Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 483
Filtrer
1.
Sci Rep ; 14(1): 17360, 2024 07 29.
Article de Anglais | MEDLINE | ID: mdl-39075089

RÉSUMÉ

Prostaglandin E2 (PGE2) is a major contributor to inflammatory pain hyperalgesia, however, the extent to which it modulates the activity of nociceptive axons is incompletely understood. We developed and characterized a microfluidic cell culture model to investigate sensitisation of the axons of dorsal root ganglia neurons. We show that application of PGE2 to fluidically isolated axons leads to sensitisation of their responses to depolarising stimuli. Interestingly the application of PGE2 to the DRG axons elicited a direct and persistent spiking activity propagated to the soma. Both the persistent activity and the membrane depolarisation in the axons are abolished by the EP4 receptor inhibitor and a blocker of cAMP synthesis. Further investigated into the mechanisms of the spiking activity showed that the PGE2 evoked depolarisation was inhibited by Nav1.8 sodium channel blockers but was refractory to the application of TTX or zatebradine. Interestingly, the depolarisation of axons was blocked by blocking ANO1 channels with T16Ainh-A01. We further show that PGE2-elicited axonal responses are altered by the changes in chloride gradient within the axons following treatment with bumetanide a Na-K-2Cl cotransporter NKCC1 inhibitor, but not by VU01240551 an inhibitor of potassium-chloride transporter KCC2. Our data demonstrate a novel role for PGE2/EP4/cAMP pathway which culminates in a sustained depolarisation of sensory axons mediated by a chloride current through ANO1 channels. Therefore, using a microfluidic culture model, we provide evidence for a potential dual function of PGE2 in inflammatory pain: it sensitises depolarisation-evoked responses in nociceptive axons and directly triggers action potentials by activating ANO1 and Nav1.8 channels.


Sujet(s)
Anoctamine-1 , Axones , Dinoprostone , Ganglions sensitifs des nerfs spinaux , Canal sodique voltage-dépendant NAV1.8 , Canal sodique voltage-dépendant NAV1.8/métabolisme , Animaux , Dinoprostone/pharmacologie , Dinoprostone/métabolisme , Axones/métabolisme , Axones/effets des médicaments et des substances chimiques , Axones/physiologie , Ganglions sensitifs des nerfs spinaux/métabolisme , Ganglions sensitifs des nerfs spinaux/effets des médicaments et des substances chimiques , Rats , Anoctamine-1/métabolisme , Cellules réceptrices sensorielles/métabolisme , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques , Potentiels d'action/effets des médicaments et des substances chimiques , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Rat Sprague-Dawley , Cellules cultivées , Membre-2 de la famille-12 des transporteurs de solutés/métabolisme , AMP cyclique/métabolisme
2.
J Cell Biol ; 223(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-38856684

RÉSUMÉ

Sonic Hedgehog (SHH) is a driver of embryonic patterning that, when corrupted, triggers developmental disorders and cancers. SHH effector responses are organized through primary cilia (PC) that grow and retract with the cell cycle and in response to extracellular cues. Disruption of PC homeostasis corrupts SHH regulation, placing significant pressure on the pathway to maintain ciliary fitness. Mechanisms by which ciliary robustness is ensured in SHH-stimulated cells are not yet known. Herein, we reveal a crosstalk circuit induced by SHH activation of Phospholipase A2α that drives ciliary E-type prostanoid receptor 4 (EP4) signaling to ensure PC function and stabilize ciliary length. We demonstrate that blockade of SHH-EP4 crosstalk destabilizes PC cyclic AMP (cAMP) equilibrium, slows ciliary transport, reduces ciliary length, and attenuates SHH pathway induction. Accordingly, Ep4-/- mice display shortened neuroepithelial PC and altered SHH-dependent neuronal cell fate specification. Thus, SHH initiates coordination between distinct ciliary receptors to maintain PC function and length homeostasis for robust downstream signaling.


Sujet(s)
Cils vibratiles , Protéines Hedgehog , Prostaglandines , Transduction du signal , Animaux , Souris , Cils vibratiles/métabolisme , AMP cyclique/métabolisme , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Souris knockout , Prostaglandines/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/génétique
3.
Zhongguo Fei Ai Za Zhi ; 27(4): 245-256, 2024 Apr 20.
Article de Chinois | MEDLINE | ID: mdl-38769827

RÉSUMÉ

BACKGROUND: Tumor microenvironment (TME) is one of the important factors in tumorigenesis and progression, in which tumor-associated macrophages (TAMs) play an important role in non-small cell lung cancer (NSCLC) progression. However, the mechanism of TAMs in NSCLC progression remains unclear, so this study aimed to investigate the role of TAMs in NSCLC progression and to find potential therapeutic targets. METHODS: Gene Expression Profiling Interactive Analysis (GEPIA) database was used to analyze the expression of prostaglandin E2 receptor 4 (EP4) mRNA in NSCLC and normal lung tissues; the protein expression levels of cyclooxygenase-2 (COX-2), EP4, cluster of differentiation 86 (CD86), CD163 and CD31 were detected by immunohistochemistry (IHC) in 120 NSCLC tissues and 24 paracancerous tissues specimens. The nude mouse lung adenocarcinoma cell A549 and macrophage RAW264.7 co-transplanted tumor model was established. And the samples were collected by gavage with EP4 inhibitor E7046, and then stained with hematoxylin-eosin (HE), IHC, and immunofluorescence (IF), and then detected by Western blot for the epithelial mesenchymal transformation (EMT) of the tumor tissues of the nude mice in each group. Western blot was used to detect the expressions of EMT related protiens in each group of nude mice; full-length transcriptome sequencing was used to screen the key genes causing liver metastasis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was performed. RESULTS: EP4 mRNA expression level in NSCLC tissues was generally lower than that in normal lung tissues (P<0.05); COX-2, EP4, CD163, CD31 proteins were differentially expressed in NSCLC tissues and adjacent tissues, and differences were observed in many clinicopathological parameters of NSCLC patients; RAW264.7 shortened the latency period of tumorigenesis of A549 and promoted the proliferation of tumors and liver metastasis of tumors, and E7046 could reduce tumor cell proliferation activity, tumor tissue vascular density and M2-type macrophage infiltration in nude mice; IF staining showed that macrophages were mainly distributed around the metastatic foci of tumors; Western blot results showed that compared with A549 alone transplantation group, the relative expression of E-cadherin protein in tumor tissues of mice in A549 and RAW264.7 co-transplantation group was significantly decreased, and the difference was statistically significant (P<0.05), while the relative expression of N-cadherin protein was up-regulated, but the difference was not statistically significant (P>0.05); the main pathways enriched in the differential genes of the full-length transcriptome were the PI3K-AKT and MAPK signaling pathways. CONCLUSIONS: During NSCLC development, the COX-2/PGE2/EP4 axis may promote tumor progression by inducing macrophage functional activation, and EP4 may be a potential new target for tumor immunotherapy. This study provides new perspectives and ideas for in-depth exploration of the mechanisms of NSCLC development, as well as a theoretical basis for the development of new therapeutic strategies for NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Cyclooxygenase 2 , Dinoprostone , Tumeurs du poumon , Sous-type EP4 des récepteurs des prostaglandines E , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/génétique , Humains , Cyclooxygenase 2/génétique , Cyclooxygenase 2/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Animaux , Dinoprostone/métabolisme , Souris , Macrophages/métabolisme , Activation des macrophages , Mâle , Femelle , Cellules A549 , Cellules RAW 264.7
4.
Life Sci ; 350: 122751, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38797363

RÉSUMÉ

AIM: To understand the mechanism of prostaglandin E2 (PGE2)-mediated immunosuppression in dendritic cells (DCs). MAIN METHODS: In vivo experiments were conducted on 4T1 tumor bearing mice (TBM). In vitro experiments were performed in bone marrow-derived DCs (BMDCs), or spleen cells. Cytokines were monitored by ELISA/ELIspot. Gene expression was monitored by RT-PCR/flow cytometry. KEY FINDINGS: In silico, in vitro, and in vivo experiments in 4T1 TBM revealed that PGE2 induced IL-6/pSTAT3 signaling through EP4 receptors in DCs, resulting in their dysfunction. These effects were reversed by EP4 antibody neutralization, EP4 antagonist, and STAT3 inhibitory peptides. PGE2 induced IL-6 was regulated by miR-365, as its mimic inhibited PGE2 induced IL-6 and the inhibitor increased lL-6 levels in DC. Bio-informatic analysis in human mammary cancers also revealed a strong compared co-relation between PGE2 and IL-6 (Correlation AnalyzeR) (R = 0.94). Mice bearing PTGS-2 KD 4T1 tumors had decreased tumor burden, PGE2, EP4, IL-6, and pSTAT3 signaling, along with improved DCs and T cell functions. Treatment of mice with a cyclooxygenase-2 (COX-2) inhibitor or EP4 antagonist decreased tumor burden, and this effect of EP4 antagonist was abrogated upon in vivo depletion of CD11c cells, indicating the crucial role of PGE2 signaling in DCs in tumor progression. SIGNIFICANCE: In summary, our data highlights the importance of dendritic cells in mediating PGE2-mediated immunosuppression and the use of EP4 or STAT3 inhibitors or miR365 mimics can restore immunogenicity in cancer.


Sujet(s)
Cellules dendritiques , Dinoprostone , Interleukine-6 , microARN , Facteur de transcription STAT-3 , Transduction du signal , Animaux , Facteur de transcription STAT-3/métabolisme , Cellules dendritiques/métabolisme , Cellules dendritiques/immunologie , microARN/génétique , microARN/métabolisme , Souris , Dinoprostone/métabolisme , Interleukine-6/métabolisme , Femelle , Humains , Souris de lignée BALB C , Lignée cellulaire tumorale , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme
5.
Pharmacol Rev ; 76(3): 388-413, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38697857

RÉSUMÉ

The involvement of the prostaglandin E2 (PGE2) system in cancer progression has long been recognized. PGE2 functions as an autocrine and paracrine signaling molecule with pleiotropic effects in the human body. High levels of intratumoral PGE2 and overexpression of the key metabolic enzymes of PGE2 have been observed and suggested to contribute to tumor progression. This has been claimed for different types of solid tumors, including, but not limited to, lung, breast, and colon cancer. PGE2 has direct effects on tumor cells and angiogenesis that are known to promote tumor development. However, one of the main mechanisms behind PGE2 driving cancerogenesis is currently thought to be anchored in suppressed antitumor immunity, thus providing possible therapeutic targets to be used in cancer immunotherapies. EP2 and EP4, two receptors for PGE2, are emerging as being the most relevant for this purpose. This review aims to summarize the known roles of PGE2 in the immune system and its functions within the tumor microenvironment. SIGNIFICANCE STATEMENT: Prostaglandin E2 (PGE2) has long been known to be a signaling molecule in cancer. Its presence in tumors has been repeatedly associated with disease progression. Elucidation of its effects on immunological components of the tumor microenvironment has highlighted the potential of PGE2 receptor antagonists in cancer treatment, particularly in combination with immune checkpoint inhibitor therapeutics. Adjuvant treatment could increase the response rates and the efficacy of immune-based therapies.


Sujet(s)
Dinoprostone , Tumeurs , Sous-type EP2 des récepteurs des prostaglandines E , Sous-type EP4 des récepteurs des prostaglandines E , Microenvironnement tumoral , Humains , Dinoprostone/métabolisme , Animaux , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/immunologie , Tumeurs/anatomopathologie , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/antagonistes et inhibiteurs , Transduction du signal
6.
Commun Biol ; 7(1): 567, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38745046

RÉSUMÉ

Lymph node metastasis, primarily caused by the migration of oral squamous cell carcinoma (OSCC) cells, stands as a crucial prognostic marker. We have previously demonstrated that EP4, a subtype of the prostaglandin E2 (PGE2) receptor, orchestrates OSCC cell migration via Ca2+ signaling. The exact mechanisms by which EP4 influences cell migration through Ca2+ signaling, however, is unclear. Our study aims to clarify how EP4 controls OSCC cell migration through this pathway. We find that activating EP4 with an agonist (ONO-AE1-473) increased intracellular Ca2+ levels and the migration of human oral cancer cells (HSC-3), but not human gingival fibroblasts (HGnF). Further RNA sequencing linked EP4 to calmodulin-like protein 6 (CALML6), whose role remains undefined in OSCC. Through protein-protein interaction network analysis, a strong connection is identified between CALML6 and calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2), with EP4 activation also boosting mitochondrial function. Overexpressing EP4 in HSC-3 cells increases experimental lung metastasis in mice, whereas inhibiting CaMKK2 with STO-609 markedly lowers these metastases. This positions CaMKK2 as a potential new target for treating OSCC metastasis. Our findings highlight CALML6 as a pivotal regulator in EP4-driven mitochondrial respiration, affecting cell migration and metastasis via the CaMKK2 pathway.


Sujet(s)
Protéines de liaison au calcium , Carcinome épidermoïde , Mouvement cellulaire , Mitochondries , Tumeurs de la bouche , Sous-type EP4 des récepteurs des prostaglandines E , Animaux , Humains , Souris , Calcium-Calmodulin-Dependent Protein Kinase Kinase/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Kinase/génétique , Calmoduline/métabolisme , Calmoduline/génétique , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique , Lignée cellulaire tumorale , Mitochondries/métabolisme , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/génétique , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/génétique , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Protéines de liaison au calcium/génétique , Protéines de liaison au calcium/métabolisme
7.
Sci Adv ; 10(22): eadk3121, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38809988

RÉSUMÉ

Regular, long-term aspirin use may act synergistically with genetic variants, particularly those in mechanistically relevant pathways, to confer a protective effect on colorectal cancer (CRC) risk. We leveraged pooled data from 52 clinical trial, cohort, and case-control studies that included 30,806 CRC cases and 41,861 controls of European ancestry to conduct a genome-wide interaction scan between regular aspirin/nonsteroidal anti-inflammatory drug (NSAID) use and imputed genetic variants. After adjusting for multiple comparisons, we identified statistically significant interactions between regular aspirin/NSAID use and variants in 6q24.1 (top hit rs72833769), which has evidence of influencing expression of TBC1D7 (a subunit of the TSC1-TSC2 complex, a key regulator of MTOR activity), and variants in 5p13.1 (top hit rs350047), which is associated with expression of PTGER4 (codes a cell surface receptor directly involved in the mode of action of aspirin). Genetic variants with functional impact may modulate the chemopreventive effect of regular aspirin use, and our study identifies putative previously unidentified targets for additional mechanistic interrogation.


Sujet(s)
Anti-inflammatoires non stéroïdiens , Tumeurs colorectales , Étude d'association pangénomique , Polymorphisme de nucléotide simple , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/traitement médicamenteux , Anti-inflammatoires non stéroïdiens/pharmacologie , Acide acétylsalicylique/pharmacologie , Sous-type EP4 des récepteurs des prostaglandines E/génétique , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Mâle , Prédisposition génétique à une maladie , Femelle , Études cas-témoins , Adulte d'âge moyen , Locus génétiques , Sujet âgé
8.
Nat Neurosci ; 27(8): 1534-1544, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38802591

RÉSUMÉ

Oligodendrocyte-lineage cells, including NG2 glia, undergo prominent changes in various neurodegenerative disorders. Here, we identify a neuroprotective role for NG2 glia against prion toxicity. NG2 glia were activated after prion infection in cerebellar organotypic cultured slices (COCS) and in brains of prion-inoculated mice. In both model systems, depletion of NG2 glia exacerbated prion-induced neurodegeneration and accelerated prion pathology. Loss of NG2 glia enhanced the biosynthesis of prostaglandin E2 (PGE2) by microglia, which augmented prion neurotoxicity through binding to the EP4 receptor. Pharmacological or genetic inhibition of PGE2 biosynthesis attenuated prion-induced neurodegeneration in COCS and mice, reduced the enhanced neurodegeneration in NG2-glia-depleted COCS after prion infection, and dampened the acceleration of prion disease in NG2-glia-depleted mice. These data unveil a non-cell-autonomous interaction between NG2 glia and microglia in prion disease and suggest that PGE2 signaling may represent an actionable target against prion diseases.


Sujet(s)
Dinoprostone , Microglie , Névroglie , Neurones , Maladies à prions , Transduction du signal , Animaux , Microglie/métabolisme , Dinoprostone/métabolisme , Souris , Transduction du signal/physiologie , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Névroglie/métabolisme , Maladies à prions/métabolisme , Maladies à prions/anatomopathologie , Souris de lignée C57BL , Antigènes/métabolisme , Protéoglycanes/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Prions/métabolisme , Prions/toxicité , Cervelet/métabolisme , Cervelet/anatomopathologie
9.
Nature ; 629(8011): 417-425, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38658748

RÉSUMÉ

Cancer-specific TCF1+ stem-like CD8+ T cells can drive protective anticancer immunity through expansion and effector cell differentiation1-4; however, this response is dysfunctional in tumours. Current cancer immunotherapies2,5-9 can promote anticancer responses through TCF1+ stem-like CD8+ T cells in some but not all patients. This variation points towards currently ill-defined mechanisms that limit TCF1+CD8+ T cell-mediated anticancer immunity. Here we demonstrate that tumour-derived prostaglandin E2 (PGE2) restricts the proliferative expansion and effector differentiation of TCF1+CD8+ T cells within tumours, which promotes cancer immune escape. PGE2 does not affect the priming of TCF1+CD8+ T cells in draining lymph nodes. PGE2 acts through EP2 and EP4 (EP2/EP4) receptor signalling in CD8+ T cells to limit the intratumoural generation of early and late effector T cell populations that originate from TCF1+ tumour-infiltrating CD8+ T lymphocytes (TILs). Ablation of EP2/EP4 signalling in cancer-specific CD8+ T cells rescues their expansion and effector differentiation within tumours and leads to tumour elimination in multiple mouse cancer models. Mechanistically, suppression of the interleukin-2 (IL-2) signalling pathway underlies the PGE2-mediated inhibition of TCF1+ TIL responses. Altogether, we uncover a key mechanism that restricts the IL-2 responsiveness of TCF1+ TILs and prevents anticancer T cell responses that originate from these cells. This study identifies the PGE2-EP2/EP4 axis as a molecular target to restore IL-2 responsiveness in anticancer TILs to achieve cancer immune control.


Sujet(s)
Lymphocytes T CD8+ , Prolifération cellulaire , Dinoprostone , Lymphocytes TIL , Tumeurs , Cellules souches , Échappement de la tumeur à la surveillance immunitaire , Animaux , Femelle , Humains , Mâle , Souris , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Différenciation cellulaire , Lignée cellulaire tumorale , Dinoprostone/métabolisme , Modèles animaux de maladie humaine , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Interleukine-2 , Noeuds lymphatiques/cytologie , Noeuds lymphatiques/immunologie , Lymphocytes TIL/cytologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Souris de lignée C57BL , Tumeurs/immunologie , Tumeurs/prévention et contrôle , Sous-type EP2 des récepteurs des prostaglandines E/déficit , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/déficit , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Transduction du signal , Cellules souches/cytologie , Cellules souches/immunologie , Cellules souches/métabolisme , Échappement de la tumeur à la surveillance immunitaire/immunologie
10.
Nature ; 629(8011): 426-434, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38658764

RÉSUMÉ

Expansion of antigen-experienced CD8+ T cells is critical for the success of tumour-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability2,3. Therefore, it is essential to comprehend mechanistic barriers to IL-2 sensing in the tumour microenvironment to implement strategies to reinvigorate IL-2 responsiveness and T cell antitumour responses. Here we report that prostaglandin E2 (PGE2), a known negative regulator of immune response in the tumour microenvironment4,5, is present at high concentrations in tumour tissue from patients and leads to impaired IL-2 sensing in human CD8+ TILs via the PGE2 receptors EP2 and EP4. Mechanistically, PGE2 inhibits IL-2 sensing in TILs by downregulating the IL-2Rγc chain, resulting in defective assembly of IL-2Rß-IL2Rγc membrane dimers. This results in impaired IL-2-mTOR adaptation and PGC1α transcriptional repression, causing oxidative stress and ferroptotic cell death in tumour-reactive TILs. Inhibition of PGE2 signalling to EP2 and EP4 during TIL expansion for ACT resulted in increased IL-2 sensing, leading to enhanced proliferation of tumour-reactive TILs and enhanced tumour control once the cells were transferred in vivo. Our study reveals fundamental features that underlie impairment of human TILs mediated by PGE2 in the tumour microenvironment. These findings have therapeutic implications for cancer immunotherapy and cell therapy, and enable the development of targeted strategies to enhance IL-2 sensing and amplify the IL-2 response in TILs, thereby promoting the expansion of effector T cells with enhanced therapeutic potential.


Sujet(s)
Lymphocytes T CD8+ , Prolifération cellulaire , Dinoprostone , Interleukine-2 , Lymphocytes TIL , Mitochondries , Transduction du signal , Animaux , Humains , Souris , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Dinoprostone/métabolisme , Régulation négative , Ferroptose , Sous-unité gamma commune aux récepteurs des interleukines/biosynthèse , Sous-unité gamma commune aux récepteurs des interleukines/déficit , Sous-unité gamma commune aux récepteurs des interleukines/métabolisme , Interleukine-2/antagonistes et inhibiteurs , Interleukine-2/immunologie , Interleukine-2/métabolisme , Sous-unité bêta du récepteur à l'interleukine-2/métabolisme , Lymphocytes TIL/cytologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Mitochondries/métabolisme , Stress oxydatif , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/antagonistes et inhibiteurs , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/métabolisme , Microenvironnement tumoral/immunologie
11.
Spine J ; 24(8): 1407-1415, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38631491

RÉSUMÉ

BACKGROUND CONTEXT: Vertebral endplate defects are often implicated in degenerative disc disorders, yet their connection to patient-reported symptoms remains unclear. COX-2 and PGE-2 are known for their roles in inflammation and pain, with EP-4 receptor involvement in pain signaling. Examining their expression in vertebral endplate tissues may provide insights into pathomechanism of low back pain. PURPOSE: To investigate the association between endplate defects and patient-reported symptoms and to further clarify the role of the COX-2/PGE-2/EP-4 axis in the pathogenesis of chronic low back pain. STUDY DESIGN/SETTING: Retrospective study. PATIENT SAMPLE: A total of 71 patients who had undergone single-level L4/5 or L5/S1 modified laminectomy decompression preserving proximal upper laminae and transforaminal lumbar interbody fusion surgery were included in this study, including 18 patients diagnosed with lumbar disc herniation, 19 with lumbar disc herniation accompanied by degenerative lumbar spinal stenosis, and 34 with degenerative spondylolisthesis. OUTCOME MEASURES: Demographic data, Pfirrmann grade, Modic changes, endplate defect score, visual analog scale (VAS) for back and leg pain, and Oswestry Disability Index (ODI) before surgery, 3-month and 6-month follow-up, and the percentage of immune-positive cells (COX-2, PGE-2, and EP-4) in endplate tissue sections. METHODS: Patients were divided into defect and nondefect groups according to endplate morphology on lumbar MR. All intraoperative endplate specimens were immediately fixed in 10% formaldehyde, and then embedded in paraffin 3 days later for tissue sections. The outcome measures were compared between the defect group and nondefect group. Data were analyzed using independent t-tests and χ² tests. Pearson's rank correlation test was used to assess correlations between patient-reported symptoms and the percentage of immune-positive cells in the groups. Multivariable logistic regression models using the forward stepwise likelihood ratio method were used to identify the factors that were independently associated with endplate defects. RESULTS: The age of Defect group was significantly higher than that of nondefect group (52.5±7.7 vs 57.2±9.1. p=.024). There were no significant differences in gender, diagnosis, BMI, comorbidities, or surgical level between the two groups. Modic changes (Type Ⅱ/Type Ⅲ) were more common in patients of Defect group than nondefect group (38.5% vs 11.1%, p<.001), and so was disc degeneration (Pfirrmann grade Ⅳ/Ⅴ) (69.2% vs 33.3%, p<.001). Defect group had significantly higher VAS-Back (6.5±2.0 vs 4.9±1.6, p<.001) and ODI scores (62.9±10.7 vs 45.2±14.8, p<.001) than nondefect group, while there was no significant differences between the two groups during the 3 and 6-month follow-up after surgery. Histologically, Defect group was characterized by upregulation of COX-2, PGE-2, and EP-4 in endplate tissue sections. Both in defect and nondefect groups, VAS-Back showed moderate positive correlations with the expressions of COX-2 (r=0.643; r=0.558, p both<.001), PGE-2 (r=0.611; r=0.640, p both<.001), and EP-4 (r=0.643; r=0.563, p both<.001). Multivariate regression analyses reveled that percentage of COX-2-positive cells was associated with endplate defects (OR=1.509, 95%CI [1.048-2.171], p=.027), as well as percentage of PGE-2-positive (OR=1.291, 95%CI [1.106-1.508], p=.001) and EP-4-positive cells (OR=1.284, 95%CI [1.048∼2.171], p=.003). CONCLUSIONS: Patients with endplate defects had worse quality of life, more severe disc degeneration and Modic changes, and up-regulated COX-2/PGE-2/EP-4 axis expression in cartilage endplates in patients with defected endplates. Inflammatory factors may significantly contribute to the onset and progression of chronic low back pain in patients with endplate defects, consequently impacting patient-reported symptoms.


Sujet(s)
Cyclooxygenase 2 , Dinoprostone , Lombalgie , Vertèbres lombales , Sous-type EP4 des récepteurs des prostaglandines E , Humains , Mâle , Femelle , Adulte d'âge moyen , Cyclooxygenase 2/métabolisme , Vertèbres lombales/chirurgie , Lombalgie/anatomopathologie , Sujet âgé , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Études rétrospectives , Dinoprostone/métabolisme , Adulte , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/anatomopathologie , Dégénérescence de disque intervertébral/chirurgie , Déplacement de disque intervertébral/métabolisme , Déplacement de disque intervertébral/chirurgie , Déplacement de disque intervertébral/anatomopathologie , Sténose du canal vertébral/chirurgie , Sténose du canal vertébral/anatomopathologie , Arthrodèse vertébrale , Spondylolisthésis/chirurgie , Spondylolisthésis/anatomopathologie , Mesures des résultats rapportés par les patients , Immunohistochimie
12.
Circ Heart Fail ; 17(4): e011089, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38525608

RÉSUMÉ

BACKGROUND: Prostaglandin E2 acts through 4 G-protein-coupled receptors (EP1-EP4). We previously reported that activation of the EP3 receptor reduces cardiac contractility, and its expression increases after a myocardial infarction (MI), mediating the reduction in cardiac function. In contrast, cardiac overexpression of the EP4 receptor in MI substantially improves cardiac function. Moreover, we recently reported that mice overexpressing EP3 have heart failure under basal conditions and worsened cardiac function after MI. Thus, the deleterious effects of the prostaglandin E2 EP receptors in the heart are mediated via its EP3 receptor. We, therefore, hypothesized that cardiomyocyte-specific knockout (CM-EP3 KO) or antagonism of the EP3 receptor protects the heart after MI. METHODS: To test our hypothesis, we made the novel CM-EP3 KO mouse and subjected CM-EP3 KO or controls to sham or MI surgery for 2 weeks. In separate experiments, C57BL/6 mice were subjected to 2 weeks of MI and treated with either the EP3 antagonist L798 106 or vehicle starting 3 days post-MI. RESULTS: CM-EP3 KO significantly prevented a decline in cardiac function after MI compared with WT animals and prevented an increase in hypertrophy and fibrosis. Excitingly, mice treated with L798 106 3 days after MI had significantly better cardiac function compared with vehicle-treated mice. CONCLUSIONS: Altogether, these data suggest that EP3 may play a direct role in regulating cardiac function, and pharmaceutical targeting of the EP3 receptor may be a therapeutic option in the treatment of heart failure.


Sujet(s)
Défaillance cardiaque , Infarctus du myocarde , Souris , Animaux , Dinoprostone/métabolisme , Dinoprostone/pharmacologie , Récepteur prostaglandine/génétique , Récepteur prostaglandine/métabolisme , Délétion de gène , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/génétique , Défaillance cardiaque/prévention et contrôle , Souris de lignée C57BL , Myocytes cardiaques/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/génétique , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP3 des récepteurs des prostaglandines E/génétique , Sous-type EP3 des récepteurs des prostaglandines E/métabolisme
13.
Article de Anglais | MEDLINE | ID: mdl-38471265

RÉSUMÉ

Prostaglandin E2 (PGE2) signals differently through 4 receptor subtypes (EP1-EP4) to elicit diverse physiologic/pathologic effects. We previously reported that PGE2 via its EP3 receptor reduces cardiac contractility and male mice with cardiomyocyte-specific deletion of the EP4 receptor (EP4 KO) develop dilated cardiomyopathy. The aim of this study was to identify pathways responsible for this phenotype. We performed ingenuity pathway analysis (IPA) and found that genes differentiating WT mice and EP4 KO mice were significantly overrepresented in mitochondrial (adj. p value = 6.28 × 10-26) and oxidative phosphorylation (adj. p value = 1.58 × 10-27) pathways. Electron microscopy from the EP4 KO hearts show substantial mitochondrial disarray and disordered cristae. Not surprisingly, isolated adult mouse cardiomyocytes (AVM) from these mice have reduced ATP levels compared to their WT littermates and reduced expression of key genes involved in the electron transport chain (ETC) in older mice. Moreover, treatment of AVM from C57Bl/6 mice with PGE2 or the EP3 agonist sulprostone resulted in changes of various genes involved in the ETC, measured by the Mitochondrial Energy Metabolism RT2-profiler assay. Lastly, the EP4 KO mice have reduced expression of superoxide dismuatse-2 (SOD2), whereas treatment of AVM with PGE2 or sulprostone increase superoxide production, suggesting increased oxidative stress levels in these EP4 KO mice. Altogether the current study supports the premise that PGE2 acting via its EP4 receptor is protective, while signaling through its other receptors, likely EP3, is deleterious.


Sujet(s)
Dinoprostone , Souris knockout , Myocytes cardiaques , Sous-type EP4 des récepteurs des prostaglandines E , Animaux , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Dinoprostone/métabolisme , Souris , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/génétique , Sous-type EP4 des récepteurs des prostaglandines E/agonistes , Mâle , Souris de lignée C57BL , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques
14.
J Leukoc Biol ; 115(6): 1183-1189, 2024 05 29.
Article de Anglais | MEDLINE | ID: mdl-38345417

RÉSUMÉ

Prostaglandin E2 (PGE2) is a recognized inhibitor of granulocyte functions. However, most of the data supporting this was obtained when available pharmacological tools mainly targeted the EP2 receptor. Herein, we revisited the inhibitory effect of PGE2 on reactive oxygen species production, leukotriene biosynthesis, and migration in human neutrophils. Our data confirm the inhibitory effect of PGE2 on these functions and unravel that the effect of PGE2 on human neutrophils is obtained by the combined action of EP2 and EP4 agonism. Accordingly, we also demonstrate that the inhibitory effect of PGE2 is fully prevented only by the combination of EP2 and EP4 receptor antagonists, underscoring the importance of targeting both receptors in the effect of PGE2. Conversely, we also show that the inhibition of ROS production by human eosinophils only involves the EP4 receptor, despite the fact that they also express the EP2 receptor.


Sujet(s)
Dinoprostone , Granulocytes neutrophiles , Sous-type EP2 des récepteurs des prostaglandines E , Sous-type EP4 des récepteurs des prostaglandines E , Humains , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Dinoprostone/métabolisme , Espèces réactives de l'oxygène/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques
15.
Front Immunol ; 15: 1355769, 2024.
Article de Anglais | MEDLINE | ID: mdl-38343540

RÉSUMÉ

Tumors educate their environment to prime the occurrence of suppressive cell subsets, which enable tumor evasion and favors tumor progression. Among these, there are the myeloid-derived suppressor cells (MDSCs), their presence being associated with the poor clinical outcome of cancer patients. Tumor-derived prostaglandin E2 (PGE2) is known to mediate MDSC differentiation and the acquisition of pro-tumor features. In myeloid cells, PGE2 signaling is mediated via E-prostanoid receptor type 2 (EP2) and EP4. Although the suppressive role of PGE2 is well established in MDSCs, the role of EP2/4 on human MDSCs or whether EP2/4 modulation can prevent MDSCs suppressive features upon exposure to tumor-derived PGE2 is poorly defined. In this study, using an in vitro model of human monocytic-MDSCs (M-MDSCs) we demonstrate that EP2 and EP4 signaling contribute to the induction of a pro-tumor phenotype and function on M-MDSCs. PGE2 signaling via EP2 and EP4 boosted M-MDSC ability to suppress T and NK cell responses. Combined EP2/4 blockade on M-MDSCs during PGE2 exposure prevented the occurrence of these suppressive features. Additionally, EP2/4 blockade attenuated the suppressive phenotype of M-MDSCs in a 3D coculture with colorectal cancer patient-derived organoids. Together, these results identify the role of tumor-derived PGE2 signaling via EP2 and EP4 in this human M-MDSC model, supporting the therapeutic value of targeting PGE2-EP2/4 axis in M-MDSCs to alleviate immunosuppression and facilitate the development of anti-tumor immunity.


Sujet(s)
Cellules myéloïdes suppressives , Humains , Cellules myéloïdes suppressives/métabolisme , Dinoprostone/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Monocytes
16.
Cytokine ; 175: 156498, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38176086

RÉSUMÉ

S. aureus isolated from bacterial bovine endometritis is common in epidemiological reports, but is often ignored as a subclinical pathogenic microorganism. In a previous study, we showed that live S. aureus (LSA) and heat killed S. aureus (HK-SA) induce different inflammatory responses in bovine endometrial tissue, and possibly being associated with the accumulation of prostaglandin E2 (PGE2). Thus, in this study, we varied PGE2 concentrations using inhibitors or agonists in HK-SA-treated bovine endometrial tissues. The results demonstrated that PGE2 has a positive relationship with IL-6, TNF-α, and damage-associated molecular patterns (DAMPs; e.g., HMGB-1 and HABP-1) expression and tissues damage, and is regulated by the EP4-p38 MAPK pathway. We concluded that lipoproteins of S. aureus are associated with PGE2 generation. To further explore the relationship between LSA and PGE2 accumulation, we used the S. aureus strain SA113 lipoprotein knockout (SA113Δlpl) to infect bovine endometrial epithelial cells (BECs). LSA decreased PGE2, cAMP, EP4, IL-6, IL-8, cAMP secretion, and the MAPK and PKA signaling pathways when infected with SA113Δlpl, as compared with SA113-infected groups. Moreover, the adhesion and invasion of BECs were similarly downregulated when lipoproteins in S. aureus were knocked out. The results of this study show that PGE2 is involved in both HK-SA- and LSA-induced inflammatory responses in the bovine endometrium. We suggest that S. aureus infection is associated with bovine endometritis, and although HK-SA and LSA induce different inflammatory responses, the strategy of decreasing PGE2 accumulation is helpful in reducing the inflammation stage caused by S. aureus.


Sujet(s)
Endométrite , Staphylococcus aureus résistant à la méticilline , Femelle , Humains , Animaux , Bovins , Dinoprostone/métabolisme , Staphylococcus aureus résistant à la méticilline/métabolisme , Staphylococcus aureus/métabolisme , Interleukine-6 , Lipoprotéines , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme
17.
Basic Clin Pharmacol Toxicol ; 134(3): 345-360, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38009541

RÉSUMÉ

Transient receptor potential vanilloid type-1 (TRPV1) channels play key roles in chronic pain conditions and are modulated by different inflammatory mediators to elicit heat sensitisation. Bradykinin is a 9-amino acid peptide chain that promotes inflammation. The aim of present study is to investigate how bradykinin and prostaglandin receptors (EP3 and EP4 ) modulate the sensitisation of TRPV1-mediated responses. Calcium imaging studies of rat dorsal root ganglion (DRG) neurons were employed to investigate the desensitizing responses of TRPV1 ion channels by capsaicin, and the re-sensitization of TRPV1 by bradykinin, then to explore the role EP3 and EP4 receptors in mediating these bradykinin-dependent effects. Immunocytochemistry was used to study the co-expression and distribution of EP4, TRPV1, COX-1 and B2 in rat DRG neurons. Desensitization was seen upon repeated capsaicin application, we show that bradykinin-mediated sensitization of capsaicin-evoked calcium responses in rat DRG neurons occurs is dependent on COX-1 activity and utilizes a pathway that involves EP4 but not EP3 receptors. Immunocytochemical techniques revealed that EP4, TRPV1, COX-1 and B2 proteins are expressed mainly in small diameter (<1000 µm2 ) cell bodies of rat DRG neurons which are typically nociceptors. The present study provides suggestive evidence for a potential signalling pathway through which bradykinin may regulate TRPV1 ion channel function via EP4 receptors. In addition to confirming existing knowledge, the anatomical distribution and colocalization of these proteins in DRG neurons as revealed by this study offer valuable insight.


Sujet(s)
Capsaïcine , Sous-type EP4 des récepteurs des prostaglandines E , Rats , Animaux , Capsaïcine/pharmacologie , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Bradykinine/pharmacologie , Rat Sprague-Dawley , Ganglions sensitifs des nerfs spinaux/métabolisme , Calcium/métabolisme , Canaux cationiques TRPV/métabolisme , Neurones/métabolisme , Cellules cultivées
18.
Cancer Res Commun ; 3(8): 1486-1500, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37559947

RÉSUMÉ

While the role of prostaglandin E2 (PGE2) in promoting malignant progression is well established, how to optimally block the activity of PGE2 signaling remains to be demonstrated. Clinical trials with prostaglandin pathway targeted agents have shown activity but without sufficient significance or dose-limiting toxicities that have prevented approval. PGE2 signals through four receptors (EP1-4) to modulate tumor progression. EP2 and EP4 signaling exacerbates tumor pathology and is immunosuppressive through potentiating cAMP production. EP1 and EP3 signaling has the opposite effect through increasing IP3 and decreasing cAMP. Using available small-molecule antagonists of single EP receptors, the cyclooxygenase-2 (COX-2) inhibitor celecoxib, or a novel dual EP2/EP4 antagonist generated in this investigation, we tested which approach to block PGE2 signaling optimally restored immunologic activity in mouse and human immune cells and antitumor activity in syngeneic, spontaneous, and xenograft tumor models. We found that dual antagonism of EP2 and EP4 together significantly enhanced the activation of PGE2-suppressed mouse and human monocytes and CD8+ T cells in vitro as compared with single EP antagonists. CD8+ T-cell activation was dampened by single EP1 and EP3 antagonists. Dual EP2/EP4 PGE2 receptor antagonists increased tumor microenvironment lymphocyte infiltration and significantly reduced disease burden in multiple tumor models, including in the adenomatous polyposis coli (APC)min+/- spontaneous colorectal tumor model, compared with celecoxib. These results support a hypothesis that redundancy of EP2 and EP4 receptor signaling necessitates a therapeutic strategy of dual blockade of EP2 and EP4. Here we describe TPST-1495, a first-in-class orally available small-molecule dual EP2/EP4 antagonist. Significance: Prostaglandin (PGE2) drives tumor progression but the pathway has not been effectively drugged. We demonstrate significantly enhanced immunologic potency and antitumor activity through blockade of EP2 and EP4 PGE2 receptor signaling together with a single molecule.


Sujet(s)
Tumeurs , Prostaglandines , Humains , Animaux , Souris , Dinoprostone/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Célécoxib/pharmacologie , Lymphocytes T CD8+/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Inhibiteurs de la cyclooxygénase 2 , Microenvironnement tumoral
19.
Am J Physiol Heart Circ Physiol ; 325(4): H687-H701, 2023 10 01.
Article de Anglais | MEDLINE | ID: mdl-37566109

RÉSUMÉ

The ductus arteriosus (DA) is a vascular shunt that allows oxygenated blood to bypass the developing lungs in utero. Fetal DA patency requires vasodilatory signaling via the prostaglandin E2 (PGE2) receptor EP4. However, in humans and mice, disrupted PGE2-EP4 signaling in utero causes unexpected patency of the DA (PDA) after birth, suggesting another role for EP4 during development. We used EP4-knockout (KO) mice and acute versus chronic pharmacological approaches to investigate EP4 signaling in DA development and function. Expression analyses identified EP4 as the primary EP receptor in the DA from midgestation to term; inhibitor studies verified EP4 as the primary dilator during this period. Chronic antagonism recapitulated the EP4 KO phenotype and revealed a narrow developmental window when EP4 stimulation is required for postnatal DA closure. Myography studies indicate that despite reduced contractile properties, the EP4 KO DA maintains an intact oxygen response. In newborns, hyperoxia constricted the EP4 KO DA but survival was not improved, and permanent remodeling was disrupted. Vasomotion and increased nitric oxide (NO) sensitivity in the EP4 KO DA suggest incomplete DA development. Analysis of DA maturity markers confirmed a partially immature EP4 KO DA phenotype. Together, our data suggest that EP4 signaling in late gestation plays a key developmental role in establishing a functional term DA. When disrupted in EP4 KO mice, the postnatal DA exhibits signaling and contractile properties characteristic of an immature DA, including impairments in the first, muscular phase of DA closure, in addition to known abnormalities in the second permanent remodeling phase.NEW & NOTEWORTHY EP4 is the primary EP receptor in the ductus arteriosus (DA) and is critical during late gestation for its development and eventual closure. The "paradoxical" patent DA (PDA) phenotype of EP4-knockout mice arises from a combination of impaired contractile potential, altered signaling properties, and a failure to remodel associated with an underdeveloped immature vessel. These findings provide new mechanistic insights into women who receive NSAIDs to treat preterm labor, whose infants have unexplained PDA.


Sujet(s)
Persistance du canal artériel , Ligament artériel , Souris , Animaux , Nouveau-né , Femelle , Grossesse , Humains , Ligament artériel/métabolisme , Dinoprostone/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/génétique , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Persistance du canal artériel/génétique , Souris knockout
20.
Front Immunol ; 14: 1209572, 2023.
Article de Anglais | MEDLINE | ID: mdl-37457723

RÉSUMÉ

Introduction: For many years, surgery, adjuvant and combination chemotherapy have been the cornerstone of pancreatic cancer treatment. Although these approaches have improved patient survival, relapse remains a common occurrence, necessitating the exploration of novel therapeutic strategies. CAR T cell therapies are now showing tremendous success in hematological cancers. However, the clinical efficacy of CAR T cells in solid tumors remained low, notably due to presence of an immunosuppressive tumor microenvironment (TME). Prostaglandin E2, a bioactive lipid metabolite found within the TME, plays a significant role in promoting cancer progression by increasing tumor proliferation, improving angiogenesis, and impairing immune cell's function. Despite the well-established impact of PGE2 signaling on cancer, its specific effects on CAR T cell therapy remain under investigation. Methods: To address this gap in knowledge the role of PGE2-related genes in cancer tissue and T cells of pancreatic cancer patients were evaluated in-silico. Through our in vitro study, we manufactured fully human functional mesoCAR T cells specific for pancreatic cancer and investigated the influence of PGE2-EP2/EP4 signaling on proliferation, cytotoxicity, and cytokine production of mesoCAR T cells against pancreatic cancer cells. Results: In-silico investigations uncovered a significant negative correlation between PGE2 expression and gene signature of memory T cells. Furthermore, in vitro experiments demonstrated that the activation of PGE2 signaling through EP2 and EP4 receptors suppressed the proliferation and major antitumor functions of mesoCAR T cells. Interestingly, the dual blockade of EP2 and EP4 receptors effectively reversed PGE2-mediated suppression of mesoCAR T cells, while individual receptor antagonists failed to mitigate the PGE2-induced suppression. Discussion: In summary, our findings suggest that mitigating PGE2-EP2/EP4 signaling may be a viable strategy for enhancing CAR T cell activity within the challenging TME, thereby improving the efficacy of CAR T cell therapy in clinical settings.


Sujet(s)
Dinoprostone , Tumeurs du pancréas , Humains , Dinoprostone/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Récidive tumorale locale , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Tumeurs du pancréas/thérapie , Immunosuppression thérapeutique , Microenvironnement tumoral , Tumeurs du pancréas
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE