Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
FASEB J ; 37(10): e23182, 2023 10.
Article in English | MEDLINE | ID: mdl-37682013

ABSTRACT

A link between increased glycolysis and vascular calcification has recently been reported, but it remains unclear how increased glycolysis contributes to vascular calcification. We therefore investigated the role of PFKFB3, a critical enzyme of glycolysis, in vascular calcification. We found that PFKFB3 expression was upregulated in calcified mouse VSMCs and arteries. We showed that expression of miR-26a-5p and miR-26b-5p in calcified mouse arteries was significantly decreased, and a negative correlation between Pfkfb3 mRNA expression and miR-26a-5p or miR-26b-5p was seen in these samples. Overexpression of miR-26a/b-5p significantly inhibited PFKFB3 expression in VSMCs. Intriguingly, pharmacological inhibition of PFKFB3 using PFK15 or knockdown of PFKFB3 ameliorated vascular calcification in vD3 -overloaded mice in vivo or attenuated high phosphate (Pi)-induced VSMC calcification in vitro. Consistently, knockdown of PFKFB3 significantly reduced glycolysis and osteogenic transdifferentiation of VSMCs, whereas overexpression of PFKFB3 in VSMCs induced the opposite effects. RNA-seq analysis and subsequent experiments revealed that silencing of PFKFB3 inhibited FoxO3 expression in VSMCs. Silencing of FoxO3 phenocopied the effects of PFKFB3 depletion on Ocn and Opg expression but not Alpl in VSMCs. Pyruvate or lactate supplementation, the product of glycolysis, reversed the PFKFB3 depletion-mediated effects on ALP activity and OPG protein expression in VSMCs. Our results reveal that blockade of PFKFB3-mediated glycolysis inhibits vascular calcification in vitro and in vivo. Mechanistically, we show that FoxO3 and lactate production are involved in PFKFB3-driven osteogenic transdifferentiation of VSMCs. PFKFB3 may be a promising therapeutic target for the treatment of vascular calcification.


Subject(s)
Forkhead Box Protein O3 , MicroRNAs , Phosphofructokinase-2 , Vascular Calcification , Animals , Mice , Glycolysis , Lactic Acid , Muscle, Smooth, Vascular , Phosphoric Monoester Hydrolases , Vascular Calcification/genetics , Phosphofructokinase-2/metabolism , Forkhead Box Protein O3/metabolism
2.
Vascul Pharmacol ; 146: 107096, 2022 10.
Article in English | MEDLINE | ID: mdl-35952961

ABSTRACT

Recent studies have shown that short-chain fatty acids (SCFAs), primarily acetate, propionate and butyrate, play a crucial role in the pathogenesis of cardiovascular disease. Whether SCFAs regulate vascular calcification, a common pathological change in cardiovascular tissues, remains unclear. This study aimed to investigate the potential role of SCFAs in vascular calcification. Using cellular and animal models of vascular calcification, we showed that butyrate significantly enhanced high phosphate (Pi)-induced calcification and osteogenic transition of vascular smooth muscle cells (VSMC) in vitro, whereas acetate and propionate had no effects. Subsequent studies confirmed that butyrate significantly promoted high Pi-induced aortic ring calcification ex vivo and high dose vitamin D3 (vD3)-induced mouse vascular calcification in vivo. Mechanistically, butyrate significantly inhibited histone deacetylase (HDAC) expression in VSMCs, and a pan HDAC inhibitor Trichostatin A showed similar inductive effects on calcification and osteogenic transition of VSMCs to butyrate. In addition, the SCFA sensing receptors Gpr41 and Gpr109a were primarily expressed by VSMCs, and butyrate induced the rapid activation of NF-κB, Wnt and Akt signaling in VSMCs. Intriguingly, the NF-κB inhibitor SC75741 significantly attenuated butyrate-induced calcification and the osteogenic gene Msx2 expression in VSMCs. We showed that knockdown of Gpr41 but not Gpr109a attenuated butyrate-induced VSMC calcification. This study reveals that butyrate accelerates vascular calcification via its dual effects on HDAC inhibition and NF-κB activation. Our data provide novel insights into the role of microbe-host interaction in vascular calcification, and may have implications for the development of potential therapy for vascular calcification.


Subject(s)
NF-kappa B , Vascular Calcification , Animals , Butyrates/metabolism , Butyrates/pharmacology , Cells, Cultured , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Phosphates , Propionates/metabolism , Propionates/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Vascular Calcification/pathology , Vitamin D
3.
J Biol Chem ; 298(5): 101887, 2022 05.
Article in English | MEDLINE | ID: mdl-35367413

ABSTRACT

Recent genome-wide association and transcriptome-wide association studies have identified an association between the PALMD locus, encoding palmdelphin, a protein involved in myoblast differentiation, and calcific aortic valve disease (CAVD). Nevertheless, the function and underlying mechanisms of PALMD in CAVD remain unclear. We herein investigated whether and how PALMD affects the pathogenesis of CAVD using clinical samples from CAVD patients and a human valve interstitial cell (hVIC) in vitro calcification model. We showed that PALMD was upregulated in calcified regions of human aortic valves and calcified hVICs. Furthermore, silencing of PALMD reduced hVIC in vitro calcification, osteogenic differentiation, and apoptosis, whereas overexpression of PALMD had the opposite effect. RNA-Seq of PALMD-depleted hVICs revealed that silencing of PALMD reduced glycolysis and nuclear factor-κB (NF-κB)-mediated inflammation in hVICs and attenuated tumor necrosis factor α-induced monocyte adhesion to hVICs. Having established the role of PALMD in hVIC glycolysis, we examined whether glycolysis itself could regulate hVIC osteogenic differentiation and inflammation. Intriguingly, the inhibition of PFKFB3-mediated glycolysis significantly attenuated osteogenic differentiation and inflammation of hVICs. However, silencing of PFKFB3 inhibited PALMD-induced hVIC inflammation, but not osteogenic differentiation. Finally, we showed that the overexpression of PALMD enhanced hVIC osteogenic differentiation and inflammation, as opposed to glycolysis, through the activation of NF-κB. The present study demonstrates that the genome-wide association- and transcriptome-wide association-identified CAVD risk gene PALMD may promote CAVD development through regulation of glycolysis and NF-κB-mediated inflammation. We propose that targeting PALMD-mediated glycolysis may represent a novel therapeutic strategy for treating CAVD.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Aortic Valve/metabolism , Aortic Valve/pathology , Aortic Valve Stenosis/metabolism , Calcinosis , Cells, Cultured , Genome-Wide Association Study , Glycolysis , Humans , Inflammation/metabolism , Membrane Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Osteogenesis
4.
Respir Res ; 23(1): 70, 2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35337337

ABSTRACT

BACKGROUND: Particular matter 2.5 (PM2.5) is one of the most important air pollutant, and it is positively associated with the development of chronic obstructive pulmonary disease (COPD). However, the precise underlying mechanisms through which PM2.5 promotes the development of COPD remains largely unknown. METHODS: Mouse alveolar destruction were determined by histological analysis of lung tissues and lung function test. Alveolar type II cells (AT2) to alveolar type I cells (AT1) transition in PM2.5-induced COPD mouse model was confirmed via immunofluorescence staining and qPCR analysis. The differentially expressed genes in PM2.5-induced COPD mouse model were identified by RNA-sequencing of alveolar epithelial organoids and generated by bioinformatics analysis. RESULTS: In this study, we found that 6 months exposure of PM2.5 induced a significantly decreased pulmonary compliance and resulted in pulmonary emphysema in mice. We showed that PM2.5 exposure significantly reduced the AT2 to AT1 cell transition in vitro and in vivo. In addition, we found a reduced expression of the intermediate AT2-AT1 cell process marker claudin 4 (CLDN4) at day 4 of differentiation in mouse alveolar organoids treated with PM2.5, suggesting that PM2.5 exposure inhibited AT2 cells from entering the transdifferentiation process. RNA-sequencing of mouse alveolar organoids showed that several key signaling pathways that involved in the AT2 to AT1 cell transition were significantly altered including the Wnt signaling, MAPK signaling and signaling pathways regulating pluripotency of stem cells following PM2.5 exposure. CONCLUSIONS: In summary, these data demonstrate a critical role of AT2 to AT1 cell transition in PM2.5-induced COPD mouse model and reveal the signaling pathways that potentially regulate AT2 to AT1 cell transition during this process. Our findings therefore advance the current knowledge of PM2.5-induced COPD and may lead to a novel therapeutic strategy to treat this disease.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Alveolar Epithelial Cells/metabolism , Animals , Lung , Mice , Particulate Matter/toxicity , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Wnt Signaling Pathway
5.
Front Physiol ; 13: 755371, 2022.
Article in English | MEDLINE | ID: mdl-35295585

ABSTRACT

Aims: Vascular calcification is a common clinical complication of chronic kidney disease (CKD), atherosclerosis (AS), and diabetes, which is associated with increased cardiovascular morbidity and mortality in patients. The transdifferentiation of vascular smooth muscle cells (VSMCs) to an osteochondrogenic phenotype is a crucial step during vascular calcification. The transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) plays an important role in regulating cell proliferation and differentiation, but whether it regulates the calcification of arteries and VSMCs remains unclear. Therefore, this study aims to understand the role of C/EBPα in the regulation of vascular calcification. Methods and Results: Both mRNA and protein expression levels of C/EBPα were significantly increased in calcified arteries from mice treated with a high dose of vitamin D3 (vD3). Upregulation of C/EBPα was also observed in the high phosphate- and calcium-induced VSMC calcification process. The siRNA-mediated knockdown of C/EBPα significantly attenuated VSMC calcification in vitro. Moreover, C/EBPα depletion in VSMCs significantly reduced the mRNA expression of the osteochondrogenic genes, e.g., sex-determining region Y-box 9 (Sox9). C/EBPα overexpression can induce SOX9 overexpression. Similar changes in the protein expression of SOX9 were also observed in VSMCs after C/EBPα depletion or overexpression. In addition, silencing of Sox9 expression significantly inhibited the phosphate- and calcium-induced VSMC calcification in vitro. Conclusion: Findings in this study indicate that C/EBPα is a key regulator of the osteochondrogenic transdifferentiation of VSMCs and vascular calcification, which may represent a novel therapeutic target for vascular calcification.

6.
Environ Pollut ; 303: 119115, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35259473

ABSTRACT

Fine particulate matter (PM2.5) exposure is a significant cause of chronic obstructive pulmonary disease (COPD), but the detailed mechanisms involved in COPD remain unclear. In this study, we established PM2.5-induced COPD rat models and showed that PM2.5 induced pulmonary microvascular injury via accelerating vascular endothelial apoptosis, increasing vascular permeability, and reducing angiogenesis, thereby contributing to COPD development. Moreover, microvascular injury in COPD was validated by measurements of plasma endothelial microparticles (EMPs) and serum VEGF in COPD patients. We then performed m6A sequencing, which confirmed that altered N6-methyladenosine (m6A) modification was induced by PM2.5 exposure. The results of a series of experiments demonstrated that the expression of methyltransferase-like protein 16 (METTL16), an m6A regulator, was upregulated in PM2.5-induced COPD rats, while the expression of other regulators did not differ upon PM2.5-induction. To clarify the regulatory effect of METTL16-mediated m6A modification induced by PM2.5 on pulmonary microvascular injury, cell apoptosis, permeability, and tube formation, the m6A level in METTL16-knockdown pulmonary microvascular endothelial cells (PMVECs) was evaluated, and the target genes of METTL16 were identified from a set of the differentially expressed and m6A-methylated genes associated with vascular injury and containing predicted sites of METTL16 methylation. The results showed that Sulfatase 2 (Sulf2) and Cytohesin-1 (Cyth1) containing the predicted METTL16 methylation sites, exhibited higher m6A methylation and were downregulated after PM2.5 exposure. Further studies demonstrated that METTL16 may regulate Sulf2 expression via m6A modification and thereby contribute to PM2.5-induced microvascular injury. These findings not only provide a better understanding of the role played by m6A modification in PM2.5-induced microvascular injury, but also identify a new therapeutic target for COPD.


Subject(s)
Lung Injury , Pulmonary Disease, Chronic Obstructive , Animals , Endothelial Cells/metabolism , Humans , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Particulate Matter/toxicity , Pulmonary Disease, Chronic Obstructive/chemically induced , Rats
7.
Front Cell Dev Biol ; 9: 629669, 2021.
Article in English | MEDLINE | ID: mdl-33634130

ABSTRACT

Mitochondria-associated ER membranes (MAMs) represent a crucial intracellular signaling hub, that regulates various cellular events including Ca2+ homeostasis, lipid metabolism, mitochondrial function, and cellular survival and death. All of these MAM-mediated cellular events contribute to carcinogenesis. Indeed, altered functions of MAMs in several types of cancers have been documented, in particular for breast cancer. Over the past years, altered expression of many MAM-resident proteins have been reported in breast cancer. These MAM-resident proteins play an important role in regulation of breast cancer initiation and progression. In the current review, we discuss our current knowledge about the functions of MAMs, and address the underlying mechanisms through which MAM-resident proteins regulate breast cancer. A fuller understanding of the pathways through which MAMs regulate breast cancer, and identification of breast cancer-specific MAM-resident proteins may help to develop novel therapeutic strategies for breast cancer.

8.
J Int Med Res ; 48(9): 300060520947917, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32972275

ABSTRACT

Retrocaval ureter is a rare disease associated with abnormal embryonic development. Here, we describe a patient who exhibited retrocaval ureter complicated by renal and ureteral calculi, which were treated by percutaneous nephrolithotomy combined with retroperitoneal laparoscopy. A 64-year-old man was admitted to our hospital because of intermittent back pain that had been present for more than 10 years. During hospitalization, he was diagnosed with retrocaval ureter, right renal calculi, and right ureteral calculi with right hydronephrosis; he underwent percutaneous nephrolithotomy combined with retroperitoneal laparoscopic surgery. After the operation, his condition was stable and he exhibited good recovery. Our findings in this case suggest that percutaneous nephrolithotomy combined with retroperitoneal laparoscopy is a suitable option for the treatment of retrocaval ureter with renal and ureteral calculi.


Subject(s)
Laparoscopy , Nephrolithotomy, Percutaneous , Retrocaval Ureter , Ureter , Ureteral Calculi , Humans , Male , Middle Aged , Retrocaval Ureter/surgery , Ureter/diagnostic imaging , Ureter/surgery , Ureteral Calculi/diagnostic imaging , Ureteral Calculi/surgery
9.
Vascul Pharmacol ; 132: 106775, 2020 09.
Article in English | MEDLINE | ID: mdl-32702412

ABSTRACT

BACKGROUNDS: Medial artery calcification (MAC) significantly contributes to the increased cardiovascular death in patients with chronic kidney disease (CKD). Previous genome-wide association studies have shown that various genetic variants of the histone deacetylase Hdac9 are associated with cardiovascular disease, but the role of Hdac9 in MAC under CKD conditions remains unclear. METHODS: High phosphate-induced vascular smooth muscle cell (VSMC) calcification and MAC in mice administered with vitamin D3 (vD) were used in the present study. Alizarin red staining, calcium quantitative assay, qPCR, western blotting and histology were performed. RESULTS: Hdac9 expression was significantly down-regulated during high phosphate-induced vascular smooth muscle cell (VSMC) calcification and MAC in mice administered with vitamin D3 (vD). Furthermore, high phosphate treatment inhibited phosphorylation of Akt, and pharmacological inhibition of Akt signaling reduced Hdac9 expression in cultured VSMCs. Knockdown of Hdac9 significantly enhanced calcium deposition in VSMCs. Conversely, adenovirus mediated-overexpression of Hdac9 inhibited high phosphate induced VSMC in vitro calcification. Our subsequent mechanistic studies revealed that the anti-calcific effect of Hdac9 was mediated through down-regulation of osteoblast-specific transcription factor Osterix. CONCLUSION: These data suggest that Hdac9 is a novel inhibitor of MAC and may represent a potential therapeutic target for MAC in CKD patients.


Subject(s)
Histone Deacetylases/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/metabolism , Repressor Proteins/metabolism , Sp7 Transcription Factor/metabolism , Vascular Calcification/enzymology , Animals , Cells, Cultured , Cholecalciferol , Disease Models, Animal , Down-Regulation , Histone Deacetylases/genetics , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/genetics , Signal Transduction , Sp7 Transcription Factor/genetics , Vascular Calcification/chemically induced , Vascular Calcification/genetics , Vascular Calcification/pathology
10.
BMC Cardiovasc Disord ; 19(1): 297, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31847835

ABSTRACT

BACKGROUND: Blood glucose (BG) is a risk factor of adverse prognosis in non-diabetic patients in several conditions. However, a limited number of studies were performed to explore the relationship between postoperative BG and adverse outcomes in non-diabetic patients with rheumatic heart disease (RHD). METHODS: We identified 1395 non-diabetic patients who diagnosed with having RHD, and underwent at least one valve replacement and preoperative coronary angiography. BG was measured at admission to the intensive care unit (ICU) after surgery. The association of postoperative BG level with in-hospital and one-year mortality was accordingly analyzed. RESULTS: Included patients were stratified into four groups according to postoperative BG level's (mmol/L) quartiles: Q1 (< 9.3 mmol/L, n = 348), Q2 (9.3-10.9 mmol/L, n = 354), Q3 (10.9-13.2 mmol/L, n = 341), and Q4 (≥ 13.2 mmol/L, n = 352). The in-hospital death (1.1% vs. 2.3% vs. 1.8% vs. 8.2%, P < 0.001) and MACEs (2.0% vs. 3.1% vs. 2.6% vs. 9.7%, P < 0.001) were significantly higher in the upper quartiles. Postoperative BG > 13.0 mmol/L was the best threshold for predicting in-hospital death (area under the curve (AUC) = 0.707, 95% confidence interval (CI): 0.634-0.780, P < 0.001). Multivariate logistic regression analysis indicated that postoperative BG > 13.0 mmol/L was an independent predictor of in-hospital mortality (adjusted odds ratio (OR) = 3.418, 95% CI: 1.713-6.821, P < 0.001). In addition, Kaplan-Meier curve analysis showed that the risk of one-year death was increased for a postoperative BG > 13.2 (log-rank = 32.762, P < 0.001). CONCLUSION: Postoperative BG, as a routine test, could be served as a risk measure for non-diabetic patients with RHD.


Subject(s)
Blood Glucose/metabolism , Heart Valve Prosthesis Implantation/adverse effects , Rheumatic Heart Disease/surgery , Biomarkers/blood , Coronary Angiography , Female , Heart Valve Prosthesis Implantation/mortality , Hospital Mortality , Humans , Male , Middle Aged , Predictive Value of Tests , Retrospective Studies , Rheumatic Heart Disease/blood , Rheumatic Heart Disease/diagnostic imaging , Rheumatic Heart Disease/mortality , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...