Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.665
Filtrer
1.
Nat Commun ; 15(1): 6538, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095358

RÉSUMÉ

Muscle invasive bladder cancers (BCs) can be divided into 2 major subgroups-basal/squamous (BASQ) tumors and luminal tumors. Since Pparg has low or undetectable expression in BASQ tumors, we tested the effects of rosiglitazone, Pparg agonist, in a mouse model of BASQ BC. We find that rosiglitazone reduces proliferation while treatment with rosiglitazone plus trametinib, a MEK inhibitor, induces apoptosis and reduces tumor volume by 91% after 1 month. Rosiglitazone and trametinib also induce a shift from BASQ to luminal differentiation in tumors, which our analysis suggests is mediated by retinoid signaling, a pathway known to drive the luminal differentiation program. Our data suggest that rosiglitazone, trametinib, and retinoids, which are all FDA approved, may be clinically active in BASQ tumors in patients.


Sujet(s)
Apoptose , Prolifération cellulaire , Modèles animaux de maladie humaine , Pyridones , Pyrimidinones , Rosiglitazone , Tumeurs de la vessie urinaire , Animaux , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Pyrimidinones/pharmacologie , Pyrimidinones/usage thérapeutique , Rosiglitazone/pharmacologie , Rosiglitazone/usage thérapeutique , Souris , Apoptose/effets des médicaments et des substances chimiques , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Invasion tumorale , Femelle , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/agonistes , Thiazolidinediones/pharmacologie , Thiazolidinediones/usage thérapeutique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Rétinoïdes/pharmacologie , Rétinoïdes/usage thérapeutique
2.
Acta Neurobiol Exp (Wars) ; 84(2): 153-164, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39087839

RÉSUMÉ

The aim of the present study was to evaluate the effect of rosiglitazone (RSG) or pioglitazone (POG) on the synaptic plasticity, neuronal apoptosis, brain-derived neurotrophic factor (BDNF), and nitric oxide (NO) metabolites in the hippocampus of juvenile hypothyroid rats. The animals were divided into four groups: control; propylthiouracil (PTU), 0.05% dose in drinking water for 42 days; PTU-POG; and PTU-RSG. The POG (20 mg/kg) and the RSG (4 mg/kg) were administered by IP injection. We conducted long­term potentiation (LTP) in the cornu ammonis 1 area of the hippocampus using high­frequency stimulation of the Schaffer collateral pathway. Then, the hippocampal tissues were collected to determine BDNF and NO levels and the degree of apoptosis. PTU administration decreased the slope (10-90%) and amplitude of the fEPSPs compared to control. Injection of RSG or POG increased the slope, slope (10-90%), and amplitude of the fEPSP in the PTU­POG or PTU­RSG groups compared to the PTU group. TUNEL­positive neurons and NO metabolites in the hippocampus of the PTU group were higher than those of the control group. RSG or POG increased BDNF content in PTU-POG or PTU-RSG groups. Treatment of the rats with POG or RSG decreased apoptotic neurons and NO metabolites in the hippocampus of PTU-POG or PTU-RSG groups, respectively, compared to the PTU group. This study's results revealed that POG or RSG normalized LTP impairment, neuronal apoptosis, and improved BDNF content in the hippocampal tissue of juvenile hypothyroid rats.


Sujet(s)
Apoptose , Facteur neurotrophique dérivé du cerveau , Hippocampe , Hypothyroïdie , Potentialisation à long terme , Récepteur PPAR gamma , Rat Wistar , Rosiglitazone , Animaux , Apoptose/effets des médicaments et des substances chimiques , Hypothyroïdie/traitement médicamenteux , Hypothyroïdie/induit chimiquement , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Mâle , Rosiglitazone/pharmacologie , Potentialisation à long terme/effets des médicaments et des substances chimiques , Récepteur PPAR gamma/agonistes , Récepteur PPAR gamma/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Thiazolidinediones/pharmacologie , Pioglitazone/pharmacologie , Rats , Propylthiouracile/pharmacologie , Modèles animaux de maladie humaine , Potentiels post-synaptiques excitateurs/effets des médicaments et des substances chimiques , Monoxyde d'azote/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme
3.
J Clin Invest ; 134(16)2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39145452

RÉSUMÉ

T cells rewire their metabolic activities to meet the demand of immune responses, but how to coordinate the immune response by metabolic regulators in activated T cells is unknown. Here, we identified autocrine VEGF-B as a metabolic regulator to control lipid synthesis and maintain the integrity of the mitochondrial inner membrane for the survival of activated T cells. Disruption of autocrine VEGF-B signaling in T cells reduced cardiolipin mass, resulting in mitochondrial damage, with increased apoptosis and reduced memory development. The addition of cardiolipin or modulating VEGF-B signaling improved T cell mitochondrial fitness and survival. Autocrine VEGF-B signaling through GA-binding protein α (GABPα) induced sentrin/SUMO-specific protease 2 (SENP2) expression, which further de-SUMOylated PPARγ and enhanced phospholipid synthesis, leading to a cardiolipin increase in activated T cells. These data suggest that autocrine VEGF-B mediates a signal to coordinate lipid synthesis and mitochondrial fitness with T cell activation for survival and immune response. Moreover, autocrine VEGF-B signaling in T cells provides a therapeutic target against infection and tumors as well as an avenue for the treatment of autoimmune diseases.


Sujet(s)
Communication autocrine , Cardiolipides , Mitochondries , Transduction du signal , Lymphocytes T , Facteur de croissance endothéliale vasculaire de type B , Mitochondries/métabolisme , Mitochondries/immunologie , Animaux , Souris , Communication autocrine/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Transduction du signal/immunologie , Cardiolipides/immunologie , Cardiolipides/métabolisme , Facteur de croissance endothéliale vasculaire de type B/génétique , Facteur de croissance endothéliale vasculaire de type B/métabolisme , Facteur de croissance endothéliale vasculaire de type B/immunologie , Activation des lymphocytes , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/immunologie , Récepteur PPAR gamma/génétique , Humains
4.
J Exp Clin Cancer Res ; 43(1): 227, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39148124

RÉSUMÉ

BACKGROUND: The failure of proper recognition of the intricate nature of pathophysiology in colorectal cancer (CRC) has a substantial effect on the progress of developing novel medications and targeted therapy approaches. Imbalances in the processes of lipid oxidation and biosynthesis of fatty acids are significant risk factors for the development of CRC. Therapeutic intervention that specifically targets the peroxisome proliferator-activated receptor gamma (PPARγ) and its downstream response element, in response to lipid metabolism, has been found to promote the growth of tumors and has shown significant clinical advantages in cancer patients. METHODS: Clinical CRC samples and extensive in vitro and in vivo experiments were carried out to determine the role of ZDHHC6 and its downstream targets via a series of biochemical assays, molecular analysis approaches and lipid metabolomics assay, etc. RESULTS: To study the effect of ZDHHC6 on the progression of CRC and identify whether ZDHHC6 is a palmitoyltransferase that regulates fatty acid synthesis, which directly palmitoylates and stabilizes PPARγ, and this stabilization in turn activates the ACLY transcription-related metabolic pathway. In this study, we demonstrate that PPARγ undergoes palmitoylation in its DNA binding domain (DBD) section. This lipid-related modification enhances the stability of PPARγ protein by preventing its destabilization. As a result, palmitoylated PPARγ inhibits its degradation induced by the lysosome and facilitates its translocation into the nucleus. In addition, we have identified zinc finger-aspartate-histidine-cysteine 6 (ZDHHC6) as a crucial controller of fatty acid biosynthesis. ZDHHC6 directly interacts with and adds palmitoyl groups to stabilize PPARγ at the Cys-313 site within the DBD domain of PPARγ. Consequently, this palmitoylation leads to an increase in the expression of ATP citrate lyase (ACLY). Furthermore, our findings reveals that ZDHHC6 actively stimulates the production of fatty acids and plays a role in the development of colorectal cancer. However, we have observed a significant reduction in the cancer-causing effects when the expression of ZDHHC6 is inhibited in in vivo trials. Significantly, in CRC, there is a strong positive correlation between the high expression of ZDHHC6 and the expression of PPARγ. Moreover, this high expression of ZDHHC6 is connected with the severity of CRC and is indicative of a poor prognosis. CONCLUSIONS: We have discovered a mechanism in which lipid biosynthesis is controlled by ZDHHC6 and includes the signaling of PPARγ-ACLY in the advancement of CRC. This finding provides a justification for targeting lipid synthesis by blocking ZDHHC6 as a potential therapeutic approach.


Sujet(s)
Acyltransferases , Récepteur PPAR gamma , Humains , Récepteur PPAR gamma/métabolisme , Souris , Animaux , Acyltransferases/métabolisme , Acyltransferases/génétique , Lipidomique/méthodes , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/génétique , Métabolisme lipidique , Mâle , Femelle , Carcinogenèse/métabolisme , Lignée cellulaire tumorale ,
5.
Eur J Med Chem ; 276: 116728, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-39089002

RÉSUMÉ

In consideration of several serious side effects induced by the classical AF-2 involved "lock" mechanism, recently disclosed PPARγ-Ser273 phosphorylation mode of action has become an alternative and mainstream mechanism for currently PPARγ-based drug discovery and development with an improved therapeutic index. In this study, by virtue of structure-based virtual high throughput screening (SB-VHTS), structurally chemical optimization by targeting the inhibition of the PPARγ-Ser273 phosphorylation as well as in vitro biological evaluation, which led to the final identification of a chrysin-based potential hit (YGT-31) as a novel selective PPARγ modulator with potent binding affinity and partial agonism. Further in vivo evaluation demonstrated that YGT-31 possessed potent glucose-lowering and relieved hepatic steatosis effects without involving the TZD-associated side effects. Mechanistically, YGT-31 presented such desired therapeutic index, mainly because it effectively inhibited the CDK5-mediated PPARγ-Ser273 phosphorylation, selectively elevated the level of insulin sensitivity-related Glut4 and adiponectin but decreased the expression of insulin-resistance-associated genes PTP1B and SOCS3 as well as inflammation-linked genes IL-6, IL-1ß and TNFα. Finally, the molecular docking study was also conducted to uncover an interesting hydrogen-bonding network of YGT-31 with PPARγ-Ser273 phosphorylation-related key residues Ser342 and Glu343, which not only gave a clear verification for our targeting modification but also provided a proof of concept for the abovementioned molecular mechanism.


Sujet(s)
Stéatose hépatique , Flavonoïdes , Récepteur PPAR gamma , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/agonistes , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Flavonoïdes/synthèse chimique , Relation structure-activité , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/métabolisme , Humains , Structure moléculaire , Diabète de type 2/traitement médicamenteux , Animaux , Hypoglycémiants/pharmacologie , Hypoglycémiants/composition chimique , Hypoglycémiants/synthèse chimique , Simulation de docking moléculaire , Relation dose-effet des médicaments , Souris , Mâle , Évaluation préclinique de médicament
6.
Sci Rep ; 14(1): 18390, 2024 08 08.
Article de Anglais | MEDLINE | ID: mdl-39117680

RÉSUMÉ

Ulcerative colitis (UC) is a chronic idiopathic inflammatory disease affecting the gastrointestinal tract. Although paeonol has been used for treating UC due to its anti-inflammatory and antioxidant effects, the underlying mechanisms remain unclear. In this study, we investigated the mechanisms of paeonol's action on UC by conducting in-vitro and in-vivo studies using NCM460 cells and RAW264.7 cells, and the DSS-induced mice colitis model. The in vitro studies demonstrate that paeonol exerts inhibitory effects on the activation of the NF-κB signaling pathway through upregulating PPARγ expression, thereby attenuating pro-inflammatory cytokine production, reducing reactive oxygen species levels, and promoting M2 macrophage polarization. These effects are significantly abrogated upon addition of the PPARγ inhibitor GW9662. Moreover, UC mice treated with paeonol showed increased PPARγ expression, which reduced inflammation and apoptosis to maintain intestinal epithelial barrier integrity. In conclusion, our findings suggest that paeonol inhibits the NF-κB signaling pathway by activating PPARγ, reducing inflammation and oxidative stress and improving Dss-induced colitis. This study provides a new insight into the mechanism of treating UC by paeonol.


Sujet(s)
Acétophénones , Rectocolite hémorragique , Facteur de transcription NF-kappa B , Récepteur PPAR gamma , Transduction du signal , Acétophénones/pharmacologie , Acétophénones/usage thérapeutique , Récepteur PPAR gamma/métabolisme , Animaux , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Humains , Cellules RAW 264.7 , Modèles animaux de maladie humaine , Mâle , Espèces réactives de l'oxygène/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Anti-inflammatoires/pharmacologie , Sulfate dextran/toxicité , Souris de lignée C57BL
7.
J Neuroinflammation ; 21(1): 194, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097742

RÉSUMÉ

Chronic neuroinflammation and microglial activation are key mediators of the secondary injury cascades and cognitive impairment that follow exposure to repetitive mild traumatic brain injury (r-mTBI). Peroxisome proliferator-activated receptor-γ (PPARγ) is expressed on microglia and brain resident myeloid cell types and their signaling plays a major anti-inflammatory role in modulating microglial responses. At chronic timepoints following injury, constitutive PPARγ signaling is thought to be dysregulated, thus releasing the inhibitory brakes on chronically activated microglia. Increasing evidence suggests that thiazolidinediones (TZDs), a class of compounds approved from the treatment of diabetes mellitus, effectively reduce neuroinflammation and chronic microglial activation by activating the peroxisome proliferator-activated receptor-γ (PPARγ). The present study used a closed-head r-mTBI model to investigate the influence of the TZD Pioglitazone on cognitive function and neuroinflammation in the aftermath of r-mTBI exposure. We revealed that Pioglitazone treatment attenuated spatial learning and memory impairments at 6 months post-injury and reduced the expression of reactive microglia and astrocyte markers in the cortex, hippocampus, and corpus callosum. We then examined whether Pioglitazone treatment altered inflammatory signaling mechanisms in isolated microglia and confirmed downregulation of proinflammatory transcription factors and cytokine levels. To further investigate microglial-specific mechanisms underlying PPARγ-mediated neuroprotection, we generated a novel tamoxifen-inducible microglial-specific PPARγ overexpression mouse line and examined its influence on microglial phenotype following injury. Using RNA sequencing, we revealed that PPARγ overexpression ameliorates microglial activation, promotes the activation of pathways associated with wound healing and tissue repair (such as: IL10, IL4 and NGF pathways), and inhibits the adoption of a disease-associated microglia-like (DAM-like) phenotype. This study provides insight into the role of PPARγ as a critical regulator of the neuroinflammatory cascade that follows r-mTBI in mice and demonstrates that the use of PPARγ agonists such as Pioglitazone and newer generation TZDs hold strong therapeutic potential to prevent the chronic neurodegenerative sequelae of r-mTBI.


Sujet(s)
Dysfonctionnement cognitif , Microglie , Récepteur PPAR gamma , Pioglitazone , Animaux , Mâle , Souris , Commotion de l'encéphale/métabolisme , Commotion de l'encéphale/traitement médicamenteux , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/traitement médicamenteux , Souris de lignée C57BL , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Pioglitazone/pharmacologie , Pioglitazone/usage thérapeutique , Récepteur PPAR gamma/métabolisme
8.
Clin Respir J ; 18(8): e13812, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39107932

RÉSUMÉ

BACKGROUND: Qi deficiency and phlegm dampness (QPD) is one of the most common traditional Chinese medicine (TCM) syndromes in lung adenocarcinoma (LUAD). This study aimed to identify syndrome-specific biomarkers for LUAD with QPD syndrome. METHODS: Peripheral blood mononuclear cells (PBMCs) from LUAD patients with QPD, LUAD patients with non-QPD (N-QPD), and healthy control (H) were collected and analyzed with RNA-seq to identify differentially expressed genes (DEGs). The area under the receiver operator characteristic curve (AUC) of each DEG was calculated, and the top 10 highest AUC DEGs were validated by qRT-PCR. Logistic regression analysis was used to develop a diagnostic model evaluated with AUC. RESULTS: A total of 135 individuals were enrolled in this study (training set: 15 QPD, 15 N-QPD, 15 H; validation set: 30 QPD, 30 N-QPD, 30 H). A total of 1480 DEGs were identified between QPD and N-QPD. The qRT-PCR results showed that the expression of DDR2 was downregulated, and PPARG was upregulated, which was in line with the finding of the training set. We developed a diagnostic model with these two genes. The AUC of the diagnostic model in the training cohort and validation cohort was 0.891 and 0.777, respectively. CONCLUSIONS: We identified the two genes (DDR2 and PPARG) as syndrome-specific biomarkers for LUAD with QPD syndrome and developed a novel diagnostic model, which may help to improve the accuracy and sensibility of clinical diagnosis and provide a new target for natural drug treatment of LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Marqueurs biologiques tumoraux , Tumeurs du poumon , Médecine traditionnelle chinoise , Humains , Mâle , Femelle , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/diagnostic , Tumeurs du poumon/génétique , Tumeurs du poumon/diagnostic , Médecine traditionnelle chinoise/méthodes , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/métabolisme , Récepteur PPAR gamma/génétique , Récepteur PPAR gamma/métabolisme , Sujet âgé , Qi , Agranulocytes/métabolisme , Courbe ROC , Études cas-témoins
9.
CNS Neurosci Ther ; 30(8): e14881, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39107960

RÉSUMÉ

BACKGROUND: Microglia and infiltrated macrophages (M/M) are integral components of the innate immune system that play a critical role in facilitating brain repair after ischemic stroke (IS) by clearing cell debris. Novel therapeutic strategies for IS therapy involve modulating M/M phenotype shifting. This study aims to elucidate the pivotal role of S100A9 in M/M and its downstream STAT6/PPARγ signaling pathway in neuroinflammation and phagocytosis after IS. METHODS: In the clinical study, we initially detected the expression pattern of S100A9 in monocytes from patients with acute IS and investigated its association with the long-term prognosis. In the in vivo study, we generated the S100A9 conditional knockout (CKO) mice and compared the stroke outcomes with the control group. We further tested the S100A9-specific inhibitor paqunimod (PQD), for its pharmaceutical effects on stroke outcomes. Transcriptomics and in vitro studies were adopted to explore the mechanism of S100A9 in modulating the M/M phenotype, which involves the regulation of the STAT6/PPARγ signaling pathway. RESULTS: S100A9 was predominantly expressed in classical monocytes and was correlated with unfavorable outcomes in patients of IS. S100A9 CKO mitigated infarction volume and white matter injury, enhanced cerebral blood flow and functional recovery, and prompted anti-inflammation phenotype and efferocytosis after tMCAO. The STAT6/PPARγ pathway, an essential signaling cascade involved in immune response and inflammation, might be the downstream target mediated by S100A9 deletion, as evidenced by the STAT6 phosphorylation inhibitor AS1517499 abolishing the beneficial effect of S100A9 inhibition in tMCAO mice and cell lines. Moreover, S100A9 inhibition by PQD treatment protected against neuronal death in vitro and brain injuries in vivo. CONCLUSION: This study provides evidence for the first time that S100A9 in classical monocytes could potentially be a biomarker for predicting IS prognosis and reveals a novel therapeutic strategy for IS. By demonstrating that S100A9-mediated M/M polarization and phagocytosis can be reversed by S100A9 inhibition in a STAT6/PPARγ pathway-dependent manner, this study opens up new avenues for drug development in the field.


Sujet(s)
Calgranuline B , Accident vasculaire cérébral ischémique , Macrophages , Souris knockout , Microglie , Récepteur PPAR gamma , Facteur de transcription STAT-6 , Transduction du signal , Animaux , Calgranuline B/génétique , Calgranuline B/métabolisme , Facteur de transcription STAT-6/métabolisme , Facteur de transcription STAT-6/déficit , Facteur de transcription STAT-6/génétique , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Souris , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Mâle , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Humains , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/génétique , Accident vasculaire cérébral ischémique/anatomopathologie , Transduction du signal/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Femelle , Adulte d'âge moyen , Sujet âgé
10.
PLoS One ; 19(8): e0306903, 2024.
Article de Anglais | MEDLINE | ID: mdl-39116155

RÉSUMÉ

Malva parviflora has shown anti-inflammatory, antihypertensive, antihyperlipidemic, and hypoglycemic effects. This study is aimed to evaluate the anti-adipogenic effect of M. parviflora on 3T3-L1 adipocytes. Fibroblast differentiation was induced either in the absence or presence of M. parviflora fractions (F3, F4, F7, F12, F13, F17, F18 and F19) for 4 days; F17 and 18 were the most effective fractions in reducing intracellular lipid accumulation (by 25.6% and 23.1%, respectively). EC50 of F17 and F18 (14 µg/mL and 17 µg/mL, respectively) were used to evaluate their anti adipogenic effect. After 10 days of inducing differentiation in the absence or presence of the extracts at the EC50 of F17 and F18, lipid accumulation, the concentration of interleukin 6 (IL-6) were measured in the culture medium; the presence of PPAR-γ, AKT, and p-AKT was also determined. In differentiated adipocytes (C2), F17 maintained intracellular lipid concentration at levels comparable to metformin, while decreasing PPAR-γ and increasing p-AKT presence; it also prevented IL-6 expression. F17 consists of alanine, valine, phenylalanine, and proline. On the other hand, F18 reduced intracellular lipid concentrations, prevented the increase of PPAR-γ and p-AKT, and maintained IL-6 expression at similar levels as metformin. F18 is mainly constituted by alanine, valine, proline, and sucrose. In conclusion, M. parviflora fractions (F17 and F18) control the process of adipogenesis, lipogenesis, and cellular dysfunction.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Récepteur PPAR gamma , Extraits de plantes , Animaux , Souris , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Adipocytes/cytologie , Adipogenèse/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Récepteur PPAR gamma/métabolisme , Interleukine-6/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme
11.
Nat Commun ; 15(1): 6622, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39103342

RÉSUMÉ

Sex steroids modulate the distribution of mammalian white adipose tissues. Moreover, WAT remodeling requires adipocyte progenitor cells. Nevertheless, the sex-dependent mechanisms regulating adipocyte progenitors remain undetermined. Here, we uncover Cxcr4 acting in a sexually dimorphic manner to affect a pool of proliferating cells leading to restriction of female fat mass. We find that deletion of Cxcr4 in Pparγ-expressing cells results in female, not male, lipodystrophy, which cannot be restored by high-fat diet consumption. Additionally, Cxcr4 deletion is associated with a loss of a pool of proliferating adipocyte progenitors. Cxcr4 loss is accompanied by the upregulation of estrogen receptor alpha in adipose-derived PPARγ-labelled cells related to estradiol hypersensitivity and stalled adipogenesis. Estrogen removal or administration of antiestrogens restores WAT accumulation and dynamics of adipose-derived cells in Cxcr4-deficient mice. These findings implicate Cxcr4 as a female adipogenic rheostat, which may inform strategies to target female adiposity.


Sujet(s)
Adipocytes , Adipogenèse , Adiposité , Récepteur PPAR gamma , Récepteurs CXCR4 , Cellules souches , Animaux , Récepteurs CXCR4/métabolisme , Récepteurs CXCR4/génétique , Femelle , Mâle , Souris , Adipocytes/métabolisme , Adipocytes/cytologie , Cellules souches/métabolisme , Cellules souches/cytologie , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Souris knockout , Tissu adipeux blanc/métabolisme , Tissu adipeux blanc/cytologie , Alimentation riche en graisse/effets indésirables , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Souris de lignée C57BL , Oestradiol/pharmacologie , Oestradiol/métabolisme , Prolifération cellulaire , Facteurs sexuels , Caractères sexuels
12.
Front Endocrinol (Lausanne) ; 15: 1401120, 2024.
Article de Anglais | MEDLINE | ID: mdl-39040675

RÉSUMÉ

Whilst western diet and sedentary lifestyles heavily contribute to the global obesity epidemic, it is likely that chemical exposure may also contribute. A substantial body of literature implicates a variety of suspected environmental chemicals in metabolic disruption and obesogenic mechanisms. Chemically induced obesogenic metabolic disruption is not yet considered in regulatory testing paradigms or regulations, but this is an internationally recognised human health regulatory development need. An early step in the development of relevant regulatory test methods is to derive appropriate minimum chemical selection lists for the target endpoint and its key mechanisms, such that the test method can be suitably optimised and validated. Independently collated and reviewed reference and proficiency chemicals relevant for the regulatory chemical universe that they are intended to serve, assist regulatory test method development and validation, particularly in relation to the OECD Test Guidelines Programme. To address obesogenic mechanisms and modes of action for chemical hazard assessment, key initiating mechanisms include molecular-level Peroxisome Proliferator-Activated Receptor (PPAR) α and γ agonism and the tissue/organ-level key event of perturbation of the adipogenesis process that may lead to excess white adipose tissue. Here we present a critical literature review, analysis and evaluation of chemicals suitable for the development, optimisation and validation of human PPARα and PPARγ agonism and human white adipose tissue adipogenesis test methods. The chemical lists have been derived with consideration of essential criteria needed for understanding the strengths and limitations of the test methods. With a weight of evidence approach, this has been combined with practical and applied aspects required for the integration and combination of relevant candidate test methods into test batteries, as part of an Integrated Approach to Testing and Assessment for metabolic disruption. The proposed proficiency and reference chemical list includes a long list of negatives and positives (20 chemicals for PPARα, 21 for PPARγ, and 11 for adipogenesis) from which a (pre-)validation proficiency chemicals list has been derived.


Sujet(s)
Adipogenèse , Obésité , Récepteur PPAR alpha , Récepteur PPAR gamma , Humains , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Adipogenèse/effets des médicaments et des substances chimiques , Obésité/métabolisme , Obésité/induit chimiquement , Activation de la transcription/effets des médicaments et des substances chimiques
13.
J Neuroimmune Pharmacol ; 19(1): 34, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38949694

RÉSUMÉ

Amorfrutin B is a selective PPARγ modulator that we demonstrated to be a promising neuroprotective compound in cellular models of stroke and perinatal asphyxia. Although neuronal mechanisms of amorfrutin B-evoked neuroprotection have been identified, none of them reflects the actions of the compound on microglia, which play a pivotal role in brain response to hypoxia/ischemia. Here, we provide evidence for amorfrutin B-induced effects on human microglia subjected to hypoxia/ischemia; the compound counteracts inflammation, and influences mitochondrial status and proliferation potential in a PPARγ-dependent manner. Post-treatment with amorfrutin B decreased the IBA1 fluorescence intensity, reduced caspase-1 activity, and downregulated IL1B/IL-1ß and TNFA but not IL10/IL-10 expression, which was upregulated. Amorfrutin B also stimulated PPARγ signaling, as evidenced by increased mRNA and/or protein levels of PPARγ and PGC1α. In addition, amorfrutin B reversed the hypoxia/ischemia-evoked effects on mitochondria-related parameters, such as mitochondrial membrane potential, BCL2/BCL2 expression and metabolic activity, which were correlated with diminished proliferation potential of microglia. Interestingly, the inhibitory effect of amorfrutin B on the proliferation potential and mitochondrial function of microglia is opposite to the stimulatory effect of amorfrutin B on mouse neuronal survival, as evidenced by increased neuronal viability and reduced neurodegeneration. In summary, this study showed for the first time that amorfrutin B compromises hypoxia/ischemia-induced activation of human microglia in a PPARγ-dependent manner, which involves inhibiting inflammation, normalizing mitochondrial status, and controlling proliferation potential. These data extend the protective potential of amorfrutin B in the pharmacotherapy of hypoxic/ischemic brain injury, targeting not only neurons but also activated microglia.


Sujet(s)
Prolifération cellulaire , Hypoxie-ischémie du cerveau , Microglie , Mitochondries , Récepteur PPAR gamma , Récepteur PPAR gamma/métabolisme , Humains , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Hypoxie-ischémie du cerveau/traitement médicamenteux , Hypoxie-ischémie du cerveau/métabolisme , Hypoxie-ischémie du cerveau/anatomopathologie , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Cellules cultivées , Neuroprotecteurs/pharmacologie
14.
Hereditas ; 161(1): 21, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38978149

RÉSUMÉ

PURPOSE: This study aims to reveal the relationship between AMIGO2 and proliferation, migration and tumorigenicity of bladder cancer, and explore the potential molecular mechanisms. METHODS: The expression level of AMIGO2 is measured by qRT-PCR and immunohistochemistry (IHC). Stable AMIGO2 knockdown cell lines T24 and 5637 were established by lentivirus transfection. Cell Counting Kit (CCK-8 assay) was produced to determine cell proliferation, flow cytometry analysis was utilized to detect cell cycle, and wound healing assay was proceeded to test migration ability of bladder cancer cells. Xenograft mouse model was established for investigating the effect of AMIGO2 on tumor formation in vivo. The RNA Sequencing technology was applied to explore the underlying mechanisms. The expression level of PPAR-γ was measured by Western Blot. RESULTS: AMIGO2 was upregulated in bladder cancer cells and tissues. Inhibited expression of AMIGO2 suppresses cell proliferation and migration. Low AMIGO2 expression inhibited tumorigenicity of 5637 in nude mice. According to RNA-Seq and bioinformatics analysis, 917 DEGs were identified. The DEGs were mainly enriched in cell-cell adhesion, peroxisome proliferators-activated receptors (PPARs) signaling pathway and some other pathways. PPAR-γ is highly expressed in bladder cancer cell lines T24 and 5637, but when AMIGO2 is knocked down in T24 and 5637, the expression level of PPAR-γ is also decreased, and overexpression of PPAR-γ could reverse the suppression effect of cell proliferation and migration caused by the inhibition of AMIGO2. CONCLUSION: AMIGO2 is overexpressed in bladder cancer cells and tissues. Knockdown of AMIGO2 suppresses bladder cancer cell proliferation and migration. These processes might be regulated by PPAR-γ signaling pathway.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Récepteur PPAR gamma , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/métabolisme , Récepteur PPAR gamma/génétique , Récepteur PPAR gamma/métabolisme , Humains , Animaux , Lignée cellulaire tumorale , Souris , Techniques de knock-down de gènes , Souris nude , Transduction du signal
15.
Cells ; 13(13)2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38994935

RÉSUMÉ

Successful pregnancy depends on precise molecular regulation of uterine physiology, especially during the menstrual cycle. Deregulated oxidative stress (OS), often influenced by inflammatory changes but also by environmental factors, represents a constant threat to this delicate balance. Oxidative stress induces a reciprocally regulated nuclear factor erythroid 2-related factor 2/peroxisome proliferator-activated receptor-gamma (Nrf2/PPARγ) pathway. However, increased PPARγ activity appears to be a double-edged sword in endometrial physiology. Activated PPARγ attenuates inflammation and attenuates OS to restore redox homeostasis. However, it also interferes with physiological processes during the menstrual cycle, such as hormonal signaling and angiogenesis. This review provides an elucidation of the molecular mechanisms that support the interplay between PPARγ and OS. Additionally, it offers fresh perspectives on the Nrf2/PPARγ pathway concerning endometrial receptivity and its potential implications for infertility.


Sujet(s)
Endomètre , Fécondité , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Récepteur PPAR gamma , Humains , Femelle , Facteur-2 apparenté à NF-E2/métabolisme , Endomètre/métabolisme , Récepteur PPAR gamma/métabolisme , Fécondité/physiologie , Transduction du signal , Animaux
16.
Nutrients ; 16(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38999751

RÉSUMÉ

To investigate the effects of rapeseed diacylglycerol oil (RDG) intake on lipid accumulation and metabolism in C57BL/6J mice, obese mice were fed a high-fat diet in which 45% of the total energy content came from RDG (RDGM group) or rapeseed triacylglycerol oil (RTGM group). This diet intervention was conducted for 12 weeks following the establishment of the obese mouse model. By the end of the experiment, the serum glucose levels of the mice in the RTGM and RDGM groups were 13.0 ± 1.3 mmol/L and 9.7 ± 1.5 mmol/L, respectively. Meanwhile, the serum triglyceride level in the RDGM group was 26.3% lower than that in the RTGM group. The weight-loss effect in the RDGM group was accompanied by a significant decrease in the white adipose tissue (WAT) index. The RDG intervention did not significantly change the antioxidant and anti-inflammatory properties of the rapeseed oil in vivo. The RDG diet improved the liver lipid metabolism abnormalities induced by a high-fat diet, leading to decreased liver damage index values (AST and ALT). Additionally, compared to that in the RTGM group, the expression of the adipogenic genes PPAR-γ and DGAT decreased in both the liver and intestine by 21.7% and 16.7% and by 38.7% and 47.2%, respectively, in the RDGM group. Further, most lipolytic genes in BAT showed no significant change after the RDG intervention. This implies that RDG regulates lipid metabolism by altering the expression of adipogenic genes in the liver, intestine, and adipose tissue, thereby reducing the accumulation of WAT. Furthermore, the RDG diet enhanced gut flora diversity, increasing the relative levels of unclassified Muribaculaceae and decreasing the levels of Dubosiella and Faecalibaculum in the mouse gut, potentially accelerating lipid metabolism. Thus, a three-month RDG diet intervention in obese mice exhibited benefits in regulating the somatotype, serum obesity-related indices, gut flora structure, and lipid metabolism in the adipose tissue, liver, and intestine.


Sujet(s)
Agents antiobésité , Alimentation riche en graisse , Diglycéride , Métabolisme lipidique , Foie , Souris de lignée C57BL , Obésité , Huile de colza , Animaux , Métabolisme lipidique/effets des médicaments et des substances chimiques , Obésité/métabolisme , Diglycéride/pharmacologie , Alimentation riche en graisse/effets indésirables , Mâle , Huile de colza/pharmacologie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Souris , Agents antiobésité/pharmacologie , Tissu adipeux blanc/métabolisme , Tissu adipeux blanc/effets des médicaments et des substances chimiques , Triglycéride/sang , Diacylglycerol O-acyltransferase/métabolisme , Diacylglycerol O-acyltransferase/génétique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Récepteur PPAR gamma/métabolisme , Souris obèse
17.
Cell Death Dis ; 15(7): 532, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39060229

RÉSUMÉ

Aquaporin 3 (AQP3), which is mostly expressed in pulmonary epithelial cells, was linked to lung adenocarcinoma (LUAD). However, the underlying functions and mechanisms of AQP3 in the tumor microenvironment (TME) of LUAD have not been elucidated. Single-cell RNA sequencing (scRNA-seq) was used to study the composition, lineage, and functional states of TME-infiltrating immune cells and discover AQP3-expressing subpopulations in five LUAD patients. Then the identifications of its function on TME were examined in vitro and in vivo. AQP3 was associated with TNM stages and lymph node metastasis of LUAD patients. We classified inter- and intra-tumor diversity of LUAD into twelve subpopulations using scRNA-seq analyses. The analysis showed AQP3 was mainly enriched in subpopulations of M2 macrophages. Importantly, mechanistic investigations indicated that AQP3 promoted M2 macrophage polarization by the PPAR-γ/NF-κB axis, which affected tumor growth and migration via modulating IL-6 production. Mixed subcutaneous transplanted tumor mice and Aqp3 knockout mice models were further utilized, and revealed that AQP3 played a critical role in mediating M2 macrophage polarization, modulating glucose metabolism in tumors, and regulating both upstream and downstream pathways. Overall, our study demonstrated that AQP3 could regulate the proliferation, migration, and glycometabolism of tumor cells by modulating M2 macrophages polarization through the PPAR-γ/NF-κB axis and IL-6/IL-6R signaling pathway, providing new insight into the early detection and potential therapeutic target of LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Aquaporine-3 , Interleukine-6 , Tumeurs du poumon , Macrophages , Facteur de transcription NF-kappa B , Récepteur PPAR gamma , Aquaporine-3/métabolisme , Aquaporine-3/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/génétique , Animaux , Interleukine-6/métabolisme , Humains , Récepteur PPAR gamma/métabolisme , Macrophages/métabolisme , Souris , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Facteur de transcription NF-kappa B/métabolisme , Microenvironnement tumoral , Évolution de la maladie , Régulation positive , Mâle , Transduction du signal , Lignée cellulaire tumorale , Souris knockout , Souris de lignée C57BL , Femelle
18.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39062809

RÉSUMÉ

The study evaluated the effects of Arthrospira maxima phycobiliproteins (PBPs), rosiglitazone (RSG), and 17ß-estradiol (E) on the differentiation process of 3T3-L1 cells and on their regulation of lipogenic and inflammatory gene expression at different stages of the process. The results showed that phycobiliproteins promoted cell proliferation after 24 h of treatment. Furthermore, for all three treatments, the regulation of the highest number of markers occurred on days 6 and 12 of differentiation, regardless of when the treatment was applied. Phycobiliproteins reduced lipid droplet accumulation on days 3, 6, 10, and 13 of the adipogenic process, while rosiglitazone showed no differences compared to the control. On day 6, both phycobiliproteins and rosiglitazone positively regulated Acc1 mRNA. Meanwhile, all three treatments negatively regulated Pparγ and C/ebpα. Phycobiliproteins and estradiol also negatively regulated Ucp1 and Glut4 mRNAs. Rosiglitazone and estradiol, on the other hand, negatively regulated Ppara and Il-6 mRNAs. By day 12, phycobiliproteins and rosiglitazone upregulated Pparγ mRNA and negatively regulated Tnfα and Il-1ß. Additionally, phycobiliproteins and estradiol positively regulated Il-6 and negatively regulated Ppara, Ucp2, Acc1, and Glut4. Rosiglitazone and estradiol upregulate C/ebpα and Ucp1 mRNAs. The regulation exerted by phycobiliproteins on the mRNA expression of the studied markers was dependent on the phase of cell differentiation. The results of this study highlight that phycobiliproteins have an anti-adipogenic and anti-inflammatory effect by reducing the expression of adipogenic, lipogenic, and inflammatory genes in 3T3-L1 cells at different stages of the differentiation process.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Différenciation cellulaire , Oestradiol , Phycobiliprotéines , Rosiglitazone , Animaux , Souris , Oestradiol/pharmacologie , Rosiglitazone/pharmacologie , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Adipocytes/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Adipogenèse/effets des médicaments et des substances chimiques , Adipogenèse/génétique , Phycobiliprotéines/pharmacologie , Phycobiliprotéines/métabolisme , Phycobiliprotéines/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Lipogenèse/effets des médicaments et des substances chimiques , Lipogenèse/génétique , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/génétique , Spirulina
19.
Biomed Pharmacother ; 177: 117073, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38981239

RÉSUMÉ

Adipogenesis is a process that differentiates new adipocytes from precursor cells and is tightly regulated by several factors, including many transcription factors and various post-translational modifications. Recently, new roles of adipogenesis have been suggested in various diseases. However, the molecular mechanisms and functional modulation of these adipogenic genes remain poorly understood. This review summarizes the regulatory factors and modulators of adipogenesis and discusses future research directions to identify novel mechanisms regulating adipogenesis and the effects of adipogenic regulators in pathological conditions. The master adipogenic transcriptional factors PPARγ and C/EBPα were identified along with other crucial regulatory factors such as SREBP, Kroxs, STAT5, Wnt, FOXO1, SWI/SNF, KLFs, and PARPs. These transcriptional factors regulate adipogenesis through specific mechanisms, depending on the adipogenic stage. However, further studies related to the in vivo role of newly discovered adipogenic regulators and their function in various diseases are needed to develop new potent therapeutic strategies for metabolic diseases and cancer.


Sujet(s)
Adipocytes , Adipogenèse , Adipogenèse/physiologie , Humains , Animaux , Adipocytes/métabolisme , Récepteur PPAR gamma/métabolisme , Facteurs de transcription/métabolisme , Régulation de l'expression des gènes
20.
J Chromatogr A ; 1730: 465141, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-38986402

RÉSUMÉ

Functional protein immobilization forms the basis for bio-detections. A series of one-point, site-specific immobilization methods have been developed, however, it still remains as a challenge how to avoid the proteins to move in all directions as well as conveniently regenerate the bio-devices. Herein, we have developed a bivalent affinity binding-inspired method for PPARγ immobilization using DNA aptamer and nickel-nitrilotriacetic acid (Ni2+-NTA) chelation. The specific DNA aptamer (Apt 2) was selected by an on-column systematic evolution of ligands by exponential enrichment (SELEX) method with affinity of (1.57 ± 0.15) × 105 M-1, determined by isothermal titration calorimetry (ITC). Apt 2 and nickel-nitrilotriacetic acid (Ni2+-NTA) were modified on macroporous silica gels via L-α-allylglycine as a linker. They respectively interacted with PPARγ and 6×His tag via bivalent affinity binding for the receptor immobilization. After comprehensive surface characterization, PPARγ was proved to be successful immobilized. Chromatographic studies revealed that the immobilized PPARγ has conformation selectivity, which discriminated agonist and antagonist of the receptor. Ligand-binding parameters (affinity and rate constant) of four agonists (rosiglitazone, pioglitazone, troglitazone, and magnolol) with PPARγ were determined. Troglitazone showed the lowest dissociation rate constant. The binding affinities (3.28 × 107, 1.91 × 106, 2.25 × 107, and 2.43 × 107 M-1) were highly consistent with the data obtained using purified receptor in solution (2.16 × 107, 4.52 × 106, 1.20 × 107, and 1.56 × 107 M-1), offering reliable bio-detection method for PPARγ and its ligands. Due to the biocompatibility of nuclear receptor with DNA, it is conceivable that the bivalent affinity-based method will be a general method for the immobilization of other nuclear receptors, which may provide selective conformation and improved ligand-binding activity for the receptors.


Sujet(s)
Aptamères nucléotidiques , Récepteur PPAR gamma , Récepteur PPAR gamma/composition chimique , Récepteur PPAR gamma/métabolisme , Ligands , Aptamères nucléotidiques/composition chimique , Aptamères nucléotidiques/métabolisme , Liaison aux protéines , Protéines immobilisées/composition chimique , Protéines immobilisées/métabolisme , Acide nitrilo-triacétique/composition chimique , Acide nitrilo-triacétique/analogues et dérivés , Humains , Calorimétrie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE