Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.042
Filtrer
1.
Mol Biomed ; 5(1): 24, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38937317

RÉSUMÉ

Chronic kidney disease (CKD) poses a significant global health dilemma, emerging from complex causes. Although our prior research has indicated that a deficiency in Reticulon-3 (RTN3) accelerates renal disease progression, a thorough examination of RTN3 on kidney function and pathology remains underexplored. To address this critical need, we generated Rtn3-null mice to study the consequences of RTN3 protein deficiency on CKD. Single-cell transcriptomic analyses were performed on 47,885 cells from the renal cortex of both healthy and Rtn3-null mice, enabling us to compare spatial architectures and expression profiles across 14 distinct cell types. Our analysis revealed that RTN3 deficiency leads to significant alterations in the spatial organization and gene expression profiles of renal cells, reflecting CKD pathology. Specifically, RTN3 deficiency was associated with Lars2 overexpression, which in turn caused mitochondrial dysfunction and increased reactive oxygen species levels. This shift induced a transition in renal epithelial cells from a functional state to a fibrogenic state, thus promoting renal fibrosis. Additionally, RTN3 deficiency was found to drive the endothelial-to-mesenchymal transition process and disrupt cell-cell communication, further exacerbating renal fibrosis. Immunohistochemistry and Western-Blot techniques were used to validate these observations, reinforcing the critical role of RTN3 in CKD pathogenesis. The deficiency of RTN3 protein in CKD leads to profound changes in cellular architecture and molecular profiles. Our work seeks to elevate the understanding of RTN3's role in CKD's narrative and position it as a promising therapeutic contender.


Sujet(s)
Évolution de la maladie , Fibrose , Analyse de profil d'expression de gènes , Insuffisance rénale chronique , Analyse sur cellule unique , Animaux , Souris , Fibrose/anatomopathologie , Fibrose/métabolisme , Fibrose/génétique , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Souris knockout , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Rein/anatomopathologie , Rein/métabolisme , Transcriptome , Espèces réactives de l'oxygène/métabolisme , Transition épithélio-mésenchymateuse/génétique , Modèles animaux de maladie humaine , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Mitochondries/génétique
2.
J Cell Biol ; 223(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-38916917

RÉSUMÉ

Context-dependent physiological remodeling of the extracellular matrix (ECM) is essential for development and organ homeostasis. On the other hand, consumption of high-caloric diet leverages ECM remodeling to create pathological conditions that impede the functionality of different organs, including the heart. However, the mechanistic basis of high caloric diet-induced ECM remodeling has yet to be elucidated. Employing in vivo molecular genetic analyses in Drosophila, we demonstrate that high dietary sugar triggers ROS-independent activation of JNK signaling to promote fatty acid oxidation (FAO) in the pericardial cells (nephrocytes). An elevated level of FAO, in turn, induces histone acetylation-dependent transcriptional upregulation of the cytokine Unpaired 3 (Upd3). Release of pericardial Upd3 augments fat body-specific expression of the cardiac ECM protein Pericardin, leading to progressive cardiac fibrosis. Importantly, this pathway is quite distinct from the ROS-Ask1-JNK/p38 axis that regulates Upd3 expression under normal physiological conditions. Our results unravel an unknown physiological role of FAO in cytokine-dependent ECM remodeling, bearing implications in diabetic fibrosis.


Sujet(s)
Protéines de Drosophila , Matrice extracellulaire , Acides gras , Oxydoréduction , Animaux , Matrice extracellulaire/métabolisme , Acides gras/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Myocarde/métabolisme , Myocarde/anatomopathologie , Cytokines/métabolisme , Cytokines/génétique , Drosophila melanogaster/métabolisme , Système de signalisation des MAP kinases , Espèces réactives de l'oxygène/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Fibrose/métabolisme , Péricarde/métabolisme , Péricarde/anatomopathologie
3.
Cell Death Dis ; 15(6): 391, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830870

RÉSUMÉ

Tissue injury causes activation of mesenchymal lineage cells into wound-repairing myofibroblasts (MFs), whose uncontrolled activity ultimately leads to fibrosis. Although this process is triggered by deep metabolic and transcriptional reprogramming, functional links between these two key events are not yet understood. Here, we report that the metabolic sensor post-translational modification O-linked ß-D-N-acetylglucosaminylation (O-GlcNAcylation) is increased and required for myofibroblastic activation. Inhibition of protein O-GlcNAcylation impairs archetypal myofibloblast cellular activities including extracellular matrix gene expression and collagen secretion/deposition as defined in vitro and using ex vivo and in vivo murine liver injury models. Mechanistically, a multi-omics approach combining proteomic, epigenomic, and transcriptomic data mining revealed that O-GlcNAcylation controls the MF transcriptional program by targeting the transcription factors Basonuclin 2 (BNC2) and TEA domain transcription factor 4 (TEAD4) together with the Yes-associated protein 1 (YAP1) co-activator. Indeed, inhibition of protein O-GlcNAcylation impedes their stability leading to decreased functionality of the BNC2/TEAD4/YAP1 complex towards promoting activation of the MF transcriptional regulatory landscape. We found that this involves O-GlcNAcylation of BNC2 at Thr455 and Ser490 and of TEAD4 at Ser69 and Ser99. Altogether, this study unravels protein O-GlcNAcylation as a key determinant of myofibroblastic activation and identifies its inhibition as an avenue to intervene with fibrogenic processes.


Sujet(s)
Myofibroblastes , Transduction du signal , Myofibroblastes/métabolisme , Animaux , Souris , Humains , Fibrose/métabolisme , Facteurs de transcription/métabolisme , Protéines de signalisation YAP/métabolisme , Souris de lignée C57BL , Facteurs de transcription à domaine TEA/métabolisme , Mâle , Maturation post-traductionnelle des protéines , Acétyl-glucosamine/métabolisme , Transcription génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique
4.
Nutrients ; 16(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892633

RÉSUMÉ

Spirulina (Arthrospira platensis) is reported to play a role in improving nonalcoholic fatty liver disease (NAFLD) and intestinal microbiota (IM). To study spirulina's effects in the improvement of NAFLD characteristics, IM, and pancreatic-renal lesions induced by a fructose-enriched diet, 40 Wistar healthy male rats, weighing 200-250 g, were randomly divided into four groups of 10, and each rat per group was assigned a diet of equal quantities (20 g/day) for 18 weeks. The first control group (CT) was fed a standardized diet, the second group received a 40% fructose-enriched diet (HFr), and the third (HFr-S5) and fourth groups (HFr-S10) were assigned the same diet composition as the second group but enriched with 5% and 10% spirulina, respectively. At week 18, the HFr-S10 group maintained its level of serum triglycerides and had the lowest liver fat between the groups. At the phylae and family level, and for the same period, the HFr-S10 group had the lowest increase in the Firmicutes/Bacteroidetes ratio and the Ruminococcaceae and the highest fecal alpha diversity compared to all other groups (p < 0.05). These findings suggest that at a 10% concentration, spirulina could be used in nutritional intervention to improve IM, fatty liver, metabolic, and inflammatory parameters associated with NAFLD.


Sujet(s)
Régime alimentaire , Compléments alimentaires , Microbiome gastro-intestinal , Stéatose hépatique non alcoolique , Spirulina , Mâle , Animaux , Rat Wistar , Spirulina/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/thérapie , Microbiome gastro-intestinal/physiologie , Fructose/métabolisme , Fibrose/métabolisme , Foie/anatomie et histologie , Rein/anatomie et histologie , Biodiversité
5.
Life Sci ; 350: 122745, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38834096

RÉSUMÉ

Fibrosis, a pathological state characterized by the excessive accumulation of extracellular matrix components, is primarily driven by the overactivation of fibroblasts. This condition becomes particularly pronounced under chronic inflammatory conditions. Fibrosis can occur in several tissues throughout the body. Among the notable discoveries in the study of fibrosis is the role of Collagen Triple Helix Repeat Containing-1 (CTHRC1), a protein that has emerged as a critical regulator in the fibrotic process. CTHRC1 is rapidly expressed on the outer membrane of fibroblasts and intimal smooth muscle cells following vascular injury, such as that induced by balloon angioplasty. This expression denotes the organism efforts to repair and restructure compromised tissue, signifying a critical component of the tissue repair mechanism in reaction to fibrosis. It plays a pivotal role in promoting cell migration and aiding tissue repair post-injury, contributing significantly to various pathophysiological processes including revascularization, bone formation, developmental morphological changes, inflammatory arthritis, and the progression of cancer. Significantly, researchers have observed marked expression of CTHRC1 across a variety of fibrotic conditions, closely associating it with the progression of the disease. Intervention with CTHRC1 can affect the occurrence and progression of fibrosis. This review aims to comprehensively explore the role and underlying mechanisms of CTHRC1 in fibrotic diseases, highlighting its potential as a key target for therapeutic interventions.


Sujet(s)
Protéines de la matrice extracellulaire , Fibrose , Humains , Fibrose/métabolisme , Protéines de la matrice extracellulaire/métabolisme , Animaux , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Matrice extracellulaire/métabolisme
6.
Int J Biol Sci ; 20(8): 3061-3075, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904010

RÉSUMÉ

Renal fibrosis is the common pathway in the progression of chronic kidney disease (CKD). Acyloxyacyl hydrolase (AOAH) is expressed in various phagocytes and is highly expressed in proximal tubular epithelial cells (PTECs). Research shows that AOAH plays a critical role in infections and chronic inflammatory diseases, although its role in kidney injury is unknown. Here, we found that AOAH deletion led to exacerbated kidney injury and fibrosis after folic acid (FA) administration, which was reversed by overexpression of Aoah in kidneys. ScRNA-seq revealed that Aoah-/- mice exhibited increased subpopulation of CD74+ PTECs, though the percentage of total PTECs were decreased compared to WT mice after FA treatment. Additionally, exacerbated kidney injury and fibrosis seen in Aoah-/- mice was attenuated via administration of methyl ester of (S, R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid (ISO-1), an inhibitor of macrophage inhibition factor (MIF) and CD74 binding. Finally, AOAH expression was found positively correlated with estimated glomerular filtration rate while negatively correlated with the degree of renal fibrosis in kidneys of CKD patients. Thus, our work indicates that AOAH protects against kidney injury and fibrosis by inhibiting renal tubular epithelial cells CD74 signaling pathways. Targeting kidney AOAH represents a promising strategy to prevent renal fibrosis progression.


Sujet(s)
Carboxylic ester hydrolases , Macrophages , Animaux , Souris , Macrophages/métabolisme , Carboxylic ester hydrolases/métabolisme , Carboxylic ester hydrolases/génétique , Humains , Antigènes de différenciation des lymphocytes B/métabolisme , Antigènes de différenciation des lymphocytes B/génétique , Insuffisance rénale chronique/métabolisme , Souris de lignée C57BL , Mâle , Antigènes d'histocompatibilité de classe II/métabolisme , Acide folique/métabolisme , Tubules rénaux/métabolisme , Tubules rénaux/anatomopathologie , Fibrose/métabolisme , Souris knockout , Cellules épithéliales/métabolisme
7.
Int J Biol Sci ; 20(8): 3185-3200, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904026

RÉSUMÉ

N6-methyladenosine (m6A) methylation plays a crucial role in various biological processes and the pathogenesis of human diseases. However, its role and mechanism in kidney fibrosis remain elusive. In this study, we show that the overall level of m6A methylated RNA was upregulated and the m6A methyltransferase METTL3 was induced in kidney tubular epithelial cells in mouse models and human kidney biopsies of chronic kidney disease (CKD). Proximal tubule-specific knockout of METTL3 in mice protected kidneys against developing fibrotic lesions after injury. Conversely, overexpression of METTL3 aggravated kidney fibrosis in vivo. Through bioinformatics analysis and experimental validation, we identified ß-catenin mRNA as a major target of METTL3-mediated m6A modification, which could be recognized by a specific m6A reader, the insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3). METTL3 stabilized ß-catenin mRNA, increased ß-catenin protein and induced its downstream profibrotic genes, whereas either knockdown of IGF2BP3 or inhibiting ß-catenin signaling abolished its effects. Collectively, these results indicate that METTL3 promotes kidney fibrosis by stimulating the m6A modification of ß-catenin mRNA, leading to its stabilization and its downstream profibrotic genes expression. Our findings suggest that targeting METTL3/IGF2BP3/ß-catenin pathway may be a novel strategy for the treatment of fibrotic CKD.


Sujet(s)
Fibrose , Methyltransferases , bêta-Caténine , bêta-Caténine/métabolisme , Animaux , Souris , Fibrose/métabolisme , Humains , Méthylation , Methyltransferases/métabolisme , Methyltransferases/génétique , Transduction du signal , Adénosine/analogues et dérivés , Adénosine/métabolisme , Rein/métabolisme , Rein/anatomopathologie , Mâle , Souris de lignée C57BL , Régulation positive , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/anatomopathologie , Souris knockout ,
8.
PeerJ ; 12: e17551, 2024.
Article de Anglais | MEDLINE | ID: mdl-38887622

RÉSUMÉ

Background: Keloid is a chronic proliferative fibrotic disease caused by abnormal fibroblasts proliferation and excessive extracellular matrix (ECM) production. Numerous fibrotic disorders are significantly influenced by ferroptosis, and targeting ferroptosis can effectively mitigate fibrosis development. This study aimed to investigate the role and mechanism of ferroptosis in keloid development. Methods: Keloid tissues from keloid patients and normal skin tissues from healthy controls were collected. Iron content, lipid peroxidation (LPO) level, and the mRNA and protein expression of ferroptosis-related genes including solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), transferrin receptor (TFRC), and nuclear factor erythroid 2-related factor 2 (Nrf2) were determined. Mitochondrial morphology was observed using transmission electron microscopy (TEM). Keloid fibroblasts (KFs) were isolated from keloid tissues, and treated with ferroptosis inhibitor ferrostatin-1 (fer-1) or ferroptosis activator erastin. Iron content, ferroptosis-related marker levels, LPO level, mitochondrial membrane potential, ATP content, and mitochondrial morphology in KFs were detected. Furthermore, the protein levels of α-smooth muscle actin (α-SMA), collagen I, and collagen III were measured to investigate whether ferroptosis affect fibrosis in KFs. Results: We found that iron content and LPO level were substantially elevated in keloid tissues and KFs. SLC7A11, GPX4, and Nrf2 were downregulated and TFRC was upregulated in keloid tissues and KFs. Mitochondria in keloid tissues and KFs exhibited ferroptosis-related pathology. Fer-1 treatment reduced iron content, restrained ferroptosis and mitochondrial dysfunction in KFs, Moreover, ferrostatin-1 restrained the protein expression of α-SMA, collagen I, and collagen III in KFs. Whereas erastin treatment showed the opposite results. Conclusion: Ferroptosis exists in keloid. Ferrostatin-1 restrained ECM deposition and fibrosis in keloid through inhibiting ferroptosis, and erastin induced ECM deposition and fibrosis through intensifying ferroptosis.


Sujet(s)
Cyclohexylamines , Ferroptose , Fibroblastes , Fibrose , Chéloïde , Facteur-2 apparenté à NF-E2 , Phénylènediamines , Phospholipid hydroperoxide glutathione peroxidase , Humains , Ferroptose/effets des médicaments et des substances chimiques , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Chéloïde/traitement médicamenteux , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Cyclohexylamines/pharmacologie , Fibrose/métabolisme , Fibrose/anatomopathologie , Phénylènediamines/pharmacologie , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Mâle , Peroxydation lipidique/effets des médicaments et des substances chimiques , Femelle , Adulte , Fer/métabolisme , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Pipérazines/pharmacologie , Actines/métabolisme , Actines/génétique , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques
9.
Life Sci ; 351: 122805, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38851422

RÉSUMÉ

AIMS: Heart failure (HF) is one of the most devastating consequences of cardiovascular diseases. Regardless of etiology, cardiac fibrosis is present and promotes the loss of heart function in HF patients. Cardiac resident fibroblasts, in response to a host of pro-fibrogenic stimuli, trans-differentiate into myofibroblasts to mediate cardiac fibrosis, the underlying mechanism of which remains incompletely understood. METHODS: Fibroblast-myofibroblast transition was induced in vitro by exposure to transforming growth factor (TGF-ß). Cardiac fibrosis was induced in mice by either transverse aortic constriction (TAC) or by chronic infusion with angiotensin II (Ang II). RESULTS: Through bioinformatic screening, we identified Kruppel-like factor 6 (KLF6) as a transcription factor preferentially up-regulated in cardiac fibroblasts from individuals with non-ischemic cardiomyopathy (NICM) compared to the healthy donors. Further analysis showed that nuclear factor kappa B (NF-κB) bound to the KLF6 promoter and mediated KLF6 trans-activation by pro-fibrogenic stimuli. KLF6 knockdown attenuated whereas KLF6 over-expression enhanced TGF-ß induced fibroblast-myofibroblast transition in vitro. More importantly, myofibroblast-specific KLF6 depletion ameliorated cardiac fibrosis and rescued heart function in mice subjected to the TAC procedure or chronic Ang II infusion. SIGNIFICANCE: In conclusion, our data support a role for KLF6 in cardiac fibrosis.


Sujet(s)
Fibroblastes , Fibrose , Facteur-6 de type krüppel , Souris de lignée C57BL , Myofibroblastes , Animaux , Facteur-6 de type krüppel/métabolisme , Facteur-6 de type krüppel/génétique , Fibrose/métabolisme , Souris , Humains , Mâle , Fibroblastes/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Angiotensine-II/pharmacologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Facteur de croissance transformant bêta/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Cellules cultivées , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Défaillance cardiaque/métabolisme , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/génétique
10.
J Biol Chem ; 300(6): 107385, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38759730

RÉSUMÉ

Non-muscle myosin 2 (NM2) is known to play an important role in myofibroblast transdifferentiation, a hallmark of fibrotic disorders. In a recent JBC article, Southern et al. demonstrate that endogenous S100A4, a calcium- and NM2-binding protein acts as a mechanoeffector in this process. Since extracellular S100A4 is also involved in fibrogenesis by triggering the inflammatory response, this small protein appears to contribute to fibrosis via at least two distinct mechanisms.


Sujet(s)
Fibrose , Protéine S100A4 liant le calcium , Protéines S100 , Humains , Protéine S100A4 liant le calcium/métabolisme , Protéine S100A4 liant le calcium/génétique , Fibrose/métabolisme , Animaux , Protéines S100/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Transdifférenciation cellulaire , Souris , Myosine de type II/métabolisme
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167267, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38810917

RÉSUMÉ

Small non-coding ribonucleic acids (sncRNAs) play an important role in cell regulation and are closely related to the pathogenesis of heart diseases. However, the role and molecular mechanism of transfer RNA-derived small RNAs (tsRNAs) in myocardial fibrosis after myocardial infarction (MI) remain unknown. In this study, we identified and validated sncRNAs (mainly miRNA and tsRNA) associated with myocardial fibrosis after MI through PANDORA sequencing of rat myocardial tissue. As a key enzyme of N4-acetylcytidine (ac4C) acetylation modification, N-acetyltransferase 10 (NAT10) plays an important role in regulating messenger RNA (mRNA) stability and translation efficiency. We found that NAT10 is highly expressed in infarcted myocardial tissue, and the results of acetylated RNA immunoprecipitation sequencing (acRIP-seq) analysis suggest that early growth response 3 (EGR3) may be an important molecule in the pathogenesis of NAT10-mediated myocardial fibrosis. Both in vivo and in vitro experiments have shown that inhibition of NAT10 can reduce the expression of EGR3 and alleviate myocardial fibrosis after MI. tsRNA can participate in gene regulation by inhibiting target genes. The expression of tsr007330 was decreased in myocardial infarction tissue. We found that overexpression of tsr007330 in rat myocardial tissue could antagonize NAT10, improve myocardial function in MI and alleviate myocardial fibrosis. In conclusion, tsRNAs (rno-tsr007330) may regulate the occurrence of myocardial fibrosis by regulating NAT10-mediated EGR3 mRNA acetylation. This study provides new insights into the improvement of myocardial fibrosis after MI by targeting tsRNA therapy.


Sujet(s)
Infarctus du myocarde , Animaux , Infarctus du myocarde/métabolisme , Infarctus du myocarde/génétique , Infarctus du myocarde/anatomopathologie , Acétylation , Rats , Mâle , ARN messager/génétique , ARN messager/métabolisme , Fibrose/métabolisme , ARN de transfert/génétique , ARN de transfert/métabolisme , Cytidine/analogues et dérivés , Cytidine/métabolisme , Myocarde/métabolisme , Myocarde/anatomopathologie , Rat Sprague-Dawley , Humains , Petit ARN non traduit/génétique , Petit ARN non traduit/métabolisme , N-terminal acetyltransferases
12.
Mol Metab ; 85: 101963, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38821174

RÉSUMÉ

OBJECTIVE: The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor regulating xenobiotic responses as well as physiological metabolism. Dietary AhR ligands activate the AhR signaling axis, whereas AhR activation is negatively regulated by the AhR repressor (AhRR). While AhR-deficient mice are known to be resistant to diet-induced obesity (DIO), the influence of the AhRR on DIO has not been assessed so far. METHODS: In this study, we analyzed AhRR-/- mice and mice with a conditional deletion of either AhRR or AhR in myeloid cells under conditions of DIO and after supplementation of dietary AhR ligands. Moreover, macrophage metabolism was assessed using Seahorse Mito Stress Test and ROS assays as well as transcriptomic analysis. RESULTS: We demonstrate that global AhRR deficiency leads to a robust, but not as profound protection from DIO and hepatosteatosis as AhR deficiency. Under conditions of DIO, AhRR-/- mice did not accumulate TCA cycle intermediates in the circulation in contrast to wild-type (WT) mice, indicating protection from metabolic dysfunction. This effect could be mimicked by dietary supplementation of AhR ligands in WT mice. Because of the predominant expression of the AhRR in myeloid cells, AhRR-deficient macrophages were analyzed for changes in metabolism and showed major metabolic alterations regarding oxidative phosphorylation and mitochondrial activity. Unbiased transcriptomic analysis revealed increased expression of genes involved in de novo lipogenesis and mitochondrial biogenesis. Mice with a genetic deficiency of the AhRR in myeloid cells did not show alterations in weight gain after high fat diet (HFD) but demonstrated ameliorated liver damage compared to control mice. Further, deficiency of the AhR in myeloid cells also did not affect weight gain but led to enhanced liver damage and adipose tissue fibrosis compared to controls. CONCLUSIONS: AhRR-deficient mice are resistant to diet-induced metabolic syndrome. Although conditional ablation of either the AhR or AhRR in myeloid cells did not recapitulate the phenotype of the global knockout, our findings suggest that enhanced AhR signaling in myeloid cells deficient for AhRR protects from diet-induced liver damage and fibrosis, whereas myeloid cell-specific AhR deficiency is detrimental.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice , Alimentation riche en graisse , Souris de lignée C57BL , Souris knockout , Obésité , Récepteurs à hydrocarbure aromatique , Animaux , Récepteurs à hydrocarbure aromatique/métabolisme , Récepteurs à hydrocarbure aromatique/génétique , Obésité/métabolisme , Souris , Alimentation riche en graisse/effets indésirables , Mâle , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Macrophages/métabolisme , Cellules myéloïdes/métabolisme , Fibrose/métabolisme , Foie/métabolisme , Transduction du signal
13.
Aging (Albany NY) ; 16(11): 9460-9469, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38814172

RÉSUMÉ

Low back pain stands as a significant factor in disability, largely resulting from intervertebral disc degeneration (IVDD). High glucose (HG) levels have been implicated in the pathogenesis of IVDD. However, the detailed mechanism of HG in IVDD is largely unknown. Our clinical results revealed that fibrosis markers such as CTGF, Col1a1, ATF4, and EIF2 are highly expressed in advanced-stage IVDD patients. Stimulation of human annulus fibrosus cells (HAFCs) with HG, but not mannitol, promotes fibrosis protein production. Ingenuity Pathway Analysis in the GSE database found that the mTOR, PKCδ, and NF-κB pathways were significantly changed during IVDD. The mTOR, PKCδ, and NF-κB inhibitors or siRNAs all abolished HG-induced fibrosis protein production. In addition, treatment of HAFCs with HG enhances the activation of mTOR, PKCδ, and NF-κB pathways. Thus, HG facilitates fibrosis in IVDD through mTOR, PKCδ, and NF-κB pathways. These results underscore the critical role of HG as a fibrotic factor in the progression of IVDD.


Sujet(s)
Anneau fibreux , Fibrose , Glucose , Facteur de transcription NF-kappa B , Protein kinase C-delta , Transduction du signal , Sérine-thréonine kinases TOR , Humains , Sérine-thréonine kinases TOR/métabolisme , Protein kinase C-delta/métabolisme , Fibrose/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Glucose/métabolisme , Anneau fibreux/métabolisme , Anneau fibreux/anatomopathologie , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Cellules cultivées , Adulte
14.
Exp Cell Res ; 440(1): 114102, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38821252

RÉSUMÉ

Renal fibrosis, apoptosis and autophagy are the main pathological manifestations of angiotensin II (Ang II)-induced renal injury. G protein-coupled receptor 39 (GPR39) is highly expressed in various tissues including the kidney, but its role in the kidney is entirely unclear. This study was performed to investigate the underlying mechanism by which knockdown of GPR39 alleviated Ang II-induced renal injury. In vivo, GPR39 knockout (KO) mice were constructed and infused with Ang II for 4 weeks, followed by renal function tests. In vitro, Ang II-induced cells were treated with si-GPR39 for 48 h. Fibrosis, apoptosis and autophagy were detected in both cells and mice. The underlying mechanism was sought by mRNA transcriptome sequencing and validated in vitro. GPR39 was upregulated in renal tissues of mice with Ang II-mediated renal injury. Knockdown of GPR39 ameliorated renal fibrosis, apoptosis, and autophagy, and decreased the expression of ribonucleotide reductase M2 (RRM2). In vitro, knockdown of GPR39 was also identified to improve the Ang II-induced cell fibrosis, apoptosis, and autophagy. mRNA transcriptome results showed that knockout of GPR39 reduced the expression of RRM2 in Ang II-induced kidney tissue. Activation of RRM2 could reverse the therapeutic effect of GPR39 knockout, and the inhibitor of RRM2 could improve the cell fibrosis, apoptosis and autophagy caused by GPR39 agonist. These results indicated that targeting of GPR39 could alleviate Ang II-induced renal fibrosis, apoptosis, and autophagy via reduction of RRM2 expression, and GPR39 may serve as a potential target for Ang II-induced renal injury.


Sujet(s)
Angiotensine-II , Apoptose , Souris knockout , Récepteurs couplés aux protéines G , Animaux , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Souris , Autophagie/génétique , Fibrose/métabolisme , Mâle , Souris de lignée C57BL , Rein/anatomopathologie , Rein/métabolisme , Maladies du rein/induit chimiquement , Maladies du rein/métabolisme , Maladies du rein/anatomopathologie , Maladies du rein/génétique
15.
J Agric Food Chem ; 72(23): 13069-13082, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38809951

RÉSUMÉ

Intestinal fibrosis is a common complication of Crohn's disease and characterized by excessive extracellular matrix (ECM) deposition. The aryl hydrocarbon receptor (AhR) detects micronutrients and microbial metabolites in diet and can attenuate intestinal fibrosis with unclear mechanisms. In this study, AhR activation was demonstrated to downregulate the transcription of collagen I and fibronectin in a Sp1- but not Sp3- or AP-1-dependent manner. A suppressed fatty acid synthesis was highlighted using untargeted metabolomics analyses, and synthetic products, palmitic acid (PA), were used as the intermediary agent. After a screening study, fatty acid synthase (FASN) was identified as the main targeted protein, and AhR activation regulated "HDAC3-acetylation" signals but not glycosylation to enhance FASN degradation. Furthermore, results of bioinformatics analysis and others showed that after being activated, AhR targeted miR-193a-3p to control HDAC3 transcription. Collectively, AhR activation inhibited ECM deposition and alleviated intestinal fibrosis by limiting fatty acid synthesis subsequent to the inhibition of "miR-193a-3p-HDAC3-FASN" signals.


Sujet(s)
Acides gras , Fibrose , Histone deacetylases , Intestins , microARN , Récepteurs à hydrocarbure aromatique , Récepteurs à hydrocarbure aromatique/métabolisme , Récepteurs à hydrocarbure aromatique/génétique , microARN/génétique , microARN/métabolisme , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Acides gras/métabolisme , Fibrose/métabolisme , Humains , Animaux , Souris , Souris de lignée C57BL , Mâle , Fatty acid synthase type I/métabolisme , Fatty acid synthase type I/génétique , Muqueuse intestinale/métabolisme , Transduction du signal
16.
FASEB J ; 38(11): e23709, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38809700

RÉSUMÉ

Brown adipose tissue (BAT) is correlated to cardiovascular health in rodents and humans, but the physiological role of BAT in the initial cardiac remodeling at the onset of stress is unknown. Activation of BAT via 48 h cold (16°C) in mice following transverse aortic constriction (TAC) reduced cardiac gene expression for LCFA uptake and oxidation in male mice and accelerated the onset of cardiac metabolic remodeling, with an early isoform shift of carnitine palmitoyltransferase 1 (CPT1) toward increased CPT1a, reduced entry of long chain fatty acid (LCFA) into oxidative metabolism (0.59 ± 0.02 vs. 0.72 ± 0.02 in RT TAC hearts, p < .05) and increased carbohydrate oxidation with altered glucose transporter content. BAT activation with TAC reduced early hypertrophic expression of ß-MHC by 61% versus RT-TAC and reduced pro-fibrotic TGF-ß1 and COL3α1 expression. While cardiac natriuretic peptide expression was yet to increase at only 3 days TAC, Nppa and Nppb expression were elevated in Cold TAC versus RT TAC hearts 2.7- and 2.4-fold, respectively. Eliminating BAT thermogenic activation with UCP1 KO mice eliminated differences between Cold TAC and RT TAC hearts, confirming effects of BAT activation rather than autonomous cardiac responses to cold. Female responses to BAT activation were blunted, with limited UCP1 changes with cold, partly due to already activated BAT in females at RT compared to thermoneutrality. These data reveal a previously unknown physiological mechanism of UCP1-dependent BAT activation in attenuating early cardiac hypertrophic and profibrotic signaling and accelerating remodeled metabolic activity in the heart at the onset of cardiac stress.


Sujet(s)
Tissu adipeux brun , Fibrose , Protéine-1 de découplage , Animaux , Tissu adipeux brun/métabolisme , Souris , Mâle , Protéine-1 de découplage/métabolisme , Fibrose/métabolisme , Carnitine O-palmitoyltransferase/métabolisme , Carnitine O-palmitoyltransferase/génétique , Souris de lignée C57BL , Cardiomégalie/métabolisme , Cardiomégalie/anatomopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Stress physiologique , Remodelage ventriculaire/physiologie , Souris knockout , Basse température
17.
Nat Aging ; 4(5): 664-680, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38760576

RÉSUMÉ

Hyaline cartilage fibrosis is typically considered an end-stage pathology of osteoarthritis (OA), which results in changes to the extracellular matrix. However, the mechanism behind this is largely unclear. Here, we found that the RNA helicase DDX5 was dramatically downregulated during the progression of OA. DDX5 deficiency increased fibrosis phenotype by upregulating COL1 expression and downregulating COL2 expression. In addition, loss of DDX5 aggravated cartilage degradation by inducing the production of cartilage-degrading enzymes. Chondrocyte-specific deletion of Ddx5 led to more severe cartilage lesions in the mouse OA model. Mechanistically, weakened DDX5 resulted in abundance of the Fn1-AS-WT and Plod2-AS-WT transcripts, which promoted expression of fibrosis-related genes (Col1, Acta2) and extracellular matrix degradation genes (Mmp13, Nos2 and so on), respectively. Additionally, loss of DDX5 prevented the unfolding Col2 promoter G-quadruplex, thereby reducing COL2 production. Together, our data suggest that strategies aimed at the upregulation of DDX5 hold significant potential for the treatment of cartilage fibrosis and degradation in OA.


Sujet(s)
Épissage alternatif , DEAD-box RNA helicases , Fibrose , G-quadruplexes , Arthrose , Animaux , DEAD-box RNA helicases/génétique , DEAD-box RNA helicases/métabolisme , Souris , Arthrose/anatomopathologie , Arthrose/génétique , Arthrose/métabolisme , Fibrose/métabolisme , Fibrose/génétique , Fibrose/anatomopathologie , Humains , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Modèles animaux de maladie humaine , Mâle
18.
J Pharm Biomed Anal ; 245: 116197, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38723558

RÉSUMÉ

The dysregulated levels of branched chain amino acids (BCAA) contribute to renal fibrosis in chronic kidney disease (CKD), yet specific analysis of BCAA contents and how they are regulated still remain unclear. It is therefore of great scientific interest to understand BCAA catabolism in CKD and develop a sensitive method for simultaneous determination of individual BCAA and their metabolites branched chain α-ketoacids (BCKA). In this work, the important role of BCAA metabolism that drives renal fibrosis in the process of CKD was first revealed by using transcriptomics. The key target genes controlling BCAA metabolism were then validated, that is, mRNA levels of BCKDHA and BCKDHB, the regulating rate-limiting enzymes during BCAA metabolism were abnormally reduced by quantitative PCR (qPCR), and a similar drop-off trend of protein expression of BCKDH, HIBCH and MCCC2 that are closely related to BCAA metabolism was also confirmed by western blotting. Furthermore, we established a novel strategy that simultaneously determines 6 individual BCAA and BCKA in serum and tissue. The method based on dansylhydrazine derivatization and ultra-high performance liquid chromatography-tandem triple quadrupole mass spectrometry (UHPLC-QQQ-MS) achieved to simultaneously determine the contents of BCAA and BCKA, which is efficient and stable. Compared with normal rats, levels of BCAA including leucine, isoleucine and valine in serum and kidney of CKD rats was decreased, while BCKA including α-ketoisocaproic acid, α-ketomethylvaleric acid and α-ketoisovaleric acid was increased. Together, these findings revealed the abnormality of BCAA metabolism in driving the course of kidney fibrosis and CKD. Our current study sheds new light on changes in BCAA metabolism during CKD, and may facilitate development of drugs to treat CKD and renal fibrosis.


Sujet(s)
Acides aminés à chaine ramifiée , Fibrose , Rein , Rat Sprague-Dawley , Insuffisance rénale chronique , Animaux , Acides aminés à chaine ramifiée/métabolisme , Rats , Mâle , Chromatographie en phase liquide à haute performance/méthodes , Fibrose/métabolisme , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/génétique , Rein/métabolisme , Rein/anatomopathologie , Cétoacides/métabolisme , Transcriptome , Spectrométrie de masse en tandem/méthodes , Analyse de profil d'expression de gènes/méthodes
19.
Clin Transl Med ; 14(5): e1686, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38769658

RÉSUMÉ

BACKGROUND: Renal allograft interstitial fibrosis/tubular atrophy (IF/TA) constitutes the principal histopathological characteristic of chronic allograft dysfunction (CAD) in kidney-transplanted patients. While renal vascular endothelial-mesenchymal transition (EndMT) has been verified as an important contributing factor to IF/TA in CAD patients, its underlying mechanisms remain obscure. Through single-cell transcriptomic analysis, we identified Rictor as a potential pivotal mediator for EndMT. This investigation sought to elucidate the role of Rictor/mTORC2 signalling in the pathogenesis of renal allograft interstitial fibrosis and the associated mechanisms. METHODS: The influence of the Rictor/mTOR2 pathway on renal vascular EndMT and renal allograft fibrosis was investigated by cell experiments and Rictor depletion in renal allogeneic transplantation mice models. Subsequently, a series of assays were conducted to explore the underlying mechanisms of the enhanced mitophagy and the ameliorated EndMT resulting from Rictor knockout. RESULTS: Our findings revealed a significant activation of the Rictor/mTORC2 signalling in CAD patients and allogeneic kidney transplanted mice. The suppression of Rictor/mTORC2 signalling alleviated TNFα-induced EndMT in HUVECs. Moreover, Rictor knockout in endothelial cells remarkably ameliorated renal vascular EndMT and allograft interstitial fibrosis in allogeneic kidney transplanted mice. Mechanistically, Rictor knockout resulted in an augmented BNIP3-mediated mitophagy in endothelial cells. Furthermore, Rictor/mTORC2 facilitated the MARCH5-mediated degradation of BNIP3 at the K130 site through K48-linked ubiquitination, thereby regulating mitophagy activity. Subsequent experiments also demonstrated that BNIP3 knockdown nearly reversed the enhanced mitophagy and mitigated EndMT and allograft interstitial fibrosis induced by Rictor knockout. CONCLUSIONS: Consequently, our study underscores Rictor/mTORC2 signalling as a critical mediator of renal vascular EndMT and allograft interstitial fibrosis progression, exerting its impact through regulating BNIP3-mediated mitophagy. This insight unveils a potential therapeutic target for mitigating renal allograft interstitial fibrosis.


Sujet(s)
Fibrose , Transplantation rénale , Complexe-2 cible mécanistique de la rapamycine , Protéines membranaires , Mitophagie , Compagnon de mTOR insensible à la rapamycine , Transduction du signal , Animaux , Compagnon de mTOR insensible à la rapamycine/métabolisme , Compagnon de mTOR insensible à la rapamycine/génétique , Souris , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Humains , Transplantation rénale/effets indésirables , Fibrose/métabolisme , Mâle , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Allogreffes , Rein/métabolisme , Rein/anatomopathologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Protéines proto-oncogènes
20.
Exp Biol Med (Maywood) ; 249: 10112, 2024.
Article de Anglais | MEDLINE | ID: mdl-38715976

RÉSUMÉ

Chronic inflammation is a key element in the progression of essential hypertension (EH). Calcium plays a key role in inflammation, so its receptor, the calcium-sensing receptor (CaSR), is an essential mediator of the inflammatory process. Compelling evidence suggests that CaSR mediates inflammation in tissues and immune cells, where it mediates their activity and chemotaxis. Macrophages (Mφs) play a major role in the inflammatory response process. This study provided convincing evidence that R568, a positive regulator of CaSR, was effective in lowering blood pressure in spontaneously hypertensive rats (SHRs), improving cardiac function by alleviating cardiac hypertrophy and fibrosis. R568 can increase the content of CaSR and M2 macrophages (M2Mφs, exert an anti-inflammatory effect) in myocardial tissue, reduce M1 macrophages (M1Mφs), which have a pro-inflammatory effect in this process. In contrast, NPS2143, a negative state regulator of CaSR, exerted the opposite effect in all of the above experiments. Following this study, R568 increased CaSR content in SHR myocardial tissue, lowered blood pressure, promoted macrophages to M2Mφs and improved myocardial fibrosis, but interestingly, both M1Mφs and M2Mφs were increased in the peritoneal cavity of SHRs, the number of M2Mφs remained lower than M1Mφs. In vitro, R568 increased CaSR content in RAW264.7 cells (a macrophage cell line), regulating intracellular Ca2+ ([Ca2+]i) inhibited NOD-like receptor family protein 3 (NLRP3) inflammasome activation and ultimately prevented its conversion to M1Mφs. The results showed that a decrease in CaSR in hypertensive rats causes further development of hypertension and cardiac damage. EH myocardial remodeling can be improved by CaSR overexpression by suppressing NLRP3 inflammasome activation and macrophage polarization toward M1Mφs and increasing M2Mφs.


Sujet(s)
Macrophages , Récepteurs-détecteurs du calcium , Remodelage ventriculaire , Animaux , Mâle , Souris , Rats , Pression sanguine , Fibrose/métabolisme , Hypertension artérielle/métabolisme , Hypertension artérielle/anatomopathologie , Macrophages/métabolisme , Myocarde/anatomopathologie , Myocarde/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Rats de lignée SHR , Récepteurs-détecteurs du calcium/métabolisme , Remodelage ventriculaire/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...