Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Genes Dis ; 11(4): 101126, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38560502

ABSTRACT

Dissecting the genetic components that contribute to the two main subphenotypes of steroid-sensitive nephrotic syndrome (SSNS) using genome-wide association studies (GWAS) strategy is important for understanding the disease. We conducted a multicenter cohort study (360 patients and 1835 controls) combined with a GWAS strategy to identify susceptibility variants associated with the following two subphenotypes of SSNS: steroid-sensitive nephrotic syndrome without relapse (SSNSWR, 181 patients) and steroid-dependent/frequent relapse nephrotic syndrome (SDNS/FRNS, 179 patients). The distribution of two single-nucleotide polymorphisms (SNPs) in ANKRD36 and ALPG was significant between SSNSWR and healthy controls, and that of two SNPs in GAD1 and HLA-DQA1 was significant between SDNS/FRNS and healthy controls. Interestingly, rs1047989 in HLA-DQA1 was a candidate locus for SDNS/FRNS but not for SSNSWR. No significant SNPs were observed between SSNSWR and SDNS/FRNS. Meanwhile, chromosome 2:171713702 in GAD1 was associated with a greater steroid dose (>0.75 mg/kg/d) upon relapse to first remission in patients with SDNS/FRNS (odds ratio = 3.14; 95% confidence interval, 0.97-9.87; P = 0.034). rs117014418 in APOL4 was significantly associated with a decrease in eGFR of greater than 20% compared with the baseline in SDNS/FRNS patients (P = 0.0001). Protein-protein intersection network construction suggested that HLA-DQA1 and HLA-DQB1 function together through GSDMA. Thus, SSNSWR belongs to non-HLA region-dependent nephropathy, and the HLA-DQA/DQB region is likely strongly associated with disease relapse, especially in SDNS/FRNS. The study provides a novel approach for the GWAS strategy of SSNS and contributes to our understanding of the pathological mechanisms of SSNSWR and SDNS/FRNS.

2.
Clin Immunol ; 260: 109914, 2024 03.
Article in English | MEDLINE | ID: mdl-38286173

ABSTRACT

OBJECTIVE: To investigate CD36 in ANCA-associated vasculitis (AAV), a condition characterized by monocyte/macrophage activation and vascular damage. METHODS: CD36 expression was assessed in AAV patients and healthy controls (HC). The impact of palmitic acid (PA) stimulation on multinucleate giant cell (MNGC) formation, macrophage, and endothelial cell activation, with or without CD36 knockdown, was examined. RESULTS: CD36 was overexpressed on AAV patients' monocytes compared to HC, regardless of disease activity. AAV patients exhibited elevated soluble CD36 levels in serum and plasma and PR3-ANCA patients' monocytes demonstrated increased MNGC formation following PA stimulation compared to HC. PA stimulation of macrophages or endothelial cells resulted in heightened CD36 expression, cell activation, increased macrophage migration inhibitory factor (MIF) production, and c-Myc expression, with attenuation upon CD36 knockdown. CONCLUSION: CD36 participates in macrophage and endothelial cell activation and MNGC formation, features of AAV pathogenesis. AAV treatment may involve targeting CD36 or MIF.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Antibodies, Antineutrophil Cytoplasmic , Humans , Antibodies, Antineutrophil Cytoplasmic/metabolism , Endothelial Cells/pathology , Macrophages/pathology , Giant Cells , Cytoplasm/pathology
3.
BMC Med ; 21(1): 417, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37924048

ABSTRACT

BACKGROUND: Accumulating evidence has suggested an oncogenic effect of diurnal disruption on cancer progression. To test whether targeting circadian rhythm by dietary strategy suppressed lung cancer progression, we adopted 6-h time-restricted feeding (TRF) paradigm to elucidate whether and how TRF impacts lung cancer progression. METHODS: This study used multiple lung cancer cell lines, two xenograft mouse models, and a chemical-treated mouse lung cancer model. Stable TIM-knockdown and TIM-overexpressing A549 cells were constructed. Cancer behaviors in vitro were determined by colony formation, EdU proliferation, wound healing, transwell migration, flow cytometer, and CCK8 assays. Immunofluorescence, pathology examinations, and targeted metabolomics were also used in tumor cells and tissues. mCherry-GFP-LC3 plasmid was used to detect autophagic flux. RESULTS: We found for the first time that compared to normal ad libitum feeding, 6-h TRF inhibited lung cancer progression and reprogrammed the rhythms of metabolites or genes involved in glycolysis and the circadian rhythm in tumors. After TRF intervention, only timeless (TIM) gene among five lung cancer-associated clock genes was found to consistently align rhythm of tumor cells to that of tumor tissues. Further, we demonstrated that the anti-tumor effect upon TRF was partially mediated by the rhythmic downregulation of the TIM and the subsequent activation of autophagy. Combining TRF with TIM inhibition further enhanced the anti-tumor effect, comparable to treatment efficacy of chemotherapy in xenograft model. CONCLUSIONS: Six-hour TRF inhibits lung cancer progression and reshapes circadian metabolism, which is partially mediated by the rhythmic downregulation of the TIM and the subsequent upregulation of autophagy.


Subject(s)
Lung Neoplasms , Humans , Mice , Animals , Lung , Circadian Rhythm/physiology , Intermittent Fasting , Disease Models, Animal
4.
Biochim Biophys Acta Mol Basis Dis ; 1869(7): 166800, 2023 10.
Article in English | MEDLINE | ID: mdl-37423141

ABSTRACT

BACKGROUND & AIMS: Fatty acid translocase CD36 (CD36/FAT) is a widely expressed membrane protein with multiple immuno-metabolic functions. Genetic CD36 deficiency is associated with increased risk of metabolic dysfunction-associated fatty liver disease (MAFLD) in patients. Liver fibrosis severity mainly affects the prognosis in patients with MAFLD, but the role of hepatocyte CD36 in liver fibrosis of MAFLD remains unclear. METHODS: A high-fat high-cholesterol diet and a high-fat diet with high-fructose drinking water were used to induce nonalcoholic steatohepatitis (NASH) in hepatocyte-specific CD36 knockout (CD36LKO) and CD36flox/flox (LWT) mice. Human hepG2 cell line was used to investigate the role of CD36 in regulating Notch pathway in vitro. RESULTS: Compared to LWT mice, CD36LKO mice were susceptible to NASH diet-induced liver injury and fibrosis. The analysis of RNA-sequencing data revealed that Notch pathway was activated in CD36LKO mice. LY3039478, an inhibitor of γ-secretase, inhibited Notch1 protein S3 cleavage and Notch1 intracellular domain (N1ICD) production, alleviating liver injury and fibrosis in CD36LKO mice livers. Likewise, both LY3039478 and knockdown of Notch1 inhibited the CD36KO-induced increase of N1ICD production, causing the decrease of fibrogenic markers in CD36KO HepG2 cells. Mechanistically, CD36 formed a complex with Notch1 and γ-secretase in lipid rafts, and hence CD36 anchored Notch1 in lipid rafts domains and blocked Notch1/γ-secretase interaction, inhibiting γ-secretase-mediated cleavage of Notch1 and the production of N1ICD. CONCLUSIONS: Hepatocyte CD36 plays a key role in protecting mice from diet-induced liver injury and fibrosis, which may provide a potential therapeutic strategy for preventing liver fibrogenesis in MAFLD.


Subject(s)
CD36 Antigens , Diet , Hepatocytes , Liver Cirrhosis , Non-alcoholic Fatty Liver Disease , Peptide Fragments , Receptor, Notch1 , Animals , Mice , Amyloid Precursor Protein Secretases/antagonists & inhibitors , CD36 Antigens/deficiency , CD36 Antigens/genetics , CD36 Antigens/metabolism , Diet/adverse effects , Gene Deletion , Hep G2 Cells , Hepatocytes/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Liver Cirrhosis/prevention & control , Membrane Microdomains , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/prevention & control , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phenotype , Receptor, Notch1/chemistry , Receptor, Notch1/metabolism , Signal Transduction , Humans
5.
Genes Dis ; 9(6): 1662-1673, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36157477

ABSTRACT

Understanding the association between the genetic and clinical phenotypes in children with nephrotic syndrome (NS) of different etiologies is critical for early clinical guidance. We employed whole-exome sequencing (WES) to detect monogenic causes of NS in a multicenter cohort of 637 patients. In this study, a genetic cause was identified in 30.0% of the idiopathic steroid-resistant nephrotic syndrome (SRNS) patients. Other than congenital nephrotic syndrome (CNS), there were no significant differences in the incidence of monogenic diseases based on the age at manifestation. Causative mutations were detected in 39.5% of patients with focal segmental glomerulosclerosis (FSGS) and 9.2% of those with minimal change disease (MCD). In terms of the patterns in patients with different types of steroid resistance, a single gene mutation was identified in 34.8% of patients with primary resistance, 2.9% with secondary resistance, and 71.4% of children with multidrug resistance. Among the various intensified immunosuppressive therapies, tacrolimus (TAC) showed the highest response rate, with 49.7% of idiopathic SRNS patients achieving complete remission. Idiopathic SRNS patients with monogenic disease showed a similar multidrug resistance pattern, and only 31.4% of patients with monogenic disease achieved a partial remission on TAC. During an average 4.1-year follow-up, 21.4% of idiopathic SRNS patients with monogenic disease progressed to end-stage renal disease (ESRD). Collectively, this study provides evidence that genetic testing is necessary for presumed steroid-resistant and idiopathic SRNS patients, especially those with primary and/or multidrug resistance.

6.
EBioMedicine ; 82: 104181, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35908416

ABSTRACT

BACKGROUND: Intestinal barrier dysfunction is crucial in alcohol-associated liver disease (ALD). The decreased beta-Klotho (KLB) expression caused by gene variation is associated with hyperpermeability in patients with irritable bowel syndrome. Here we investigated the roles of intestinal KLB in maintaining the intestinal epithelial barrier in ALD and the underlying mechanisms. METHODS: We constructed the intestine-specific overexpression KLB mice to investigate the role of KLB on alcohol-induced intestinal barrier dysfunction and liver injury in an ALD mouse model. To investigate the molecular mechanism in vitro, Caco2 cells were cultured and infected with the KLB overexpression lentivirus, or transfected with KLB/TRPV6 siRNA, or TRPV6/FXR1 overexpression plasmid, and treated with or without ethanol. FINDINGS: The upregulation of KLB in enterocytes effectively protected mice from alcohol-induced intestinal barrier hyperpermeability, thereby ameliorating hepatic steatosis and inflammation. KLB competitively suppressed FXR1 binding to the TRPV6 mRNA, increasing TRPV6 mRNA stability and protein abundance in intestinal epithelial cells. Furthermore, KLB formed a complex with TRPV6 and tight junction (TJ) proteins, protecting against alcohol-induced TJ proteins endocytosis and degradation as well as intestinal barrier impairment. INTERPRETATION: This work suggested that KLB attenuated alcohol-induced intestinal epithelial barrier dysfunction and liver injury through FXR1/TRPV6/TJ proteins pathway. FUNDING: National Natural Science Foundation of China, Chongqing Natural Science Foundation, Talent Project of Chongqing and the Science and Technology Research Program of Chongqing Municipal Education Commission.


Subject(s)
Klotho Proteins/metabolism , Liver Diseases, Alcoholic , Animals , Caco-2 Cells , Ethanol/metabolism , Ethanol/toxicity , Humans , Intestinal Mucosa/metabolism , Intestines , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/metabolism , Mice , Mice, Inbred C57BL , Protective Factors , RNA-Binding Proteins/metabolism
7.
Sheng Li Xue Bao ; 73(5): 805-812, 2021 Oct 25.
Article in Chinese | MEDLINE | ID: mdl-34708237

ABSTRACT

This study aimed to investigate the effects and the underlying mechanism of CD36 gene on glucose and lipid metabolism disorder induced by high-fat diet in mice. Wild type (WT) mice and systemic CD36 knockout (CD36-/-) mice were fed with high-fat diet for 14 weeks (n = 12). Mice were intraperitoneally injected with glucose (1 g/kg) or insulin (5 units/kg) to perform glucose tolerance test (GTT) or insulin tolerance test (ITT). Liver lipid deposition was observed by HE staining, and the contents of total triglyceride (TG), free fatty acid (FFA), aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in the serum were determined by automatic biochemical analyzer. Real-time PCR and Western blot were used to detect insulin signaling pathways in liver and muscle tissues of mice. The mRNA levels of genes encoding phosphoenolpyruvate carboxykinase (PEPCK) in primary hepatocytes of mice were detected by real-time PCR, and glucose detection kit was used to detect gluconeogenesis. Co-immunoprecipitation (Co-IP) and ELISA were used to detect insulin receptor ß (IRß) tyrosine phosphorylation in mouse muscle. Real-time PCR and immunofluorescence staining (IF) were used to detect the expression and location of glucose transporter 4 (GLUT4) in muscle of mice. After high-fat diet feeding, serum FFA, TG, AST and ALT levels of CD36-/- mice were significantly higher than WT mice (P < 0.05). The appearance of CD36-/- mouse liver presented fatty degeneration, and HE staining results showed increased lipid accumulation in the liver, suggesting that CD36 knockout promoted the occurrence of fatty liver. However, CD36-/- mice showed decreased fasting glucose levels, increased glucose tolerance, and decreased insulin tolerance compared with WT mice (P < 0.05), suggesting that CD36 knockout protects against the abnormal glucose metabolism induced by high-fat diet. Compared with WT mice, there was no significant difference in insulin signaling pathway in CD36-/- mouse liver, and there were no significant differences in PEPCK expression and gluconeogenesis between the two groups of primary hepatocytes. In muscle tissue, Co-IP and ELISA experiments showed that the phosphorylation level of IRß tyrosine was significantly increased in CD36-/- mice compared with that in WT mice. Besides, the levels of p-AKT in CD36-/- mouse muscle were significantly increased (P < 0.05). At the same time, IF experiment indicated that GLUT4 localization in cell membrane was enhanced in the muscle of CD36-/- mice, indicating that insulin sensitivity and glucose utilization ability were enhanced in CD36-/- mouse muscle. The results suggested that deletion of CD36 gene increased lipid accumulation in liver of mice with high-fat diet, but had no significant effect on liver gluconeogenesis. CD36 deficiency improves the abnormal glucose metabolism in mice with high-fat diet mainly through improving insulin sensitivity of muscle tissue and promoting GLUT4-mediated glucose utilization.


Subject(s)
Fatty Liver , Insulin Resistance , Animals , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Glucose/metabolism , Insulin/metabolism , Lipid Metabolism , Liver , Mice , Triglycerides
8.
Sheng Li Xue Bao ; 73(5): 813-820, 2021 Oct 25.
Article in Chinese | MEDLINE | ID: mdl-34708238

ABSTRACT

This study aimed to investigate the effect of lipopolysaccharide (LPS) on lipophagy in hepatocytes and the underlying mechanism. Human hepatoma cell line HepG2 was cultured in vitro, treated with 0.1 mmol/L palmitic acid (PA), and then divided into control group (0 µg/mL LPS), LPS group (10 µg/mL LPS), LPS+DMSO group and LPS+RAPA (rapamycin, 10 µmol/L) group. Lipid accumulation in hepatocytes was observed by oil red O staining. The autophagic flux of the cells was assessed using confocal laser scanning microscope after being transfected with autophagy double-labeled adenovirus (mRFP-GFP-LC3). The level of intracellular lipophagy was visualized by the colocalization of lipid droplets (BODIPY 493/503 staining) and lysosomes (lysosome marker, lysosomal associated membrane protein 1, LAMP1). The expression levels of mammalian target of rapamycin (mTOR), phosphorylated mTOR (p-mTOR), ribosome protein subunit 6 kinase 1 (S6K1), p-S6K1, LC3II/I and P62 protein were examined by Western blot. The results showed that the number of red lipid droplets stained with oil red O was significantly increased in LPS group compared with that in control group (P < 0.001). Moreover, in LPS group, the number of autophagosomes was increased, while the number of autophagolysosomes and the colocalization rate of LAMP1 and BODIPY were significantly decreased (P < 0.05). Meanwhile, the ratios of p-mTOR/mTOR and p-S6K1/S6K1, the ratio of LC3II/LC3I and the protein expression of P62 were significantly increased (P < 0.05) in LPS group. Furthermore, compared with LPS+DMSO group, RAPA treatment obviously reduced the number of lipid droplets and autophagosomes, and raised the number of autophagolysosomes and the colocalization rate of LAMP1 and BODIPY (P < 0.05). In conclusion, the results demonstrate that LPS inhibits lipophagy in HepG2 cells via activating mTOR signaling pathway, thereby aggravating intracellular lipid accumulation.


Subject(s)
Lipopolysaccharides , TOR Serine-Threonine Kinases , Autophagy , Hep G2 Cells , Humans , Palmitic Acid , Signal Transduction
9.
Front Cell Dev Biol ; 9: 644363, 2021.
Article in English | MEDLINE | ID: mdl-33748139

ABSTRACT

Hyperphosphatemia or even serum phosphate levels within the "normal laboratory range" are highly associated with increased cardiovascular disease risk and mortality in the general population and patients suffering from chronic kidney disease (CKD). As the kidney function declines, serum phosphate levels rise and subsequently induce the development of hypertension, vascular calcification, cardiac valvular calcification, atherosclerosis, left ventricular hypertrophy and myocardial fibrosis by distinct mechanisms. Therefore, phosphate is considered as a promising therapeutic target to improve the cardiovascular outcome in CKD patients. The current therapeutic strategies are based on dietary and pharmacological reduction of serum phosphate levels to prevent hyperphosphatemia in CKD patients. Large randomized clinical trials with hard endpoints are urgently needed to establish a causal relationship between phosphate excess and cardiovascular disease (CVD) and to determine if lowering serum phosphate constitutes an effective intervention for the prevention and treatment of CVD.

10.
Acta Pharmacol Sin ; 41(8): 1111-1118, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32203081

ABSTRACT

Some studies have shown that gut microbiota along with its metabolites is closely associated with diabetic mellitus (DM). In this study we explored the relationship between gut microbiota and kidney injuries of early diabetic nephropathy (DN) and its underlying mechanisms. Male SD rats were intraperitoneally injected with streptozotocin to induce DM. DM rats were orally administered compound broad-spectrum antibiotics for 8 weeks. After the rats were sacrificed, their blood, urine, feces, and renal tissues were harvested for analyses. We found that compared with the control rats, DM rats had abnormal intestinal microflora, increased plasma acetate levels, increased proteinuria, thickened glomerular basement membrane, and podocyte foot process effacement in the kidneys. Furthermore, the protein levels of angiotensin II, angiotensin-converting enzyme, and angiotensin II type 1 receptor in the kidneys of DM rats were significantly increased. Administration of broad-spectrum antibiotics in DM rats not only completely killed most intestinal microflora, but also significantly lowered the plasma acetate levels, inhibited intrarenal RAS activation, and attenuated kidney damage. Finally, we showed that plasma acetate levels were positively correlated with intrarenal angiotensin II protein expression (r = 0.969, P < 0.001). In conclusion, excessive acetate produced by disturbed gut microbiota might be involved in the kidney injuries of early DN through activating intrarenal RAS.


Subject(s)
Acetates/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/physiopathology , Dysbiosis/physiopathology , Gastrointestinal Microbiome/physiology , Renin-Angiotensin System/physiology , Acetates/blood , Animals , Anti-Bacterial Agents/pharmacology , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Gastrointestinal Microbiome/drug effects , Kidney/pathology , Male , Rats, Sprague-Dawley
11.
Front Pharmacol ; 10: 1040, 2019.
Article in English | MEDLINE | ID: mdl-31607907

ABSTRACT

Background: Metabolic syndrome (MS) is one of the major causes of coronary artery diseases (CAD). Gut microbiome diversity and its natural fermentation products are not only correlated with MS and CAD, but their correlations also appear to be stronger than the associations with traditional risk factors. Therefore, the aim of this study was to provide a new potential pathway for the natural fermentation product butyrate to improve MS and to examine whether it is associated with serum metabolic profiles and gut flora composition. Methods: C57BL/6J mice fed a high-fat diet (HFD) were treated with 400 mg/kg of sodium butyrate for 16 weeks. Blood and fecal samples were collected, and the metabolite concentrations and 16s rRNA were measured with liquid chromatography-MS and Illumina platform, respectively. The plasma differential metabolites and gut microbiome composition were analyzed with XCMS online and QIIME 2, respectively. Results: Gut microbiome-derived butyrate reduced glucose intolerance and insulin resistance, resisting HFD-induced increase in the relative abundance of f_Lachnospiraceae, f_Rikenellaceae, and f_Paraprevotellaceae. Meanwhile, sodium butyrate increased the levels of α-linolenate, all-trans-retinal, resolvin E1, and leukotriene in the plasma, and the differential pathways showed enrichment in mainly resolvin E biosynthesis, histidine degradation, lipoxin biosynthesis, and leukotriene biosynthesis. Moreover, sodium butyrate increased the levels of phosphorylated-adenosine 5'-monophosphate-activated protein kinase (p-AMPK) and facilitated glucose transporter member 4 (GLUT4) in the adipose tissue. Conclusion: Butyrate can induce AMPK activation and GLUT4 expression in the adipose tissue, improving cardiovascular disease (CVD)-related metabolic disorder, resisting HFD-induced gut microbiome dysbiosis, and promoting resolvin E1 and lipoxin biosynthesis. Oral supplement of the natural fermentation product butyrate can be a potential strategy for preventing CVD.

12.
Adv Exp Med Biol ; 1165: 525-541, 2019.
Article in English | MEDLINE | ID: mdl-31399983

ABSTRACT

Since the lipid nephrotoxicity hypothesis was proposed in 1982, increasing evidence has supported the hypothesis that lipid abnormalities contributed to the progression of glomerulosclerosis. In this chapter, we will discuss the general promises of the original hypothesis, focusing especially on the role of lipids and metabolic inflammation accompanying CKD in renal fibrosis and potential new strategies of prevention.


Subject(s)
Kidney Diseases/physiopathology , Lipid Metabolism Disorders/physiopathology , Disease Progression , Fibrosis , Humans , Inflammation , Lipids
13.
J Cell Physiol ; 234(12): 22352-22364, 2019 12.
Article in English | MEDLINE | ID: mdl-31074042

ABSTRACT

C1q/tumor necrosis factor-related protein-3 (CTRP3) has been extensively reported as an important role involved in antifibrosis, antiapoptosis, and anti-inflammation. However, the role of CTRP3 involved in renal fibrosis remains unclear. Our current study explored the role of CTRP3 in renal fibrosis and its underlying mechanisms by using serums and renal biopsy specimens from renal fibrosis patients and control subjects, rats models with the surgery of unilateral ureteral obstruction (UUO) and human renal proximal tubular epithelial cells (HRPTEpiCs). We found that circulating levels of CTRP3 had no significant difference between renal fibrosis patients and healthy subjects; however, renal CTRP3 expression was markedly downregulated in the fibrotic region with an abundant expression of collagen-I. In UUO rat models, circulating levels of CTRP3 have not changed with the prolonged obstruction of the kidney; renal CTRP3 expression was decreased with the severity of renal fibrosis; adenovirus-mediated CTRP3 treatment inhibited renal interstitial fibrosis. In vitro experiments revealed that CTRP3 attenuates TGF-ß1 induced tubular epithelial cells fibrotic changes; CTRP3 knockdown facilitates the expression of fibrotic markers in TGF-ß1-induced HRPTEpiCs; recombinant CTRP3 or adenovirus-mediated CTRP3 overexpression significantly inhibited the Notch signaling pathway-associated factors, and knockdown of CTRP3 increased TGF-ß1-mediated activation of the Notch signaling pathways. Collectively, our current study found that CTRP3 could improve renal fibrosis, to some extent, through inhibiting the Notch pathway.


Subject(s)
Kidney/pathology , Receptors, Notch/metabolism , Signal Transduction , Tumor Necrosis Factors/metabolism , Adenoviridae/metabolism , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Gene Silencing , Humans , Kidney Tubules, Proximal/pathology , Male , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factors/blood , Ureteral Obstruction/blood , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
14.
Nat Metab ; 1(11): 1141-1156, 2019 11.
Article in English | MEDLINE | ID: mdl-32694864

ABSTRACT

The central pacemaker in the hypothalamic suprachiasmatic nucleus (SCN) synchronizes peripheral oscillators to coordinate physiological and behavioural activities throughout the body. How circadian phase coherence between the SCN and the periphery is controlled is not well understood. Here, we identify hepatic SIRT7 as an early responsive element to light that ensures circadian phase coherence in the mouse liver. The SCN-driven body temperature (BT) oscillation induces rhythmic expression of HSP70, which promotes SIRT7 ubiquitination and proteasomal degradation. Acute temperature challenge dampens the BT oscillation and causes an advanced liver circadian phase. Further, hepatic SIRT7 deacetylates CRY1, promotes its FBXL3-mediated degradation and regulates the hepatic clock and glucose homeostasis. Loss of Sirt7 in mice leads to an advanced liver circadian phase and rapid entrainment of the hepatic clock upon daytime-restricted feeding. These data identify a BT-HSP70-SIRT7-CRY1 axis that couples the mouse hepatic clock to the central pacemaker and ensures circadian phase coherence and glucose homeostasis.


Subject(s)
Body Temperature , Circadian Rhythm , Gluconeogenesis , Light , Liver/metabolism , Sirtuins/metabolism , Animals , Homeostasis , Mice
15.
Acta Pharmacol Sin ; 40(4): 468-476, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30446735

ABSTRACT

Platelet microparticles (PMPs) are closely associated with diabetic macrovascular complications. The present study aimed to investigate the effects of PMPs in diabetes on aortic vascular endothelial injury and to explore the underlying mechanisms. Peritoneal injection of streptozotocin was used to generate a diabetic rat model in vivo, and human umbilical vein endothelial cells (HUVECs) treated with PMPs were used in vitro. PMP levels in the circulation and aorta tissues were time-dependently increased in streptozotocin-induced diabetic rats at weeks 4, 8, and 12 (P < 0.05). Aspirin significantly inhibited the PMP levels at each time point (P < 0.05). In diabetic rats, the endothelial nitric oxide levels were decreased significantly combined with increased endothelial permeability. PMPs were internalized by HUVECs and primarily accumulated around the nuclei. PMPs inhibited endothelial nitric oxide levels to about 50% and caused approximately twofold increase in reactive oxygen species production. Furthermore, PMPs significantly decreased the endothelial glycocalyx area and expression levels of glypican-1 and occludin (P < 0.05). Interestingly, the PMP-induced endothelial injuries were prevented by raptor siRNA and rapamycin. In conclusion, increased PMPs levels contribute to aortic vascular endothelial injuries in diabetes through activating the mTORC1 pathway.


Subject(s)
Blood Platelets/chemistry , Cell-Derived Microparticles/metabolism , Diabetes Mellitus, Experimental/metabolism , Endothelium, Vascular/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Animals , Cell-Derived Microparticles/chemistry , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Endothelium, Vascular/pathology , Humans , Male , Rats , Rats, Sprague-Dawley , Streptozocin
16.
Int Urol Nephrol ; 50(11): 2049-2059, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30073617

ABSTRACT

PURPOSE: The heart and kidney are of utmost importance for the maintenance of cardiovascular (CV) homeostasis. The relationship between cardiac remodeling, especially the left ventricular hypertrophy (LVH) and renal damage reflected by the estimated glomerular filtration rate (eGFR), decline in type 2 diabetes mellitus (T2DM) patients is unclear. And it is also unknown whether cardiac remodeling can be used to assess the eGFR decline in T2DM patients. METHODS: We retrospectively analyzed the relationship between cardiac remodeling especially the LVH and the eGFR decline for 265 patients with T2DM, who were diagnosed between 2011 and 2015 and followed for ≥ 3 months. The parameters of cardiac remodeling were determined using Doppler echocardiography. RESULTS: In the Cox regression model, the parameters of cardiac remodeling were associated with the composite endpoint in different models. These associations were independent of age, body mass index (BMI), history of hypertension, duration of diabetes, the baseline eGFR, 24-h urinary protein, or using angiotensin-converting enzyme inhibitors (ACEI) and (or) angiotensin receptor blockers (ARB). The risk of composite endpoint in patients with T2DM was higher (hazard ratio, 10.832; p < 0.001 for trend) in the group with the highest number of abnormal echocardiographic parameters, than in the group with no abnormal echocardiographic parameters. In receiver operating characteristics (ROC) curve analyses, the parameter of left ventricular posterior wall (LVPW) thickness was superior to the other parameters of cardiac remodeling as represented by the higher area under the curve (AUC) values generated according to the sensitivity and specificity. CONCLUSION: Echocardiographic parameters are strongly correlated with the eGFR decline in patients with T2DM. Moreover, the severity of cardiac remodeling, especially the LVH is closely associated with the eGFR decline in patients with T2DM. Therefore, the recognition of cardiac structural alterations in patients with T2DM may evaluate renal damage at an early stage.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glomerular Filtration Rate , Hypertrophy, Left Ventricular/etiology , Adult , Aged , Diabetes Mellitus, Type 2/complications , Echocardiography, Doppler , Female , Humans , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/physiopathology , Male , Middle Aged , Predictive Value of Tests , Retrospective Studies , Risk Factors , Ventricular Remodeling
17.
Lipids Health Dis ; 17(1): 153, 2018 Jul 17.
Article in English | MEDLINE | ID: mdl-30016988

ABSTRACT

BACKGROUND: Accumulating evidence suggests that activated hepatocytes are involved in the deposition of the excess extracellular matrix during liver fibrosis via the epithelial to mesenchymal transition. Lipid accumulation in hepatocytes are implicated in the pathogenesis of chronic liver injury. CD36 is known to mediate long-chain fatty acid (LCFA) uptake and lipid metabolism. However, it is unclear whether LCFA directly promotes hepatocyte activation and the involved mechanisms have not been fully clarified. METHODS: Mice were fed with a high fat diet (HFD) and normal hepatocyte cells (Chang liver cells) were treated with palmitic acid (PA) in vivo and in vitro. Real-time polymerase chain reaction (RT-PCR) and western blotting were used to examine the gene and protein expression of molecules involved in hepatic fibrogenesis and hepatocyte activation. CD36 was knocked down by transfecting CD36 siRNA into hepatocyte cells. Hydrogen peroxide (H2O2) and reactive oxygen species (ROS) levels were detected using commercial kits. RESULTS: HFD induced a profibrogenic response and up-regulated CD36 expression in vivo. Analogously, PA increased lipid accumulation and induced human hepatocyte activation in vitro, which was also accompanied by increased CD36 expression. Interestingly, knockdown of CD36 resulted in a reduction of hepatocyte lipid deposition and decreased expression of Acta2 (34% decrease), Vimentin (29% decrease), Desmin (60% decrease), and TGF-ß signaling pathway related genes. In addition, HFD and PA increased the production of H2O2 in vivo (48% increase) and in vitro (385% increase), and the antioxidant, NAC, ameliorated PA-induced hepatocyte activation. Furthermore, silencing of CD36 in vitro markedly attenuated PA-induced oxidative stress (H2O2: 41% decrease; ROS: 39% decrease), and the anti-activation effects of CD36 knockdown could be abolished by pretreatment with H2O2. CONCLUSIONS: Our study demonstrated that LCFA facilitates hepatocyte activation by up-regulating oxidative stress through CD36, which could be an important mechanism in the development of hepatic fibrosis.


Subject(s)
CD36 Antigens/genetics , Diet, High-Fat/adverse effects , Liver Cirrhosis/genetics , Non-alcoholic Fatty Liver Disease/genetics , Oxidative Stress/drug effects , Palmitic Acid/pharmacology , Actins/genetics , Actins/metabolism , Animals , CD36 Antigens/antagonists & inhibitors , CD36 Antigens/metabolism , Cell Line , Desmin/genetics , Desmin/metabolism , Gene Expression Regulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Hydrogen Peroxide/agonists , Hydrogen Peroxide/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vimentin/genetics , Vimentin/metabolism
18.
Kidney Blood Press Res ; 43(1): 206-219, 2018.
Article in English | MEDLINE | ID: mdl-29490300

ABSTRACT

BACKGROUND/AIMS: Chronic kidney disease (CKD) is often accompanied by hyperlipidemia, which accelerates progression of the disease. Podocyte injury can lead to dysfunction of the glomerular filtration barrier, which is associated with proteinuria, a risk marker for the progression of CKD. Our previous studies demonstrated that palmitic acid (PA) can induce podocyte apoptosis; however, the underlying mechanisms are unclear. In the present study, we investigated the specific molecular mechanisms of PA-induced apoptosis in cultured podocytes. METHODS: We cultured mouse podocytes and treated them with PA. Then, cell viability was measured using the Cell Counting Kit-8 colorimetric assay, lipid uptake was assessed by Oil Red O staining and boron-dipyrromethene staining, apoptosis was measured by flow cytometry, mitochondrial injury was assessed by JC-1 staining and transmission electron microscopy, and mitochondrial production of reactive oxygen species (ROS) was evaluated by fluorescence microscopy using the MitoSOX Red reagent. The effects of PA on the mitochondria-mediated caspase activation pathway were investigated by examining the expression of caspase-8, cleaved caspase-9, cleaved caspase-3, cleaved poly (ADP-ribose) polymerase (PARP), B-cell lymphoma 2 (Bcl-2), Bax, Bid, cytochrome c, and Fas-associated protein with death domain (FADD) using western blotting. The translocation of Bax and cytochrome c were detected by immunofluorescence. RESULTS: PA treatment significantly increased lipid accumulation and induced podocyte apoptosis. We investigated whether the two primary apoptosis signaling pathways (death receptor-mediated pathway and mitochondria-mediated pathway) were involved in the execution of PA-induced podocyte apoptosis, and found that the levels of FADD, caspase-8, and Bid did not significantly change during this process. Meanwhile, PA treatment induced an increase in Bax protein expression and a decrease in Bcl-2 protein expression, with Bax translocation to the mitochondria. Furthermore, PA treatment induced mitochondrial impairment, and triggered the release of cytochrome c from the mitochondria to cytosol, with a concomitant dose-dependent increase in the levels of cleaved caspase-9, cleaved caspase-3, and PARP. Meanwhile, PA treatment increased mitochondrial production of ROS, and the mitochondria-targeted antioxidant mitoTEMPO significantly ameliorated PA-induced podocyte apoptosis. CONCLUSION: Our findings indicated that PA induced caspase-dependent podocyte apoptosis through the mitochondrial pathway, and mitochondrial ROS production participated in this process, thus potentially contributing to podocyte injury.


Subject(s)
Apoptosis/drug effects , Mitochondria/metabolism , Palmitic Acid/pharmacology , Podocytes/cytology , Reactive Oxygen Species/metabolism , Animals , Caspases/metabolism , Cells, Cultured , Mice , Podocytes/drug effects
19.
Med Sci Monit ; 24: 1484-1492, 2018 Mar 12.
Article in English | MEDLINE | ID: mdl-29528039

ABSTRACT

BACKGROUND Increased lipid accumulation in renal tubular epithelial cells (TECs) contributes to their injury and dysfunction and progression of tubulointerstitial fibrosis. Berberine (BBR), a natural plant alkaloid isolated from traditional medicine herbs, is effective in lowing serum lipid, and has a protective effect on chronic kidney disease (CKD) with dyslipidemia, including diabetic nephropathy. The aim of this study was to investigate the effect of BBR on palmitate (PA)-induced lipid accumulation and apoptosis in TECs. MATERIAL AND METHODS Human kidney proximal tubular epithelial cell line (HK-2) cells were treated with PA, BBR, and/or palmitoyltransferase 1A (CPT1A) inhibitor Etomoxir. Intracellular lipid content was assessed by Oil Red O and Nile Red staining. Cell apoptosis rate was evaluated by flow cytometry assay. The expression of apoptosis-related protein cleaved-caspase3 and fatty acid oxidation (FAO)-regulating proteins, including CPT1A, peroxisome proliferator-activated receptor α (PPARα), and PPARγ co-activator-1α (PGC1α), was measured by Western blot analysis and immunofluorescence. RESULTS In the present study, PA treatment increased intracellular lipid deposition accompanied by elevated apoptosis in TECs compared with control group, whereas the protein expression of CPT1A, PPARα, and PGC1α, did not correspondingly increase in TECs. BBR significantly up-regulated the protein expression of CPT1A, PPARα, and PGC1α in TECs treated with or without PA, and reversed PA-induced intracellular lipid accumulation and apoptosis. Moreover, the CPT1A inhibitor Etomoxir counteracted the protective effect of BBR in TECs. CONCLUSIONS These in vitro findings suggest that PA can induce intracellular lipid accumulation and apoptosis in TECs, and the mechanism may be associated with inducing defective FAO, whereas BBR can protect TECs against PA-induced intracellular lipid accumulation and apoptosis by promoting FAO.


Subject(s)
Apoptosis/drug effects , Berberine/pharmacology , Epithelial Cells/pathology , Kidney Tubules/pathology , Palmitates/toxicity , Protective Agents/pharmacology , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/metabolism , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lipid Metabolism/drug effects , Oxidation-Reduction/drug effects
20.
Circ Res ; 122(5): 730-741, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29301853

ABSTRACT

RATIONALE: An elevated level of plasma LDL (low-density lipoprotein) is an established risk factor for cardiovascular disease. Recently, we reported that the (pro)renin receptor ([P]RR) regulates LDL metabolism in vitro via the LDLR (LDL receptor) and SORT1 (sortilin-1), independently of the renin-angiotensin system. OBJECTIVES: To investigate the physiological role of (P)RR in lipid metabolism in vivo. METHODS AND RESULTS: We used N-acetylgalactosamine modified antisense oligonucleotides to specifically inhibit hepatic (P)RR expression in C57BL/6 mice and studied the consequences this has on lipid metabolism. In line with our earlier report, hepatic (P)RR silencing increased plasma LDL-C (LDL cholesterol). Unexpectedly, this also resulted in markedly reduced plasma triglycerides in a SORT1-independent manner in C57BL/6 mice fed a normal- or high-fat diet. In LDLR-deficient mice, hepatic (P)RR inhibition reduced both plasma cholesterol and triglycerides, in a diet-independent manner. Mechanistically, we found that (P)RR inhibition decreased protein abundance of ACC (acetyl-CoA carboxylase) and PDH (pyruvate dehydrogenase). This alteration reprograms hepatic metabolism, leading to reduced lipid synthesis and increased fatty acid oxidation. As a result, hepatic (P)RR inhibition attenuated diet-induced obesity and hepatosteatosis. CONCLUSIONS: Collectively, our study suggests that (P)RR plays a key role in energy homeostasis and regulation of plasma lipids by integrating hepatic glucose and lipid metabolism.


Subject(s)
Fatty Liver/metabolism , Hepatocytes/metabolism , Lipid Metabolism , Obesity/metabolism , Receptors, Cell Surface/metabolism , Acetyl-CoA Carboxylase/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Gene Silencing , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Pyruvate Dehydrogenase Complex/metabolism , Receptors, Cell Surface/genetics , Prorenin Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...