Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Cell Mol Life Sci ; 80(6): 168, 2023 May 30.
Article in English | MEDLINE | ID: mdl-37249651

ABSTRACT

Hepatic glucose and lipid metabolism disorders promote the development and progression of type 2 diabetes mellitus (T2DM), yet the underlying mechanisms are not fully understood. Here, we identify tripartite motif-containing protein 21 (TRIM21), a class IV TRIM family member, as a pivotal regulator of hepatic metabolism in T2DM for the first time. Bioinformatic analysis suggests that TRIM21 expression is significantly reduced in T2DM patients. Intriguingly, in a mouse model of obese diabetes, TRIM21 expression is predominantly reduced in the liver rather than in other metabolic organs. It is further demonstrated that hepatic overexpression of TRIM21 significantly ameliorates glucose intolerance, insulin resistance, hepatic steatosis, and dyslipidemia in obese diabetic mice. In contrast, the knockdown of TRIM21 promotes glucose intolerance, insulin resistance, and triglyceride accumulation. Mechanistically, both phosphoenolpyruvate carboxykinase 1 (PEPCK1) and fatty acid synthase (FASN) are the hepatic targets of TRIM21. We revealed that TRIM21 promotes the degradation of PEPCK1 and FASN through a direct protein-protein interaction mediated K48-linked ubiquitination. Notably, overexpression of PEPCK1 and FASN essentially abolished the beneficial effects achieved by TRIM21 overexpression in obese diabetic mice. Overall, our data demonstrate that TRIM21 is a novel regulator of hepatic metabolic disorder, and suggest TRIM21 as a promising therapeutic target for T2DM.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Glucose Intolerance , Insulin Resistance , Lipid Metabolism Disorders , Animals , Mice , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/therapeutic use , Glucose/metabolism , Glucose Intolerance/metabolism , Lipid Metabolism Disorders/metabolism , Lipids , Liver/metabolism , Obesity/metabolism , Ubiquitination , Humans
2.
Cell Mol Life Sci ; 79(3): 184, 2022 Mar 12.
Article in English | MEDLINE | ID: mdl-35279748

ABSTRACT

The pathogenesis of acute kidney injury (AKI) is associated with the activation of multiple signaling pathways, including Wnt/ß-catenin signaling. However, the mechanism of Wnt/ß-catenin pathway activation in renal interstitial fibroblasts during AKI is unclear. S100 calcium-binding protein A16 (S100A16), a new member of calcium-binding protein S100 family, is a multi-functional signaling factor involved in various pathogenies, including tumors, glycolipid metabolism disorder, and chronic kidney disease (CKD). We investigated the potential participation of S100A16 in Wnt/ß-catenin pathway activation during AKI by subjecting wild-type (WT) and S100A16 knockout (S100A16+/-) mice to the ischemia-reperfusion injury (IRI), and revealed S100A16 upregulation in this model, in which knockout of S100A16 impeded the Wnt/ß-catenin signaling pathway activation and recovered the expression of downstream hepatocyte growth factor (HGF). We also found that S100A16 was highly expressed in Platelet-derived growth factor receptor beta (PDGFRß) positive renal fibroblasts in vivo. Consistently, in rat renal interstitial fibroblasts (NRK-49F cells), both hypoxia/reoxygenation and S100A16 overexpression exacerbated fibroblasts apoptosis and inhibited HGF secretion; whereas S100A16 knockdown or Wnt/ß-catenin pathway inhibitor ICG-001 reversed these changes. Mechanistically, we showed that S100A16 promoted Wnt/ß-catenin signaling activation via the ubiquitylation and degradation of ß-catenin complex members, glycogen synthase kinase 3ß (GSK3ß) and casein kinase 1α (CK1α), mediated by E3 ubiquitin ligase, the HMG-CoA reductase degradation protein 1 (HRD1). Our study identified the S100A16 as a key regulator in the activation of Wnt/ß-catenin signaling pathway in AKI.


Subject(s)
Acute Kidney Injury/pathology , Casein Kinase Ialpha/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , S100 Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Acute Kidney Injury/metabolism , Animals , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/metabolism , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Knockout , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , S100 Proteins/antagonists & inhibitors , S100 Proteins/deficiency , S100 Proteins/genetics , Ubiquitination , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , bcl-2-Associated X Protein/metabolism
3.
Pak J Med Sci ; 39(4): 950-955, 2023.
Article in English | MEDLINE | ID: mdl-37492335

ABSTRACT

Objective: To explore the diagnostic value of contrast-enhanced ultrasound (CEUS) combined with magnetic resonance imaging (MRI) for cervical abnormal lymph nodes. Methods: We retrospectively reviewed the clinical records of 150 patients undergoing lymph node examinations at Hangzhou Chest Hospital from January 2017 to December 2019. According to the characteristics of lymph nodes, the patients were divided into three groups: 45 patients had hyperplastic lymph nodes (HLNs; Group-A), 55 had lymph node tuberculosis (LNTB; Group-B), 50 had metastatic lymph nodes (MLN; Group-C). We compared the ultrasonic examination and MRI results between the groups, and compared the diagnostic value of CEUS alone and CEUS plus MRI. Results: Lower resistance indexes (RI) for Groups-A and B than Group-C(P<0.05). Mixed blood flow type was predominant in Group-A, while the lymphohilum type was predominant in Group-B, and the marginal type was predominant in Group-C(P<0.05). The proportion of non-uniform types in Group-B was significantly higher than that in Groups-A and C(P<0.05). After enhancement, the proportions of non-uniform types in Groups-A and B were higher than Group-C(P<0.05). The results of MRI examination showed that positive reinforcement integral (PEI) and maximum slope of increase (MSI) values increased sequentially from Group-B to Group-A, and then to Group-C(P<0.05); while time to peak (TTP) values increased sequentially from Group-C to Group-A, and then to Group-B(P<0.05). The diagnosis accuracy of CEUS combined with MRI was significantly higher than that of CEUS alone(P<0.05). RI-PEI, RI-MSI, and RI-TTP showed high specificity and sensitivity in the diagnosis and differentiation of HLNs, LNTB, and MLNs(P<0.05). Conclusion: CEUS combined with MRI can significantly facilitate the differential diagnosis between HLNs, LNTB, and MLNs. The two diagnosis methods combined improve the diagnosis accuracy of cervical lymph node diseases.

4.
Immunology ; 163(4): 478-492, 2021 08.
Article in English | MEDLINE | ID: mdl-33786850

ABSTRACT

Age-related thymic atrophy results in reduced output of naïve conventional T (Tcon) cells. However, its impact on regulatory T (Treg) cells is insufficiently understood. Given evidence that thymic Treg (tTreg) cell generation is enhanced in the aged, atrophy thymus and that the aged periphery accumulates peripheral Treg (pTreg) cells, we asked why these Treg cells are unable to effectively attenuate increased autoreactivity-induced chronic inflammation in the elderly. We designed a mock-self-antigen chimera mouse model, in which membrane-bound ovalbumin (mOVA) transgenic mice, bearing a FoxN1-floxed gene for induction of conditional thymic atrophy, received OVA-specific (OT-II) T-cell receptor (TCR) transgenic progenitor cells. The chimeric mice with thymic atrophy exhibited a significant decrease in OVA-specific tTreg and pTreg cells but not polyclonal (pan)-Treg cells. These OVA-specific pTreg cells were significantly less able to suppress OVA-specific stimulation-induced proliferation in vitro and exhibited lower FoxP3 expression. Additionally, we conducted preliminary TCR repertoire diversity sequencing for Treg cells among recent thymic emigrants (RTEs) from RagGFP -FoxP3RFP dual-reporter mice and observed a trend for decreased diversity in mice with thymic atrophy compared to littermates with normal thymus. These data indicate that although the effects of age-related thymic atrophy do not affect pan-Treg generation, certain tissue-specific Treg clones may experience abnormal agonist selection. This, combined with enhanced pan-pTreg cells, may greatly contribute to age-related chronic inflammation, even in the absence of acute autoimmune disease in the elderly.


Subject(s)
Aging/physiology , Autoimmune Diseases/immunology , Inflammation/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/pathology , Aged , Animals , Atrophy , Autoantigens/immunology , Clonal Selection, Antigen-Mediated , Clone Cells , Humans , Immunomodulation , Mice , Mice, Transgenic , Ovalbumin/immunology , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity , Transplantation Chimera
5.
Mol Cancer ; 20(1): 25, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33530981

ABSTRACT

BACKGROUND: E3 ubiquitin ligase HRD1 (HMG-CoA reductase degradation protein 1, alias synoviolin with SYVN1 as the official gene symbol) was found downregulated and acting as a tumor suppressor in breast cancer, while the exact expression profile of HRD1 in different breast cancer subtypes remains unknown. Recent studies characterized circular RNAs (circRNAs) playing an regulatory role as miRNA sponge in tumor progression, presenting a new viewpoint for the post-transcriptional regulation of cancer-related genes. METHODS: Examination of the expression of HRD1 protein and mRNA was implemented using public microarray/RNA-sequencing datasets and breast cancer tissues/cell lines. Based on public RNA-sequencing results, online databases and enrichment/clustering analyses were used to predict the specific combinations of circRNA/miRNA that potentially govern HRD1 expression. Gain-of-function and rescue experiments in vitro and in vivo were executed to evaluate the suppressive effects of circNR3C2 on breast cancer progression through HRD1-mediated proteasomal degradation of Vimentin, which was identified using immunoblotting, immunoprecipitation, and in vitro ubiquitination assays. RESULTS: HRD1 is significantly underexpressed in triple-negative breast cancer (TNBC) against other subtypes and has an inverse correlation with Vimentin, inhibiting the proliferation, migration, invasion and EMT (epithelial-mesenchymal transition) process of breast cancer cells via inducing polyubiquitination-mediated proteasomal degradation of Vimentin. CircNR3C2 (hsa_circ_0071127) is also remarkably downregulated in TNBC, negatively correlated with the distant metastasis and lethality of invasive breast carcinoma. Overexpressing circNR3C2 in vitro and in vivo leads to a crucial enhancement of the tumor-suppressive effects of HRD1 through sponging miR-513a-3p. CONCLUSIONS: Collectively, we elucidated a bona fide circNR3C2/miR-513a-3p/HRD1/Vimentin axis that negatively regulates the metastasis of TNBC, suggesting that circNR3C2 and HRD1 can act as potential prognostic biomarkers. Our study may facilitate the development of therapeutic agents targeting circNR3C2 and HRD1 for patients with aggressive breast cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA Interference , RNA, Circular/genetics , Receptors, Mineralocorticoid/genetics , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , 3' Untranslated Regions , Animals , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Female , Heterografts , Humans , Mice , Models, Biological , Neoplasm Grading , Neoplasm Staging , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Vimentin/genetics , Vimentin/metabolism
6.
FASEB J ; 34(3): 4591-4601, 2020 03.
Article in English | MEDLINE | ID: mdl-32017279

ABSTRACT

Injury of renal tubular epithelial cells is a key feature of the pathogenicity associated with tubulointerstitial fibrosis and other kidney diseases. HUWE1, an E3 ubiquitin ligase, acts by participating in ubiquitination and degradation of its target proteins. However, the detailed mechanisms by which HUWE1 might regulate fibrosis in renal tubular epithelial cells have not been established. Here, the possible regulation of renal tubulointerstitial fibrosis by HUWE1 was investigated by examining the expression of HUWE1 and EGFR in unilateral ureteral obstruction (UUO) mice. Markedly consistent reciprocal changes in HUWE1 and EGFR expression were observed at the protein and mRNA levels in the kidney after UUO injury. Expression of HUWE1 inhibited TGF-ß-induced injury to HK-2 cells, while HUWE1 overexpression decreased the expression of EGFR. Further analysis indicated that HUWE1 physically interacted with EGFR and promoted its ubiquitination and degradation. HUWE1 expression also showed clinical relevance in renal disease, as it notably decreased in multiple types of clinical nephropathy, while EGFR expression significantly increased when compared to the normal kidney. Therefore, this study demonstrated that HUWE1, which serves as an E3 ubiquitin ligase specific for EGFR, promotes EGFR ubiquitination and degradation, thereby regulating EGFR expression and providing protection against kidney injury.


Subject(s)
Fibrosis/metabolism , Fibrosis/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney/metabolism , Kidney/pathology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Western , Cell Line , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Kidney Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Signal Transduction/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/physiology , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism
7.
Cell Commun Signal ; 19(1): 18, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33588886

ABSTRACT

BACKGROUND: Our previous studies have shown that the E3 ubiquitin ligase of HMG-CoA reductase degradation 1 (HRD1) functions as a tumor suppressor, as overexpression of HRD1 suppressed breast cancer proliferation and invasion. However, its role in breast cancer cell glucose metabolism was unclear. Here, our aim was to uncover the role and molecular mechanisms of HRD1 in regulating aerobic glycolysis in breast cancer. METHODS: The effect of HRD1 on robic glycolysis in breast cancer cells were assessed. Then the proliferation, colony formation ability, invasion and migration of breast cancer cells were evaluated. The relationship between HRD1 and PFKP was validated by Mass spectrometry analysis, immunofluorescence and co-immunoprecipitation. The level of PFKP ubiquitination was measured using ubiquitylation assay. Furthermore, the tumor growth and metastasis in mice xenografts were observed. RESULTS: We found that upregulation of HRD1 clearly decreased aerobic glycolysis, and subsequently inhibited breast cancer proliferation and invasion. Mass spectrometry analysis results revealed a large HRD1 interactome, which included PFKP (platelet isoform of phosphofructokinase), a critical enzyme involved in the Warburg Effect in breast cancer. Mechanistically, HRD1 interacted and colocalized with PFKP in the cytoplasm, targeted PFKP for ubiquitination and degradation, and ultimately reduced PFKP expression and activity in breast cancer cells. HRD1 inhibited breast cancer growth and metastasis in vivo through a PFKP-dependent way CONCLUSIONS: Our findings reveal a new regulatory role of HRD1 in Warburg effect and provide a key contributor in breast cancer metabolism. Video abstract.


Subject(s)
Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Phosphofructokinase-1, Type C/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphate/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Movement , Cell Proliferation , Disease Progression , Female , Glucose/metabolism , Glycolysis , Humans , Lactic Acid/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Nude , Phosphofructokinase-1, Type C/genetics , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Up-Regulation
8.
Mol Ther ; 28(4): 1200-1213, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32097602

ABSTRACT

In mammals, resting primordial follicles serve as the ovarian reserve. The decline in ovarian function with aging is characterized by a gradual decrease in both the quantity and quality of the oocytes residing within the primordial follicles. Many reports show that mesenchymal stem cells have the ability to recover ovarian function in premature ovarian insufficiency (POI) or natural aging animal models; however, the underlying mechanism remains unclear. In this study, using exosomes derived from human umbilical cord mesenchymal stem cells (HucMSC-exos), we found the specific accumulation of exosomes in primordial oocytes. The stimulating effects of exosomes on primordial follicles were manifested as the activation of the oocyte phosphatidylinositol 3-kinase (PI3K)/mTOR signaling pathway and the acceleration of follicular development after kidney capsule transplantation. Further analysis revealed the stimulatory effects of HucMSC-exos on primordial follicles were through carrying functional microRNAs, such as miR-146a-5p or miR-21-5p. In aged female mice, the intrabursal injection of HucMSC-exos demonstrated the recovery of decreased fertility with increased oocyte production and improved oocyte quality. Although assisted reproductive technologies have been widely used to treat infertility, their overall success rates remain low, especially for women in advanced maternal age. We propose HucMSC-exos as a new approach to mitigate the age-related retardation of fertility in women.


Subject(s)
Exosomes/transplantation , Infertility, Female/therapy , Oocytes/metabolism , Umbilical Cord/cytology , Aging/physiology , Animals , Exosomes/genetics , Female , Infertility, Female/genetics , Mesenchymal Stem Cells/cytology , Mice , MicroRNAs/genetics , Signal Transduction
9.
Biochem Biophys Res Commun ; 529(2): 277-282, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703423

ABSTRACT

BACKGROUND: Tangeretin, a flavonoid derived from citrus peel, showed anti-diabetic effects. However, the role of tangeretin on liver, the organ that act as target of insulin and play the central role in maintaining the blood glucose level control, is still largely unknown. The current study was designed to assess the effect of tangeretin on liver insulin sensitivity in vitro and in vivo. METHODS: Primary hepatocytes and mice were treated with different dose of tangeretin, parameters of insulin sensitivity, such as blood glucose levels, serum insulin levels, glucose tolerate test (GTT), insulin tolerate test (ITT), insulin stimulated IR-AKT pathway were analyzed. RESULTS: Primary hepatocytes treated with 10/20 µM tangeretin showed up-regulated insulin signaling pathway as well as the glycogen content, while the glucose output were reduced. Intragastric administration of tangeretin (25/50 mg/kg) also ameliorated the liver insulin sensitivity and improved the glucose homeostasis, both in wild type C57 mice and in db/db mice, a diabetic model. Tangeretin treatment dose-dependently suppressed the MEK-ERK1/2 pathway, while forced activation of p-ERK1/2 reversed the insulin sensitized effect of tangeretin. CONCLUSION: These results indicated that tangeretin enhanced the liver insulin sensitivity in vitro and in vivo, through suppressing the MEK-ERK1/2 pathway.


Subject(s)
Flavones/pharmacology , Hepatocytes/drug effects , Insulin Resistance , Insulin/metabolism , MAP Kinase Signaling System/drug effects , Animals , Cells, Cultured , Citrus/chemistry , Flavones/chemistry , Hepatocytes/metabolism , Male , Mice, Inbred C57BL
10.
PLoS Biol ; 15(11): e2003352, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29117183

ABSTRACT

Postnatal thymic epithelial cell (TEC) homeostatic defect- or natural aging-induced thymic atrophy results in a decline in central T-cell tolerance establishment, which is constituted by thymocyte negative selection and cluster of differentiation (CD) 4+ thymic regulatory T (tTreg) cell generation. Emerging evidence shows this decline mainly results from defects in negative selection, but there is insufficient evidence regarding whether tTreg cell generation is also impaired. We mechanistically studied tTreg cell generation in the atrophied thymus by utilizing both postnatal TEC-defective (resulting from FoxN1-floxed conditional knockout [cKO]) and naturally aged mouse models. We found that the capacity of tTreg cell generation was not impaired compared to CD4+ thymic conventional T cells, suggesting thymic atrophy positively influences tTreg cell generation. This is potentially attributed to decreased T cell receptor (TCR) signaling strength due to inefficiency in promiscuous expression of self-antigens or presenting a neo-self-antigen by medullary TECs, displaying decreased negative selection-related marker genes (Nur77 and CD5high) in CD4 single positive (SP) thymocytes. Our results provide evidence that the atrophied thymus attempts to balance the defective negative selection by enhancing tTreg cell generation to maintain central T-cell tolerance in the elderly. Once the balance is broken, age-related diseases could take place.


Subject(s)
Atrophy/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Atrophy/metabolism , Atrophy/pathology , Autoantigens/immunology , Cell Differentiation , Cells, Cultured , Forkhead Transcription Factors/physiology , Homeodomain Proteins/physiology , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Thymus Gland/metabolism , Thymus Gland/pathology
11.
Immun Ageing ; 17: 2, 2020.
Article in English | MEDLINE | ID: mdl-31988649

ABSTRACT

Immune system aging is characterized by the paradox of immunosenescence (insufficiency) and inflammaging (over-reaction), which incorporate two sides of the same coin, resulting in immune disorder. Immunosenescence refers to disruption in the structural architecture of immune organs and dysfunction in immune responses, resulting from both aged innate and adaptive immunity. Inflammaging, described as a chronic, sterile, systemic inflammatory condition associated with advanced age, is mainly attributed to somatic cellular senescence-associated secretory phenotype (SASP) and age-related autoimmune predisposition. However, the inability to reduce senescent somatic cells (SSCs), because of immunosenescence, exacerbates inflammaging. Age-related adaptive immune system deviations, particularly altered T cell function, are derived from age-related thymic atrophy or involution, a hallmark of thymic aging. Recently, there have been major developments in understanding how age-related thymic involution contributes to inflammaging and immunosenescence at the cellular and molecular levels, including genetic and epigenetic regulation, as well as developments of many potential rejuvenation strategies. Herein, we discuss the research progress uncovering how age-related thymic involution contributes to immunosenescence and inflammaging, as well as their intersection. We also describe how T cell adaptive immunity mediates inflammaging and plays a crucial role in the progression of age-related neurological and cardiovascular diseases, as well as cancer. We then briefly outline the underlying cellular and molecular mechanisms of age-related thymic involution, and finally summarize potential rejuvenation strategies to restore aged thymic function.

12.
J Cell Physiol ; 234(11): 21395-21406, 2019 11.
Article in English | MEDLINE | ID: mdl-31069793

ABSTRACT

To investigate the role of S100 calcium-binding protein A16 (S100A16) in hepatic lipid metabolism, S100a16 transgenic, S100a16 knockdown, and wildtype C57BL/6 mice were fed either a high-fat diet (HFD) or normal-fat diet (NFD) for 16 weeks. The results showed that for HFD-fed mice, S100a16 transgenic mice showed significantly more severe fatty liver than other HFD-fed mice, with a significant increase in serum triglyceride (TG) concentration, with more and larger lipid droplets in the liver, whereas S100a16 knockdown mice were completely opposite, with liver fat lesions and TG serological changes being the mildest; for NFD-fed mice, liver fat accumulation and serum TG concentrations were significantly lower than those fed HFD, and no significant lipid droplets were found in the liver. Further, we found that calmodulin (CaM) interacts with S100A16, a member of the AMP-activated protein kinase (AMPK) pathway. Our research found that S100A16 regulates the AMPK pathway-associated protein by interacting with CaM to regulate liver lipid synthesis. S100A16 regulates liver lipid metabolism through the CaM/CAMKK2/AMPK pathway. Overexpression of S100A16 promotes the deterioration of fatty liver induced by HFD, and low expression of S100A16 can attenuate fatty liver.


Subject(s)
Hepatocytes/metabolism , Lipogenesis/physiology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , S100 Proteins/metabolism , Animals , Diet, High-Fat , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
13.
J Cell Sci ; 130(6): 1027-1036, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28137758

ABSTRACT

Ubiquitylation of the epithelial Na+ channel (ENaC) plays a critical role in cellular functions, including transmembrane transport of Na+, Na+ and water balance, and blood pressure stabilization. Published studies have suggested that ENaC subunits are targets of ER-related degradation (ERAD) in yeast systems. However, the molecular mechanism underlying proteasome-mediated degradation of ENaC subunits remains to be established. Derlin-1, an E3 ligase mediator, links recognized target proteins to ubiquitin-mediated proteasomal degradation in the cytosol. In the present study, we found that derlin-1 suppressed the expression of ENaC at the protein level and that the subunit α-ENaC (also known as SCNN1A) physically interacted with derlin-1 at the membrane-anchored domains or the loop regions, and that derlin-1 initiated α-ENaC retrotranslocation. In addition, HUWE1, an endoplasmic reticulum (ER)-resident E3 ubiquitin ligase, was recruited and promoted K11-linked polyubiquitylation of α-ENaC and, hence, formation of an α-ENaC ubiquitin-mediated degradation complex. These findings suggest that derlin-1 promotes ENaC ubiquitylation and enhances ENaC ubiquitin- mediated proteasome degradation. The derlin-1 pathway therefore may represent a significant early checkpoint in the recognition and degradation of ENaC in mammalian cells.


Subject(s)
Epithelial Sodium Channels/metabolism , Membrane Proteins/metabolism , Proteolysis , Ubiquitination , Animals , Cell Membrane/metabolism , HEK293 Cells , Humans , Lysine/metabolism , Mice , Models, Biological , Polyubiquitin/metabolism , Protein Binding , Protein Domains , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/metabolism
14.
Biochem Biophys Res Commun ; 517(3): 538-544, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31376936

ABSTRACT

Doxorubicin (DOX) is an antitumor drug widely used in hematological tumors and various solid tumors. However, the cardiotoxicity elicited by DOX severely limits its clinical treatment. Dimethyl itaconate (DI), a common form of itaconate, is found many potential targets for prevent heart injury. Here we employed wild type and Nrf2 knockout mice and induced a cardiotoxicity model by administration of DOX to clarify the effects of DI. After treatment with DI, we found that it could effectively alleviate the cardiotoxicity by analyzing morphology, LDH levels and heart weight/body weight ratio changes. Meanwhile we demonstrated that RIP3, a key protein of necrosis, was significantly decreased in DI treated group. Further we observed that treatment with DI could suppress oxidative stress by altering Nrf2/HO-1. Compared with vehicle group, DI could increase the tissue SOD and GSH, and reduce MDA levels, then DHE staining revealed that the level of ROS in DI group reduced by half. Finally, transmission electron microscope (TEM) data showed that treatment with DI obviously decreased the mitochondrial damage. While Nrf2 was ablated in mice, the protective effects of DI were vanished and SOD, GSH, MDA became unchanged related to vehicle group. This report provides the evidence for the protective effects of DI treatment in cardiotoxicity induced by DOX. On mechanisms, DI could reduce the oxidative stress by altering Nrf2/HO-1 pathway and prevent mitochondrial from damage. Taken together, these findings of this paper will afford the new therapeutic targets in DOX related cardiotoxicity.


Subject(s)
Cardiotonic Agents/pharmacology , Cardiotoxicity/prevention & control , Doxorubicin/antagonists & inhibitors , Myocytes, Cardiac/drug effects , NF-E2-Related Factor 2/genetics , Succinates/pharmacology , Acute Disease , Animals , Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/etiology , Cardiotoxicity/genetics , Cardiotoxicity/pathology , Doxorubicin/adverse effects , Gene Expression , Glutathione/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Male , Malondialdehyde/antagonists & inhibitors , Malondialdehyde/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-E2-Related Factor 2/deficiency , Oxidative Stress/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
15.
FASEB J ; : fj201800059R, 2018 May 21.
Article in English | MEDLINE | ID: mdl-29782203

ABSTRACT

Biologic aging results in a chronic inflammatory condition, termed inflammaging, which establishes a risk for such age-related diseases as neurocardiovascular diseases; therefore, it is of great importance to develop rejuvenation strategies that are able to attenuate inflammaging as a means of intervention for age-related diseases. A promising rejuvenation factor that is present in young blood has been found that can make aged neurons younger; however, the component in the young blood and its mechanism of action are poorly elucidated. We assessed rejuvenation in naturally aged mice with extracellular vesicles (EVs) or exosomes extracted from young murine serum on the basis of different spectrums of microRNAs in these vesicles from young and old sera. We found that EVs extracted from young donor mouse serum, rather than EVs extracted from old donor mouse serum or non-EV supernatant extracted from young donor mouse serum, were able to attenuate inflammaging in old mice. Inflammaging is attributed to multiple factors, one of which is thymic aging-released self-reactive T cell-induced pathology. We found that the attenuation of inflammaging after treatment with EVs from young serum partially contributed to the rejuvenation of thymic aging, which is characterized by partially reversed thymic involution, enhancement of negative selection signals, and reduced autoreactions in the periphery. Our results provide evidence for understanding of the potential rejuvenation factor in the young donor serum, which holds great promise for the development of novel therapeutics to reduce morbidity and mortality caused by age-related inflammatory diseases.-Wang, W., Wang, L., Ruan, L., Oh, J., Dong, X., Zhuge, Q., Su, D.-M. Extracellular vesicles extracted from young donor serum attenuate inflammaging via partially rejuvenating aged T-cell immunotolerance.

16.
Sheng Li Xue Bao ; 71(2): 279-286, 2019 Apr 25.
Article in Zh | MEDLINE | ID: mdl-31008487

ABSTRACT

The aim of this study was to investigate the role of S100 calcium binding protein A16 (S100A16) in lipid metabolism in hepatocytes and its possible biological mechanism. HepG2 cells (human hepatoma cell line) were cultured with fatty acid to establish fatty acid culture model. The control model was cultured without fatty acid. Each model was divided into three groups and transfected with S100a16 over-expression, shRNA and vector plasmids, respectively. The concentration of triglyceride (TG) in the cells was measured by kit, and the lipid droplets was observed by oil red O staining. Immunoprecipitation and mass spectrometry were used to find the interesting proteins interacting with S100A16, and the interaction was verified by immunoprecipitation. The further mechanism was studied by Western blot and qRT-PCR. The results showed that the intracellular lipid droplet and TG concentrations in the fatty acid culture model were significantly higher than those in the control model. The accumulation of intracellular fat in the S100a16 over-expression group was significantly higher than that in the vector plasmid transfection group. There was an interaction between heat shock protein A5 (HSPA5) and S100A16. Over-expression of S100A16 up-regulated protein expression levels of HSPA5, inositol-requiring enzyme 1α (IRE1α) and pIREα1, which belong to endoplasmic reticulum stress HSPA5/IRE1α-XBP1 pathway. Meanwhile, over-expression of S100A16 up-regulated the mRNA expression levels of adipose synthesis-related gene Srebp1c, Acc and Fas. In the S100a16 shRNA plasmid transfection group, the above-mentioned protein and mRNA levels were lower than those of vector plasmid transfection group. These results suggest that S100A16 may promote lipid synthesis in HepG2 cells through endoplasmic reticulum stress HSPA5/IRE1α-XBP1 pathway.


Subject(s)
Endoplasmic Reticulum Stress , Lipid Metabolism , S100 Proteins/physiology , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/physiology , Heat-Shock Proteins/physiology , Hep G2 Cells , Humans , Protein Serine-Threonine Kinases/physiology , Triglycerides/biosynthesis , X-Box Binding Protein 1/physiology
17.
Cancer Cell Int ; 18: 59, 2018.
Article in English | MEDLINE | ID: mdl-29686537

ABSTRACT

OBJECTIVE: Recently, long non-coding RNA (lncRNA) MIAT has been demonstrated as an oncogenic gene in several types of cancer. However, the role and mechanism of MIAT in colorectal cancer (CRC) have not been investigated. METHODS: Real-time PCR was used to measure MIAT expression in CRC tissues and cells. Small interfering RNA specific for MIAT (si-MIAT) was used to down-regulate MIAT expression in CRC cells. The interaction of MIAT and miR-132 was measured by RNA pull-down assay. The effect of si-MIAT on CRC cells apoptosis and metastasis were measured by flow cytometry assay, invasion and migration assay, respectively. RESULTS: In present study, we found that MIAT was highly expressed in CRC tissues and cells. MIAT knockdown inhibited proliferation, migration and invasion and enhanced apoptosis of CRC cells. Further, we demonstrated that MIAT acted as a competing endogenous RNA for miR-132, antagonized its functions, and resulted in the de-repression of its target gene Derlin-1, which acted as an oncogene in promoting growth and metastasis of CRC cells. In LOVO and SW480 cells with si-MIAT, miR-132 inhibitor resulted in an increase of cell proliferation, migration and invasion and a decrease of cell apoptosis, which was partially abolished by transfection of Derlin-1 shRNA. CONCLUSIONS: Our data indicated that highly expressed MIAT was an oncogenic lncRNA that promoted the growth and metastasis of CRC through miR-132/Derlin-1 axis.

18.
J Cell Sci ; 128(19): 3621-30, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26272921

ABSTRACT

The epithelial Na(+) channel (ENaC), regulated by insulin, is of fundamental importance in the control of Na(+) reabsorption in the distal nephron. The potential role of Forkhead box O1 (FoxO1), downstream of insulin signaling, in the regulation of ENaC remains to be investigated. Here, we found that the overexpression of a constitutively active form of FoxO1 (ADA-FoxO1) suppressed the mRNA level of the ENaC α subunit (α-ENaC; also known as SCCN1A) and the apical density of ENaC in mouse cortical collecting duct (mCCD) cells. Conversely, knockdown of FoxO1 increased the apical membrane levels of α-ENaC and Na(+) transport under basal conditions. Insulin elevated α-ENaC expression and induced FoxO1 phosphorylation; however, the increase in α-ENaC and phosphorylated FoxO1 expression observed with insulin treatment was blunted ∼ 60% in cells expressing ADA-FoxO1. Moreover, insulin induced the interaction between phosphorylated FoxO1 and 14-3-3ε, indicating that FoxO1 phosphorylation promotes ENaC membrane trafficking by binding to 14-3-3ε. FoxO1 also suppressed activity of the α-ENaC promoter, and the putative FoxO1 target site is located in the -500 to -200 nt region of the α-ENaC promoter. These findings indicate that FoxO1 is a key negative regulatory factor in the insulin-dependent control of ENaC expression and forward trafficking in mCCD epithelia.


Subject(s)
Epithelial Sodium Channels/metabolism , Forkhead Transcription Factors/metabolism , Animals , Cell Line, Tumor , Epithelial Sodium Channels/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Immunoprecipitation , Protein Transport/genetics , Protein Transport/physiology , Signal Transduction/genetics , Signal Transduction/physiology
19.
J Immunol ; 194(12): 5825-37, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25957168

ABSTRACT

Thymic involution and the subsequent amplified release of autoreactive T cells increase the susceptibility toward developing autoimmunity, but whether they induce chronic inflammation with advanced age remains unclear. The presence of chronic low-level proinflammatory factors in elderly individuals (termed inflammaging) is a significant risk factor for morbidity and mortality in virtually every chronic age-related disease. To determine how thymic involution leads to the persistent release and activation of autoreactive T cells capable of inducing inflammaging, we used a Foxn1 conditional knockout mouse model that induces accelerated thymic involution while maintaining a young periphery. We found that thymic involution leads to T cell activation shortly after thymic egress, which is accompanied by a chronic inflammatory phenotype consisting of cellular infiltration into non-lymphoid tissues, increased TNF-α production, and elevated serum IL-6. Autoreactive T cell clones were detected in the periphery of Foxn1 conditional knockout mice. A failure of negative selection, facilitated by decreased expression of Aire rather than impaired regulatory T cell generation, led to autoreactive T cell generation. Furthermore, the young environment can reverse age-related regulatory T cell accumulation in naturally aged mice, but not inflammatory infiltration. Taken together, these findings identify thymic involution and the persistent activation of autoreactive T cells as a contributing source of chronic inflammation (inflammaging).


Subject(s)
Autoimmunity , Clonal Selection, Antigen-Mediated , Inflammation/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Age Factors , Animals , Cellular Microenvironment , Chronic Disease , Clonal Deletion/genetics , Clonal Deletion/immunology , Disease Models, Animal , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , Immunophenotyping , Inflammation/genetics , Inflammation/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Phenotype , T-Lymphocyte Subsets/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
20.
Diabetologia ; 59(7): 1524-1532, 2016 07.
Article in English | MEDLINE | ID: mdl-27003684

ABSTRACT

AIM/HYPOTHESIS: MicroRNA-9 (miR-9) is involved in the regulation of pancreatic beta cell function. However, its role in gluconeogenesis is still unclear. Our objective was to investigate the role of miR-9 in hepatic glucose production (HGP). METHODS: MiR-9 expression was measured in livers of high-fat diet (HFD) mice and ob/ob mice. The methylation status of the miR-9-3 promoter regions in hepatocytes was determined by the methylation-specific PCR procedure. The binding activity of DNA methyltransferase (DNMT)1, DNMT3a and DNMT3b on the miR-9-3 promoter was detected by chromatin immunoprecipitation (ChIP) and quantitative real-time PCR assays. HGP was evaluated in vitro and in vivo. Glucose tolerance, insulin tolerance and pyruvate tolerance tests were also performed. RESULTS: Reduced miR-9 expression and hypermethylation of the miR-9-3 promoter were observed in the livers of obese mice. Further study showed that the binding of DNMT1, but not of DNMT3a and DNMT3b, to the miR-9-3 promoter was increased in hepatocytes from ob/ob mice. Knockdown of DNMT1 alleviated the decrease in hepatic miR-9 expression in vivo and in vitro. Overexpression of hepatic miR-9 improved insulin sensitivity in obese mice and inhibited HGP. In addition, deletion of hepatic miR-9 led to an increase in random and fasting blood glucose levels in lean mice. Importantly, silenced forkhead box O1 (FOXO1) expression reversed the gluconeogenesis and glucose production in hepatocytes induced by miR-9 deletion. CONCLUSIONS/INTERPRETATION: Our observations suggest that the decrease in miR-9 expression contributes to an inappropriately activated gluconeogenesis in obese mice.


Subject(s)
Forkhead Box Protein O1/metabolism , Gluconeogenesis/physiology , MicroRNAs/metabolism , Animals , Chromatin Immunoprecipitation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA Methylation/physiology , DNA Methyltransferase 3A , Diet, High-Fat , Forkhead Box Protein O1/genetics , Gluconeogenesis/genetics , Glucose/metabolism , Liver/metabolism , Mice , Mice, Obese , MicroRNAs/genetics , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL