Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.394
Filtrer
1.
Article de Chinois | MEDLINE | ID: mdl-38964904

RÉSUMÉ

Objective: To investigate the mechanism of Sulfo-N-succinimidyloleate (SSO) regulating lipid metabolism disorder induced by silicon dioxide (SiO(2)) . Methods: In March 2023, Rat alveolar macrophages NR8383 were cultured in vitro and randomly divided into control group (C), SSO exposure group (SSO), SiO(2) exposure group (SiO(2)) and SiO(2)+SSO exposure group (SiO(2)+SSO). NR8383 cells were exposure separately or jointly by SSO and SiO(2) for 36 h to construct cell models. Immunofluorescence and BODIPY 493/ 503 staining were used to detect cluster of differentiation (CD36) and intracellular lipid levels, the protein expression levels of CD36, liver X receptors (LXR), P-mammalian target of rapamycin (P-mTOR) and cholinephosphotransferase 1 (CHPT1) were detected by Western blot, respectively, and lipid metabolomics was used to screen for different lipid metabolites and enrichment pathways. Single-factor ANOVA was used for multi-group comparison, and LSD test was used for pair-to-group comparison. Results: SiO(2) caused the expression of CD36 and P-mTOR to increase (P=0.012, 0.020), the expression of LXR to decrease (P=0.005), and the intracellular lipid level to increase. After SSO treatment, CD36 expression decreased (P=0.023) and LXR expression increased (P=0.000) in SiO(2)+SSO exposure group compared with SiO(2) exposure group. Metabolomics identified 87 different metabolites in the C group and SiO(2) exposure group, 19 different metabolites in the SiO(2) exposure group and SiO(2)+SSO group, and 5 overlaps of different metabolites in the two comparison groups, they are PS (22∶1/14∶0), DG (O-16∶0/18∶0/0∶0), PGP (i-13∶0/i-20∶0), PC (18∶3/16∶0), and Sphinganine. In addition, the differential metabolites of the two comparison groups were mainly concentrated in the glycerophospholipid metabolism and sphingolipid metabolism pathways. The differential gene CHPT1 in glycerophospholipid metabolic pathway was verified, and the expression of CHPT1 decreased after SiO(2) exposure. Conclusion: SSO may improve SiO(2)-induced lipid metabolism disorders by regulating PS (22∶1/14∶0), DG (O-16∶0/18∶0/0∶0), PGP (i-13∶0/i-20∶0), PC (18∶3/16∶0), SPA, glycerophospholipid metabolism and sphingolipid metabolism pathways.


Sujet(s)
Antigènes CD36 , Métabolisme lipidique , Silice , Animaux , Rats , Silice/toxicité , Métabolisme lipidique/effets des médicaments et des substances chimiques , Antigènes CD36/métabolisme , Métabolomique , Troubles du métabolisme lipidique/métabolisme , Troubles du métabolisme lipidique/induit chimiquement , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Récepteurs hépatiques X/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Lipides
2.
J Agric Food Chem ; 72(29): 16287-16297, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-38986018

RÉSUMÉ

Variances in the biological functions of astaxanthin geometric isomers (i.e., all-E, Z) are related to their intestinal absorption, but the mechanism of isomer absorption mediated by transporters remains unclear. Here, models of in vitro cell overexpression, in situ intestinal perfusion, and in vivo mouse inhibition were employed to investigate the impact of cluster of differentiation 36 (CD36) on the absorption of astaxanthin isomers. Cells overexpressing CD36 notably enhanced the uptake of Z-astaxanthin, particularly the 9-Z-isomer (47.76%). The absorption rate and permeability of Z-astaxanthin surpassed that of the all-E-isomer by the in situ model. Furthermore, the addition of the CD36-specific inhibitor sulfo-N-succinimidyl oleate significantly reduced the absorption of Z-astaxanthin in the mouse duodenum and jejunum, especially the 9-Z-isomer (57.66%). Molecular docking and surface plasmon resonance techniques further validated that 9-Z-astaxanthin binds to more amino acids of CD36 with higher affinity and in a fast-binding, fast-dissociating mode, thus favoring transport. Our findings elucidate, for the first time, the mechanism of the CD36-mediated transmembrane transport of astaxanthin geometric isomers.


Sujet(s)
Antigènes CD36 , Absorption intestinale , Simulation de docking moléculaire , Xanthophylles , Xanthophylles/métabolisme , Xanthophylles/composition chimique , Animaux , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Souris , Absorption intestinale/effets des médicaments et des substances chimiques , Mâle , Humains , Isomérie , Souris de lignée C57BL , Jéjunum/métabolisme , Liaison aux protéines
3.
Acta Biochim Pol ; 71: 13004, 2024.
Article de Anglais | MEDLINE | ID: mdl-39041003

RÉSUMÉ

CD36 is a type 2 cell surface scavenger receptor expressed in various tissues. In macrophages, CD36 recognizes oxidized low-density lipoprotein (ox-LDL), which promotes the formation of foam cells, the first step toward an atherosclerotic arterial lesion. CD36 possesses a variety of posttranslational modifications, among them N-glycosylation and O-GlcNAc modification. Some of the roles of these modifications on CD36 are known, such as N-linked glycosylation, which provides proper folding and trafficking to the plasma membrane in the human embryonic kidney. This study aimed to determine whether variations in the availability of UDP-GlcNAc could impact Rab-5-mediated endocytic trafficking and, therefore, the cellular localization of CD36. These preliminary results suggest that the availability of the substrate UDP-GlcNAc, modulated in response to treatment with Thiamet G (TMG), OSMI-1 (O-GlcNAcylation enzymes modulators) or Azaserine (HBP modulator), influences the localization of CD36 in J774 macrophages, and the endocytic trafficking as evidenced by the regulatory protein Rab-5, between the plasma membrane and the cytoplasm.


Sujet(s)
Antigènes CD36 , Macrophages , Antigènes CD36/métabolisme , Macrophages/métabolisme , Animaux , Souris , Lignée cellulaire , Glycosylation , Membrane cellulaire/métabolisme , Humains , Lipoprotéines LDL/métabolisme , Hexosamine/métabolisme , Hexosamine/biosynthèse , Protéines G rab5/métabolisme , Transport des protéines , Voies de biosynthèse , Maturation post-traductionnelle des protéines
4.
Eur J Pharmacol ; 977: 176745, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38880220

RÉSUMÉ

High fat diet (HFD) consumption can cause dysregulation of glucose and lipid metabolism, coupled with increased ectopic lipid deposition in renal tissue leading to steatosis and dysfunction. Sitagliptin is a dipeptidyl peptidase-4 (DPP-4) inhibitor clinically used for type II diabetes therapy; however its effect on renal steatosis in obese state is still uncertain. Herein, obesity was induced by feeding male Wistar rats HFD for 18 weeks, thereafter received either drug vehicle, or sitagliptin (10 mg/kg, PO) along with HFD for further 6 weeks and compared with age-matched rats receiving normal chow diet (NCD). After 24 weeks, serum and kidneys were collected for histological and biochemical assessments. Compared to NCD-fed group, HFD-fed rats displayed marked weight gain, increased fat mass, insulin resistance, dyslipidemia, impaired kidney functions and renal histological alterations. Sitagliptin effectively ameliorated obesity and related metabolic perturbations and improved kidney architecture and function. There were increased levels of triglycerides and cluster of differentiation 36 (CD36) in kidneys of obese rats, that were lowered by sitagliptin therapy. Sitagliptin significantly repressed the expression of lipogenesis genes, while up-regulated genes involved in mitochondrial biogenesis and fatty acid oxidation in kidneys of HFD-fed rats. Sitagliptin was found to induce down-regulation of endoplasmic reticulum (ER) stress and apoptotic markers in kidneys of obese rats. These findings together may emphasize a novel concept that sitagliptin can be an effective therapeutic approach for halting obesity-related renal steatosis and CKD.


Sujet(s)
Antigènes CD36 , Alimentation riche en graisse , Stress du réticulum endoplasmique , Rein , Obésité , Transduction du signal , Phosphate de sitagliptine , Animaux , Mâle , Rats , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Alimentation riche en graisse/effets indésirables , Inhibiteurs de la dipeptidyl-peptidase IV/pharmacologie , Inhibiteurs de la dipeptidyl-peptidase IV/usage thérapeutique , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Maladies du rein/traitement médicamenteux , Maladies du rein/métabolisme , Maladies du rein/étiologie , Maladies du rein/anatomopathologie , Maladies du rein/prévention et contrôle , Obésité/traitement médicamenteux , Obésité/métabolisme , Obésité/complications , Rat Wistar , Transduction du signal/effets des médicaments et des substances chimiques , Phosphate de sitagliptine/pharmacologie , Phosphate de sitagliptine/usage thérapeutique
5.
Sci Rep ; 14(1): 14723, 2024 06 26.
Article de Anglais | MEDLINE | ID: mdl-38926392

RÉSUMÉ

Invasive candidiasis (IC) is a notable healthcare-associated fungal infection, characterized by high morbidity, mortality, and substantial treatment costs. Candida albicans emerges as a principal pathogen in this context. Recent academic advancements have shed light on the critical role of exosomes in key biological processes, such as immune responses and antigen presentation. This burgeoning body of research underscores the potential of exosomes in the realm of medical diagnostics and therapeutics, particularly in relation to fungal infections like IC. The exploration of exosomal functions in the pathophysiology of IC not only enhances our understanding of the disease but also opens new avenues for innovative therapeutic interventions. In this investigation, we focus on exosomes (Exos) secreted by macrophages, both uninfected and those infected with C. albicans. Our objective is to extract and analyze these exosomes, delving into the nuances of their protein compositions and subgroups. To achieve this, we employ an innovative technique known as Proximity Barcoding Assay (PBA). This methodology is pivotal in our quest to identify novel biological targets, which could significantly enhance the diagnostic and therapeutic approaches for C. albicans infection. The comparative analysis of exosomal contents from these two distinct cellular states promises to yield insightful data, potentially leading to breakthroughs in understanding and treating this invasive fungal infection. In our study, we analyzed differentially expressed proteins in exosomes from macrophages and C. albicans -infected macrophages, focusing on proteins such as ACE2, CD36, CAV1, LAMP2, CD27, and MPO. We also examined exosome subpopulations, finding a dominant expression of MPO in the most prevalent subgroup, and a distinct expression of CD36 in cluster14. These findings are crucial for understanding the host response to C. albicans and may inform targeted diagnostic and therapeutic approaches. Our study leads us to infer that MPO and CD36 proteins may play roles in the immune escape mechanisms of C. albicans. Additionally, the CD36 exosome subpopulations, identified through our analysis, could serve as potential biomarkers and therapeutic targets for C. albicans infection. This insight opens new avenues for understanding the infection's pathology and developing targeted treatments.


Sujet(s)
Marqueurs biologiques , Antigènes CD36 , Candida albicans , Candidose , Exosomes , Macrophages , Exosomes/métabolisme , Marqueurs biologiques/métabolisme , Macrophages/métabolisme , Macrophages/microbiologie , Macrophages/immunologie , Antigènes CD36/métabolisme , Candidose/diagnostic , Candidose/microbiologie , Candidose/métabolisme , Candidose/immunologie , Humains , Animaux , Souris
6.
BMC Cancer ; 24(1): 768, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926671

RÉSUMÉ

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a 'difficult-to-treat' entity. To forecast its prognosis, we introduced a new biomarker, SARIFA (stroma areactive invasion front areas), which are areas at the tumour invasion front lacking desmoplastic stroma reaction upon malignant invasion in the surrounding tissue, leading to direct contact between tumour cells and adipocytes. SARIFA showed its significance in gastric and colorectal carcinoma, revealing lipid metabolism alternations that promote tumour progression. METHODS: We reviewed the SARIFA status of 166 PDAC cases on all available H&E-stained tumour slides from archival Whipple-resection specimens. SARIFA positivity was defined as SARIFA detection in at least 66% of the available slides. To investigate alterations in tumour metabolism and microenvironment, we performed immunohistochemical staining for FABP4, CD36 and CD68. To verify and quantify a supposed delipidation of adipocytes, adipose tissue was digitally morphometrised. RESULTS: In total, 53 cases (32%) were classified as SARIFA positive and 113 (68%) as SARIFA negative. Patients with SARIFA-positive PDAC showed a significantly worse overall survival compared with SARIFA-negative cases (median overall survival: 11.0 months vs. 22.0 months, HR: 1.570 (1.082-2.278), 95% CI, p = 0.018), which was independent from other prognostic markers (p = 0.014). At the invasion front of SARIFA-positive PDAC, we observed significantly higher expression of FABP4 (p < 0.0001) and higher concentrations of CD68+ macrophages (p = 0.031) related to a higher risk of tumour progression. CD36 staining showed no significant expression differences. The adipocyte areas at the invasion front were significantly smaller, with mean values of 4021 ± 1058 µm2 and 1812 ± 1008 µm2 for the SARIFA-negative and -positive cases, respectively (p < 0.001). CONCLUSIONS: SARIFA is a promising prognostic biomarker for PDAC. Its assessment is characterised by simplicity and low effort. The mechanisms behind SARIFA suggest a tumour-promoting increased lipid metabolism and altered immune background, both showing new therapeutic avenues.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome du canal pancréatique , Protéines de liaison aux acides gras , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/mortalité , Femelle , Mâle , Marqueurs biologiques tumoraux/métabolisme , Pronostic , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/mortalité , Sujet âgé , Adulte d'âge moyen , Protéines de liaison aux acides gras/métabolisme , Invasion tumorale , Microenvironnement tumoral , Métabolisme lipidique , Antigènes de différenciation des myélomonocytes/métabolisme , Antigènes CD/métabolisme , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie , Antigènes CD36/métabolisme , Adipocytes/métabolisme , Adipocytes/anatomopathologie , Adulte , Sujet âgé de 80 ans ou plus ,
7.
EBioMedicine ; 105: 105198, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38889480

RÉSUMÉ

BACKGROUND: Disease susceptibility and progression of Mycobacterium avium complex pulmonary disease (MAC-PD) is associated with multiple factors, including low body mass index (BMI). However, the specific impact of low BMI on MAC-PD progression remains poorly understood. This study aims to examine the progression of MAC-PD in the context of low BMI, utilising a disease-resistant mouse model. METHODS: We employed a MAC infection-resistant female A/J mouse model to compare the progression of MAC-PD under two dietary conditions: one group was fed a standard protein diet, representing protein-energy unrestricted conditions, and the other was fed a low protein diet (LPD), representing protein-energy restriction. FINDINGS: Our results reveal that protein-energy restriction significantly exacerbates MAC-PD progression by disrupting lipid metabolism. Mice fed an LPD showed elevated fatty acid levels and related gene expressions in lung tissues, similar to findings of increased fatty acids in the serum of patients who exhibited the MAC-PD progression. These mice also exhibited increased CD36 expression and lipid accumulation in macrophages upon MAC infection. In vitro experiments emphasised the crucial role of CD36-mediated palmitic acid uptake in bacterial proliferation. Importantly, in vivo studies demonstrated that administering anti-CD36 antibody to LPD-fed A/J mice reduced macrophage lipid accumulation and impeded bacterial growth, resulting in remarkable slowing disease progression. INTERPRETATION: Our findings indicate that the metabolic status of host immune cells critically influences MAC-PD progression. This study highlights the potential of adequate nutrient intake in preventing MAC-PD progression, suggesting that targeting CD36-mediated pathways might be a host-directed therapeutic strategy to managing MAC infection. FUNDING: This research was funded by the National Research Foundation of Korea, the Korea Research Institute of Bioscience and Biotechnology, and the Korea National Institute of Health.


Sujet(s)
Modèles animaux de maladie humaine , Évolution de la maladie , Métabolisme lipidique , Infection due à Mycobacterium avium-intracellulare , Animaux , Femelle , Souris , Infection due à Mycobacterium avium-intracellulare/microbiologie , Infection due à Mycobacterium avium-intracellulare/métabolisme , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Macrophages/métabolisme , Humains , Complexe Mycobacterium avium , Poumon/métabolisme , Poumon/microbiologie , Poumon/anatomopathologie , Acides gras/métabolisme , Mycobacterium avium , Prédisposition aux maladies
8.
Cell Rep Med ; 5(6): 101592, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38843841

RÉSUMÉ

Environmental lipids are essential for fueling tumor energetics, but whether these exogenous lipids transported into cancer cells facilitate immune escape remains unclear. Here, we find that CD36, a transporter for exogenous lipids, promotes acute myeloid leukemia (AML) immune evasion. We show that, separately from its established role in lipid oxidation, CD36 on AML cells senses oxidized low-density lipoprotein (OxLDL) to prime the TLR4-LYN-MYD88-nuclear factor κB (NF-κB) pathway, and exogenous palmitate transfer via CD36 further potentiates this innate immune pathway by supporting ZDHHC6-mediated MYD88 palmitoylation. Subsequently, NF-κB drives the expression of immunosuppressive genes that inhibit anti-tumor T cell responses. Notably, high-fat-diet or hypomethylating agent decitabine treatment boosts the immunosuppressive potential of AML cells by hijacking CD36-dependent innate immune signaling, leading to a dampened therapeutic effect. This work is of translational interest because lipid restriction by US Food and Drug Administration (FDA)-approved lipid-lowering statin drugs improves the efficacy of decitabine therapy by weakening leukemic CD36-mediated immunosuppression.


Sujet(s)
Antigènes CD36 , Décitabine , Leucémie aigüe myéloïde , Métabolisme lipidique , Lipoprotéines LDL , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Décitabine/pharmacologie , Décitabine/usage thérapeutique , Lipoprotéines LDL/métabolisme , Animaux , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire tumorale , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/métabolisme , Acyltransferases/génétique , Immunité innée/effets des médicaments et des substances chimiques , Souris de lignée C57BL
9.
Nat Commun ; 15(1): 4814, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862469

RÉSUMÉ

A detailed understanding of how spaceflight affects human health is essential for long-term space exploration. Liquid biopsies allow for minimally-invasive multi-omics assessments that can resolve the molecular heterogeneity of internal tissues. Here, we report initial results from the JAXA Cell-Free Epigenome Study, a liquid biopsy study with six astronauts who resided on the International Space Station (ISS) for more than 120 days. Analysis of plasma cell-free RNA (cfRNA) collected before, during, and after spaceflight confirms previously reported mitochondrial dysregulation in space. Screening with 361 cell surface marker antibodies identifies a mitochondrial DNA-enriched fraction associated with the scavenger receptor CD36. RNA-sequencing of the CD36 fraction reveals tissue-enriched RNA species, suggesting the plasma mitochondrial components originated from various tissues. We compare our plasma cfRNA data to mouse plasma cfRNA data from a previous JAXA mission, which had used on-board artificial gravity, and discover a link between microgravity and the observed mitochondrial responses.


Sujet(s)
Antigènes CD36 , Acides nucléiques acellulaires , ADN mitochondrial , Vol spatial , Impesanteur , ADN mitochondrial/génétique , ADN mitochondrial/sang , Humains , Acides nucléiques acellulaires/sang , Animaux , Souris , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Mitochondries/métabolisme , Mitochondries/génétique , Mâle , Astronaute , ARN/métabolisme , ARN/génétique , Biopsie liquide/méthodes , ARN mitochondrial/métabolisme , ARN mitochondrial/génétique , Femelle , Adulte d'âge moyen , Adulte
10.
Front Immunol ; 15: 1413947, 2024.
Article de Anglais | MEDLINE | ID: mdl-38881887

RÉSUMÉ

CD36 is a scavenger receptor that has been reported to function as a signaling receptor that responds to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) and could integrate metabolic pathways and cell signaling through its dual functions. Thereby influencing activation to regulate the immune response and immune cell differentiation. Recent studies have revealed that CD36 plays critical roles in the process of lipid metabolism, inflammatory response and immune process caused by Mycobacterium tuberculosis infection. This review will comprehensively investigate CD36's functions in lipid uptake and processing, inflammatory response, immune response and therapeutic targets and biomarkers in the infection process of M. tuberculosis. The study also raised outstanding issues in this field to designate future directions.


Sujet(s)
Antigènes CD36 , Mycobacterium tuberculosis , Tuberculose , Humains , Antigènes CD36/métabolisme , Mycobacterium tuberculosis/immunologie , Tuberculose/immunologie , Tuberculose/métabolisme , Tuberculose/microbiologie , Animaux , Métabolisme lipidique , Transduction du signal , Marqueurs biologiques , Interactions hôte-pathogène/immunologie
11.
J Neuroinflammation ; 21(1): 148, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38840180

RÉSUMÉ

BACKGROUND: White matter injury (WMI) represents a significant etiological factor contributing to neurological impairment subsequent to Traumatic Brain Injury (TBI). CD36 receptors are recognized as pivotal participants in the pathogenesis of neurological disorders, including stroke and spinal cord injury. Furthermore, dynamic fluctuations in the phenotypic polarization of microglial cells have been intimately associated with the regenerative processes within the injured tissue following TBI. Nevertheless, there is a paucity of research addressing the impact of CD36 receptors on WMI and microglial polarization. This investigation aims to elucidate the functional role and mechanistic underpinnings of CD36 in modulating microglial polarization and WMI following TBI. METHODS: TBI models were induced in murine subjects via controlled cortical impact (CCI). The spatiotemporal patterns of CD36 expression were examined through quantitative polymerase chain reaction (qPCR), Western blot analysis, and immunofluorescence staining. The extent of white matter injury was assessed via transmission electron microscopy, Luxol Fast Blue (LFB) staining, and immunofluorescence staining. Transcriptome sequencing was employed to dissect the molecular mechanisms underlying CD36 down-regulation and its influence on white matter damage. Microglial polarization status was ascertained using qPCR, Western blot analysis, and immunofluorescence staining. In vitro, a Transwell co-culture system was employed to investigate the impact of CD36-dependent microglial polarization on oligodendrocytes subjected to oxygen-glucose deprivation (OGD). RESULTS: Western blot and qPCR analyses revealed that CD36 expression reached its zenith at 7 days post-TBI and remained sustained at this level thereafter. Immunofluorescence staining exhibited robust CD36 expression in astrocytes and microglia following TBI. Genetic deletion of CD36 ameliorated TBI-induced white matter injury, as evidenced by a reduced SMI-32/MBP ratio and G-ratio. Transcriptome sequencing unveiled differentially expressed genes enriched in processes linked to microglial activation, regulation of neuroinflammation, and the TNF signaling pathway. Additionally, bioinformatics analysis pinpointed the Traf5-p38 axis as a critical signaling pathway. In vivo and in vitro experiments indicated that inhibition of the CD36-Traf5-MAPK axis curtailed microglial polarization toward the pro-inflammatory phenotype. In a Transwell co-culture system, BV2 cells treated with LPS + IFN-γ exacerbated the damage of post-OGD oligodendrocytes, which could be rectified through CD36 knockdown in BV2 cells. CONCLUSIONS: This study illuminates that the suppression of CD36 mitigates WMI by constraining microglial polarization towards the pro-inflammatory phenotype through the down-regulation of the Traf5-MAPK signaling pathway. Our findings present a potential therapeutic strategy for averting neuroinflammatory responses and ensuing WMI damage resulting from TBI.


Sujet(s)
Antigènes CD36 , Souris de lignée C57BL , Microglie , Animaux , Microglie/métabolisme , Microglie/anatomopathologie , Souris , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Souris knockout , Substance blanche/anatomopathologie , Substance blanche/métabolisme , Système de signalisation des MAP kinases/physiologie , Mâle , Polarité de la cellule/physiologie , Lésions traumatiques de l'encéphale/métabolisme , Lésions traumatiques de l'encéphale/anatomopathologie , Transduction du signal/physiologie
12.
BMC Cancer ; 24(1): 710, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858612

RÉSUMÉ

BACKGROUND: Papillary thyroid carcinoma (PTC) is globally prevalent and associated with an increased risk of lymph node metastasis (LNM). The role of cancer-associated fibroblasts (CAFs) in PTC remains unclear. METHODS: We collected postoperative pathological hematoxylin-eosin (HE) slides from 984 included patients with PTC to analyze the density of CAF infiltration at the invasive front of the tumor using QuPath software. The relationship between CAF density and LNM was assessed. Single-cell RNA sequencing (scRNA-seq) data from GSE193581 and GSE184362 datasets were integrated to analyze CAF infiltration in PTC. A comprehensive suite of in vitro experiments, encompassing EdU labeling, wound scratch assays, Transwell assays, and flow cytometry, were conducted to elucidate the regulatory role of CD36+CAF in two PTC cell lines, TPC1 and K1. RESULTS: A significant correlation was observed between high fibrosis density at the invasive front of the tumor and LNM. Analysis of scRNA-seq data revealed metastasis-associated myoCAFs with robust intercellular interactions. A diagnostic model based on metastasis-associated myoCAF genes was established and refined through deep learning methods. CD36 positive expression in CAFs can significantly promote the proliferation, migration, and invasion abilities of PTC cells, while inhibiting the apoptosis of PTC cells. CONCLUSION: This study addresses the significant issue of LNM risk in PTC. Analysis of postoperative HE pathological slides from a substantial patient cohort reveals a notable association between high fibrosis density at the invasive front of the tumor and LNM. Integration of scRNA-seq data comprehensively analyzes CAF infiltration in PTC, identifying metastasis-associated myoCAFs with strong intercellular interactions. In vitro experimental results indicate that CD36 positive expression in CAFs plays a promoting role in the progression of PTC. Overall, these findings provide crucial insights into the function of CAF subset in PTC metastasis.


Sujet(s)
Fibroblastes associés au cancer , Analyse sur cellule unique , Cancer papillaire de la thyroïde , Tumeurs de la thyroïde , Humains , Cancer papillaire de la thyroïde/anatomopathologie , Cancer papillaire de la thyroïde/génétique , Cancer papillaire de la thyroïde/métabolisme , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Analyse sur cellule unique/méthodes , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/métabolisme , Prolifération cellulaire , Mâle , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Mouvement cellulaire , Femelle , Lignée cellulaire tumorale , Métastase lymphatique , Invasion tumorale , Adulte d'âge moyen , Apoptose
13.
Mol Cell Biol ; 44(7): 261-272, 2024.
Article de Anglais | MEDLINE | ID: mdl-38828991

RÉSUMÉ

The protein tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP-1) plays an important role in modulating glucose and lipid homeostasis. We previously suggested a potential role of SHP-1 in the regulation of peroxisome proliferator-activated receptor γ2 (PPARγ2) expression and activity but the mechanisms were unexplored. PPARγ2 is the master regulator of adipogenesis, but how its activity is regulated by tyrosine phosphorylation is largely unknown. Here, we found that SHP-1 binds to PPARγ2 primarily via its N-terminal SH2-domain. We confirmed the phosphorylation of PPARγ2 on tyrosine-residue 78 (Y78), which was reduced by SHP-1 in vitro resulting in decreased PPARγ2 stability. Loss of SHP-1 led to elevated, agonist-induced expression of the classical PPARγ2 targets FABP4 and CD36, concomitant with increased lipid content in cells expressing PPARγ2, an effect blunted by abrogation of PPARγ2 phosphorylation. Collectively, we discovered that SHP-1 affects the stability of PPARγ2 through dephosphorylation thereby influencing adipogenesis.


Sujet(s)
Adipogenèse , Récepteur PPAR gamma , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Récepteur PPAR gamma/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 6/métabolisme , Phosphorylation , Humains , Animaux , Souris , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Cellules HEK293 , Protéines de liaison aux acides gras/métabolisme , Protéines de liaison aux acides gras/génétique , Stabilité protéique , Cellules 3T3-L1 , Domaine d'homologie SRC , Liaison aux protéines
14.
Obesity (Silver Spring) ; 32(7): 1349-1361, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38816990

RÉSUMÉ

OBJECTIVE: Metabolic dysfunction-associated steatotic liver disease (MASLD) is becoming an escalating health problem in pediatric populations. This study aimed to investigate the role of N-acetyltransferase 10 (NAT10) in maternal high-fat diet (HFD)-induced MASLD in offspring at early life. METHODS: We generated male hepatocyte-specific NAT10 knockout (Nat10HKO) mice and mated them with female Nat10fl/fl mice under chow or HFD feeding. Body weight, liver histopathology, and expression of lipid metabolism-associated genes (Srebp1c, Fasn, Pparα, Cd36, Fatp2, Mttp, and Apob) were assessed in male offspring at weaning. Lipid uptake assays were performed both in vivo and in vitro. The mRNA stability assessment and RNA immunoprecipitation were performed to determine NAT10-regulated target genes. RESULTS: NAT10 deletion in hepatocytes of male offspring alleviated perinatal lipid accumulation induced by maternal HFD, decreasing expression levels of Srebp1c, Fasn, Cd36, Fatp2, Mttp, and Apob while enhancing Pparα expression. Furthermore, Nat10HKO male mice exhibited reduced lipid uptake. In vitro, NAT10 promoted lipid uptake by enhancing the mRNA stability of CD36 and FATP2. RNA immunoprecipitation assays exhibited direct interactions between NAT10 and CD36/FATP2 mRNA. CONCLUSIONS: NAT10 deletion in offspring hepatocytes ameliorates maternal HFD-induced hepatic steatosis through decreasing mRNA stability of CD36 and FATP2, highlighting NAT10 as a potential therapeutic target for pediatric MASLD.


Sujet(s)
Alimentation riche en graisse , Stéatose hépatique , Hépatocytes , Métabolisme lipidique , Foie , Souris knockout , Animaux , Alimentation riche en graisse/effets indésirables , Mâle , Femelle , Souris , Grossesse , Foie/métabolisme , Foie/anatomopathologie , Hépatocytes/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/métabolisme , Acetyltransferases/génétique , Acetyltransferases/métabolisme , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Effets différés de l'exposition prénatale à des facteurs de risque , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Souris de lignée C57BL , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie
15.
Biosci Biotechnol Biochem ; 88(7): 759-767, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38719485

RÉSUMÉ

Our previous studies have demonstrated that Mito-Tempol (also known as 4-hydroxy-Tempo), a mitochondrial reactive oxygen species scavenger, alleviates oxidized low-density lipoprotein (ox-LDL)-triggered foam cell formation. Given the effect of oxidative stress on activating the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome, which promotes foam cell formation, we aimed to explore whether Mito-Tempo inhibits ox-LDL-triggered foam cell formation by regulating NLRP3 inflammasome. The results revealed that Mito-Tempo re-activated Nrf2 and alleviated macrophage foam cell formation induced by ox-LDL, whereas the effects were reversed by ML385 (a specific Nrf2 inhibitor). Mito-Tempo restored the expression and nuclear translocation of Nrf2 by decreasing ox-LDL-induced ubiquitination. Furthermore, Mito-Tempo suppressed ox-LDL-triggered NLRP3 inflammasome activation and subsequent pyroptosis, whereas the changes were blocked by ML385. Mito-Tempo decreased lipoprotein uptake by inhibiting CD36 expression and suppressed foam cell formation by regulating the NLRP3 inflammasome. Taken together, Mito-Tempo exhibits potent anti-atherosclerotic effects by regulating Nrf2/NLRP3 signaling.


Sujet(s)
Cellules spumeuses , Lipoprotéines LDL , Facteur-2 apparenté à NF-E2 , Protéine-3 de la famille des NLR contenant un domaine pyrine , Transduction du signal , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Lipoprotéines LDL/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Cellules spumeuses/effets des médicaments et des substances chimiques , Cellules spumeuses/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Animaux , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Pyroptose/effets des médicaments et des substances chimiques , Humains , Cellules RAW 264.7 , N-oxydes cycliques/pharmacologie , Antigènes CD36/métabolisme , Composés organiques du phosphore , Pipéridines
16.
Phytomedicine ; 130: 155668, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38776739

RÉSUMÉ

BACKGROUND: Baoyuan decoction (BYD) has been widely utilized as a traditional prescription for the treatment of various conditions such as coronary heart disease, aplastic anemia, and chronic renal failure. However, its potential efficacy in improving atherosclerosis has not yet been investigated. PURPOSE: Our research aimed to assess the potential of BYD as an inhibitor of atherosclerosis and uncover the underlying mechanism by which it acts on foam cell formation. STUDY DESIGN AND METHODS: High-fat diet-induced ApoE-/- mice were employed to explore the effect of BYD on atherosclerosis. The differential metabolites in feces were identified and analyzed by LC-Qtrap-MS. In addition, we utilized pharmacological inhibition of BYD on foam cell formation induced by oxLDL in THP-1 cells to elucidate the underlying mechanisms specifically in macrophages. RESULTS: The atherosclerotic plaque burden in the aortic sinus of ApoE-/- mice was notably reduced with BYD treatment, despite no significant alterations in plasma lipids. Metabolomic analysis revealed that BYD suppressed the increased levels of peroxidized fatty acids, specifically 9/13-hydroxyoctadecadienoic acid (9/13-HODE), in the feces of mice. As a prominent peroxidized fatty acid found in oxLDL, we confirmed that 9/13-HODE induced the overexpression of CD36 in THP-1 macrophages by upregulating PPARγ. In subsequent experiments, the decreased levels of CD36 triggered by oxLDL were observed after BYD treatment. This decrease occurred through the regulation of the Src/MMK4/JNK pathway, resulting in the suppression of lipid deposition in THP-1 macrophages. CONCLUSIONS: These results illustrate that BYD exhibits potential anti-atherosclerotic effects by inhibiting CD36 expression to prevent foam cell formation.


Sujet(s)
Athérosclérose , Antigènes CD36 , Médicaments issus de plantes chinoises , Cellules spumeuses , Lipoprotéines LDL , Animaux , Athérosclérose/traitement médicamenteux , Antigènes CD36/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Humains , Mâle , Souris , Cellules spumeuses/effets des médicaments et des substances chimiques , Cellules spumeuses/métabolisme , Lipoprotéines LDL/métabolisme , Alimentation riche en graisse , Acides gras , Souris de lignée C57BL , Cellules THP-1 , Plaque d'athérosclérose/traitement médicamenteux , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Apolipoprotéines E
17.
Nat Metab ; 6(6): 1161-1177, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38698281

RÉSUMÉ

Diabetic cardiomyopathy is characterized by myocardial lipid accumulation and cardiac dysfunction. Bile acid metabolism is known to play a crucial role in cardiovascular and metabolic diseases. Takeda G-protein-coupled receptor 5 (TGR5), a major bile acid receptor, has been implicated in metabolic regulation and myocardial protection. However, the precise involvement of the bile acid-TGR5 pathway in maintaining cardiometabolic homeostasis remains unclear. Here we show decreased plasma bile acid levels in both male and female participants with diabetic myocardial injury. Additionally, we observe increased myocardial lipid accumulation and cardiac dysfunction in cardiomyocyte-specific TGR5-deleted mice (both male and female) subjected to a high-fat diet and streptozotocin treatment or bred on the diabetic db/db genetic background. Further investigation reveals that TGR5 deletion enhances cardiac fatty acid uptake, resulting in lipid accumulation. Mechanistically, TGR5 deletion promotes localization of CD36 on the plasma membrane through the upregulation of CD36 palmitoylation mediated by the palmitoyl acyltransferase DHHC4. Our findings indicate that the TGR5-DHHC4 pathway regulates cardiac fatty acid uptake, which highlights the therapeutic potential of targeting TGR5 in the management of diabetic cardiomyopathy.


Sujet(s)
Cardiomyopathies diabétiques , Acides gras , Récepteurs couplés aux protéines G , Animaux , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Cardiomyopathies diabétiques/métabolisme , Souris , Mâle , Femelle , Acides gras/métabolisme , Humains , Souris knockout , Acides et sels biliaires/métabolisme , Alimentation riche en graisse , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Myocarde/métabolisme , Métabolisme lipidique , Myocytes cardiaques/métabolisme , Diabète expérimental/métabolisme
18.
J Cell Biochem ; 125(7): e30574, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38704688

RÉSUMÉ

Altered energy metabolism is an emerging hallmark of cancer and plays a pivotal in cell survival, proliferation, and biosynthesis. In a rapidly proliferating cancer, energy metabolism acts in synergism with epithelial-to-mesenchymal transition (EMT), enabling cancer stemness, dissemination, and metastasis. In this study, an interconnected functional network governing energy metabolism and EMT signaling pathways was targeted through the concurrent inhibition of IR, ITGB1, and CD36 activity. A novel multicomponent MD simulation approach was employed to portray the simultaneous inhibition of IR, ITGB1, and CD36 by a 2:1 combination of Pimozide and Ponatinib. Further, in-vitro studies revealed the synergistic anticancer efficacy of drugs against monolayer as well as tumor spheroids of breast cancer cell lines (MCF-7 and MDA-MB-231). In addition, the combination therapy exerted approximately 40% of the apoptotic population and more than 1.5- to 3-fold reduction in the expression of ITGB1, IR, p-IR, IRS-1, and p-AKT in MCF-7 and MDA-MB-231 cell lines. Moreover, the reduction in fatty acid uptake, lipid droplet accumulation, cancer stemness, and migration properties were also observed. Thus, targeting IR, ITGB1, and CD36 in the interconnected network with the combination of Pimozide and Ponatinib represents a promising therapeutic approach for breast cancer.


Sujet(s)
Tumeurs du sein , Antigènes CD36 , Métabolisme énergétique , Transition épithélio-mésenchymateuse , Antigènes CD29 , Humains , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Antigènes CD29/métabolisme , Antigènes CD36/métabolisme , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Femelle , Métabolisme énergétique/effets des médicaments et des substances chimiques , Cellules MCF-7 , Imidazoles/pharmacologie , Pyridazines/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
19.
Biochim Biophys Acta Mol Cell Res ; 1871(6): 119748, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38723678

RÉSUMÉ

The Ser/Thr kinase protein kinase-D1 (PKD1) is involved in induction of various cell physiological processes in the heart such as myocellular hypertrophy and inflammation, which may turn maladaptive during long-term stimulation. Of special interest is a key role of PKD1 in the regulation of cardiac substrate metabolism. Glucose and fatty acids are the most important substrates for cardiac energy provision, and the ratio at which they are utilized determines the health status of the heart. Cardiac glucose uptake is mainly regulated by translocation of the glucose transporter GLUT4 from intracellular stores (endosomes) to the sarcolemma, and fatty acid uptake via a parallel translocation of fatty acid transporter CD36 from endosomes to the sarcolemma. PKD1 is involved in the regulation of GLUT4 translocation, but not CD36 translocation, giving it the ability to modulate glucose uptake without affecting fatty acid uptake, thereby altering the cardiac substrate balance. PKD1 would therefore serve as an attractive target to combat cardiac metabolic diseases with a tilted substrate balance, such as diabetic cardiomyopathy. However, PKD1 activation also elicits cardiac hypertrophy and inflammation. Therefore, identification of the events upstream and downstream of PKD1 may provide superior therapeutic targets to alter the cardiac substrate balance. Recent studies have identified the lipid kinase phosphatidylinositol 4-kinase IIIß (PI4KIIIß) as signaling hub downstream of PKD1 to selectively stimulate GLUT4-mediated myocardial glucose uptake without inducing hypertrophy. Taken together, the PKD1 signaling pathway serves a pivotal role in cardiac glucose metabolism and is a promising target to selectively modulate glucose uptake in cardiac disease.


Sujet(s)
Transporteur de glucose de type 4 , Glucose , Myocarde , Protéine kinase C , Transport des protéines , Transduction du signal , Transporteur de glucose de type 4/métabolisme , Humains , Myocarde/métabolisme , Animaux , Protéine kinase C/métabolisme , Protéine kinase C/génétique , Glucose/métabolisme , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Acides gras/métabolisme
20.
Cell Mol Biol Lett ; 29(1): 76, 2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38762740

RÉSUMÉ

BACKGROUND: The role of the scavenger receptor CD36 in cell metabolism and the immune response has been investigated mainly in macrophages, dendritic cells, and T cells. However, its involvement in B cells has not been comprehensively examined. METHODS: To investigate the function of CD36 in B cells, we exposed Cd36fl/flMB1cre mice, which lack CD36 specifically in B cells, to apoptotic cells to trigger an autoimmune response. To validate the proteins that interact with CD36 in primary B cells, we conducted mass spectrometry analysis following anti-CD36 immunoprecipitation. Immunofluorescence and co-immunoprecipitation were used to confirm the protein interactions. RESULTS: The data revealed that mice lacking CD36 in B cells exhibited a reduction in germinal center B cells and anti-DNA antibodies in vivo. Mass spectrometry analysis identified 30 potential candidates that potentially interact with CD36. Furthermore, the interaction between CD36 and the inhibitory Fc receptor FcγRIIb was first discovered by mass spectrometry and confirmed through immunofluorescence and co-immunoprecipitation techniques. Finally, deletion of FcγRIIb in mice led to decreased expression of CD36 in marginal zone B cells, germinal center B cells, and plasma cells. CONCLUSIONS: Our data indicate that CD36 in B cells is a critical regulator of autoimmunity. The interaction of CD36-FcγRIIb has the potential to serve as a therapeutic target for the treatment of autoimmune disorders.


Sujet(s)
Maladies auto-immunes , Lymphocytes B , Antigènes CD36 , Récepteurs du fragment Fc des IgG , Animaux , Souris , Maladies auto-immunes/métabolisme , Maladies auto-immunes/immunologie , Auto-immunité , Lymphocytes B/métabolisme , Lymphocytes B/immunologie , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Centre germinatif/métabolisme , Centre germinatif/immunologie , Souris de lignée C57BL , Souris knockout , Liaison aux protéines , Récepteurs du fragment Fc des IgG/métabolisme , Récepteurs du fragment Fc des IgG/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE