Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.890
Filtrer
1.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000165

RÉSUMÉ

Chronic kidney disease (CKD) is a noncommunicable condition that has become a major healthcare burden across the globe, often underdiagnosed and associated with low awareness. The main cause that leads to the development of renal impairment is diabetes mellitus and, in contrast to other chronic complications such as retinopathy or neuropathy, it has been suggested that intensive glycemic control is not sufficient in preventing the development of diabetic kidney disease. Nevertheless, a novel class of antidiabetic agents, the sodium-glucose cotransporter-2 inhibitors (SGLT2i), have shown multiple renoprotective properties that range from metabolic and hemodynamic to direct renal effects, with a major impact on reducing the risk of occurrence and progression of CKD. Thus, this review aims to summarize current knowledge regarding the renoprotective mechanisms of SGLT2i and to offer a new perspective on this innovative class of antihyperglycemic drugs with proven pleiotropic beneficial effects that, after decades of no significant progress in the prevention and in delaying the decline of renal function, start a new era in the management of patients with CKD.


Sujet(s)
Insuffisance rénale chronique , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Humains , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/prévention et contrôle , Insuffisance rénale chronique/métabolisme , Animaux , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/anatomopathologie , Néphropathies diabétiques/traitement médicamenteux , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/prévention et contrôle , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Hypoglycémiants/usage thérapeutique , Hypoglycémiants/pharmacologie , Transporteur-2 sodium-glucose/métabolisme
2.
Vet Q ; 44(1): 1-9, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39028217

RÉSUMÉ

Hair cortisol concentration (HCC) and a questionnaire were used as indicators of chronic stress status and quality of life (QoL), respectively, in cats. To date, there has been limited research on the simultaneous application of both indicators in unwell cats. Our aim was to evaluate HCC and questionnaire data obtained from a healthy cat cohort (n = 61) and cat cohorts with either chronic kidney disease (CKD) (n = 78) or suspected feline infectious peritonitis (FIP) (n = 24). Furthermore, we also investigated the correlation between HCC and clinical pathological data. For this study, hair from the abdomen of cats was collected and analyzed for HCC using a commercial ELISA kit. Owners also completed a questionnaire, from which average-item-weighted-impact-scores (AWISs) were calculated. Cats with late-stage-CKD (median, HCC = 330.15 pg/mg, AWIS = -0.43) presented with a significantly higher HCC (p < 0.01) and a significantly lower AWIS (p < 0.01) than cats with early-stage-CKD (HCC = 183.56 pg/mg, AWIS = 1.08). Similarly, there were significant differences in both HCC (p < 0.001) and AWIS (p < 0.001) between cats with suspected FIP (HCC = 896.27 pg/mg, AWIS = -1.97) and healthy cats (HCC = 181.24 pg/mg, AWIS = 1.24). The degree of consistency between the HCC results and the questionnaire results reminds us that the severity of a chronic disease or the presence of a life-threatening disease can significantly increase stress and thus can affect the QoL of cats.


Sujet(s)
Maladies des chats , Péritonite infectieuse féline , Poils , Hydrocortisone , Qualité de vie , Insuffisance rénale chronique , Animaux , Chats , Enquêtes et questionnaires , Insuffisance rénale chronique/médecine vétérinaire , Insuffisance rénale chronique/métabolisme , Hydrocortisone/analyse , Mâle , Poils/composition chimique , Femelle , Stress physiologique , Stress psychologique
3.
J Transl Med ; 22(1): 649, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992710

RÉSUMÉ

BACKGROUND: Renal interstitial fibrosis (RIF) is a progressive, irreversible terminal kidney disease with a poor prognosis and high mortality. Angiopoietin-like 4 (ANGPTL4) is known to be associated with fibrosis in various organs, but its impact on the RIF process remains unclear. This study aimed to elucidate the role and underlying mechanisms of ANGPTL4 in the progression of RIF. METHODS: In vivo, a chronic kidney disease (CKD) rat model of renal interstitial fibrosis was established via intragastric administration of adenine at different time points (4 and 6 weeks). Blood and urine samples were collected to assess renal function and 24-h urinary protein levels. Kidney tissues were subjected to HE and Masson staining for pathological observation. Immunohistochemistry and real-time quantitative PCR (qRT‒PCR) were performed to evaluate the expression of ANGPTL4 and hypoxia-inducible factor-1α (HIF-1α), followed by Pearson correlation analysis. Subsequently, kidney biopsy tissues from 11 CKD patients (6 with RIF and 5 without RIF) were subjected to immunohistochemical staining to validate the expression of ANGPTL4. In vitro, a fibrosis model of human renal tubular epithelial cells (HK2) was established through hypoxic stimulation. Subsequently, an HIF-1α inhibitor (2-MeOE2) was used, and ANGPTL4 was manipulated using siRNA or plasmid overexpression. Changes in ANGPTL4 and fibrosis markers were analyzed through Western blotting, qRT‒PCR, and immunofluorescence. RESULTS: ANGPTL4 was significantly upregulated in the CKD rat model and was significantly positively correlated with renal injury markers, the fibrotic area, and HIF-1α. These results were confirmed by clinical samples, which showed a significant increase in the expression level of ANGPTL4 in CKD patients with RIF, which was positively correlated with HIF-1α. Further in vitro studies indicated that the expression of ANGPTL4 is regulated by HIF-1α, which in turn is subject to negative feedback regulation by ANGPTL4. Moreover, modulation of ANGPTL4 expression influences the progression of fibrosis in HK2 cells. CONCLUSION: Our findings indicate that ANGPTL4 is a key regulatory factor in renal fibrosis, forming a loop with HIF-1α, potentially serving as a novel therapeutic target for RIF.


Sujet(s)
Protéine-4 similaire à l'angiopoïétine , Fibrose , Sous-unité alpha du facteur-1 induit par l'hypoxie , Rein , Rat Sprague-Dawley , Animaux , Protéine-4 similaire à l'angiopoïétine/métabolisme , Protéine-4 similaire à l'angiopoïétine/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Humains , Mâle , Rein/anatomopathologie , Rein/métabolisme , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Rats , Lignée cellulaire , Maladies du rein/anatomopathologie , Maladies du rein/métabolisme , Adulte d'âge moyen
4.
Int J Biol Sci ; 20(9): 3412-3425, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993571

RÉSUMÉ

Chronic kidney disease (CKD) is linked to greater prevalence and rapid progression of calcific aortic valve disease (CAVD) characterized by valvular leaflet fibrosis and calcification. Fibroblast growth factor 23 (FGF23) level is elevated, and anti-aging protein Klotho is reduced in CKD patients. However, the roles of FGF23 and Klotho in the mechanism of aortic valve fibrosis and calcification remain unclear. We hypothesized that FGF23 mediates CKD-induced CAVD by enhancing aortic valve interstitial cell (AVIC) fibrosis and calcification, while soluble Klotho inhibits FGF23 effect. Methods and Results: In an old mouse model of CKD, kidney damages were accompanied by aortic valve thickening and calcification. FGF23 levels in plasma and aortic valve were increased, while Klotho levels were decreased. Recombinant FGF23 elevated the inflammatory, fibrogenic, and osteogenic activities in AVICs. Neutralizing antibody or shRNA targeting FGF23 suppressed the pathobiological activities in AVICs from valves affected by CAVD. FGF23 exerts its effects on AVICs via FGF receptor (FGFR)/Yes-associated protein (YAP) signaling, and inhibition of FGFR/YAP reduced FGF23's potency in AVICs. Recombinant Klotho downregulated the pathobiological activities in AVICs exposed to FGF23. Incubation of FGF23 with Klotho formed complexes and decreased FGF23's potency. Further, treatment of CKD mice with recombinant Klotho attenuated aortic valve lesions. Conclusion: This study demonstrates that CKD induces FGF23 accumulation, Klotho insufficiency and aortic valve lesions in old mice. FGF23 upregulates the inflammatory, fibrogenic and osteogenic activities in AVICs via the FGFR/YAP signaling pathway. Soluble Klotho suppresses FGF23 effect through molecular interaction and is capable of mitigating CKD-induced CAVD.


Sujet(s)
Valve aortique , Facteur-23 de croissance des fibroblastes , Facteurs de croissance fibroblastique , Glucuronidase , Protéines Klotho , Insuffisance rénale chronique , Protéines Klotho/métabolisme , Facteur-23 de croissance des fibroblastes/métabolisme , Animaux , Insuffisance rénale chronique/métabolisme , Glucuronidase/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Souris , Valve aortique/métabolisme , Valve aortique/anatomopathologie , Calcinose/métabolisme , Mâle , Transduction du signal , Souris de lignée C57BL , Humains , Sténose aortique/métabolisme , Modèles animaux de maladie humaine
5.
Kidney Int ; 106(2): 183-185, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39032962

RÉSUMÉ

Persistent enhancement of glycolysis in kidney tubular epithelial cells has been linked to the progression of chronic kidney disease, although the underlying mechanisms are largely unknown. In this issue of Kidney International, Wang et al. report that the glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 plays a crucial role in kidney fibrosis by enhancing histone H4 lysine 12 lactylation through lactate accumulation. This increases the transcription of nuclear factor-κB-related genes and promotes inflammation and fibrosis. Inhibiting 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 reduces these effects, indicating therapeutic potential for kidney fibrosis.


Sujet(s)
Évolution de la maladie , Acide lactique , Insuffisance rénale chronique , Humains , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Acide lactique/métabolisme , Animaux , Fibrose , Inflammation/métabolisme , Glycolyse , Histone/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Rein/anatomopathologie , Rein/métabolisme
6.
Ren Fail ; 46(2): 2367708, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38973391

RÉSUMÉ

BACKGROUND: Cellular senescence, macrophages infiltration, and vascular smooth muscle cells (VSMCs) osteogenic transdifferentiation participate in the pathophysiology of vascular calcification in chronic kidney disease (CKD). Senescent macrophages are involved in the regulation of inflammation in pathological diseases. In addition, senescent cells spread senescence to neighboring cells via Interferon-induced transmembrane protein3 (IFITM3). However, the role of senescent macrophages and IFITM3 in VSMCs calcification remains unexplored. AIMS: To explore the hypothesis that senescent macrophages contribute to the calcification and senescence of VSMCs via IFITM3. METHODS: Here, the macrophage senescence model was established using Lipopolysaccharides (LPS). The VSMCs were subjected to supernatants from macrophages (MCFS) or LPS-induced macrophages (LPS-MCFS) in the presence or absence of calcifying media (CM). Senescence-associated ß-galactosidase (SA-ß-gal), Alizarin red (AR), immunofluorescent staining, and western blot were used to identify cell senescence and calcification. RESULTS: The expression of IFITM3 was significantly increased in LPS-induced macrophages and the supernatants. The VSMCs transdifferentiated into osteogenic phenotype, expressing higher osteogenic differentiation markers (RUNX2) and lower VSMCs constructive makers (SM22α) when cultured with senescent macrophages supernatants. Also, senescence markers (p16 and p21) in VSMCs were significantly increased by senescent macrophages supernatants treated. However, IFITM3 knockdown inhibited this process. CONCLUSIONS: Our study showed that LPS-induced senescence of macrophages accelerated the calcification of VSMCs via IFITM3. These data provide a new perspective linking VC and aging, which may provide clues for diagnosing and treating accelerated vascular aging in patients with CKD.


Sujet(s)
Vieillissement de la cellule , Lipopolysaccharides , Macrophages , Protéines membranaires , Muscles lisses vasculaires , Protéines de liaison à l'ARN , Calcification vasculaire , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Lipopolysaccharides/pharmacologie , Calcification vasculaire/anatomopathologie , Calcification vasculaire/métabolisme , Macrophages/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines de liaison à l'ARN/métabolisme , Humains , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Cellules cultivées , Animaux , Ostéogenèse , Transdifférenciation cellulaire
7.
Ren Fail ; 46(2): 2375033, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38967135

RÉSUMÉ

The Astragalus mongholicus Bunge and Panax notoginseng formula (A&P) has been clinically shown to effectively slow down the progression of chronic kidney disease (CKD) and has demonstrated significant anti-fibrosis effects in experimental CKD model. However, the specific active ingredients and underlying mechanism are still unclear. The active ingredients of A&P were analyzed by Ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-HR-MS). A mouse model of CKD was constructed by 5/6 nephrectomy. Renal function was assessed by creatinine and urea nitrogen. Real-time PCR and Western Blot were performed to detect the mRNA and protein changes in kidney and cells. An in vitro fibrotic cell model was constructed by TGF-ß induction in TCMK-1 cells. The results showed that thirteen active ingredients of A&P were identified by UPLC-HR-MS, nine of which were identified by analysis with standards, among which the relative percentage of NOB was high. We found that NOB treatment significantly improved renal function, pathological damage and reduced the expression level of fibrotic factors in CKD mice. The results also demonstrated that Lgals1 was overexpressed in the interstitial kidney of CKD mice, and NOB treatment significantly reduced its expression level, while inhibiting PI3K and AKT phosphorylation. Interestingly, overexpression of Lgals1 significantly increased fibrosis in TCMK1 cells and upregulated the activity of PI3K and AKT, which were strongly inhibited by NOB treatment. NOB is one of the main active components of A&P. The molecular mechanism by which NOB ameliorates renal fibrosis in CKD may be through the inhibition of Lgals1/PI3K/AKT signaling pathway.


Sujet(s)
Modèles animaux de maladie humaine , Médicaments issus de plantes chinoises , Fibrose , Flavones , Rein , Panax notoginseng , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Insuffisance rénale chronique , Transduction du signal , Animaux , Souris , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Mâle , Panax notoginseng/composition chimique , Flavones/pharmacologie , Flavones/usage thérapeutique , Rein/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Astragalus/composition chimique , Souris de lignée C57BL , Spectrométrie de masse en tandem , Chromatographie en phase liquide à haute performance
8.
Medicine (Baltimore) ; 103(30): e33705, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39058890

RÉSUMÉ

Chronic kidney disease (CKD) inevitably progresses to end-stage renal disease if intervention does not occur timely. However, there are limitations in predicting the progression of CKD by solely relying on changes in renal function. A biomarker with high sensitivity and specificity that can predict CKD progression early is required. We used the online Gene Expression Omnibus microarray dataset GSE45980 to identify differentially expressed genes (DEGs) in patients with progressive and stable CKD. We then performed functional enrichment and protein-protein interaction network analysis on DEGs and identified key genes. Finally, the expression patterns of key genes were verified using the GSE60860 dataset, and the receiver operating characteristic curve analysis was performed to clarify their predictive ability of progressive CKD. Ultimately, we verified the expression profiles of these hub genes in an in vitro renal interstitial fibrosis model by real-time PCR and western blot analysis. Differential expression analysis identified 50 upregulated genes and 47 downregulated genes. The results of the functional enrichment analysis revealed that upregulated DEGs were mainly enriched in immune response, inflammatory response, and NF-κB signaling pathways, whereas downregulated DEGs were mainly related to angiogenesis and the extracellular environment. Protein-protein interaction network and key gene analysis identified CCR7 as the most important gene. CCR7 mainly plays a role in immune response, and its only receptors, CCL19 and CCL21, have also been identified as DEGs. The receiver operating characteristic curve analysis of CCR7, CCL19, and CCL21 found that CCR7 and CCL19 present good disease prediction ability. CCR7 may be a stable biomarker for predicting CKD progression, and the CCR7-CCL19/CCL21 axis may be a therapeutic target for end-stage renal disease. However, further experiments are needed to explore the relationship between these genes and CKD.


Sujet(s)
Marqueurs biologiques , Biologie informatique , Évolution de la maladie , Cartes d'interactions protéiques , Récepteurs CCR7 , Insuffisance rénale chronique , Récepteurs CCR7/génétique , Récepteurs CCR7/métabolisme , Humains , Biologie informatique/méthodes , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/métabolisme , Marqueurs biologiques/métabolisme , Cartes d'interactions protéiques/génétique , Analyse de profil d'expression de gènes , Courbe ROC
10.
Respir Res ; 25(1): 288, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080603

RÉSUMÉ

BACKGROUND: Chronic kidney disease (CKD) is a significant risk factor for pulmonary hypertension (PH), a complication that adversely affects patient prognosis. However, the mechanisms underlying this association remain poorly understood. A major obstacle to progress in this field is the lack of a reliable animal model replicating CKD-PH. METHODS: This study aimed to establish a stable rat model of CKD-PH. We employed a combined approach, inducing CKD through a 5/6 nephrectomy and concurrently exposing the rats to a high-salt diet. The model's hemodynamics were evaluated dynamically, alongside a comprehensive assessment of pathological changes in multiple organs. Lung tissues and serum samples were collected from the CKD-PH rats to analyze the expression of angiotensin-converting enzyme 2 (ACE2), evaluate the activity of key vascular components within the renin-angiotensin-aldosterone system (RAAS), and characterize alterations in the serum metabolic profile. RESULTS: At 14 weeks post-surgery, the CKD-PH rats displayed significant changes in hemodynamic parameters indicative of pulmonary arterial hypertension. Additionally, right ventricular hypertrophy was observed. Notably, no evidence of pulmonary vascular remodeling was found. Further analysis revealed RAAS dysregulation and downregulated ACE2 expression within the pulmonary vascular endothelium of CKD-PH rats. Moreover, the serum metabolic profile of these animals differed markedly from the sham surgery group. CONCLUSIONS: Our findings suggest that the development of pulmonary arterial hypertension in CKD-PH rats is likely a consequence of a combined effect: RAAS dysregulation, decreased ACE2 expression in pulmonary vascular endothelial cells, and metabolic disturbances.


Sujet(s)
Angiotensine-II , Hypertension pulmonaire , Néphrectomie , Chlorure de sodium alimentaire , Animaux , Mâle , Rats , Angiotensine-II/sang , Angiotensin-converting enzyme 2/métabolisme , Modèles animaux de maladie humaine , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/induit chimiquement , Rein/métabolisme , Rein/anatomopathologie , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Système rénine-angiotensine/physiologie , Chlorure de sodium alimentaire/effets indésirables
11.
Funct Integr Genomics ; 24(4): 131, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39078513

RÉSUMÉ

BACKGROUND: Macrophages are the main inflammatory cells involved in kidney injury and play a significant role in the development of acute kidney injury (AKI) and progression of chronic kidney disease (CKD). Emodin is believed to stabilize macrophage homeostasis under pathological conditions. The objective of this study aimed to explore the underlying mechanisms and effects of Emodin on M1 macrophages. METHODS: Network pharmacology methods were used to predict target proteins associated with renal injury and identify the pathways affected by emodin. RAW264.7 macrophages were induced into M1 polarization using LPS and then treated with emodin at 20, 40, and 80 µM. The effects of emodin on cell viability, cytokines (IL-1ß, IL-6, TNF-α), M1 macrophage markers (F4/80 + CD86+), and the EGFR/MAPK pathway were evaluated. Additionally, we transfected RAW264.7 cells with an EGFR shRNA interference lentivirus to assess its effects on RAW264.7 cells function and MAPK pathway. After RAW264.7 cells were passaged to expanded culture and transfected with EGFR-interfering plasmid, macrophages were induced to polarize towards M1 with LPS and then treated with 80 µM emodin. CKD modeling was performed to test how emodin is regulated during CKD. RESULTS: There are 15 common targets between emodin and kidney injury, of which the EGFR/MAPK pathway is the pathway through which emodin affects macrophage function. Emodin significantly reduced the levels of IL-6, IL-1ß and TNF-α (p < 0.05) and the ratio of M1 macrophage surface markers F4/80 + CD86+ (p < 0.01) in the supernatant of RAW264.7 cells in a dose-dependent manner. Furthermore, the inhibitory effect of emodin on RAW264.7 cells was achieved by interfering with the EGFR/MAPK pathway. Moreover, emodin also affected the mRNA and protein expression of EGFR and Ras, leading to a decrease in the rate of M1 macrophages, thus inhibiting the pro-inflammatory effect of M1 macrophages. The addition of emodin reduced the rate of M1 macrophages in CKD and inhibited the further polarization of M1 macrophages, thus maintaining the pro-inflammatory and anti-inflammatory homeostasis in CKD, and these effects were achieved by emodin through the control of the EGRF/ERK pathway. CONCLUSION: Emodin attenuates M1 macrophage polarization and pro-inflammatory responses via the EGFR/MAPK signalling pathway. And the addition of emodin maintains pro- and anti-inflammatory homeostasis, which is important for maintaining organ function and tissue repair.


Sujet(s)
Atteinte rénale aigüe , Émodine , Récepteurs ErbB , Système de signalisation des MAP kinases , Activation des macrophages , Macrophages , Insuffisance rénale chronique , Animaux , Souris , Émodine/pharmacologie , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Cellules RAW 264.7 , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/anatomopathologie , Activation des macrophages/effets des médicaments et des substances chimiques , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/anatomopathologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Cytokines/génétique
12.
Drug Des Devel Ther ; 18: 2693-2712, 2024.
Article de Anglais | MEDLINE | ID: mdl-38974121

RÉSUMÉ

Background: Chronic kidney disease (CKD) is a significant worldwide health concern that leads to high mortality rates. The bioactive substance costunolide (CTD) has demonstrated several pharmacological effects and holds promise as a CKD treatment. This study aims to investigate the impact of CTD on CKD and delve into its mechanisms of action. Methods: Unilateral ureteral obstruction (UUO) methods and renal fibrosis mice models were created. Various concentrations of CTD were injected into UUO mice models to investigate the therapeutic effects of CTD on renal fibrosis of mice. Then, renal morphology, pathological changes, and the expression of genes related to fibrosis, inflammation and ferroptosis were analysed. RNA sequencing was utilized to identify the main biological processes and pathways involved in renal injury. Finally, both overexpression and inhibition of IKKß were studied to examine their respective effects on fibrosis and inflammation in both in vitro and in vivo models. Results: CTD treatment was found to significantly alleviate fibrosis, inflammation and ferroptosis in UUO-induced renal fibrosis mice models. The results of RNA sequencing suggested that the IKKß acted as key regulatory factor in renal injury and the expression of IKKß was increased in vitro and in vivo renal fibrosis model. Functionally, down-regulated IKKß expression inhibits ferroptosis, inflammatory cytokine production and collagen deposition. Conversely, IKKß overexpression exacerbates progressive renal fibrosis. Mechanistically, CTD alleviated renal fibrosis and inflammation by inhibiting the expression of IKKß and attenuating IKKß/NF-κB pathway. Conclusion: This study demonstrates that CTD could mitigate renal fibrosis, ferroptosis and inflammation in CKD by modulating the IKKß/NF-κB pathway, which indicates targeting IKKß has an enormous potential for treating CKD.


Sujet(s)
Insuffisance rénale chronique , Sesquiterpènes , Animaux , Humains , Mâle , Souris , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Fibrose/traitement médicamenteux , I-kappa B Kinase/métabolisme , I-kappa B Kinase/antagonistes et inhibiteurs , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Souris de lignée C57BL , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Sesquiterpènes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Obstruction urétérale/traitement médicamenteux , Obstruction urétérale/métabolisme
13.
Front Endocrinol (Lausanne) ; 15: 1331603, 2024.
Article de Anglais | MEDLINE | ID: mdl-39027471

RÉSUMÉ

Background: The association of Remnant cholesterol (RC) with renal function and its progression in patients with Type 2 diabetes (T2DM) related chronic kidney disease (CKD) remains unclear. Methods: 8,678 patients with T2DM-related CKD were included in cross-sectional analysis, and 6,165 patients were enrolled in longitudinal analysis and followed up for a median of 36.0 months. The outcomes were renal composite endpoint event and rapid progression of renal function. Results: 24.54% developed a renal composite endpoint event, and 27.64% rapid progression of renal function. RC levels above 0.56 mmol/L independently increased the risk of both renal composite endpoint (HR, 1.17; 95% CIs, 1.03-1.33) and rapid progression of renal function (OR, 1.17; 95% CIs, 1.01- 1.37). TG levels above 1.65 mmol/L only increased the risk of renal composite endpoint (HR, 1.16; 95% CIs, 1.02 -1.32). TC levels above 5.21 mmol/L increased the risk of renal composite endpoint (HR, 1.14; 95% CIs, 1.01-1.29) only in patients with proteinuria≥0.5g/d. Conversely, HDL-C levels below 1.20 mmol/L or above 1.84 mmol/L increased the risk of rapid progression of renal function (OR, 0.88; 95% CIs, 0.70 -0.99) in patients with proteinuria<0.5g/d (all P<0.05). Conclusion: In patients with T2DM-related CKD, RC was an independent risk factor for progression of renal function, and maintaining it below 0.56 mmol/L could reduce the risk of renal function progression.


Sujet(s)
Cholestérol , Diabète de type 2 , Néphropathies diabétiques , Évolution de la maladie , Insuffisance rénale chronique , Humains , Diabète de type 2/complications , Diabète de type 2/métabolisme , Diabète de type 2/sang , Mâle , Femelle , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/sang , Insuffisance rénale chronique/physiopathologie , Insuffisance rénale chronique/métabolisme , Adulte d'âge moyen , Cholestérol/sang , Études transversales , Sujet âgé , Néphropathies diabétiques/sang , Néphropathies diabétiques/étiologie , Néphropathies diabétiques/physiopathologie , Débit de filtration glomérulaire , Études longitudinales , Études de suivi , Rein/physiopathologie , Rein/métabolisme , Facteurs de risque
14.
Biomed Pharmacother ; 177: 117079, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38968801

RÉSUMÉ

Macrophages are widely distributed throughout various tissues of the body, and mounting evidence suggests their involvement in regulating the tissue microenvironment, thereby influencing disease onset and progression through direct or indirect actions. In chronic kidney disease (CKD), disturbances in renal functional homeostasis lead to inflammatory cell infiltration, tubular expansion, glomerular atrophy, and subsequent renal fibrosis. Macrophages play a pivotal role in this pathological process. Therefore, understanding their role is imperative for investigating CKD progression, mitigating its advancement, and offering novel research perspectives for fibrosis treatment from an immunological standpoint. This review primarily delves into the intrinsic characteristics of macrophages, their origins, diverse subtypes, and their associations with renal fibrosis. Particular emphasis is placed on the transition between M1 and M2 phenotypes. In late-stage CKD, there is a shift from the M1 to the M2 phenotype, accompanied by an increased prevalence of M2 macrophages. This transition is governed by the activation of the TGF-ß1/SMAD3 and JAK/STAT pathways, which facilitate macrophage-to-myofibroblast transition (MMT). The tyrosine kinase Src is involved in both signaling cascades. By thoroughly elucidating macrophage functions and comprehending the modes and molecular mechanisms of macrophage-fibroblast interaction in the kidney, novel, tailored therapeutic strategies for preventing or attenuating the progression of CKD can be developed.


Sujet(s)
Fibrose , Macrophages , Insuffisance rénale chronique , Humains , Macrophages/anatomopathologie , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/immunologie , Insuffisance rénale chronique/métabolisme , Animaux , Transduction du signal , Rein/anatomopathologie , Rein/métabolisme , Évolution de la maladie , Phénotype
15.
J Am Heart Assoc ; 13(14): e034066, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38979792

RÉSUMÉ

BACKGROUND: Atherosclerosis is highly prevalent in people with chronic kidney disease (CKD), including those receiving peritoneal dialysis (PD). Although it is lifesaving, PD induces profound systemic inflammation, which may aggravate atherosclerosis. Therefore, the hypothesis is that this PD-induced inflammation aggravates atherosclerosis via immune cell activation. METHODS AND RESULTS: ApoE-/- mice were subjected to a 5/6 nephrectomy to induce CKD. Three weeks later, mice were fed a high-cholesterol diet. Half of the nephrectomized mice then received daily peritoneal infusions of 3.86% Physioneal for 67 further days (CKD+PD) until the end of the experiment, and were compared with mice without CKD. Sham operated and PD-only mice were additional controls. CKD+PD mice displayed more severe atherosclerotic disease than control mice. Plaque area increased, and plaques were more advanced with a vulnerable phenotype typified by decreased collagen content and decreased fibrous cap thickness. Increased CD3+ T-cell numbers were present in plaques and perivascular adipose tissue of CKD and CKD+PD mice. Plaques of CKD+PD mice contained more iNOS+ immune cells. Spleens of CKD+PD mice showed more CD4+ central memory, terminally differentiated type 1 T-helper (Th1), Th17, and CX3C motif chemokine receptor 1+ (CX3CR1) CD4+ T-cells with less regulatory and effector T-cells. CONCLUSIONS: PD-fluid exposure in uremic mice potentiates systemic and vascular T-cell-driven inflammation and aggravates atherosclerosis. PD polarized CD4+ T-cells toward an inflammatory Th1/Th17 phenotype, and increased CX3CR1+ CD4+ T-cells, which are associated with vascular homing in CKD-associated atherosclerosis. Targeting CD4+ T-cell activation and CX3CR1+ polarization has the potential to attenuate atherosclerosis in PD patients.


Sujet(s)
Athérosclérose , Modèles animaux de maladie humaine , Dialyse péritonéale , Insuffisance rénale chronique , Urémie , Animaux , Athérosclérose/anatomopathologie , Athérosclérose/étiologie , Athérosclérose/immunologie , Athérosclérose/métabolisme , Athérosclérose/génétique , Urémie/immunologie , Urémie/métabolisme , Dialyse péritonéale/effets indésirables , Insuffisance rénale chronique/immunologie , Insuffisance rénale chronique/métabolisme , Souris invalidées pour les gènes ApoE , Souris , Plaque d'athérosclérose , Mâle , Souris de lignée C57BL , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Néphrectomie
16.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-39062887

RÉSUMÉ

Chronic kidney disease (CKD) causes specific hormonal disturbances, such as variations in leptin and testosterone levels and function. These disturbances can promote errors in signaling interaction and cellular information processing and can be implicated in the pathogenesis of atherosclerosis. This study investigates the factors that affect leptin in CKD patients and examines how leptin is related to markers of vascular disease. We conducted a cross-sectional study of 162 patients with CKD in pre-dialysis and dialysis stages. We recorded clinical and laboratory data, including leptin, testosterone, and subclinical atherosclerosis markers like brachial-ankle pulse wave velocity (ba PWV) in pre-dialysis CKD patients and flow-mediated vasodilation (FMD) and nitroglycerin-mediated vasodilation (NMD) in hemodialysis (HD) patients. Leptin was significantly correlated with testosterone in CKD pre-dialysis stages (p < 0.001) and also in HD (p = 0.026), with adipose tissue mass in pre-dialysis stages (p < 0.001), and also in HD (p < 0.001). In women HD patients, leptin correlated with NMD (p = 0.039; r = -0.379); in all HD patients, leptin correlated with C reactive protein (p = 0.007; r = 0.28) and parathormone (p = 0.039; r = -0.220). Our research emphasizes the connection between leptin, adipose tissue, and testosterone in all stages of CKD. Leptin was associated with NMD in HD women and correlated with inflammatory syndrome and parathyroid hormone in all HD patients.


Sujet(s)
Marqueurs biologiques , Leptine , Insuffisance rénale chronique , Testostérone , Humains , Leptine/sang , Leptine/métabolisme , Insuffisance rénale chronique/sang , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/physiopathologie , Insuffisance rénale chronique/complications , Femelle , Testostérone/sang , Testostérone/métabolisme , Mâle , Adulte d'âge moyen , Marqueurs biologiques/sang , Études transversales , Sujet âgé , Dialyse rénale/effets indésirables , Adulte , Vasodilatation , Analyse de l'onde de pouls
17.
Sci Rep ; 14(1): 16589, 2024 07 18.
Article de Anglais | MEDLINE | ID: mdl-39025899

RÉSUMÉ

Chronic exposure to heavy metals as aluminum chloride (AlCl3) could result in severe health hazards such as chronic renal injury. The present study aimed to evaluate the therapeutic potential of adipose tissue-derived stem cells (ASCs) in comparison to their microvesicles (MV) in AlCl3-induced chronic renal injury. Forty-eight adult male Wistar rats were divided into four groups: Control group, AlCl3-treated group, AlCl3/ASC-treated group, and AlCl3/MV-treated group. Biochemical studies included estimation of serum urea and creatinine levels, oxidative biomarkers assay, antioxidant biomarkers, serum cytokines (IL-1ß, IL-8, IL-10, and IL-33), real time-PCR analysis of renal tissue MALT1, TNF-α, IL-6, and serum miR-150-5p expression levels. Histopathological studies included light and electron microscopes examination of renal tissue, Mallory trichrome stain for fibrosis, Periodic acid Schiff (PAS) stain for histochemical detection of carbohydrates, and immunohistochemical detection of Caspase-3 as apoptosis marker, IL-1B as a proinflammatory cytokine and CD40 as a marker of MVs. AlCl3 significantly deteriorated kidney function, enhanced renal MDA and TOS, and serum cytokines concentrations while decreased the antioxidant parameters (SOD, GSH, and TAC). Moreover, serum IL-10, TNF-α, miR-150-5p, and renal MALT1 expression values were significantly higher than other groups. Kidney sections showed marked histopathological damage in both renal cortex and medulla in addition to enhanced apoptosis and increased inflammatory cytokines immunoexpression than other groups. Both ASCs and MVs administration ameliorated the previous parameters levels with more improvement was detected in MVs-treated group. In conclusion: ASCs-derived MVs have a promising ameliorating effect on chronic kidney disease.


Sujet(s)
Rat Wistar , Animaux , Mâle , Rats , Microparticules membranaires/métabolisme , Insuffisance rénale chronique/thérapie , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Cytokines/métabolisme , Cytokines/sang , Rein/anatomopathologie , Rein/métabolisme , Chlorure d'aluminium/effets indésirables , Stress oxydatif , Cellules souches/métabolisme , Tissu adipeux/métabolisme , Transplantation de cellules souches , Marqueurs biologiques/sang
18.
Molecules ; 29(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38893451

RÉSUMÉ

Chronic kidney disease (CKD) presents a formidable global health concern, affecting one in six adults over 25. This review explores the potential of phenolic compounds in managing CKD and its complications. By examining the existing research, we highlight their diverse biological activities and potential to combat CKD-related issues. We analyze the nutritional benefits, bioavailability, and safety profile of these compounds. While the clinical evidence is promising, preclinical studies offer valuable insights into underlying mechanisms, optimal dosages, and potential side effects. Further research is crucial to validate the therapeutic efficacy of phenolic compounds for CKD. We advocate for continued exploration of their innovative applications in food, pharmaceuticals, and nutraceuticals. This review aims to catalyze the scientific community's efforts to leverage phenolic compounds against CKD-related challenges.


Sujet(s)
Phénols , Insuffisance rénale chronique , Humains , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/métabolisme , Phénols/composition chimique , Phénols/usage thérapeutique , Phénols/pharmacologie , Animaux , Compléments alimentaires , Biodisponibilité
19.
Int J Mol Sci ; 25(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892221

RÉSUMÉ

Chronic kidney disease (CKD) presents a significant global health challenge, characterized by complex pathophysiology. This study utilized a multi-omic approach, integrating genomic data from the CKDGen consortium alongside transcriptomic, metabolomic, and proteomic data to elucidate the genetic underpinnings and identify therapeutic targets for CKD and kidney function. We employed a range of analytical methods including cross-tissue transcriptome-wide association studies (TWASs), Mendelian randomization (MR), summary-based MR (SMR), and molecular docking. These analyses collectively identified 146 cross-tissue genetic associations with CKD and kidney function. Key Golgi apparatus-related genes (GARGs) and 41 potential drug targets were highlighted, with MAP3K11 emerging as a significant gene from the TWAS and MR data, underscoring its potential as a therapeutic target. Capsaicin displayed promising drug-target interactions in molecular docking analyses. Additionally, metabolome- and proteome-wide MR (PWMR) analyses revealed 33 unique metabolites and critical inflammatory proteins such as FGF5 that are significantly linked to and colocalized with CKD and kidney function. These insights deepen our understanding of CKD pathogenesis and highlight novel targets for treatment and prevention.


Sujet(s)
Simulation de docking moléculaire , Insuffisance rénale chronique , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/traitement médicamenteux , Humains , Étude d'association pangénomique , Rein/métabolisme , Rein/anatomopathologie , Transcriptome , Protéomique/méthodes , Analyse de randomisation mendélienne , Prédisposition génétique à une maladie , Métabolomique/méthodes , Protéome/métabolisme , Métabolome , Multi-omique
20.
FASEB J ; 38(11): e23726, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38847773

RÉSUMÉ

Calcitriol and calcimimetics are used to treat hyperparathyroidism secondary to chronic kidney disease (CKD). Calcitriol administration and the subsequent increase in serum calcium concentration decrease parathyroid hormone (PTH) levels, which should reduce bone remodeling. We have previously reported that, when maintaining a given concentration of PTH, the addition of calcimimetics is associated with an increased bone cell activity. Whether calcitriol administration affects bone cell activity while PTH is maintained constant should be evaluated in an animal model of renal osteodystrophy. The aim of the present study was to compare in CKD PTH-clamped rats the bone effects of calcitriol and calcimimetic administration. The results show that the administration of calcitriol and calcimimetic at doses that induced a similar reduction in PTH secretion produced dissimilar effects on osteoblast activity in 5/6 nephrectomized (Nx) rats with secondary hyperparathyroidism and in Nx rats with clamped PTH. Remarkably, in both rat models, the administration of calcitriol decreased osteoblastic activity, whereas calcimimetic increased bone cell activity. In vitro, calcitriol supplementation inhibited nuclear translocation of ß-catenin and reduced proliferation, osteogenesis, and mineralization in mesenchymal stem cells differentiated into osteoblasts. In conclusion, besides the action of calcitriol and calcimimetics at parathyroid level, these treatments have specific effects on bone cells that are independent of the PTH level.


Sujet(s)
Calcimimétiques , Calcitriol , Ostéoblastes , Hormone parathyroïdienne , Animaux , Calcitriol/pharmacologie , Rats , Calcimimétiques/pharmacologie , Calcimimétiques/usage thérapeutique , Hormone parathyroïdienne/pharmacologie , Mâle , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Hyperparathyroïdie secondaire/traitement médicamenteux , Hyperparathyroïdie secondaire/étiologie , Hyperparathyroïdie secondaire/métabolisme , Os et tissu osseux/métabolisme , Os et tissu osseux/effets des médicaments et des substances chimiques , Rat Wistar , Insuffisance rénale/traitement médicamenteux , Insuffisance rénale/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/complications , Différenciation cellulaire/effets des médicaments et des substances chimiques , Calcium/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE