalein in inhibiting the migration and invasion of OSCC cells (p < 0.05). CONCLUSION: Baicalein abates the migration, invasion, and metastasis of OSCC cells through the ERK/ELK-1/Snail signaling pathway. This study provides a basis for the development of baicalein as a compound for the treatment of OSCC.


Sujet(s)
Carcinome épidermoïde , Mouvement cellulaire , Prolifération cellulaire , Flavanones , Tumeurs de la bouche , Transduction du signal , Facteurs de transcription de la famille Snail , Protéine Elk-1 à domaine ets , Flavanones/pharmacologie , Flavanones/usage thérapeutique , Humains , Protéine Elk-1 à domaine ets/métabolisme , Facteurs de transcription de la famille Snail/métabolisme , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/traitement médicamenteux , Tumeurs de la bouche/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Métastase tumorale , Extracellular Signal-Regulated MAP Kinases/métabolisme
12.
Asian Pac J Cancer Prev ; 25(6): 2193-2201, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38918683

RÉSUMÉ

The alterations of EGFR and HER2/neu as growth factor receptors and the cytoplasmic signal transduction proteins of RAS/RAF/MAP kinases including its end effector molecule (ERK) are important in the carcinogenesis of many tumors. The activation of these protooncogenes in prostate cancer is still under investigation. The aim of this work was to study EGFR, HER2- neu, inactive (non-phosphorylated) and active (phosphorylated) ERK expression in prostatic adenocarcinomas in correlation to the clinical and pathological parameters. METHODS: Immunohistochemistry- using tissue microarrays- for EGFR, HER2/neu, non-phosphorylated, and phosphor-ERK, was performed on tissues from 166 patients- with primary prostatic adenocarcinoma with no prior treatment-. The results of different markers expression were correlated with the clinical and pathological parameters and were analyzed statistically. RESULTS: The prostatic tissue showed EGFR, HER2 neu, phosphorylated and non-phosphorylated ERK expression in 8.4%, 1.4%, 78.2%, and 83.4% respectively whether low (patchy) or high expression (diffuse).  There were no significant correlations found between patient characteristics and expression of the tested markers. The negative immune reactivity for non-phosphorylated ERK and EGFR- was significantly correlated with high tumor stage (p values 0.03 and 0.01, respectively). CONCLUSION: EGFR and HER2/neu may play a limited role in prostatic adenocarcinoma as they showed positive expression in a limited number of the examined tissues specifically HER2neu. The expression of non-phosphorylated ERK (mostly weak to moderate) and phosphorylated ERK (mostly moderate to strong)- was appreciated in most cases. Thus, we suggest that anti-EGFR drugs may have a limited role in the treatment of castrate-resistant prostate cancer, but anti-MEK/ERK drugs may have more promising role as a target therapy. It is recommended to perform further molecular testing to elucidate the exact mechanism and significance of these markers.


Sujet(s)
Adénocarcinome , Marqueurs biologiques tumoraux , Récepteurs ErbB , Tumeurs de la prostate , Récepteur ErbB-2 , Humains , Mâle , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Récepteurs ErbB/métabolisme , Récepteur ErbB-2/métabolisme , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Adénocarcinome/traitement médicamenteux , Marqueurs biologiques tumoraux/métabolisme , Sujet âgé , Adulte d'âge moyen , Pronostic , Phosphorylation , Kinases raf/métabolisme , Études de suivi , Système de signalisation des MAP kinases , Protéines G ras/métabolisme , Sujet âgé de 80 ans ou plus , Extracellular Signal-Regulated MAP Kinases/métabolisme , Transduction du signal
13.
Elife ; 132024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38921956

RÉSUMÉ

BRAFV600E mutation is a driver mutation in the serrated pathway to colorectal cancers. BRAFV600E drives tumorigenesis through constitutive downstream extracellular signal-regulated kinase (ERK) activation, but high-intensity ERK activation can also trigger tumor suppression. Whether and how oncogenic ERK signaling can be intrinsically adjusted to a 'just-right' level optimal for tumorigenesis remains undetermined. In this study, we found that FAK (Focal adhesion kinase) expression was reduced in BRAFV600E-mutant adenomas/polyps in mice and patients. In Vil1-Cre;BRAFLSL-V600E/+;Ptk2fl/fl mice, Fak deletion maximized BRAFV600E's oncogenic activity and increased cecal tumor incidence to 100%. Mechanistically, our results showed that Fak loss, without jeopardizing BRAFV600E-induced ERK pathway transcriptional output, reduced EGFR (epidermal growth factor receptor)-dependent ERK phosphorylation. Reduction in ERK phosphorylation increased the level of Lgr4, promoting intestinal stemness and cecal tumor formation. Our findings show that a 'just-right' ERK signaling optimal for BRAFV600E-induced cecal tumor formation can be achieved via Fak loss-mediated downregulation of ERK phosphorylation.


Sujet(s)
Tumeurs du caecum , Focal adhesion kinase 1 , Protéines proto-oncogènes B-raf , Animaux , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes B-raf/génétique , Phosphorylation , Souris , Humains , Tumeurs du caecum/métabolisme , Tumeurs du caecum/génétique , Tumeurs du caecum/anatomopathologie , Focal adhesion kinase 1/métabolisme , Focal adhesion kinase 1/génétique , Extracellular Signal-Regulated MAP Kinases/métabolisme , Extracellular Signal-Regulated MAP Kinases/génétique , Système de signalisation des MAP kinases , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Mâle
14.
Science ; 384(6700): eadk0775, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38843331

RÉSUMÉ

How the KRAS oncogene drives cancer growth remains poorly understood. Therefore, we established a systemwide portrait of KRAS- and extracellular signal-regulated kinase (ERK)-dependent gene transcription in KRAS-mutant cancer to delineate the molecular mechanisms of growth and of inhibitor resistance. Unexpectedly, our KRAS-dependent gene signature diverges substantially from the frequently cited Hallmark KRAS signaling gene signature, is driven predominantly through the ERK mitogen-activated protein kinase (MAPK) cascade, and accurately reflects KRAS- and ERK-regulated gene transcription in KRAS-mutant cancer patients. Integration with our ERK-regulated phospho- and total proteome highlights ERK deregulation of the anaphase promoting complex/cyclosome (APC/C) and other components of the cell cycle machinery as key processes that drive pancreatic ductal adenocarcinoma (PDAC) growth. Our findings elucidate mechanistically the critical role of ERK in driving KRAS-mutant tumor growth and in resistance to KRAS-ERK MAPK targeted therapies.


Sujet(s)
Carcinome du canal pancréatique , Extracellular Signal-Regulated MAP Kinases , Régulation de l'expression des gènes tumoraux , Système de signalisation des MAP kinases , Mutation , Tumeurs du pancréas , Protéines proto-oncogènes p21(ras) , Transcriptome , Animaux , Humains , Souris , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Extracellular Signal-Regulated MAP Kinases/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Cellules HEK293
15.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891846

RÉSUMÉ

Tumor recurrence and drug resistance are responsible for poor prognosis in colorectal cancer (CRC). DNA mismatch repair (MMR) deficiency or elevated interleukin-8 (IL-8) levels are characteristics of CRCs, which have been independently correlated with treatment resistance to common therapies. We recently demonstrated significantly impaired therapeutical response and increased IL-8 release of CRC cell lines with reduced expression of MMR protein MLH1 as well as cytoskeletal non-erythrocytic spectrin alpha II (SPTAN1). In the present study, decreased intratumoral MLH1 and SPTAN1 expression in CRCs could be significantly correlated with enhanced serum IL-8. Furthermore, using stably reduced SPTAN1-expressing SW480, SW620 or HT-29 cell lines, the RAS-mediated RAF/MEK/ERK pathway was analyzed. Here, a close connection between low SPTAN1 expression, increased IL-8 secretion, enhanced extracellular-signal-regulated kinase (ERK) phosphorylation and a mesenchymal phenotype were detected. The inhibition of ERK by U0126 led to a significant reduction in IL-8 secretion, and the combination therapy of U0126 with FOLFOX optimizes the response of corresponding cancer cell lines. Therefore, we hypothesize that the combination therapy of FOLFOX and U0126 may have great potential to improve drug efficacy on this subgroup of CRCs, showing decreased MLH1 and SPTAN1 accompanied with high serum IL-8 in affected patients.


Sujet(s)
Butadiènes , Tumeurs colorectales , Fluorouracil , Interleukine-8 , Nitriles , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Interleukine-8/métabolisme , Interleukine-8/génétique , Fluorouracil/pharmacologie , Fluorouracil/usage thérapeutique , Butadiènes/pharmacologie , Nitriles/pharmacologie , Lignée cellulaire tumorale , Composés organiques du platine/pharmacologie , Composés organiques du platine/usage thérapeutique , Leucovorine/usage thérapeutique , Leucovorine/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Femelle , Mâle , Extracellular Signal-Regulated MAP Kinases/métabolisme , Cellules HT29 , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Protéine-1 homologue de MutL/métabolisme , Protéine-1 homologue de MutL/génétique , Adulte d'âge moyen , Sujet âgé , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques
16.
Cells ; 13(11)2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38891045

RÉSUMÉ

Porcine astrovirus (PAstV) has a potential zoonotic risk, with a high proportion of co-infection occurring with porcine epidemic diarrhea virus (PEDV) and other diarrheal pathogens. Despite its high prevalence, the cellular mechanism of PAstV pathogenesis is ill-defined. Previous proteomics analyses have revealed that the differentially expressed protein NOD-like receptor X1 (NLRX1) located in the mitochondria participates in several important antiviral signaling pathways in PAstV-4 infection, which are closely related to mitophagy. In this study, we confirmed that PAstV-4 infection significantly up-regulated NLRX1 and mitophagy in Caco-2 cells, while the silencing of NLRX1 or the treatment of mitophagy inhibitor 3-MA inhibited PAstV-4 replication. Additionally, PAstV-4 infection triggered the activation of the extracellular regulated protein kinases/ myosin light-chain kinase (ERK/MLCK) pathway, followed by the down-regulation of tight-junction proteins (occludin and ZO-1) as well as MUC-2 expression. The silencing of NLRX1 or the treatment of 3-MA inhibited myosin light-chain (MLC) phosphorylation and up-regulated occludin and ZO-1 proteins. Treatment of the ERK inhibitor PD98059 also inhibited MLC phosphorylation, while MLCK inhibitor ML-7 mitigated the down-regulation of mucosa-related protein expression induced by PAstV-4 infection. Yet, adding PD98059 or ML-7 did not affect NLRX1 expression. In summary, this study preliminarily explains that NLRX1 plays an important role in the disruption of intestinal mucosal function triggered by PAstV-4 infection via the ERK/MLC pathway. It will be helpful for further antiviral drug target screening and disease therapy.


Sujet(s)
Muqueuse intestinale , Myosin-Light-Chain Kinase , Animaux , Muqueuse intestinale/métabolisme , Muqueuse intestinale/virologie , Muqueuse intestinale/anatomopathologie , Cellules Caco-2 , Humains , Suidae , Myosin-Light-Chain Kinase/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Infections à Astroviridae/virologie , Mamastrovirus/physiologie , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Maladies des porcs/virologie , Maladies des porcs/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
17.
J Clin Invest ; 134(15)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38885336

RÉSUMÉ

Osteogenesis imperfecta (OI) type V is the second most common form of OI, distinguished by hyperplastic callus formation and calcification of the interosseous membranes, in addition to the bone fragility. It is caused by a recurrent, dominant pathogenic variant (c.-14C>T) in interferon-induced transmembrane protein 5 (IFITM5). Here, we generated a conditional Rosa26-knockin mouse model to study the mechanistic consequences of the recurrent mutation. Expression of the mutant Ifitm5 in osteo-chondroprogenitor or chondrogenic cells resulted in low bone mass and growth retardation. Mutant limbs showed impaired endochondral ossification, cartilage overgrowth, and abnormal growth plate architecture. The cartilage phenotype correlates with the pathology reported in patients with OI type V. Surprisingly, expression of mutant Ifitm5 in mature osteoblasts caused no obvious skeletal abnormalities. In contrast, earlier expression in osteo-chondroprogenitors was associated with an increase in the skeletal progenitor cell population within the periosteum. Lineage tracing showed that chondrogenic cells expressing the mutant Ifitm5 had decreased differentiation into osteoblastic cells in diaphyseal bone. Moreover, mutant IFITM5 disrupted early skeletal homeostasis in part by activating ERK signaling and downstream SOX9 protein, and inhibition of these pathways partially rescued the phenotype in mutant animals. These data identify the contribution of a signaling defect altering osteo-chondroprogenitor differentiation as a driver in the pathogenesis of OI type V.


Sujet(s)
Différenciation cellulaire , Système de signalisation des MAP kinases , Ostéoblastes , Ostéogenèse imparfaite , Facteur de transcription SOX-9 , Animaux , Femelle , Mâle , Souris , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris transgéniques , Mutation , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Ostéogenèse/génétique , Ostéogenèse imparfaite/génétique , Ostéogenèse imparfaite/anatomopathologie , Ostéogenèse imparfaite/métabolisme , Facteur de transcription SOX-9/génétique , Facteur de transcription SOX-9/métabolisme , Cellules souches/métabolisme , Cellules souches/anatomopathologie , Extracellular Signal-Regulated MAP Kinases
18.
J Ethnopharmacol ; 332: 118341, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-38754646

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: The extracellular regulated protein kinase (ERK) plays a crucial role in the mitogen-activated protein kinase (MAPK) family, influencing apoptosis, proliferation, and differentiation. It connection to the insulin (INS) signaling cascade and the development of type 2 diabetes mellitus (T2DM) has been established. Rubus irritans Focke, an indispensable herb in Chinese Tibetan medicine for diabetes mellitus treatment, lacks a comprehensive understanding of its effects and pharmacological mechanisms in T2DM. AIM OF THE STUDY: This study aimed to elucidate the effects of Rubus irritans Focke extract (Rife) on a T2DM rat model, exploring its impact on glycemic and lipid metabolism, histopathological changes, and its potential targeting of the extracellular regulated protein kinase/insulin receptor substrate-1 (ERK/IRS-1) signaling pathway. MATERIALS AND METHODS: A T2DM rat model was induced by streptozotocin (STZ) injection (40 mg/kg) in high-fat diet-fed (HFD) male Wistar rats. Rife and metformin (Met) were administered for 4 weeks, and glycemic, lipid metabolism indices, and histopathological changes were assessed. Protein expression of ERK, IRS-1 in rat liver tissues was examined to evaluate the impact on the ERK/IRS-1 pathway. RESULTS: Rife reducing hepatic ERK and IRS-1 protein expression in T2DM rats. Untargeted metabolomics identified 13 potential biomarkers and 4 differential metabolic pathways related to glycolipid metabolism disorders. CONCLUSIONS: Rife demonstrated improved glycolipid metabolism in T2DM rats by inhibiting the ERK/IRS-1 related signaling pathway and influencing multiple metabolic pathways. This study provides valuable insights into the potential therapeutic mechanisms of Rife in the context of T2DM.


Sujet(s)
Diabète expérimental , Diabète de type 2 , Glycolipides , Hypoglycémiants , Substrats du récepteur à l'insuline , Extraits de plantes , Rat Wistar , Animaux , Mâle , Substrats du récepteur à l'insuline/métabolisme , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Extraits de plantes/pharmacologie , Glycolipides/pharmacologie , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Rats , Hypoglycémiants/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Métabolisme lipidique/effets des médicaments et des substances chimiques , Glycémie/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Streptozocine
19.
Curr Opin Cell Biol ; 88: 102368, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38754355

RÉSUMÉ

The mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling network is a key transducer of signals from various receptors, including receptor tyrosine kinases (RTKs). It controls cell-cycle entry, survival, motility, differentiation, as well as other fates. After four decades of studying this pathway with biochemical methods, the use of fluorescent biosensors has revealed dynamic behaviors such as ERK pulsing, oscillations, and amplitude-modulated activity. Different RTKs equip the MAPK network with specific feedback mechanisms to encode these different ERK dynamics, which are then subsequently decoded into cytoskeletal events and transcriptional programs, actuating cellular fates. Recently, collective ERK wave behaviors have been observed in multiple systems to coordinate cytoskeletal dynamics with fate decisions within cell collectives. This emphasizes that a correct understanding of this pathway requires studying it at multiple scales.


Sujet(s)
Système de signalisation des MAP kinases , Animaux , Humains , Mitogen-Activated Protein Kinases/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Cytosquelette/métabolisme
20.
Mol Pain ; 20: 17448069241261940, 2024.
Article de Anglais | MEDLINE | ID: mdl-38818809

RÉSUMÉ

This study investigated the ERK pathway of the peripheral nervous system and discovered a gender-specific pattern of ERK activation in the dorsal root ganglion of an acid-induced chronic widespread muscular pain model. We employed a twice acid-induced chronic musculoskeletal pain model in rats to evaluate mechanical pain behavior in both male and female groups. We further conducted protein analysis of dissected dorsal root ganglions from both genders. Both male and female rats exhibited a similar pain behavior trend, with females demonstrating a lower pain threshold. Protein analysis of the dorsal root ganglion (DRG) showed a significant increase in phosphorylated ERK after the second acid injection in all groups. However, phosphorylation of ERK was observed in the dorsal root ganglion, with higher levels in the male ipsilateral group compared to the female group. Moreover, there was a no difference between the left and right sides in males, whereas the significant difference was observed in females. In conclusions, the administration of acid injections induced painful behavior in rats, and concurrent with this, a significant upregulation of pERK was observed in the dorsal root ganglia, with a greater magnitude of increase in males than females, and in the contralateral side compared to the ipsilateral side. Our findings shed light on the peripheral mechanisms underlying chronic pain disorders and offer potential avenues for therapeutic intervention.


Sujet(s)
Extracellular Signal-Regulated MAP Kinases , Fibromyalgie , Ganglions sensitifs des nerfs spinaux , Rat Sprague-Dawley , Caractères sexuels , Animaux , Mâle , Femelle , Fibromyalgie/métabolisme , Ganglions sensitifs des nerfs spinaux/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Rats , Seuil nociceptif , Modèles animaux de maladie humaine , Douleur/métabolisme , Douleur/physiopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE