Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.300
Filtrer
1.
Nat Commun ; 15(1): 8308, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39333075

RÉSUMÉ

Intracellular infections by Gram-negative bacteria are a significant global health threat. The nuclear receptor Nur77 (also called TR3, NGFI-B, or NR4A1) was recently shown to sense cytosolic bacterial lipopolysaccharide (LPS). However, the potential role for Nur77 in controlling intracellular bacterial infection has not been examined. Here we show that Nur77 protects against intracellular infection in the bladder by uropathogenic Escherichia coli (UPEC), the leading cause of urinary tract infections (UTI). Nur77 deficiency in mice promotes the formation of UPEC intracellular bacterial communities (IBCs) in the cells lining the bladder lumen, leading to persistent infection in bladder tissue. Conversely, treatment with a small-molecule Nur77 agonist, cytosporone B, inhibits invasion and enhances the expulsion of UPEC from human urothelial cells in vitro, and significantly reduces UPEC IBC formation and bladder infection in mice. Our findings reveal a new role for Nur77 in control of bacterial infection and suggest that pharmacologic agonism of Nur77 function may represent a promising antibiotic-sparing therapeutic approach for UTI.


Sujet(s)
Infections à Escherichia coli , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Vessie urinaire , Infections urinaires , Escherichia coli uropathogène , Urothélium , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Animaux , Urothélium/microbiologie , Urothélium/métabolisme , Urothélium/effets des médicaments et des substances chimiques , Urothélium/anatomopathologie , Escherichia coli uropathogène/effets des médicaments et des substances chimiques , Escherichia coli uropathogène/pathogénicité , Humains , Infections urinaires/microbiologie , Vessie urinaire/microbiologie , Vessie urinaire/anatomopathologie , Vessie urinaire/métabolisme , Vessie urinaire/effets des médicaments et des substances chimiques , Infections à Escherichia coli/microbiologie , Infections à Escherichia coli/traitement médicamenteux , Infections à Escherichia coli/métabolisme , Souris , Femelle , Souris de lignée C57BL , Souris knockout , Phénylacétates
2.
Int J Biol Sci ; 20(11): 4458-4475, 2024.
Article de Anglais | MEDLINE | ID: mdl-39247823

RÉSUMÉ

This study investigated the mechanism by which NR4A1 regulates mitochondrial fission factor (Mff)-related mitochondrial fission and FUN14 domain 1 (FUNDC1)-mediated mitophagy following cardiac ischemia-reperfusion injury(I/R). Our findings showed that the damage regulation was positively correlated with the pathological fission and pan-apoptosis of myocardial cell mitochondria. Compared with wild-type mice (WT), NR4A1-knockout mice exhibited resistance to myocardial ischemia-reperfusion injury and mitochondrial pathological fission, characterized by mitophagy activation. Results showed that ischemia-reperfusion injury increased NR4A1 expression level, activating mitochondrial fission mediated by Mff and restoring the mitophagy phenotype mediated by FUNDC1. The inactivation of FUNDC1 phosphorylation could not mediate the normalization of mitophagy in a timely manner, leading to an excessive stress response of unfolded mitochondrial proteins and an imbalance in mitochondrial homeostasis. This process disrupted the normalization of the mitochondrial quality control network, leading to accumulation of damaged mitochondria and the activation of pan-apoptotic programs. Our data indicate that NR4A1 is a novel and critical target in myocardial I/R injury that exertsand negative regulatory effects by activating Mff-mediated mito-fission and inhibiting FUNDC1-mediated mitophagy. Targeting the crosstalk balance between NR4A1-Mff-FUNDC1 is a potential approach for treating I/R.


Sujet(s)
Souris knockout , Dynamique mitochondriale , Protéines mitochondriales , Mitophagie , Lésion de reperfusion myocardique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Animaux , Lésion de reperfusion myocardique/métabolisme , Souris , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Mâle , Souris de lignée C57BL , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Mitochondries/métabolisme , Apoptose , Mitochondries du myocarde/métabolisme
3.
Theranostics ; 14(15): 5809-5825, 2024.
Article de Anglais | MEDLINE | ID: mdl-39346541

RÉSUMÉ

Introduction: Ionizing radiation has been widely used in industry, medicine, military and agriculture. Radiation-induced skin injury is a significant concern in the context of radiotherapy and accidental exposure to radiation. The molecular changes at the single-cell level and intercellular communications during radiation-induced skin injury are not well understood. Methods: This study aims to illustrate this information in a murine model and human skin samples from a radiation accident using single-cell RNA sequencing (scRNA-Seq). We further characterize the functional significance of key molecule, which may provide a potential therapeutic target. ScRNA-Seq was performed on skin samples from a nuclear accident patient and rats exposed to ionizing radiation. Bioinformatic tools were used to analyze the cellular heterogeneity and preferential mRNAs. Comparative analysis was performed to identify dysregulated pathways, regulators, and ligand-receptor interactions in fibroblasts. The function of key molecule was validated in skin cells and in three mouse models of radiation-induced skin injury. Results: 11 clusters in human skin and 13 clusters of cells in rat skin were depicted respectively. Exposure to ionizing radiation caused changes in the cellular population (upregulation of fibroblasts and endothelial cells, downregulation of keratinocytes). Fibroblasts and keratinocytes possessed the most interaction pairs with other cell lineages. Among the five DEGs common to human and rat skins, Nur77 was highly expressed in fibroblasts, which mediated radiosensitivity by cell apoptosis and modulated crosstalk between macrophages, keratinocytes and endothelial cells in radiation-induced skin injury. In animal models, Nur77 knock-out mice (Nur77 -/-) showed more severe injury after radiation exposure than wild-type counterparts in three models of radiation-induced skin injury with complex mechanisms. Conclusion: The study reveals a single-cell transcriptional framework during radiation-induced skin injury, which provides a useful resource to uncover key events in its progression. Nur77 is a novel target in radiation-induced skin injury, which provides a potential therapeutic strategy against this disease.


Sujet(s)
Kératinocytes , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , RNA-Seq , Analyse sur cellule unique , Peau , Animaux , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Humains , Souris , Rats , Peau/effets des radiations , Peau/anatomopathologie , Peau/métabolisme , Peau/traumatismes , Kératinocytes/effets des radiations , Kératinocytes/métabolisme , Fibroblastes/effets des radiations , Fibroblastes/métabolisme , Mâle , Souris knockout , Rayonnement ionisant , Lésions radiques/génétique , Lésions radiques/anatomopathologie , Analyse de l'expression du gène de la cellule unique
4.
J Immunother Cancer ; 12(8)2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39151930

RÉSUMÉ

BACKGROUND: Antitumor effect of chimeric antigen receptor (CAR)-T cells against solid tumors is limited due to various factors, such as low infiltration rate, poor expansion capacity, and exhaustion of T cells within the tumor. NR4A transcription factors have been shown to play important roles in T-cell exhaustion in mice. However, the precise contribution of each NR4a factor to human T-cell differentiation remains to be clarified. METHODS: In this study, we deleted NR4A family factors, NR4A1, NR4A2, and NR4A3, in human CAR-T cells recognizing human epidermal growth factor receptor type 2 (HER2) by using the CRISPR/Cas9 system. We induced T-cell exhaustion in these cells in vitro through repeated co-culturing of CAR-T cells with Her2+A549 lung adenocarcinoma cells and evaluated cell surface markers such as memory and exhaustion phenotypes, proliferative capacity, cytokine production and metabolic activity. We validated the antitumor toxicity of NR4A1/2/3 triple knockout (TKO) CAR-T cells in vivo by transferring CAR-T cells into A549 tumor-bearing immunodeficient mice. RESULTS: Human NR4A-TKO CAR-T cells were resistant against exhaustion induced by repeated antigen stimulation in vitro, and maintained higher tumor-killing activity both in vitro and in vivo compared with control CAR-T cells. A comparison of the effectiveness of NR4A single, double, and TKOs demonstrated that triple KO was the most effective in avoiding exhaustion. Furthermore, a strong enhancement of antitumor effects by NR4A TKO was also observed in T cells from various donors including aged persons. Mechanistically, NR4A TKO CAR-T cells showed enhanced mitochondrial oxidative phosphorylation, therefore could persist for longer periods within the tumors. CONCLUSIONS: NR4A factors regulate CAR-T cell persistence and stemness through mitochondrial gene expression, therefore NR4A is a highly promising target for the generation of superior CAR-T cells against solid tumors.


Sujet(s)
Immunothérapie adoptive , Mitochondries , Récepteurs chimériques pour l'antigène , Humains , Animaux , Souris , Mitochondries/métabolisme , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Récepteurs des hormones thyroïdiennes/métabolisme , Récepteurs des hormones thyroïdiennes/génétique , Tumeurs/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Femelle , Protéines de liaison à l'ADN , Récepteurs aux stéroïdes
5.
Eur J Pharmacol ; 982: 176945, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-39182547

RÉSUMÉ

The nuclear receptor Nur77 is a transcription factor belonging to the NR4A subfamily. Upon activation, it regulates a wide array of biological and pathophysiological processes by modulating the expression of its target genes. Previous findings have classified Nur77 as an orphan receptor because of the discovery of a structurally atypical ligand-binding domain and the lack of identification of an endogenous ligand. Nevertheless, recent studies have uncovered several endogenous, natural, and small synthetic molecules that can bind to and activate Nur77. However, developing selective and potent Nur77 activators remains a significant challenge. The discovery of novel and potential small synthetic molecules that modulate Nur77 activity will facilitate therapeutic interventions of Nur77 against several human diseases. In this study, we have reported the development of a novel and effective Nur77 ligand. Our data show that (1E,4E)-1,5-di(pyrazin-2-yl)penta-1,4-dien-3-one (PB) induces Nur77 transcriptional activity by interacting with a putative Nur77 ligand binding site by forming solid hydrogen bonding. Calculated chemical parameters denote that PB has sophisticated chemical features that will enhance its interaction with the Nur77 ligand-binding domain. Molecular docking simulations showed that PB fits in the Nur77 putative ligand binding site with solid hydrogen bonding, and molecular dynamics simulations indicate that PB binding would stabilize the Nur77 ligand binding domain. Further, in vitro studies revealed that PB increases Nur77 nuclear expression and activity, inhibits cigarette smoke-induced inflammatory phenotype of airway epithelial cells, and protects against apoptosis. These findings provide insights into developing an effective Nur77 small-molecule activator which could be developed into a therapeutic agent against inflammatory diseases.


Sujet(s)
Simulation de docking moléculaire , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Humains , Ligands , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Animaux , Sites de fixation
6.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119813, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39142522

RÉSUMÉ

INTRODUCTION: Angiogenesis is closely related to renal fibrosis; however, its basic mechanism remains unclear. In our study, we found that nuclear receptor 4A1 (NR4A1) inhibits vascular endothelial growth factor A (VEGFA)-induced angiogenesis, ameliorating renal fibrosis. METHODS: We prepared a renal fibrosis animal model with unilateral ureteral obstruction (UUO) and NR4A1 knockdown UUO mice model, Using Human umbilical vein endothelial cells (HUVECs) to conduct all in vitro experiments. We then detected and analyzed the expression levels of NR4A1 and other genes related to angiogenesis and fibrosis. RESULTS: The angiogenesis related genes, such as VEGFA, vascular endothelial growth factor receptor-2 (VEGFR-2), endoglin (CD105), as well as the expression of fibrosis related genes that included, α-smooth muscle actin (α-SMA), Vimentin, and Collagen I are all significantly increased in the UUO rat model. In addition, the expression of NR4A1 of the kidney tissue of UUO rats was significantly reduced. Therefore, according to the above results, we speculated that angiogenesis may exacerbate renal fibrosis and NR4A1 may repress renal fibrosis by inhibiting angiogenesis. To further verify the above results, we used VEGFA to stimulate HUVECs with (or without) overexpression or knockdown of NR4A1. The results showed that with prolonged stimulation using VEGFA, the expression of NR4A1 decreases. Overexpression of NR4A1 significantly inhibits the expression of related indicators of angiogenesis and renal fibrosis. Furthermore, knockdown of NR4A1 induces endothelial cell proliferation and migration; therefore, exacerbating angiogenesis and fibrosis. Finally, the results of NR4A1 knockdown UUO mice showed that knockdown of NR4A1 can aggravating kidney damage and induce the expression of angiogenesis and renal fibrosis related indicators, while UUO can significantly induce kidney damage, angiogenesis and renal fibrosis. When knockdown of NR4A1, renal kidney damage, angiogenesis and fibrosis becomes more severe than UUO. Thus, all of these results indicate that NR4A1 can ameliorate renal fibrosis by inhibiting angiogenesis. CONCLUSIONS: NR4A1 can inhibit angiogenesis to ameliorate renal fibrosis.


Sujet(s)
Fibrose , Cellules endothéliales de la veine ombilicale humaine , Maladies du rein , Rein , Néovascularisation pathologique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Obstruction urétérale , Facteur de croissance endothéliale vasculaire de type A , Animaux , Humains , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Rats , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Obstruction urétérale/anatomopathologie , Obstruction urétérale/métabolisme , Obstruction urétérale/génétique , Obstruction urétérale/complications , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie , Rein/anatomopathologie , Rein/métabolisme , Rein/vascularisation , Maladies du rein/métabolisme , Maladies du rein/anatomopathologie , Maladies du rein/génétique , Mâle , Modèles animaux de maladie humaine , Souris , Rat Sprague-Dawley , Prolifération cellulaire ,
7.
Phytomedicine ; 133: 155925, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39173278

RÉSUMÉ

BACKGROUND: Nur77, an orphan member of the nuclear receptor superfamily, regulates inflammatory diseases and is a therapeutic target for treating inflammation. Phthalides in Angelica sinensis exhibit anti-inflammatory activity. PURPOSE: This study aimed to screen compounds from A. sinensis phthalide extract that could exert anti-inflammatory activity by targeting Nur77. To provide new theoretical support for better elucidation of Chinese medicine targeting mitochondria to achieve multiple clinical efficacies. METHODS: The anti-inflammatory capacity of phthalides was assessed in tumor necrosis factor-alpha (TNF-α)-stimulated HepG2 cells using western blotting. The interaction between phthalides and Nur77 was verified by molecular docking, surface plasmon resonance, and cellular thermal shift assay. Co-immunoprecipitation, western blotting, and immunostaining were performed to determine the molecular mechanisms. The in vivo anti-inflammatory activity of the phthalides was evaluated in a lipopolysaccharide (LPS)/d-galactosamine (d-GalN)-induced acute hepatitis and liver injury mouse model of acute hepatitis and liver injury. Finally, the toxicity of phthalide toxicity was assessed in zebrafish experiments. RESULTS: Among the 27 phthalide compounds isolated from A. sinensis, tokinolide B (TB) showed the best Nur77 binding capacity and, the best anti-inflammatory activity, which was induced without apoptosis. In vivo and in vitro experiments showed that TB promoted Nur77 translocation from the nucleus to the mitochondria and interacted with tumor necrosis factor receptor-associated factor 2 (TRAF2) and sequestosome 1 (p62) to induce mitophagy for anti-inflammatory functions. TB substantially inhibited LPS/d-GalN-induced acute hepatitis and liver injury in mice. TB also exhibited significantly lower toxicity than celastrol in zebrafish experiments. CONCLUSION: These findings suggested that TB inhibits inflammation by promoting Nur77 interaction with TRAF2 and p62, thereby inducing mitophagy. These findings offer promising directions for developing novel anti-inflammatory agents, enhance the understanding of phthalide compounds, and highlight the therapeutic potential of traditional Chinese herbs.


Sujet(s)
Angelica sinensis , Anti-inflammatoires , Benzofuranes , Simulation de docking moléculaire , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Danio zébré , Animaux , Angelica sinensis/composition chimique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Humains , Anti-inflammatoires/pharmacologie , Benzofuranes/pharmacologie , Souris , Cellules HepG2 , Mâle , Lipopolysaccharides , Facteur de nécrose tumorale alpha/métabolisme , Lésions hépatiques dues aux substances/traitement médicamenteux , Modèles animaux de maladie humaine
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(4): 1078-1084, 2024 Aug.
Article de Chinois | MEDLINE | ID: mdl-39192401

RÉSUMÉ

OBJECTIVE: To investigate the anti- chronic myelogenous leukemia (CML) activity of Nur77-specific agonist Csn-B combined with imatinib by promoting Nur77 expression, and explore the potential role of its signaling pathway. METHODS: Firstly, CCK-8 and Transwell assay were used to detect the inhibitory effects of Csn-B, imatinib, and their combination on the proliferation and migration of K562 cells. Furthermore, the apoptosis rate of K562 cells treated with Csn-B, imatinib, and their combination was detected by flow cytometry. The expression levels of Nur77, Pim-1, Drp1, p-Drp1 S616, Bcl-2 and Bax in K562 cells were detected by Western blot. Finally, the expression levels of reactive oxygen species (ROS) in K562 cells treated with Csn-B, imatinib and their combination were detected by immunofluorescence assay. RESULTS: The level of Nur77 in CML patients decreased significantly compared with normal population in dataset of GSE43754 (P < 0.001). Csn-B combined with imatinib could significantly inhibit the proliferation and migration of K562 cells (both P < 0.001), and induce apoptosis (P < 0.001). Csn-B promoted Nur77 expression in K562 cells, and synergistically enhanced imatinib sensitivity when combined with imatinib. Csn-B combined with imatinib could significantly enhanced ROS levels in K562 cells and mitochondria compared with single-drug treatment (both P < 0.001). CONCLUSION: Csn-B combined with imatinib can enhance ROS expression and induce apoptosis of K562 cells through Nur77/Pim-1/Drp1 pathway.


Sujet(s)
Apoptose , Prolifération cellulaire , Mésilate d'imatinib , Leucémie myéloïde chronique BCR-ABL positive , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Protéines proto-oncogènes c-pim-1 , Humains , Mésilate d'imatinib/pharmacologie , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Apoptose/effets des médicaments et des substances chimiques , Cellules K562 , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-pim-1/métabolisme , Dynamines , Transduction du signal , Espèces réactives de l'oxygène/métabolisme , Mouvement cellulaire
9.
Int Immunopharmacol ; 139: 112705, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39029235

RÉSUMÉ

Fibrosis is not a disease but rather an outcome of the pathological tissue repair response. Many myofibroblasts are activated which lead to the excessive accumulation of extracellular matrix components such as collagen and fibronectin with fibrosis. A variety of organs, including kidney, liver, lung, heart and skin, can undergo fibrosis under the stimulation of exogenous or endogenous pathogenic factors. The orphan nuclear receptor 4 group A1 (NR4A1) and nuclear receptor 4 group A2(NR4A2)are belong to the nuclear receptor subfamily and inhibit the occurrence and development of fibrosis. NR4A1 is an inhibitory factor of TGF-ß signaling transduction. Overexpression of NR4A1 in fibroblasts can reduce TGF-ß induced collagen deposition and fibrosis related gene expression. Here, we summarize the current research progress on the NR4A1/2 and fibrosis, providing reference for the treatment of fibrosis.


Sujet(s)
Fibrose , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires , Humains , Animaux , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie
10.
Reprod Biol Endocrinol ; 22(1): 86, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39044215

RÉSUMÉ

Reproductive aging not only affects the fertility and physical and mental health of women but also accelerates the aging process of other organs. There is an urgent need newfor novel mechanisms, targets, and drugs to break the vicious cycle of mitochondrial dysfunction, redox imbalance, and germ cell apoptosis associated with ovarian aging. Autophagy, recognized as a longevity mechanism, has recently become a focal point in anti-aging research. Although mitophagy is a type of autophagy, its role and regulatory mechanisms in ovarian aging, particularly in age-related ovarian function decline, remain unclear. Nerve growth factor inducible gene B (Nur77) is an early response gene that can be stimulated by oxidative stress, DNA damage, metabolism, and inflammation. Recent evidence recommends that decreased expression of Nur77 is associated with age-related myocardial fibrosis, renal dysfunction, and Parkinson's disease; however, its association with ovarian aging has not been studied yet. We herein identified Nur77 as a regulator of germ cell senescence, apoptosis, and mitophagy and found that overexpression of Nur77 can activate mitophagy, improve oxidative stress, reduce apoptosis, and ultimately enhance ovarian reserve in aged mice ovaries. Furthermore, we discovered an association between Nur77 and the AKT pathway through String and molecular docking analyses. Experimental confirmation revealed that the AKT/mTOR signaling pathway is involved in the regulation of Nur77 in ovarian function. In conclusion, our results suggest Nur77 as a promising target for preventing and treating ovarian function decline related to reproductive aging.


Sujet(s)
Vieillissement , Apoptose , Mitophagie , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Ovaire , Animaux , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Femelle , Mitophagie/physiologie , Souris , Apoptose/physiologie , Apoptose/génétique , Ovaire/métabolisme , Vieillissement/physiologie , Vieillissement/génétique , Stress oxydatif/physiologie , Transduction du signal/physiologie , Réserve ovarienne/physiologie , Reproduction/physiologie , Protéines proto-oncogènes c-akt/métabolisme , Souris de lignée C57BL
11.
J Immunol ; 213(4): 506-518, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38940624

RÉSUMÉ

Monocytes and macrophages (Mos/Mϕs) play diverse roles in wound healing by adopting a spectrum of functional phenotypes; however, the regulation of such heterogeneity remains poorly defined. We enhanced our previously published Bayesian inference TF activity model, incorporating both single-cell RNA sequencing and single-cell ATAC sequencing data to infer transcription factor (TF) activity in Mos/Mϕs during skin wound healing. We found that wound Mos/Mϕs clustered into early-stage Mos/Mϕs, late-stage Mϕs, and APCs, and that each cluster showed differential chromatin accessibility and differential predicted TF activity that did not always correlate with mRNA or protein expression. Network analysis revealed two highly connected large communities involving a total of 19 TFs, highlighting TF cooperation in regulating wound Mos/Mϕs. This analysis also revealed a small community populated by NR4A1 and NFKB1, supporting a proinflammatory link between these TFs. Importantly, we validated a proinflammatory role for NR4A1 activity during wound healing, showing that Nr4a1 knockout mice exhibit decreased inflammatory gene expression in early-stage wound Mos/Mϕs, along with delayed wound re-epithelialization and impaired granulation tissue formation. In summary, our study provides insight into TF activity that regulates Mo/Mϕ heterogeneity during wound healing and provides a rational basis for targeting Mo/Mϕ TF networks to alter phenotypes and improve healing.


Sujet(s)
Macrophages , Souris knockout , Peau , Cicatrisation de plaie , Animaux , Cicatrisation de plaie/génétique , Cicatrisation de plaie/immunologie , Macrophages/immunologie , Souris , Peau/immunologie , Peau/anatomopathologie , Peau/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Sous-unité p50 de NF-kappa B/génétique , Sous-unité p50 de NF-kappa B/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Souris de lignée C57BL , Monocytes/immunologie
12.
Nat Commun ; 15(1): 4757, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38834564

RÉSUMÉ

Semaglutide, a glucagon-like peptide-1 receptor agonist, is clinically used as a glucose-lowering and weight loss medication due to its effects on energy metabolism. In heart failure, energy production is impaired due to altered mitochondrial function and increased glycolysis. However, the impact of semaglutide on cardiomyocyte metabolism under pressure overload remains unclear. Here we demonstrate that semaglutide improves cardiac function and reduces hypertrophy and fibrosis in a mouse model of pressure overload-induced heart failure. Semaglutide preserves mitochondrial structure and function under chronic stress. Metabolomics reveals that semaglutide reduces mitochondrial damage, lipid accumulation, and ATP deficiency by promoting pyruvate entry into the tricarboxylic acid cycle and increasing fatty acid oxidation. Transcriptional analysis shows that semaglutide regulates myocardial energy metabolism through the Creb5/NR4a1 axis in the PI3K/AKT pathway, reducing NR4a1 expression and its translocation to mitochondria. NR4a1 knockdown ameliorates mitochondrial dysfunction and abnormal glucose and lipid metabolism in the heart. These findings suggest that semaglutide may be a therapeutic agent for improving cardiac remodeling by modulating energy metabolism.


Sujet(s)
Métabolisme énergétique , Peptides glucagon-like , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Animaux , Mâle , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Métabolisme énergétique/effets des médicaments et des substances chimiques , Souris , Peptides glucagon-like/pharmacologie , Peptides glucagon-like/usage thérapeutique , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Souris de lignée C57BL , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Modèles animaux de maladie humaine , Myocarde/métabolisme , Myocarde/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Cardiomégalie/traitement médicamenteux , Cardiomégalie/métabolisme
13.
Science ; 384(6700): eadh8697, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38843327

RÉSUMÉ

After antigen stimulation, naïve T cells display reproducible population-level responses, which arise from individual T cells pursuing specific differentiation trajectories. However, cell-intrinsic predeterminants controlling these single-cell decisions remain enigmatic. We found that the subcellular architectures of naïve CD8 T cells, defined by the presence (TØ) or absence (TO) of nuclear envelope invaginations, changed with maturation, activation, and differentiation. Upon T cell receptor (TCR) stimulation, naïve TØ cells displayed increased expression of the early-response gene Nr4a1, dependent upon heightened calcium entry. Subsequently, in vitro differentiation revealed that TØ cells generated effector-like cells more so compared with TO cells, which proliferated less and preferentially adopted a memory-precursor phenotype. These data suggest that cellular architecture may be a predeterminant of naïve CD8 T cell fate.


Sujet(s)
Lymphocytes T CD8+ , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Récepteurs aux antigènes des cellules T , Animaux , Souris , Calcium/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/ultrastructure , Différenciation cellulaire , Mémoire immunologique , Activation des lymphocytes , Souris de lignée C57BL , Enveloppe nucléaire/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Microscopie de fluorescence , Technique d'immunofluorescence , Humains
14.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 233-237, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836656

RÉSUMÉ

Nur77 is a member of the NR4A subfamily of orphan nuclear receptors that is expressed and has a function within the immune system. This study aimed to investigate the role of Nur77 in hypoxic pulmonary hypertension. SPF male SD rats were exposed in hypobaric chamber simulating 5000 m high altitude for 0, 3, 7, 14, 21 or 28 days. Rat pulmonary artery smooth muscle cells (RPASMCs) were cultured under normoxic conditions (5% CO2-95% ambient air) or hypoxic conditions (5% O2 for 6 h, 12 h, 24 h, 48 h). Hypoxic rats developed pulmonary arterial remodeling and right ventricular hypertrophy with significantly increased pulmonary arterial pressure. The levels of Nur77, HIF-1α and PNCA were upregulated in pulmonary arterial smooth muscle from hypoxic rats. Silencing of either Nur77 or HIF-1α attenuated hypoxia-induced proliferation. Silencing of HIF-1α down-regulated Nur77 protein level, but Nur77 silence did not reduce HIF-1α. Nur77 was not con-immunoprecipitated with HIF-1α. This study demonstrated that Nur77 acted as a downstream regulator of HIF-1α under hypoxia, and plays a critical role in the hypoxia-induced pulmonary vascular remodeling, which is regulated by HIF-1α. Nur77 maybe a novel target of HPH therapy.


Sujet(s)
Hypertension pulmonaire , Sous-unité alpha du facteur-1 induit par l'hypoxie , Hypoxie , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Artère pulmonaire , Rat Sprague-Dawley , Remodelage vasculaire , Animaux , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Remodelage vasculaire/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Mâle , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/génétique , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Hypoxie/métabolisme , Prolifération cellulaire , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Rats , Hypertrophie ventriculaire droite/métabolisme , Hypertrophie ventriculaire droite/anatomopathologie , Hypertrophie ventriculaire droite/physiopathologie , Hypertrophie ventriculaire droite/génétique , Cellules cultivées
15.
Cell Rep ; 43(6): 114301, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38823016

RÉSUMÉ

CD8+ T cells are rendered exhausted in tumor and chronic infection. Among heterogeneous exhausted T cells, a subpopulation of progenitor-like (Tpex) cells have been found important for long-term tumor or pathogen control and are also the main responders in immunotherapy. Using an RFP reporter mouse for the orphan nuclear receptor NR4A1, originally characterized as critical in T cell dysfunction, we discover that the reporter is highly expressed in Tpex cells in tumor and chronic infection. Enforced expression of Nr4a1 promotes Tpex cell accumulation, whereas tumor control is improved after Nr4a1 deletion, associated with increased effector function but decreased long-term maintenance of CD8+ T cells. Integrating chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq) analysis, NR4A1 is found to bind and promote the expression of Tpex-related genes, as well as suppress terminal differentiation-associated genes. This study therefore has identified a key role of NR4A1 in Tpex regulation and provides a promising target for immunotherapy.


Sujet(s)
Lymphocytes T CD8+ , Différenciation cellulaire , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Microenvironnement tumoral , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Souris , Microenvironnement tumoral/immunologie , Souris de lignée C57BL , Transcription génétique , Cellules souches/métabolisme , Humains
16.
Eur J Pharmacol ; 977: 176697, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38823760

RÉSUMÉ

Gastric cancer (GC) remains a global challenge due to the lack of early detection and precision therapies. Genkwadaphnin (DD1), a natural diterpene isolated from the bud of Flos GenkWa (Thymelaeaceae), serves as a Karyopherin ß1 (KPNB1) inhibitor. In this study, we investigated the anti-tumor effect of DD1 in both cell culture and animal models. Our findings reveal that KPNB1, a protein involved in nuclear import, was highly expressed in GC tissues and associated with a poor prognosis in patients. We demonstrated that DD1, alongside the established KPNB1 inhibitor importazole (IPZ), inhibited GC cell proliferation and tumor growth by enhancing both genomic and non-genomic activity of Nur77. DD1 and IPZ reduced the interaction between KPNB1 and Nur77, resulting in Nur77 cytoplasmic accumulation and triggering mitochondrial apoptosis. The inhibitors also increased the expression of the Nur77 target apoptotic genes ATF3, RB1CC1 and PMAIP1, inducing apoptosis in GC cell. More importantly, loss of Nur77 effectively rescued the inhibitory effect of DD1 and IPZ on GC cells in both in vitro and in vivo experiments. In this study, we for the first time explored the relationship between KPNB1 and Nur77, and found KPNB1 inhibition could significantly increase the expression of Nur77. Moreover, we investigated the function of KPNB1 in GC for the first time, and the results suggested that KPNB1 could be a potential target for cancer therapy, and DD1 might be a prospective therapeutic candidate.


Sujet(s)
Apoptose , Prolifération cellulaire , Diterpènes , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Transduction du signal , Tumeurs de l'estomac , Caryophérines bêta , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme , Humains , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Animaux , Diterpènes/pharmacologie , Diterpènes/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Souris , Caryophérines bêta/métabolisme , Caryophérines bêta/génétique , Évolution de la maladie , Mâle , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Femelle , Souris de lignée BALB C
17.
FEBS Lett ; 598(14): 1715-1729, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38825601

RÉSUMÉ

Mitochondrial biogenesis requires precise regulation of both mitochondrial-encoded and nuclear-encoded genes. Nuclear receptor Nur77 is known to regulate mitochondrial metabolism in macrophages and skeletal muscle. Here, we compared genome-wide Nur77 binding site and target gene expression in these two cell types, which revealed conserved regulation of mitochondrial genes and enrichment of motifs for the transcription factor Yin-Yang 1 (YY1). We show that Nur77 and YY1 interact, that YY1 increases Nur77 activity, and that their binding sites are co-enriched at mitochondrial ribosomal protein gene loci in macrophages. Nur77 and YY1 co-expression synergistically increases Mrpl1 expression as well as mitochondrial abundance and activity in macrophages but not skeletal muscle. As such, we identify a macrophage-specific Nur77-YY1 interaction that enhances mitochondrial metabolism.


Sujet(s)
Macrophages , Mitochondries , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Facteur de transcription YY1 , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Macrophages/métabolisme , Animaux , Mitochondries/métabolisme , Mitochondries/génétique , Souris , Facteur de transcription YY1/métabolisme , Facteur de transcription YY1/génétique , Humains , Sites de fixation , Régulation de l'expression des gènes , Protéines de transport de la membrane mitochondriale/métabolisme , Protéines de transport de la membrane mitochondriale/génétique , Liaison aux protéines , Muscles squelettiques/métabolisme , Muscles squelettiques/cytologie , Protéines ribosomiques/métabolisme , Protéines ribosomiques/génétique
18.
Biol Reprod ; 111(3): 640-654, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-38936833

RÉSUMÉ

Nuclear receptor NR4A1 is a key factor in glycolipid metabolism and steroidogenesis, while lipid droplets serve as crucial dynamic organelles for lipid metabolism in luteal cells. To investigate the effects of NR4A1 on lipid droplet metabolism and progesterone (P4) synthesis in goat corpus luteum in vitro, luteal cells from the middle-cyclic corpus luteum were isolated and treated with Cytosporone B (CSNB, an agonist) or siRNA of NR4A1. Results showed that both low (1 µM) and high (50 µM) concentrations of CSNB promoted lipid droplet accumulation, while NR4A1 knockdown reduced lipid droplet content. CSNB increased while siNR4A1 decreased total cholesterol content; however, CSNB and siNR4A1 did not change triglyceride content. CSNB increased the expression of perilipins at mRNA and protein levels, also increased LDLR, SCARB1, SREBFs, and HMGCR mRNA abundance. Treatment with siNR4A1 revealed opposite results of CSNB, except for HMCGR and SREBF2. For steroidogenesis, 1 µM CSNB increased, but 50 µM CSNB inhibited P4 synthesis, NR4A1 knockdown also reduced the P4 level. Further analysis demonstrated that 1 µM CSNB increased the protein levels of StAR, HSD3B, and P-HSL, while 50 µM CSNB decreased StAR, HSD3B, and CYP11A1 protein levels. Moreover, 50 µM CSNB impaired active mitochondria, reduced the BCL2, and increased DRP1, Caspase 3, and cleaved-Caspase 3 protein levels. siNR4A1 consistently downregulated the P-HSL/HSL ratio and the steroidogenic protein levels. In conclusion, NR4A1-mediated lipid droplets are involved in the regulation of progesterone synthesis in goat luteal cells.


Sujet(s)
Capra , Gouttelettes lipidiques , Cellules lutéales , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Progestérone , Animaux , Femelle , Progestérone/métabolisme , Progestérone/biosynthèse , Cellules lutéales/métabolisme , Cellules lutéales/effets des médicaments et des substances chimiques , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Gouttelettes lipidiques/métabolisme , Métabolisme lipidique/physiologie , Cellules cultivées
19.
Pancreas ; 53(9): e760-e773, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38710022

RÉSUMÉ

OBJECTIVES: Diabetes secondary to chronic pancreatitis (CP) presents clinical challenges due to lack of understanding on factor(s) triggering insulin secretory defects. Therefore, we aimed to delineate the molecular mechanism of ß-cell dysfunction in CP. MATERIALS AND METHODS: Transcriptomic analysis was conducted to identify endocrine-specific receptor expression in mice and human CP on microarray. The identified receptor (NR4A1) was overexpressed in MIN6 cells using PEI linear transfection. RNA-Seq analysis of NR4A1-overexpressed (OE) MIN6 cells on NovaSeq6000 identified aberrant metabolic pathways. Upstream trigger for NR4A1OE was studied by InBio Discover and cytokine exposure, whereas downstream effect was examined by Fura2 AM-based fluorimetric and imaging studies. Mice with CP were treated with IFN-γ-neutralizing monoclonal antibodies to assess NR4A1 expression and insulin secretion. RESULTS: Increased expression of NR4A1 associated with decreased insulin secretion in islets (humans: controls 9 ± 0.2, CP 3.7 ± 0.2, mice: controls 8.5 ± 0.2, CP 2.1 ± 0.1 µg/L). NR4A1OE in MIN6 cells (13.2 ± 0.1) showed reduction in insulin secretion (13 ± 5 to 0.2 ± 0.1 µg/mg protein per minute, P = 0.001) and downregulation of calcium and cAMP signaling pathways. IFN-γ was identified as upstream signal for NR4A1OE in MIN6. Mice treated with IFN-γ-neutralizing antibodies showed decreased NR4A1 expression 3.4 ± 0.11-fold ( P = 0.03), showed improved insulin secretion (4.4 ± 0.2-fold, P = 0.01), and associated with increased Ca 2+ levels (2.39 ± 0.06-fold, P = 0.009). CONCLUSIONS: Modulating NR4A1 expression can be a promising therapeutic strategy to improve insulin secretion in CP.


Sujet(s)
Modèles animaux de maladie humaine , Sécrétion d'insuline , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Pancréatite chronique , Animaux , Pancréatite chronique/métabolisme , Pancréatite chronique/génétique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Humains , Souris , Mâle , Cellules à insuline/métabolisme , Souris de lignée C57BL , Insuline/métabolisme , Interféron gamma/métabolisme , Lignée cellulaire
20.
Neuron ; 112(12): 2031-2044.e7, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38754414

RÉSUMÉ

The patterns of synaptic connectivity and physiological properties of diverse neuron types are shaped by distinct gene sets. Our study demonstrates that, in the mouse forebrain, the transcriptional profiles of inhibitory GABAergic interneurons are regulated by Nr4a1, an orphan nuclear receptor whose expression is transiently induced by sensory experiences and is required for normal learning. Nr4a1 exerts contrasting effects on the local axonal wiring of parvalbumin- and somatostatin-positive interneurons, which innervate different subcellular domains of their postsynaptic partners. The loss of Nr4a1 activity in these interneurons results in bidirectional, cell-type-specific transcriptional switches across multiple gene families, including those involved in surface adhesion and repulsion. Our findings reveal that combinatorial synaptic organizing codes are surprisingly flexible and highlight a mechanism by which inducible transcription factors can influence neural circuit structure and function.


Sujet(s)
Neurones GABAergiques , Interneurones , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Animaux , Interneurones/métabolisme , Neurones GABAergiques/métabolisme , Neurones GABAergiques/physiologie , Souris , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Somatostatine/métabolisme , Somatostatine/génétique , Parvalbumines/métabolisme , Souris knockout , Mâle , Synapses/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE