215 results on '"SREBP-2"'
Search Results
2. Luteolin Mitigates Diabetic Dyslipidemia in Rats by Modulating ACAT-2, PPARα, SREBP-2 Proteins, and Oxidative Stress.
- Author
-
Shehnaz, Syed Ilyas, Roy, Anitha, Vijayaraghavan, Rajagopalan, and Sivanesan, Senthilkumar
- Abstract
Diabetic dyslipidemia is a crucial link between type-2 diabetes mellitus (T2DM) and atherosclerotic cardiovascular diseases (ASCVD). Natural biologically active substances have been advocated as complementary remedies for ASCVD and T2DM. Luteolin, a flavonoid, exhibits antioxidant, hypolipidemic, and antiatherogenic effects. Hence, we aimed to determine influence of luteolin on lipid homeostasis and hepatic damage in rats with T2DM induced by high-fat-diet (HFD) and streptozotocin (STZ). After being fed HFD for 10 days, male Wistar rats received 40 mg/kg STZ intraperitoneal injection on 11th day. Seventy-two hours later, hyperglycemic rats (fasting glucose > 200 mg/dL) were randomized into groups, and oral hydroxy-propyl-cellulose, atorvastatin (5 mg/kg), or luteolin (50 mg/kg or 100 mg/kg) administered daily, while continuing HFD for 28 days. Luteolin significantly ameliorated dyslipidemia levels and concomitantly improved atherogenic index of plasma in a dose-dependent manner. Increased levels of malondialdehyde and diminished levels of superoxide dismutase, catalase, and glutathione in HFD-STZ-diabetic rats were significantly regulated by luteolin. Luteolin significantly intensified PPARα expression while decreasing expression of acyl-coenzyme A:cholesterol acyltransferase-2 (ACAT-2) and sterol regulatory element binding protein-2 (SREBP-2) proteins. Moreover, luteolin effectively alleviated hepatic impairment in HFD-STZ-diabetic rats to near-normal control levels. The findings of the present study expound mechanisms by which luteolin mitigated diabetic dyslipidemia and alleviated hepatic impairment in HFD-STZ-diabetic rats by amelioration of oxidative stress, modulation of PPARα expression, and downregulation of ACAT-2 and SREBP-2. In conclusion, our results imply that luteolin may be efficacious in management of dyslipidemia in T2DM, and future research may be essential to substantiate our findings. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. The role of Indonesianin-5 in HMG CoA reductase and sterol regulatory element binding protein-2 expressions of hypercholesterolemic rat.
- Author
-
Gama, Noviyanty Indjar, Prasetyastuti, and Sunarti
- Subjects
- *
GENE expression , *LDL cholesterol , *RATTUS norvegicus , *FLAVONOIDS , *RATS , *LOW density lipoproteins , *SIMVASTATIN - Abstract
Increased cholesterol level increases the risk of heart disease and stroke. Sterol Regulatory Element Binding Protein-2 (SREBP-2) is a transcription factor for the synthesis of cholesterol genes. SREBP-2 can induce the expression of HMG CoA Reductase (HMGR). HMGR is a key cholesterol synthesis enzyme. Indonesianin-5, derived from Mahogany seeds (Swietenia macrophylla K), is a flavonoid. Indonesianin-5 is known to have a hypocholesterolemic effect. This study aimed to analyze the mechanism of Indonesianin-5 to decrease LDL and cholesterol levels and the expression of hepatic SREBP-2 and HMGR. Rattus norvegicus were grouped into 5 groups: normal group (N), Hypercholesterolemia group (HC), Hypercholesterolemic groups given simvastatin (HC+Sim), 10 mg of Indonesianin-5 (HC+ 10 Ind-5) and 90 mg of Indonesianin-5 (HC+ 90 Ind-5). In silico analysis was used to determine the effects of Indonesianin-5. The expressions of SREBP-2 and HMGR were analyzed by the RT-PCR method. Indonesianin-5 inhibited the activity of the HMGR enzyme. Expression of SREBP-2 was high after induction of Indonesianin-5 (p<0.05). The expression of HMGR was high after induction of Indonesianin-5 (p<0.05). Indonesianis-5 inhibits HMG CoA reductase and increases gene expression of SREBP-2 and HMG CoA reductase. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. Effects of Static Magnetic Field (SMF) Exposure on Coronary Heart Disease (CHD) Risk Indicators
- Author
-
Rahma Nur Istiqomah, Luluk Yunaini, Umiatin Umiatin, and Puji Sari
- Subjects
CHD ,Gen LDLR ,LDL ,SMF ,SREBP-2 ,Biotechnology ,TP248.13-248.65 ,Medicine - Abstract
Dyslipidemia is a condition of abnormalities in blood lipids, such as increased LDL and decreased HDL. Exposure to intracellular static magnetic fields (SMFs) can affect membranes to modulate the number of Ca2+ ions, which can affect cell biological effects. Increasing the number of Ca2+ ions will affect the expression of genes that help reduce LDL levels as an indicator of CHD. This study aims to see whether the influence of SMFs can affect calcium levels, lipid profiles, SREBP-2 protein excretion, and LDLR gene expression, which affects the process of breaking down cholesterol. We used obese C57BL/6J male mice divided into standard and HFD feed groups. The HFD feed group was split into Obes0, Obes2, Obes7, Obes14, and Obes21 groups based on the day of exposure. The exposure was carried out with an intensity of Bmax = 2 mT for 1 h daily. After exposure to SMFs, there were changes in blood cholesterol and LDL levels, which tended to decrease in the Obes2 and Obes7 groups and increase in the Obes14 and Obes21 groups. In addition, the exposed group tended to have an increase in the expression of the LDLR gene, SREBP-2 protein, and calcium ion compared to the control. Although it managed to increase after exposure on days 14 and 21, there was a decrease in the expression of the LDLR gene and SREBP-2 protein; hence, the effect of SMF exposure can reduce the risk of CHD. However, research still needs to be done regarding the effective duration of exposure.
- Published
- 2023
- Full Text
- View/download PDF
5. ABCA9, an ER cholesterol transporter, inhibits breast cancer cell proliferation via SREBP‐2 signaling.
- Author
-
Hwang, Hyeon‐Ji, Lee, Kang‐Hoon, and Cho, Je‐Yoel
- Abstract
The association between cholesterol metabolism and cancer development and progression has been recently highlighted. However, the role and function of many cholesterol transporters remain largely unknown. Here, we focused on the ATP‐binding cassette subfamily A member 9 (ABCA9) transporter given that its expression is significantly downregulated in both canine mammary tumors and human breast cancers, which in breast cancer patients correlates with poor prognosis. We found that ABCA9 is mainly present in the endoplasmic reticulum (ER) and is responsible for promoting cholesterol accumulation in this structure. Accordingly, ABCA9 inhibited sterol‐regulatory element binding protein‐2 (SREBP‐2) translocation from the ER to the nucleus, a crucial step for cholesterol synthesis, resulting in the downregulation of cholesterol synthesis gene expression. ABCA9 expression in breast cancer cells attenuated cell proliferation and reduced their colony‐forming abilities. We identified ABCA9 expression to be regulated by Forkhead box O1 (FOXO1). Inhibition of PI3K induced enhanced ABCA9 expression through the activation of the PI3K–Akt–FOXO1 pathway in breast cancer cells. Altogether, our study suggests that ABCA9 functions as an ER cholesterol transporter that suppresses cholesterol synthesis via the inhibition of SREBP‐2 signaling and that its restoration halts breast cancer cell proliferation. Our findings provide novel insight into the vital role of ABCA9 in breast cancer progression. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Effects of Static Magnetic Field (SMF) Exposure on Coronary Heart Disease (CHD) Risk Indicators.
- Author
-
Istiqomah, Rahma Nur, Yunaini, Luluk, Umiatin, Umiatin, and Sari, Puji
- Subjects
- *
MAGNETIC fields , *CORONARY disease , *CALCIUM , *LIPID metabolism , *OBESITY - Abstract
Dyslipidemia is a condition of abnormalities in blood lipids, such as increased LDL and decreased HDL. Exposure to intracellular static magnetic fields (SMFs) can affect membranes to modulate the number of Ca2+ ions, which can affect cell biological effects. Increasing the number of Ca2+ ions will affect the expression of genes that help reduce LDL levels as an indicator of CHD. This study aims to see whether the influence of SMFs can affect calcium levels, lipid profiles, SREBP-2 protein excretion, and LDLR gene expression, which affects the process of breaking down cholesterol. We used obese C57BL/6J male mice divided into standard and HFD feed groups. The HFD feed group was split into Obes0, Obes2, Obes7, Obes14, and Obes21 groups based on the day of exposure. The exposure was carried out with an intensity of Bmax = 2 mT for 1 h daily. After exposure to SMFs, there were changes in blood cholesterol and LDL levels, which tended to decrease in the Obes2 and Obes7 groups and increase in the Obes14 and Obes21 groups. In addition, the exposed group tended to have an increase in the expression of the LDLR gene, SREBP-2 protein, and calcium ion compared to the control. Although it managed to increase after exposure on days 14 and 21, there was a decrease in the expression of the LDLR gene and SREBP-2 protein; hence, the effect of SMF exposure can reduce the risk of CHD. However, research still needs to be done regarding the effective duration of exposure. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. SKI-1/S1P Facilitates SARS-CoV-2 Spike Induced Cell-to-Cell Fusion via Activation of SREBP-2 and Metalloproteases, Whereas PCSK9 Enhances the Degradation of ACE2.
- Author
-
Essalmani, Rachid, Andréo, Ursula, Evagelidis, Alexandra, Le Dévéhat, Maïlys, Pereira Ramos, Oscar Henrique, Fruchart Gaillard, Carole, Susan-Resiga, Delia, Cohen, Éric A., and Seidah, Nabil G.
- Subjects
- *
PROPROTEIN convertases , *ANGIOTENSIN converting enzyme , *METALLOPROTEINASES , *SARS-CoV-2 , *CATALYTIC domains , *VIRAL envelope proteins - Abstract
Proprotein convertases activate various envelope glycoproteins and participate in cellular entry of many viruses. We recently showed that the convertase furin is critical for the infectivity of SARS-CoV-2, which requires cleavage of its spike protein (S) at two sites: S1/S2 and S2′. This study investigates the implication of the two cholesterol-regulating convertases SKI-1 and PCSK9 in SARS-CoV-2 entry. The assays used were cell-to-cell fusion in HeLa cells and pseudoparticle entry into Calu-3 cells. SKI-1 increased cell-to-cell fusion by enhancing the activation of SREBP-2, whereas PCSK9 reduced cell-to-cell fusion by promoting the cellular degradation of ACE2. SKI-1 activity led to enhanced S2′ formation, which was attributed to increased metalloprotease activity as a response to enhanced cholesterol levels via activated SREBP-2. However, high metalloprotease activity resulted in the shedding of S2′ into a new C-terminal fragment (S2″), leading to reduced cell-to-cell fusion. Indeed, S-mutants that increase S2″ formation abolished S2′ and cell-to-cell fusion, as well as pseudoparticle entry, indicating that the formation of S2″ prevents SARS-CoV-2 cell-to-cell fusion and entry. We next demonstrated that PCSK9 enhanced the cellular degradation of ACE2, thereby reducing cell-to-cell fusion. However, different from the LDLR, a canonical target of PCSK9, the C-terminal CHRD domain of PCSK9 is dispensable for the PCSK9-induced degradation of ACE2. Molecular modeling suggested the binding of ACE2 to the Pro/Catalytic domains of mature PCSK9. Thus, both cholesterol-regulating convertases SKI-1 and PCSK9 can modulate SARS-CoV-2 entry via two independent mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Butyrate Lowers Cellular Cholesterol through HDAC Inhibition and Impaired SREBP-2 Signalling.
- Author
-
Bridgeman, Stephanie, Woo, Hon Chiu, Newsholme, Philip, and Mamotte, Cyril
- Subjects
- *
BUTYRATES , *SODIUM butyrate , *CHOLESTEROL , *CHOLESTEROL metabolism , *LDL cholesterol , *BLOOD cholesterol - Abstract
In animal studies, HDAC inhibitors such as butyrate have been reported to reduce plasma cholesterol, while conferring protection from diabetes, but studies on the underlying mechanisms are lacking. This study compares the influence of butyrate and other HDAC inhibitors to that of statins on cholesterol metabolism in multiple cell lines, but primarily in HepG2 hepatic cells due to the importance of the liver in cholesterol metabolism. Sodium butyrate reduced HepG2 cholesterol content, as did sodium valproate and the potent HDAC inhibitor trichostatin A, suggesting HDAC inhibition as the exacting mechanism. In contrast to statins, which increase SREBP-2 regulated processes, HDAC inhibition downregulated SREBP-2 targets such as HMGCR and the LDL receptor. Moreover, in contrast to statin treatment, butyrate did not increase cholesterol uptake by HepG2 cells, consistent with its failure to increase LDL receptor expression. Sodium butyrate also reduced ABCA1 and SRB1 protein expression in HepG2 cells, but these effects were not consistent across all cell types. Overall, the underlying mechanism of cell cholesterol lowering by sodium butyrate and HDAC inhibition is consistent with impaired SREBP-2 signalling, and calls into question the possible use of butyrate for lowering of serum LDL cholesterol in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. Amomum villosum Lour. Fruit extract mitigates hyperlipidemia through SREBP-2/LDLR/HMGCR signaling in high-cholesterol diet-fed mice
- Author
-
Ye-Seul Kim, Ha-Rim Kim, Paulrayer Antonisamy, Young-Rae Lee, Guemsan Lee, Hyun-Jong Jung, and Kang-Beom Kwon
- Subjects
Amomum villosum ,LDL receptor ,SREBP-2 ,HMGCR ,LDL cholesterol ,Science (General) ,Q1-390 - Abstract
Amomum villosum Lour. (Zingiberaceae) is an herbal medicine used in Asian countries for various ailments. In the current experiment, the effect of AV water extract (AVE) on cholesterol-lowering capabilities along with its essential bio-molecular mechanisms have been examined. The efficiency of AVE against high-cholesterol diet (HCD)-generated hyperlipidemia was investigated using C57BL/6 mice. Mice have been divided to six categories: control group (normal diet), high-cholesterol diet (HCD), and HCD treated with AVE at 100, 200, and 500 mg/kg, or simvastatin 40 mg/kg for four weeks. AVE treated animal groups (100, 200, 500 mg/kg) had markedly lessened body mass increase (p
- Published
- 2022
- Full Text
- View/download PDF
10. Cholesterol metabolism is a potential therapeutic target in Duchenne muscular dystrophy
- Author
-
Fatima Amor, Ai Vu Hong, Guillaume Corre, Mathilde Sanson, Laurence Suel, Stephanie Blaie, Laurent Servais, Thomas Voit, Isabelle Richard, and David Israeli
- Subjects
Duchenne muscular dystrophy ,Host gene ,Biological interpretation of miRNA dysregulation ,SREBP‐1 ,SREBP‐2 ,Lipid metabolism ,Diseases of the musculoskeletal system ,RC925-935 ,Human anatomy ,QM1-695 - Abstract
Abstract Background Duchenne muscular dystrophy (DMD) is a lethal muscle disease detected in approximately 1:5000 male births. DMD is caused by mutations in the DMD gene, encoding a critical protein that links the cytoskeleton and the extracellular matrix in skeletal and cardiac muscles. The primary consequence of the disrupted link between the extracellular matrix and the myofibre actin cytoskeleton is thought to involve sarcolemma destabilization, perturbation of Ca2+ homeostasis, activation of proteases, mitochondrial damage, and tissue degeneration. A recently emphasized secondary aspect of the dystrophic process is a progressive metabolic change of the dystrophic tissue; however, the mechanism and nature of the metabolic dysregulation are yet poorly understood. In this study, we characterized a molecular mechanism of metabolic perturbation in DMD. Methods We sequenced plasma miRNA in a DMD cohort, comprising 54 DMD patients treated or not by glucocorticoid, compared with 27 healthy controls, in three groups of the ages of 4–8, 8–12, and 12–20 years. We developed an original approach for the biological interpretation of miRNA dysregulation and produced a novel hypothesis concerning metabolic perturbation in DMD. We used the mdx mouse model for DMD for the investigation of this hypothesis. Results We identified 96 dysregulated miRNAs (adjusted P‐value
- Published
- 2021
- Full Text
- View/download PDF
11. 25-Hydroxycholesterol attenuates tumor necrosis factor alpha-induced blood-brain barrier breakdown in vitro.
- Author
-
Loiola, Rodrigo Azevedo, Nguyen, Cindy, Dib, Shiraz, Saint-Pol, Julien, Dehouck, Lucie, Sevin, Emmanuel, Naudot, Marie, Landry, Christophe, Pahnke, Jens, Pot, Caroline, and Gosselet, Fabien
- Subjects
- *
CHOLESTEROL metabolism , *TUMOR necrosis factors , *TIGHT junctions , *BLOOD-brain barrier , *CELL metabolism , *ENCEPHALITIS - Abstract
Intracellular cholesterol metabolism is regulated by the SREBP-2 and LXR signaling pathways. The effects of inflammation on these molecular mechanisms remain poorly studied, especially at the blood-brain barrier (BBB) level. Tumor necrosis factor α (TNFα) is a proinflammatory cytokine associated with BBB dysfunction. Therefore, the aim of our study was to investigate the effects of TNFα on BBB cholesterol metabolism, focusing on its underlying signaling pathways. Using a human in vitro BBB model composed of human brain-like endothelial cells (hBLECs) and brain pericytes (HBPs), we observed that TNFα increases BBB permeability by degrading the tight junction protein CLAUDIN-5 and activating stress signaling pathways in both cell types. TNFα also promotes cholesterol release and decreases cholesterol accumulation and APOE secretion. In hBLECs, the expression of SREBP-2 targets (LDLR and HMGCR) is increased, while ABCA1 expression is decreased. In HBPs, only LDLR and ABCA1 expression is increased. TNFα treatment also induces 25-hydroxycholesterol (25-HC) production, a cholesterol metabolite involved in the immune response and intracellular cholesterol metabolism. 25-HC pretreatment attenuates TNFα-induced BBB leakage and partially alleviates the effects of TNFα on ABCA1, LDLR, and HMGCR expression. Overall, our results suggest that TNFα favors cholesterol efflux via an LXR/ABCA1-independent mechanism at the BBB, while it activates the SREBP-2 pathway. Treatment with 25-HC partially reversed the effect of TNFα on the LXR/SREBP-2 pathways. Our study provides novel perspectives for better understanding cerebrovascular signaling events linked to BBB dysfunction and cholesterol metabolism in neuroinflammatory diseases. • TNFα increases BBB permeability via CLAUDIN-5 degradation • TNFα deeply alters cholesterol metabolism of BBB cells • TNFα promotes 25-hydroxycholesterol production • 25-HC partially abolishes TNFα effect on BBB cells [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Hempseed (Cannabis sativa) Peptide H3 (IGFLIIWV) Exerts Cholesterol-Lowering Effects in Human Hepatic Cell Line.
- Author
-
Li, Jianqiang, Bollati, Carlotta, Bartolomei, Martina, Mazzolari, Angelica, Arnoldi, Anna, Vistoli, Giulio, and Lammi, Carmen
- Abstract
Hempseed (Cannabis sativa) protein is an important source of bioactive peptides. H3 (IGFLIIWV), a transepithelial transported intestinal peptide obtained from the hydrolysis of hempseed protein with pepsin, carries out antioxidant and anti-inflammatory activities in HepG2 cells. In this study, the main aim was to assess its hypocholesterolemic effects at a cellular level and the mechanisms behind this health-promoting activity. The results showed that peptide H3 inhibited the 3-hydroxy-3-methylglutaryl co-enzyme A reductase (HMGCoAR) activity in vitro in a dose-dependent manner with an IC
50 value of 59 μM. Furthermore, the activation of the sterol regulatory element binding proteins (SREBP)-2 transcription factor, followed by the increase of low-density lipoprotein (LDL) receptor (LDLR) protein levels, was observed in human hepatic HepG2 cells treated with peptide H3 at 25 µM. Meanwhile, peptide H3 regulated the intracellular HMGCoAR activity through the increase of its phosphorylation by the activation of AMP-activated protein kinase (AMPK)-pathways. Consequently, the augmentation of the LDLR localized on the cellular membranes led to the improved ability of HepG2 cells to uptake extracellular LDL with a positive effect on cholesterol levels. Unlike the complete hempseed hydrolysate (HP), peptide H3 can reduce the proprotein convertase subtilisin/kexin 9 (PCSK9) protein levels and its secretion in the extracellular environment via the decrease of hepatic nuclear factor 1-α (HNF1-α). Considering all these evidences, H3 may represent a new bioactive peptide to be used for the development of dietary supplements and/or peptidomimetics for cardiovascular disease (CVD) prevention. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
13. PCSK9 mediates dyslipidemia induced by olanzapine treatment in schizophrenia patients.
- Author
-
Huang, Jing, Xiao, Jingmei, Peng, Zhuang, Shao, Ping, Sun, Mengxi, Long, Yujun, Wang, Xiaoyi, Shen, Manjun, Kang, Dongyu, Yang, Ye, Peng, Xingjie, Wang, Weiyan, Xie, Peng, Shao, Tiannan, Zhao, Jingping, and Wu, Renrong
- Subjects
- *
OLANZAPINE , *PEOPLE with schizophrenia , *LDL cholesterol , *DYSLIPIDEMIA , *LIPOPROTEIN receptors , *DOPAMINE receptors , *LIPID metabolism , *SCHIZOPHRENIA treatment , *HOMEOSTASIS , *ANIMAL experimentation , *HYPERLIPIDEMIA , *GENE expression , *WEIGHT gain , *MICE , *CARRIER proteins - Abstract
Rationale: It is controversial whether dyslipidemia induced by antipsychotics in schizophrenia patients is due to weight gain or direct effects of drug treatment. However, recent evidence showed that olanzapine can cause acute dyslipidemia independent of weight change, and the underlying mechanism remains unclear. Objective: To study the role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in olanzapine-induced dyslipidemia, we analyzed in schizophrenic patients and in experimental models involving mice and cells to understand the mechanism. Methods: Disturbances in lipid homeostasis caused by 8-week olanzapine treatment were prospectively evaluated in first-episode schizophrenic patients. Additionally, mice were administered olanzapine for 5 or 8 weeks to delineate liver actions for PCSK9 contributing to olanzapine-induced dyslipidemia. Results: Olanzapine directly affected lipid metabolism, suggesting dyslipidemia is independent of weight gain in schizophrenia patients. Olanzapine administration significantly increased plasma PCSK9, which was positively correlated with the increment in low-density lipoprotein cholesterol (LDL-C) (r=0.77, p<0.001). Increased expression of PCSK9 in liver tissue of olanzapine-treated mice occurred prior to olanzapine-induced LDL-C abnormality. Hepatic sterol regulatory element binding protein-2 (SREBP-2) protein levels increased in mice treated with olanzapine but largely declined in olanzapine (10μM) treated HepG2 cells, which suggested high concentration of olanzapine-induced PCSK9 increase was not SREBP-2-dependent. However, expressions of sterol regulatory element binding protein-1c (SREBP-1c) significantly increased in the higher dose treated groups, which was consistent with PCSK9 increases. Activation of SREBP-1c after high-dose olanzapine treatment promotes PSCK9 expression, and consequently the degradation of low-density lipoprotein receptors results in LDL-C increase. Conclusions: Lipid disturbances caused by olanzapine are independent of weight gain. The study explored the relationship between SREBP-1c and PCSK9 in regulating lipoprotein metabolism after olanzapine treatment in vitro and in vivo. Further exploration of olanzapine-induced PCSK9 regulatory mechanisms may help identify control points for inhibition of olanzapine-mediated dyslipidemia. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Butyrate Lowers Cellular Cholesterol through HDAC Inhibition and Impaired SREBP-2 Signalling
- Author
-
Stephanie Bridgeman, Hon Chiu Woo, Philip Newsholme, and Cyril Mamotte
- Subjects
HDAC inhibitors ,butyrate ,cholesterol ,LDL ,statins ,SREBP-2 ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
In animal studies, HDAC inhibitors such as butyrate have been reported to reduce plasma cholesterol, while conferring protection from diabetes, but studies on the underlying mechanisms are lacking. This study compares the influence of butyrate and other HDAC inhibitors to that of statins on cholesterol metabolism in multiple cell lines, but primarily in HepG2 hepatic cells due to the importance of the liver in cholesterol metabolism. Sodium butyrate reduced HepG2 cholesterol content, as did sodium valproate and the potent HDAC inhibitor trichostatin A, suggesting HDAC inhibition as the exacting mechanism. In contrast to statins, which increase SREBP-2 regulated processes, HDAC inhibition downregulated SREBP-2 targets such as HMGCR and the LDL receptor. Moreover, in contrast to statin treatment, butyrate did not increase cholesterol uptake by HepG2 cells, consistent with its failure to increase LDL receptor expression. Sodium butyrate also reduced ABCA1 and SRB1 protein expression in HepG2 cells, but these effects were not consistent across all cell types. Overall, the underlying mechanism of cell cholesterol lowering by sodium butyrate and HDAC inhibition is consistent with impaired SREBP-2 signalling, and calls into question the possible use of butyrate for lowering of serum LDL cholesterol in humans.
- Published
- 2022
- Full Text
- View/download PDF
15. SIRT-1 Activity Sustains Cholesterol Synthesis in the Brain.
- Author
-
Paulazo, María A. and Sodero, Alejandro O.
- Subjects
- *
CHOLESTEROL , *CHOLESTEROL metabolism , *NEUROTRANSMITTER receptors , *APOLIPOPROTEIN E , *ADENOSINE triphosphate , *HYDROXYCHOLESTEROLS - Abstract
[Display omitted] • SIRT-1 inhibition with EX-527 downregulates SREBP-2 and HMGCR expression, and reduces cholesterol levels. • Cholesterol reduction occurs in neurons and astrocytes, and affects the synaptic content of this lipid. • SIRT-1 activity is required to sustain normal levels of cholesterol synthesis in the adult brain. SIRT-1 is a potent energy regulator that has been implicated in the aging of different tissues, and cholesterol synthesis demands high amounts of cellular adenosine triphosphate. An efficient synaptic transmission depends on processes that are highly influenced by cholesterol levels, like endocytosis, exocytosis and membrane lateral diffusion of neurotransmitter receptors. We set out to investigate whether SIRT-1 activity affects brain cholesterol metabolism. We found that pharmacological inhibition of SIRT-1 with EX-527 reduces the mRNA amounts of 3-hydroxy-3-methylglutaryl-Coenzyme A reductase (HMGCR), Cytochrome P450 46A1 (CYP46A1) and Apolipoprotein E (APO-E) in rat primary cortical cultures. The decreased expression of these genes was paralleled by a significant reduction of the cholesterol levels in this type of neuronal culture. Interestingly, a cholesterol decrease of similar extent was observed in mouse astroglial cultures after EX-527 treatment. In agreement, mice administered with EX-527 for 5 days showed a down-regulation of cholesterol synthesis in the cortex, with significant reductions in the mRNA amounts of the transcription factor Sterol Regulatory Element Binding Protein 2 (SREBP-2) and the enzyme HMGCR, two key regulators of the cholesterol synthesis. These transcriptional changes were paralleled by reduced cholesterol levels at cortical synapses. SIRT-1 inhibition also reduced the amount of cholesterol in the hippocampus but without affecting the HMGCR expression levels. Altogether, these results uncover a role for SIRT-1 in the regulation of cholesterol metabolism, and demonstrate that SIRT-1 is required to sustain adequate levels of cholesterol synthesis in the adult brain. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
16. Cholesterol metabolism is a potential therapeutic target in Duchenne muscular dystrophy.
- Author
-
Amor, Fatima, Vu Hong, Ai, Corre, Guillaume, Sanson, Mathilde, Suel, Laurence, Blaie, Stephanie, Servais, Laurent, Voit, Thomas, Richard, Isabelle, and Israeli, David
- Subjects
DUCHENNE muscular dystrophy ,DRUG target ,CHOLESTEROL metabolism ,LIPID metabolism ,HOMEOSTASIS ,LABORATORY mice ,CYTOSKELETON ,ACTIN - Abstract
Background: Duchenne muscular dystrophy (DMD) is a lethal muscle disease detected in approximately 1:5000 male births. DMD is caused by mutations in the DMD gene, encoding a critical protein that links the cytoskeleton and the extracellular matrix in skeletal and cardiac muscles. The primary consequence of the disrupted link between the extracellular matrix and the myofibre actin cytoskeleton is thought to involve sarcolemma destabilization, perturbation of Ca2+ homeostasis, activation of proteases, mitochondrial damage, and tissue degeneration. A recently emphasized secondary aspect of the dystrophic process is a progressive metabolic change of the dystrophic tissue; however, the mechanism and nature of the metabolic dysregulation are yet poorly understood. In this study, we characterized a molecular mechanism of metabolic perturbation in DMD. Methods: We sequenced plasma miRNA in a DMD cohort, comprising 54 DMD patients treated or not by glucocorticoid, compared with 27 healthy controls, in three groups of the ages of 4–8, 8–12, and 12–20 years. We developed an original approach for the biological interpretation of miRNA dysregulation and produced a novel hypothesis concerning metabolic perturbation in DMD. We used the mdx mouse model for DMD for the investigation of this hypothesis. Results: We identified 96 dysregulated miRNAs (adjusted P‐value <0.1), of which 74 were up‐regulated and 22 were down‐regulated in DMD. We confirmed the dysregulation in DMD of Dystro‐miRs, Cardio‐miRs, and a large number of the DLK1‐DIO3 miRNAs. We also identified numerous dysregulated miRNAs yet unreported in DMD. Bioinformatics analysis of both target and host genes for dysregulated miRNAs predicted that lipid metabolism might be a critical metabolic perturbation in DMD. Investigation of skeletal muscles of the mdx mouse uncovered dysregulation of transcription factors of cholesterol and fatty acid metabolism (SREBP‐1 and SREBP‐2), perturbation of the mevalonate pathway, and the accumulation of cholesterol in the dystrophic muscles. Elevated cholesterol level was also found in muscle biopsies of DMD patients. Treatment of mdx mice with Simvastatin, a cholesterol‐reducing agent, normalized these perturbations and partially restored the dystrophic parameters. Conclusions: This investigation supports that cholesterol metabolism and the mevalonate pathway are potential therapeutic targets in DMD. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
17. Hempseed (Cannabis sativa) Peptide H3 (IGFLIIWV) Exerts Cholesterol-Lowering Effects in Human Hepatic Cell Line
- Author
-
Jianqiang Li, Carlotta Bollati, Martina Bartolomei, Angelica Mazzolari, Anna Arnoldi, Giulio Vistoli, and Carmen Lammi
- Subjects
hempseed protein ,LDLR ,SREBP-2 ,cholesterol metabolism ,PCSK9 ,Nutrition. Foods and food supply ,TX341-641 - Abstract
Hempseed (Cannabis sativa) protein is an important source of bioactive peptides. H3 (IGFLIIWV), a transepithelial transported intestinal peptide obtained from the hydrolysis of hempseed protein with pepsin, carries out antioxidant and anti-inflammatory activities in HepG2 cells. In this study, the main aim was to assess its hypocholesterolemic effects at a cellular level and the mechanisms behind this health-promoting activity. The results showed that peptide H3 inhibited the 3-hydroxy-3-methylglutaryl co-enzyme A reductase (HMGCoAR) activity in vitro in a dose-dependent manner with an IC50 value of 59 μM. Furthermore, the activation of the sterol regulatory element binding proteins (SREBP)-2 transcription factor, followed by the increase of low-density lipoprotein (LDL) receptor (LDLR) protein levels, was observed in human hepatic HepG2 cells treated with peptide H3 at 25 µM. Meanwhile, peptide H3 regulated the intracellular HMGCoAR activity through the increase of its phosphorylation by the activation of AMP-activated protein kinase (AMPK)-pathways. Consequently, the augmentation of the LDLR localized on the cellular membranes led to the improved ability of HepG2 cells to uptake extracellular LDL with a positive effect on cholesterol levels. Unlike the complete hempseed hydrolysate (HP), peptide H3 can reduce the proprotein convertase subtilisin/kexin 9 (PCSK9) protein levels and its secretion in the extracellular environment via the decrease of hepatic nuclear factor 1-α (HNF1-α). Considering all these evidences, H3 may represent a new bioactive peptide to be used for the development of dietary supplements and/or peptidomimetics for cardiovascular disease (CVD) prevention.
- Published
- 2022
- Full Text
- View/download PDF
18. Curcumin inhibits the proteolytic process of SREBP‐2 by first inhibiting the expression of S1P rather than directly inhibiting SREBP‐2 expression.
- Author
-
Li, Yongnan and Wu, Shuodong
- Subjects
- *
STEROL regulatory element-binding proteins , *CURCUMIN - Abstract
Many studies have demonstrated that curcumin can downregulate mRNA levels of sterol regulatory element‐binding proteins (SREBP‐2); however, our study did not find similar results. This study was designed to demonstrate that curcumin inhibits the proteolytic process of SREBP‐2 by first inhibiting the expression of membrane‐bound transcription factor site‐1 protease (S1P) rather than directly inhibiting SREBP‐2 expression. After curcumin treatment, Caco‐2 cells were collected to observe the dose‐ and time‐dependent dynamics of precursor and mature SREBP‐2, transcription factor‐specific protein 1 (SP‐1), and SREBP cleavage‐activating protein (SCAP). After curcumin treatment, SREBP‐2 distribution was detected in the cells and S1P protein expression was examined. Curcumin could downregulate mRNA levels of SREBP2, SP‐1 and SCAP, but it did not simultaneously downregulate the expression of precursor SREBP‐2 (pSREBP‐2) and SCAP. Curcumin can inhibit the proteolytic process of SREBP‐2, reduce the production of mature SREBP‐2 (mSREBP‐2), and change the cellular distribution of SREBP‐2. The inhibitory effect of curcumin on SP‐1 protein expression is short‐acting. Curcumin could downregulate the mRNA and protein expression of S1P, but has no obvious inhibitory effect on the mRNA and protein expression of S2P (site‐2 protease). Curcumin can inhibit the SREBP‐2 proteolytic process to reduce mSREBP‐2 which functions as a transcription factor, affecting the regulation of cholesterol metabolism‐related genes. Curcumin does not directly inhibit the expression of mSREBP‐2 protein, and it has no such inhibitory effect for at least a short period of time, although curcumin does reduce the amount of mSREBP‐2 protein. S1P is a key protease in the hydrolysis of pSREBP‐2 into mSREBP‐2. Therefore, curcumin may decrease the amount of mSREBP‐2 by directly inhibiting the expression of S1P mRNA and protein. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
19. Targeting SREBP-2-Regulated Mevalonate Metabolism for Cancer Therapy
- Author
-
Linyuan Xue, Hongyu Qi, He Zhang, Lu Ding, Qingxia Huang, Daqing Zhao, Boyang Jason Wu, and Xiangyan Li
- Subjects
SREBP-2 ,HMG-CoA reductase ,mevalonate ,cholesterol ,cancer therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Recently, targeting metabolic reprogramming has emerged as a potential therapeutic approach for fighting cancer. Sterol regulatory element binding protein-2 (SREBP-2), a basic helix-loop-helix leucine zipper transcription factor, mainly regulates genes involved in cholesterol biosynthesis and homeostasis. SREBP-2 binds to the sterol regulatory elements (SREs) in the promoters of its target genes and activates the transcription of mevalonate pathway genes, such as HMG-CoA reductase (HMGCR), mevalonate kinase and other key enzymes. In this review, we first summarized the structure of SREBP-2 and its activation and regulation by multiple signaling pathways. We then found that SREBP-2 and its regulated enzymes, including HMGCR, FPPS, SQS, and DHCR4 from the mevalonate pathway, participate in the progression of various cancers, including prostate, breast, lung, and hepatocellular cancer, as potential targets. Importantly, preclinical and clinical research demonstrated that fatostatin, statins, and N-BPs targeting SREBP-2, HMGCR, and FPPS, respectively, alone or in combination with other drugs, have been used for the treatment of different cancers. This review summarizes new insights into the critical role of the SREBP-2-regulated mevalonate pathway for cancer and its potential for targeted cancer therapy.
- Published
- 2020
- Full Text
- View/download PDF
20. miR-33a Mediates the Anti-Tumor Effect of Lovastatin in Osteosarcoma by Targeting CYR61
- Author
-
Yazeng Huang, Jun Zhang, Haiyu Shao, Jianwen Liu, Mengran Jin, Jinping Chen, and Hongying Zhao
- Subjects
Osteosarcoma ,Lovastatin ,CYR61 ,SREBP-2 ,miR-33a ,Cell invasion ,Epithelial-to-mesenchymal transition ,Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Abstract
Background/Aims: Preventing cell metastasis is an effective therapeutic strategy to treat osteosarcoma and improve prognosis. Statins have been found to have anticancer effects in addition to their cholesterol-lowering action. As a new target of statins, cysteine-rich 61 (CYR61) was recently identified to promote cell migration and metastasis in osteosarcoma. However, the underlying mechanisms mediating the regulation of CYR61 expression by statins remain unknown. Methods: Human osteosarcoma cell lines MG63 and SaOS2 were used to clarify the effect of lovastatin on CYR61 expression. Real-time PCR was performed to detect mRNA or microRNA (miRNA) levels and western blot was performed to detect protein levels. Cell invasive ability was determined using Transwell assays. Lentivirus encoding CYR61 cDNA or sterol regulatory element-binding protein 2 (SREBP-2) shRNA was used to upregulate CYR61 expression or downregulate SREBP-2 expression. Binding of the CYR61 3’ untranslated region (UTR) and miR-33a was analyzed by luciferase reporter assay. Results: We found that lovastatin treatment decreased CYR61 expression, inhibited cell invasion and altered epithelial-to-mesenchymal-transition (EMT)-related protein expression, while CYR61 overexpression abolished the effect of lovastatin. Moreover, lovastatin increased the expression of SREBP-2 and miR-33a, which were then downregulated by SREBP-2 silencing. Bioinformatics analysis indicated that the CYR61 3′UTR harbored a potential miR-33a binding site and luciferase reporter assay demonstrated that CYR61 was a target of miR-33a in osteosarcoma cells. Furthermore, miR-33a could inhibit cell invasion and alter EMT-related protein expression. SREBP-2 silencing or miR-33a inhibitor upregulated CYR61 expression and reversed the effects of lovastatin on cell invasion and EMT-related proteins. Conclusion: Our findings suggest lovastatin suppresses osteosarcoma cell invasion through the SREBP-2/miR-33a/CYR61 pathway.
- Published
- 2018
- Full Text
- View/download PDF
21. Targeting SREBP-2-Regulated Mevalonate Metabolism for Cancer Therapy.
- Author
-
Xue, Linyuan, Qi, Hongyu, Zhang, He, Ding, Lu, Huang, Qingxia, Zhao, Daqing, Wu, Boyang Jason, and Li, Xiangyan
- Subjects
CANCER treatment ,MEVALONATE kinase ,LEUCINE zippers ,TRANSCRIPTION factors ,LIVER cancer - Abstract
Recently, targeting metabolic reprogramming has emerged as a potential therapeutic approach for fighting cancer. Sterol regulatory element binding protein-2 (SREBP-2), a basic helix-loop-helix leucine zipper transcription factor, mainly regulates genes involved in cholesterol biosynthesis and homeostasis. SREBP-2 binds to the sterol regulatory elements (SREs) in the promoters of its target genes and activates the transcription of mevalonate pathway genes, such as HMG-CoA reductase (HMGCR), mevalonate kinase and other key enzymes. In this review, we first summarized the structure of SREBP-2 and its activation and regulation by multiple signaling pathways. We then found that SREBP-2 and its regulated enzymes, including HMGCR, FPPS, SQS, and DHCR4 from the mevalonate pathway, participate in the progression of various cancers, including prostate, breast, lung, and hepatocellular cancer, as potential targets. Importantly, preclinical and clinical research demonstrated that fatostatin, statins, and N-BPs targeting SREBP-2, HMGCR, and FPPS, respectively, alone or in combination with other drugs, have been used for the treatment of different cancers. This review summarizes new insights into the critical role of the SREBP-2-regulated mevalonate pathway for cancer and its potential for targeted cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. Neoagarooligosaccharides enhance the level and efficiency of LDL receptor and improve cholesterol homeostasis
- Author
-
Ji Hye Yang, Sam Seok Cho, Kyu Min Kim, Ji Young Kim, Eun Joo Kim, Eun Young Park, Je Hyeon Lee, and Sung Hwan Ki
- Subjects
LDL cholesterol ,LDL receptor ,Neoagarooligosaccharides ,PCSK9 ,SREBP-2 ,Nutrition. Foods and food supply ,TX341-641 - Abstract
Neoagarooligosaccharides (NAOS) are produced by β-agarases through the cleavage of β-1,4 linkage in agarose. Here, we investigated the cholesterol-lowering properties of NAOS and its underlying molecular mechanism. Administration of NAOS in mice fed a high-cholesterol diet (HCD) inhibited hepatic fat accumulation and serum LDL cholesterol. Mice fed an HCD showed a decrease in LDL receptor (LDLR) expression, which was reversed by NAOS. Moreover, NAOS enhanced LDLR expression and LDL uptake in hepatocytes. NAOS increased nuclear translocation of SREBP-2, a crucial transcription factor for LDLR gene expression. Increased LDLR luciferase activity, induced by NAOS, was abolished on deletion of SRE in the LDLR promoter. NAOS upregulated LDLR levels via PI3K/Akt-mediated activation of SREBP-2. Furthermore, we demonstrated that NAOS reduced the nuclear HNF-1α, leading to decreased levels of PCSK9, a natural inhibitor of LDLR. These findings indicate that NAOS exhibits hypolipidemic activity, and may serve as a new hypolipidemic drug.
- Published
- 2017
- Full Text
- View/download PDF
23. The sterol regulatory element‐binding protein 2 is dysregulated by tau alterations in Alzheimer disease.
- Author
-
Wang, Chunyu, Zhao, Fanpeng, Shen, Katie, Wang, Wenzhang, Siedlak, Sandra L., Lee, Hyoung‐gon, Phelix, Clyde F., Perry, George, Shen, Lu, Tang, Beisha, Yan, Riqiang, and Zhu, Xiongwei
- Subjects
- *
STEROL regulatory element-binding proteins , *NEUROFIBRILLARY tangles , *ALZHEIMER'S disease , *PYRAMIDAL neurons - Abstract
Disturbed neuronal cholesterol homeostasis has been observed in Alzheimer disease (AD) and contributes to the pathogenesis of AD. As the master switch of cholesterol biosynthesis, the sterol regulatory element‐binding protein 2 (SREBP‐2) translocates to the nucleus after cleavage/activation, but its expression and activation have not been studied in AD which is the focus of the current study. We found both a significant decrease in the nuclear translocation of N‐terminal SREBP‐2 accompanied by a significant accumulation of C‐terminal SREBP‐2 in NFT‐containing pyramidal neurons in AD. N‐terminal‐ SREBP‐2 is also found in dystrophic neurites around plaques in AD brain. Western blot confirmed a significantly reduced nuclear translocation of mature SREBP‐2 (mSREBP‐2) in AD brain. Interestingly, reduced nuclear mSREBP‐2 was only found in animal models of tauopathies such as 3XTg AD mice and P301L Tau Tg mice but not in CRND8 APP transgenic mice, suggesting that tau alterations likely are involved in the changes of mSREBP‐2 distribution and activation in AD. Altogether, our study demonstrated disturbed SREBP‐2 signaling in AD and related models, and proved for the first time that tau alterations contribute to disturbed cholesterol homeostasis in AD likely through modulation of nuclear mSREBP‐2 translocation. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
24. SREBP-2 aggravates breast cancer associated osteolysis by promoting osteoclastogenesis and breast cancer metastasis.
- Author
-
Jie, Zhiwei, Xie, Ziang, Xu, Wenbin, Zhao, Xiangde, Jin, Gu, Sun, Xuewu, Huang, Bao, Tang, Pan, Wang, Gangliang, Shen, Shuying, Qin, An, and Fan, Shunwu
- Subjects
- *
BREAST cancer , *BONE resorption , *OSTEOCLASTOGENESIS , *METASTASIS , *CREB protein - Abstract
Abstract Bone is one of the most common sites of breast cancer metastasis and a major cause of high mortality in these patients. Thus, further understanding the molecular mechanisms regulating breast cancer-induced osteolysis is critical for the development of more effective treatments. In this study, we demonstrated that important roles sterol regulatory element-binding protein 2 (SREBP-2) play in osteoclast formation a function, and in breast cancer metastasis. SREBP-2 expression was found to be induced during the early stages of osteoclast formation under the control of the RANKL/cAMP-response element binding protein (CREB) signaling cascade. SREBP-2 is subsequently translocated into the nucleus where it participates with other transcriptional factors to induce the expression of NFATc1 required for mature osteoclast formation. Additionally, SREBP-2 was also found to be highly expressed in breast cancer tissues and correlated with a poor prognosis. SREBP-2 was similarly under the transcriptional control of CREB and its induction regulates the expression of matrix metalloproteinases (MMPs), key degradative enzymes involved in bone metastases by breast cancer cells. Accordingly, targeting of SREBP-2 with Fatostatin which specifically inhibits SCAP (SREBP cleavage-activating protein) and prevents SREBP activation, attenuated breast cancer-induced osteolysis in vivo. Collectively, our results suggest that SREBP-2 plays a critical role in regulating osteoclastogenesis and contributes to breast cancer-induced osteolysis. Thus, SREBP-2 inhibition is a potential therapeutic approach for breast cancer patients with osteolytic bone lesions. Highlights • CREB/SREBP-2/NFATc1 is a novel signaling axis regulating RANKL-induced osteoclastogenesis. • Elevated SREBP-2 expression contributes to breast cancer metastasis and is a predictor of poor prognosis in patients. • SREBP-2 is a novel molecular target that can alleviate breast cancer-induced osteolytic lesions in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
25. miR-33a Mediates the Anti-Tumor Effect of Lovastatin in Osteosarcoma by Targeting CYR61.
- Author
-
Huang, Yazeng, Zhang, Jun, Shao, Haiyu, Liu, Jianwen, Jin, Mengran, Chen, Jinping, and Zhao, Hongying
- Subjects
- *
METASTASIS , *LOVASTATIN , *ANTINEOPLASTIC agents , *OSTEOSARCOMA , *POLYMERASE chain reaction , *CYSTEINE - Abstract
Background/Aims: Preventing cell metastasis is an effective therapeutic strategy to treat osteosarcoma and improve prognosis. Statins have been found to have anticancer effects in addition to their cholesterol-lowering action. As a new target of statins, cysteine-rich 61 (CYR61) was recently identified to promote cell migration and metastasis in osteosarcoma. However, the underlying mechanisms mediating the regulation of CYR61 expression by statins remain unknown. Methods: Human osteosarcoma cell lines MG63 and SaOS2 were used to clarify the effect of lovastatin on CYR61 expression. Real-time PCR was performed to detect mRNA or microRNA (miRNA) levels and western blot was performed to detect protein levels. Cell invasive ability was determined using Transwell assays. Lentivirus encoding CYR61 cDNA or sterol regulatory element-binding protein 2 (SREBP-2) shRNA was used to upregulate CYR61 expression or downregulate SREBP-2 expression. Binding of the CYR61 3' untranslated region (UTR) and miR-33a was analyzed by luciferase reporter assay. Results: We found that lovastatin treatment decreased CYR61 expression, inhibited cell invasion and altered epithelial-to-mesenchymal-transition (EMT)-related protein expression, while CYR61 overexpression abolished the effect of lovastatin. Moreover, lovastatin increased the expression of SREBP-2 and miR-33a, which were then downregulated by SREBP-2 silencing. Bioinformatics analysis indicated that the CYR61 3′UTR harbored a potential miR-33a binding site and luciferase reporter assay demonstrated that CYR61 was a target of miR-33a in osteosarcoma cells. Furthermore, miR-33a could inhibit cell invasion and alter EMT-related protein expression. SREBP-2 silencing or miR-33a inhibitor upregulated CYR61 expression and reversed the effects of lovastatin on cell invasion and EMT-related proteins. Conclusion: Our findings suggest lovastatin suppresses osteosarcoma cell invasion through the SREBP-2/miR-33a/CYR61 pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
26. Genistein upregulates LDLR levels via JNK-mediated activation of SREBP-2
- Author
-
Medicia Kartawijaya, Hye Won Han, Yunhye Kim, and Seung-Min Lee
- Subjects
genistein ,JNK ,LDL receptor ,cholesterol ,SREBP-2 ,Nutrition. Foods and food supply ,TX341-641 - Abstract
Background: Genistein has been proved in vitro and in vivo to lower LDLR level. It is also widely consumed and implicated for its anti-atherogenic effects. However, the molecular mechanism by which genistein lowers the LDL level is still unknown. Objective: To understand the anti-atherogenic molecular mechanism of action, genistein was investigated for its impact on the expression of LDLR, the receptor for LDL cholesterol, and related signaling pathways in a human hepatoma cell line. Design: HepG2 cell was used for the experiments. Genistein with different concentrations was diluted in media and was incubated for 24 h or more as indicated. Protein levels were measured by western blotting, and mRNA expression was detected by RT-qPCR. Chromatin immunoprecipitation assay (CHIP) assay was used to determine protein binding levels, and luciferase assay was used to measure promoter activity. Result: Genistein increased the mRNA and protein levels of LDLR in a time-dependent manner. Genistein increased the transcriptional activity of the LDLR promoter containing the reporter gene (pLDLR-luc, −805 to +50). But the sterol regulatory element deletion mutant construct failed to be activated by genistein. Genistein increased the nuclear fraction of SREBP-2 and the DNA-binding activity of SREBP-2 to LDLR promoter, as assessed by CHIP. The genistein-phosphorylated JNK inhibitor (SP600126) abolished the genistein-stimulated levels of LDLR and the nuclear SREBP-2. The addition of cholesterol up to 5 µg/mL for 24 h did not affect the effect of genistein on LDLR protein expression. Even the addition of 40 µM genistein increased the cholesterol uptake by more than 10% in the human hepatoma cell line. Conclusion: Our data support the idea that genistein may have anti-atherogenic effects by activating JNK signals and SREBP-2 processing, which is followed by the upregulation of LDLR.
- Published
- 2016
- Full Text
- View/download PDF
27. Serum sdLDL-C and cellular SREBP2-dependent cholesterol levels: Is there a challenge on targeting PCSK9?
- Author
-
Soltanmohammadi Elham, Piran Sadegh, Mohammadi Asghar, Hosseni Bita, Naseri Faezeh, Shabani Mohammad, and Najafi Mohammad
- Subjects
sdldl-c ,ldl-c ,pcsk9 ,srebp-2 ,Biochemistry ,QD415-436 - Abstract
Background: Serum small dense LDL-cholesterol (sdLDLC) value is suggested to be an important risk factor for atherosclerosis. Since sdLDL-C changes may be related to PCSK9 and SREBP-2 functions, the aim of this study was to investigate correlations between sdLDL-C, circulating PCSK9, SREBP-2 expression and some lipid parameters in serum and buffy coat fraction of healthy subjects. Methods: One hundred and twenty-four subjects were randomly included in the study. The lipid profile was measured using routine laboratory methods. The serum sdLDL-C level was calculated by a heparin-related precipitation technique. The cellular LDL-C/protein and cholesterol/protein values were measured after lysing of cells with methanol/ch loroform binary solvent. The circulating PCSK9 level was measured using ELISA technique. The SREBP-2 expression level was estimated using the RT-qPCR technique. Results: Data showed significant correlations between LDLC, TG and sdLDL-C levels (r=0.34, p=0.001; r=0.2, p=0.04). The circulating PCSK9 level was correlated to LDL-C (r=0.29, p=0.04), but not to sdLDL-C (r=-0.08, p=0.57). Also, cellular LDL-C value was not related to serum LDL-C level (r=-0.12, p=0.39). Furthermore, there was an inverse correlation between cellular LDL-C/protein value and estimated de novo cholesterol/protein value (r= -0.5, p=0.001). Similar results were observed for cellular LDL-C/protein value and SREBP-2 expression level (r= -0.52, p=0.004). Conclusions: We concluded that the serum sdLDL-C value is not related to circulating PCSK9. Furthermore, SREBP-2 regulatory system was able to elevate the cellular cholesterol level after reducing LDL influx. We suggest to investigate the cellular sdLDL fate and lipid synthesis pathways in PCSK9-targeting studies.
- Published
- 2016
28. SKI-1/S1P Facilitates SARS-CoV-2 Spike Induced Cell-to-Cell Fusion via Activation of SREBP-2 and Metalloproteases, Whereas PCSK9 Enhances the Degradation of ACE2
- Author
-
Rachid Essalmani, Ursula Andréo, Alexandra Evagelidis, Maïlys Le Dévéhat, Oscar Henrique Pereira Ramos, Carole Fruchart Gaillard, Delia Susan-Resiga, Éric A. Cohen, and Nabil G. Seidah
- Subjects
PCSK9 ,SREBP-2 ,shedding ,Infectious Diseases ,SARS-CoV-2 ,Virology ,cell-to-cell fusion ,proprotein convertases ,protease inhibitors ,biochemistry ,SKI-1/S1P ,metalloproteases ,mutagenesis - Abstract
Proprotein convertases activate various envelope glycoproteins and participate in cellular entry of many viruses. We recently showed that the convertase furin is critical for the infectivity of SARS-CoV-2, which requires cleavage of its spike protein (S) at two sites: S1/S2 and S2′. This study investigates the implication of the two cholesterol-regulating convertases SKI-1 and PCSK9 in SARS-CoV-2 entry. The assays used were cell-to-cell fusion in HeLa cells and pseudoparticle entry into Calu-3 cells. SKI-1 increased cell-to-cell fusion by enhancing the activation of SREBP-2, whereas PCSK9 reduced cell-to-cell fusion by promoting the cellular degradation of ACE2. SKI-1 activity led to enhanced S2′ formation, which was attributed to increased metalloprotease activity as a response to enhanced cholesterol levels via activated SREBP-2. However, high metalloprotease activity resulted in the shedding of S2′ into a new C-terminal fragment (S2″), leading to reduced cell-to-cell fusion. Indeed, S-mutants that increase S2″ formation abolished S2′ and cell-to-cell fusion, as well as pseudoparticle entry, indicating that the formation of S2″ prevents SARS-CoV-2 cell-to-cell fusion and entry. We next demonstrated that PCSK9 enhanced the cellular degradation of ACE2, thereby reducing cell-to-cell fusion. However, different from the LDLR, a canonical target of PCSK9, the C-terminal CHRD domain of PCSK9 is dispensable for the PCSK9-induced degradation of ACE2. Molecular modeling suggested the binding of ACE2 to the Pro/Catalytic domains of mature PCSK9. Thus, both cholesterol-regulating convertases SKI-1 and PCSK9 can modulate SARS-CoV-2 entry via two independent mechanisms.
- Published
- 2023
- Full Text
- View/download PDF
29. Unripe Rubus coreanus Miquel Extract Containing Ellagic Acid Regulates AMPK, SREBP-2, HMGCR, and INSIG-1 Signaling and Cholesterol Metabolism In Vitro and In Vivo
- Author
-
Ki Hoon Lee, Eui-Seon Jeong, Goeun Jang, Ju-Ryun Na, Soyi Park, Wan Seok Kang, Eun Kim, Hakjoon Choi, Jin Seok Kim, and Sunoh Kim
- Subjects
unripe rubus coreanus ,cholesterol ,hypercholesterolemia ,ampk ,srebp-2 ,hmgcr ,Nutrition. Foods and food supply ,TX341-641 - Abstract
Our previous study demonstrated that a 5% ethanol extract of unripe Rubus coreanus (5-uRCK) has hypo-cholesterolemic and anti-obesity activity. However, the molecular mechanisms of its effects are poorly characterized. We hypothesized that 5-uRCK and one of its major bioactive compounds, ellagic acid, decrease cellular and plasma cholesterol levels. Thus, we investigated the hypocholesterolemic activity and mechanism of 5-uRCK in both hepatocytes and a high-cholesterol diet (HCD)-induced rat model. Cholesterol in the liver and serum was significantly reduced by 5-uRCK and ellagic acid. The hepatic activities of HMG-CoA and CETP were reduced, and the hepatic activity of LCAT was increased by both 5-uRCK extract and ellagic acid, which also caused histological improvements. The MDA content in the aorta and serum was significantly decreased after oral administration of 5-uRCK or ellagic acid. Further immunoblotting analysis showed that AMPK phosphorylation in the liver was induced by 5-uRCK and ellagic acid, which activated AMPK, inhibiting the activity of HMGCR by inhibitory phosphorylation. In contrast, 5-uRCK and ellagic acid suppressed the nuclear translocation and activation of SREBP-2, which is a key transcription factor in cholesterol biosynthesis. In conclusion, our results suggest that 5-uRCK and its bioactive compound, ellagic acid, are useful alternative therapeutic agents to regulate blood cholesterol.
- Published
- 2020
- Full Text
- View/download PDF
30. MicroRNA-98 regulates hepatic cholesterol metabolism via targeting sterol regulatory element-binding protein 2.
- Author
-
Geng, Chong, Dong, Tianyi, Jin, Weilong, Yu, Bo, Yin, Fuhui, Peng, Fengqiang, Chen, Guanmin, Ji, Chuncai, and Ding, Feng
- Subjects
- *
MICRORNA , *CORONARY disease , *CHOLESTEROL , *PROTEINS , *NEOPLASTIC cell transformation - Abstract
Abstract Hypercholesterolemia is an important risk factor for coronary heart disease. Although a lot of research has been conducted, the regulation of cholesterol metabolism is still largely unknown. Some miRNAs have been found to play critical role in the cholesterol metabolism. MiR-98 is a miRNA whose function has been reported mainly in tumorigenesis. In this study, we elucidate a novel role of miR-98 in cholesterol metabolism. We found that the expression of miR-98 was decreased significantly in hypercholesterolemic patients compared with healthy control subjects. Furthermore, we identified that SREBP-2, an important transcriptional factor in cholesterol metabolism, was a direct target of miR-98. Overexpression of miR-98 significantly repressed the 3′-UTR reporter activities of SREBP-2 in a dose-dependent manner in HepG2 cells, while the effect of miR-98 was blocked when the binding site of miR-98 within the SREBP-2 3′-UTR was mutated. And overexpression of miR-98 reduced both the mRNA and protein levels of HMGCR and LDLR significantly in vitro, which are two target genes of SREBP-2. Furthermore, MiR-98 overexpression reduced the intracellular total cholesterol levels dramatically. Moreover, we overexpressed the miR-98 by lentiviral tail vein injection in vivo. Compared with the control mice, the miR-98 overexpression mice showed lower serum cholesterol level and decreased SREBP-2, HMGCR as well as LDLR expression. Our data confirmed that reduced expression of miR-98 potentially contributes to disturbance of cholesterol metabolism. MiR-98 might be a novel therapeutic target to hypercholesterolemia. Highlights • The expression of miR-98 decreased while the expression of SREBP-2 increased in hypercholesterolemic patients. • SREBP-2 is a direct target gene of miR-98. • The effect of miR-98 on cholesterol metabolism is mediated by SREBP-2. • Overexpression of miR-98 reduced cholesterol level in vitro and in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
31. Long noncoding RNA lncARSR promotes hepatic cholesterol biosynthesis via modulating Akt/SREBP-2/HMGCR pathway.
- Author
-
Huang, Jiabin, Chen, Shangjun, Cai, Dongliang, Bian, Deqiang, and Wang, Fengling
- Subjects
- *
PHYSIOLOGICAL effects of cholesterol , *HOMEOSTASIS , *BIOSYNTHESIS , *STEROL regulatory element-binding proteins , *ATHEROSCLEROSIS , *HYPERCHOLESTEREMIA - Abstract
Aims Disruption of cholesterol homeostasis has been identified as a major factor in the pathogenesis of atherosclerosis, myocardial infarction, and strokes. Long noncoding RNAs (lncRNAs) have emerged as critical players in cellular cholesterol metabolism, but their functions are still largely unknown. Materials and methods C57BL6/j mice were fed with high cholesterol diet (containing 4% cholesterol) or chow diet. Adenoviruses-lncARSR and lncARSR shRNA were used to overexpress or knockdown lncARSR expression. Key findings The expression of lncARSR were increased both in patients with hypercholesterolemia and mice with high cholesterol diet feeding. Overexpression of lncARSR in mice resulted in elevated lipid levels in both serum and liver fragments. However, knockdown of lncARSR in mice fed with high cholesterol diet showed decreased lipid levels in serum and liver fragments compared with control mice. Furthermore, we found that the expression of HMG-CoA reductase (HMGCR), the rate-limiting enzyme of cholesterol synthesis was increased with lncARSR overexpression, which was accompanied with the increase of hepatic de novo cholesterol synthesis rate. Mechanistically, we found that lncARSR increased the expression of mature SREBP-2, which is a primary transcription factor of HMGCR. And lncARSR activated the PI3K/Akt pathway. When PI3K/Akt pathway was blocked by LY294002, the inhibitor of PI3K, the effect of lncARSR on SREBP-2 and HMGCR disappeared. Significance Our data indicated upregulated lncARSR promotes hepatic cholesterol biosynthesis via modulating Akt/SREBP-2/HMGCR pathway, and implied that lncARSR may serve as a therapeutic target for cholesterol disorder. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
32. SREBP-2 expression pattern contributes to susceptibility of Mongolian gerbils to hypercholesterolemia.
- Author
-
Li, Changlong, Guo, Honggang, Dai, Fangwei, Huo, Xueyun, Li, Zhenkun, Zhang, Shuangyue, Fu, Rui, He, Zhengming, Guo, Meng, Du, Xiaoyan, and Chen, Zhenwen
- Subjects
- *
GERBILS , *HYPERCHOLESTEREMIA , *FATTY liver , *LOW density lipoproteins , *MESSENGER RNA - Abstract
Gerbils are susceptible to dietary cholesterol and prone to hypercholesterolemia and non-alcoholic fatty liver disease. The present study aimed to explore the role of sterol regulatory element binding protein (SREBP)-2 and 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR) in hypercholesterolemia susceptibility in gerbils. Male gerbils were fed the normal diet or a high-fat diet (HFD) for 2 weeks, or the HFD for 2 weeks followed with the normal diet for an additional 2 weeks. Serum lipid levels and hepatic fat deposition were measured, and mRNA and protein levels of SREBP-2 and HMGCR were evaluated by quantitative polymerase chain reaction and Western blotting. In addition, the role of SREBP-2 function in cholesterol synthesis from the gerbil primary hepatic cells was also investigated by modulation of SERBP-2 expression via the transfection of SREBP-2 overexpression and knockdown plasmids, respectively. The data demonstrated that the total cholesterol and low-density lipoprotein cholesterol levels in the gerbil serum samples were rapidly and significantly elevated in response to HFD. In addition, the effect of the HFD was rapidly attenuated in the gerbils following a return to the normal diet. HMGCR expression and activation were not altered by dietary cholesterol consumption in the livers from the gerbils in model or recovery groups. HMGCR expression and activation were effectively regulated in cultured hepatic cells from the gerbils. These results indicated that the activation of SREBP-2 to HMGCR was not terminated in gerbil livers during cholesterol intake. Therefore, stable SREBP-2 expression contributes to the susceptibility of gerbils to hypercholesterolemia. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
33. Does Sirt2 Regulate Cholesterol Biosynthesis During Oligodendroglial Differentiation In Vitro and In Vivo?
- Author
-
Thangaraj, Merlin P., Furber, Kendra L., Sobchishin, LaRhonda, Ji, Shaoping, Doucette, J. Ronald, and Nazarali, Adil J.
- Subjects
- *
SIRTUINS , *PHYSIOLOGICAL effects of cholesterol , *NEUROGLIA , *BIOSYNTHESIS , *CELL differentiation , *OLIGODENDROGLIA , *IN vitro studies - Abstract
Sirtuin2 (SIRT2) is a deacetylase enzyme predominantly expressed in myelinating glia of the central nervous system (CNS). We have previously demonstrated that Sirt2 expression enhances oligodendrocyte (OL) differentiation and arborization in vitro, but the molecular targets of SIRT2 in OLs remain speculative. SIRT2 has been implicated in cholesterol biosynthesis by promoting the nuclear translocation of sterol regulatory element binding protein (SREBP)-2. We investigated this further in CNS myelination by examining the role of Sirt2 in cholesterol biosynthesis in vivo and in vitro employing Sirt2 mice, primary OL cells and CG4-OL cells. Our results demonstrate that expression of cholesterol biosynthetic genes in the CNS white matter or cholesterol content in purified myelin fractions did not differ between Sirt2 and age-matched wild-type mice. Cholesterol biosynthetic gene expression profiles and total cholesterol content were not altered in primary OLs from Sirt2 mice and in CG4-OLs when Sirt2 was either down-regulated with RNAi or overexpressed. In addition, Sirt2 knockdown or overexpression in CG4-OLs had no effect on SREBP-2 nuclear translocation. Our results indicate that Sirt2 does not impact the expression of genes encoding enzymes involved in cholesterol biosynthesis, total cholesterol content, or nuclear translocation of SREBP-2 during OL differentiation and myelination. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
34. Regulation of genes related to cholesterol metabolism in rainbow trout (Oncorhynchus mykiss) fed a plant-based diet.
- Author
-
Tengfei Zhu, Corraze, Geneviève, Plagnes-Juan, Elisabeth, Quillet, Edwige, Dupont-Nivet, Mathilde, and Skiba-Cassy, Sandrine
- Subjects
- *
CHOLESTEROL metabolism , *GENETIC regulation , *RAINBOW trout - Abstract
When compared with fish meal and fish oil, plant ingredients differ not only in their protein content and amino acid and fatty acid profiles but are also devoid of cholesterol, the major component of cell membrane and precursor of several bioactive compounds. Based on these nutritional characteristics, plant-based diets can affect fish physiology and cholesterol metabolism. To investigate the mechanisms underlying cholesterol homeostasis, rainbow trout were fed from 1 g body wt for 6 mo with a totally plant-based diet (V), a marine diet (M), and a marine-restricted diet (MR), with feed intake adjusted to that of the V group. The expression of genes involved in cholesterol synthesis, esterification, excretion, bile acid synthesis, and cholesterol efflux was measured in liver. Results showed that genes involved in cholesterol synthesis were upregulated in trout fed the V diet, whereas expression of genes related to bile acid synthesis (cyp7a1) and cholesterol elimination (abcg8) were reduced. Feeding trout the V diet also enhanced the expression of srebp-2 while reducing that of lxrα and miR-223. Overall, these data suggested that rainbow trout coped with the altered nutritional characteristics and absence of dietary cholesterol supply by increasing cholesterol synthesis and limiting cholesterol efflux through molecular mechanisms involving at least srebp-2, lxrα, and miR-223. However, plasma and body cholesterol levels in trout fed the V diet were lower than in fish fed the M diet, raising the question of the role of cholesterol in the negative effect of plant-based diet on growt [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
35. Effect of dietary eicosapentaenoic and docosahexaenoic acid on expression of rat liver genes controlling cholesterol homeostasis and on plasma cholesterol level
- Author
-
T. Komprda, G. Zorníková, A. Knoll, Z. Vykoukalová, V. Rozíková, O. Škultéty, and R. Krobot
- Subjects
pparα ,srebp-2 ,insig-1 ,cholesterol ,pufan-3 ,rats ,Animal culture ,SF1-1100 - Abstract
A hypothesis that eicosapentaenoic acid + docosahexaenoic acid (EPA+DHA) lower plasma cholesterol via increased expression of the Insig-1 gene with ensuing decrease of expression of genes coding for 3-hydroxy-3-methyl-glutaryl-CoA reductase (Hmgcr) and low density lipoprotein receptor (Ldlr) was tested in rats fed a diet with 3% of fish oil (FO). Expression of the Insig-1 gene in the liver of the FO-fed rats was 730% (P < 0.05) of the control. However, contrary to the hypothesis, expression of the Hmgcr gene and Ldlr gene was 165% and 210% of the control (P > 0.05). Nevertheless, FO in the diet decreased (P < 0.05) plasma cholesterol of rats by 10% (from 1.19 to 1.07 mmol/l); it was therefore concluded that the cholesterol-lowering effect of EPA+DHA is at least partly based on mechanisms other than tested in the present experiment.
- Published
- 2014
- Full Text
- View/download PDF
36. Curcumin inhibits the proteolytic process of SREBP‐2 by first inhibiting the expression of S1P rather than directly inhibiting SREBP‐2 expression
- Author
-
Yongnan Li and Shuodong Wu
- Subjects
0301 basic medicine ,medicine.medical_treatment ,NPC1L1 ,lcsh:TX341-641 ,S1P ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Downregulation and upregulation ,Transcription (biology) ,medicine ,curcumin ,SREBP‐2 ,Transcription factor ,Gene ,Original Research ,Messenger RNA ,cholesterol gallstones ,Protease ,Chemistry ,food and beverages ,Cell biology ,Sterol regulatory element-binding protein ,030104 developmental biology ,030220 oncology & carcinogenesis ,Curcumin ,lipids (amino acids, peptides, and proteins) ,lcsh:Nutrition. Foods and food supply ,Food Science - Abstract
Many studies have demonstrated that curcumin can downregulate mRNA levels of sterol regulatory element‐binding proteins (SREBP‐2); however, our study did not find similar results. This study was designed to demonstrate that curcumin inhibits the proteolytic process of SREBP‐2 by first inhibiting the expression of membrane‐bound transcription factor site‐1 protease (S1P) rather than directly inhibiting SREBP‐2 expression. After curcumin treatment, Caco‐2 cells were collected to observe the dose‐ and time‐dependent dynamics of precursor and mature SREBP‐2, transcription factor‐specific protein 1 (SP‐1), and SREBP cleavage‐activating protein (SCAP). After curcumin treatment, SREBP‐2 distribution was detected in the cells and S1P protein expression was examined. Curcumin could downregulate mRNA levels of SREBP2, SP‐1 and SCAP, but it did not simultaneously downregulate the expression of precursor SREBP‐2 (pSREBP‐2) and SCAP. Curcumin can inhibit the proteolytic process of SREBP‐2, reduce the production of mature SREBP‐2 (mSREBP‐2), and change the cellular distribution of SREBP‐2. The inhibitory effect of curcumin on SP‐1 protein expression is short‐acting. Curcumin could downregulate the mRNA and protein expression of S1P, but has no obvious inhibitory effect on the mRNA and protein expression of S2P (site‐2 protease). Curcumin can inhibit the SREBP‐2 proteolytic process to reduce mSREBP‐2 which functions as a transcription factor, affecting the regulation of cholesterol metabolism‐related genes. Curcumin does not directly inhibit the expression of mSREBP‐2 protein, and it has no such inhibitory effect for at least a short period of time, although curcumin does reduce the amount of mSREBP‐2 protein. S1P is a key protease in the hydrolysis of pSREBP‐2 into mSREBP‐2. Therefore, curcumin may decrease the amount of mSREBP‐2 by directly inhibiting the expression of S1P mRNA and protein., This study found for the first time that although curcumin could indeed downregulate the expression of SREBP‐2 mRNA, it did not down‐regulate the expression of SREBP‐2 protein. This discovery is different from all previous studies on the relationship between curcumin and SREBP‐2, which is a new discovery. In addition, this study also found that curcumin inhibited the process of SREBP‐2 proteolysis, possibly by inhibiting the expression of S1P, which is also a new discovery and has not been reported.
- Published
- 2021
37. Neoagarooligosaccharides enhance the level and efficiency of LDL receptor and improve cholesterol homeostasis.
- Author
-
Yang, Ji Hye, Cho, Sam Seok, Kim, Kyu Min, Kim, Ji Young, Kim, Eun Joo, Park, Eun Young, Lee, Je Hyeon, and Ki, Sung Hwan
- Abstract
Neoagarooligosaccharides (NAOS) are produced by β-agarases through the cleavage of β-1,4 linkage in agarose. Here, we investigated the cholesterol-lowering properties of NAOS and its underlying molecular mechanism. Administration of NAOS in mice fed a high-cholesterol diet (HCD) inhibited hepatic fat accumulation and serum LDL cholesterol. Mice fed an HCD showed a decrease in LDL receptor (LDLR) expression, which was reversed by NAOS. Moreover, NAOS enhanced LDLR expression and LDL uptake in hepatocytes. NAOS increased nuclear translocation of SREBP-2, a crucial transcription factor for LDLR gene expression. Increased LDLR luciferase activity, induced by NAOS, was abolished on deletion of SRE in the LDLR promoter. NAOS upregulated LDLR levels via PI3K/Akt-mediated activation of SREBP-2. Furthermore, we demonstrated that NAOS reduced the nuclear HNF-1α, leading to decreased levels of PCSK9, a natural inhibitor of LDLR. These findings indicate that NAOS exhibits hypolipidemic activity, and may serve as a new hypolipidemic drug. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
38. Cholesterol-lowering effect of Aralia elata (Miq.) Seem via the activation of SREBP-2 and the LDL receptor.
- Author
-
Hwang, Kyung-A., Hwang, Yu-Jin, and Song, Jin
- Subjects
ANTICHOLESTEREMIC agents ,HYPERLIPIDEMIA ,CORONARY disease ,LOW density lipoproteins ,HIGH-fat diet ,ACETYLTRANSFERASES ,CHOLESTEROL metabolism - Abstract
Background Hyperlipidemia causes arteriosclerosis, a risk factor for coronary heart disease. Prevention of hyperlipidemia by improving dietary habits has recently attracted attention. In this regard, we investigated whether Aralia elata (Miq.) Seem (AE) extract inhibits hepatic cholesterol accumulation and modulate the cellular signaling pathway. Methods To determine AE's cholesterol regulating mechanism, we measured cholesterol level, 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase activity and cholesterol regulating-related gene expression in HepG2 cells and in high-fat diet (HFD)-induced mice using ELISA and RT-PCR assay. Results The AE extract reduced cholesterol levels and HMG-CoA reductase activity in hepatocellular carcinoma HepG2 cells. In addition, it also reduced the plasma cholesterol concentrations in HFD-induced mice. Furthermore, the AE extract increased the gene expression of the LDL-receptor (LDL-R); sterol-regulatory-element binding protein-2 (SREBP-2); ATP-binding cassette, sub-family A, member 1 (ABCA1); and scavenger receptor class B member 1 (SR-B1) in a dose-dependent manner. However, the AE extract did not affect the gene expression of acetyl-coenzyme A acetyltransferase (ACAT) in either the HepG2 cells or mice. Conclusion We demonstrated that the AE extract activated genes related to cholesterol metabolism, such as SREBP-2 and LDL-R, which resulted in hypocholesterolemic activities. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
39. mTORC1 activates SREBP-2 by suppressing cholesterol trafficking to lysosomes in mammalian cells.
- Author
-
Eid, Walaa, Dauner, Kristin, Courtney, Kevin C., Gagnon, AnneMarie, Parks, Robin J., Sorisky, Alexander, and Xiaohui Zha
- Subjects
- *
STEROL regulatory element-binding proteins , *CHOLESTEROL , *LYSOSOMES , *MTOR protein , *SMALL interfering RNA - Abstract
mTORC1 is known to activate sterol regulatory element-binding proteins (SREBPs) including SREBP-2, a master regulator of cholesterol synthesis. Through incompletely understood mechanisms, activated mTORC1 triggers translocation of SREBP-2, an endoplasmic reticulum (ER) resident protein, to the Golgi where SREBP-2 is cleaved to translocate to the nucleus and activate gene expression for cholesterol synthesis. Low ER cholesterol is a well-established trigger for SREBP-2 activation. We thus investigated whether mTORC1 activates SREBP-2 by reducing cholesterol delivery to the ER. We report here that mTORC1 activation is accompanied by low ER cholesterol and an increase of SREBP-2 activation. Conversely, a decrease in mTORC1 activity coincides with a rise in ER cholesterol and a decrease in SERBP-2 activity. This rise in ER cholesterol is of lysosomal origin: blocking the exit of cholesterol from lysosomes by U18666A or NPC1 siRNA prevents ER cholesterol from increasing and, consequently, SREBP-2 is activated without mTORC1 activation. Furthermore, when mTORC1 activity is low, cholesterol is delivered to lysosomes through two membrane trafficking pathways: autophagy and rerouting of endosomes to lysosomes. Indeed, with dual blockade of both pathways by Atg5-/- and dominant-negative rab5, ER cholesterol fails to increase when mTORC1 activity is low, and SREBP-2 is activated. Conversely, overexpressing constitutively active Atg7, which forces autophagy and raises ER cholesterol even when mTORC1 activity is high, suppresses SREBP-2 activation. We conclude that mTORC1 actively suppresses autophagy and maintains endosomal recycling, thereby preventing endosomes and autophagosomes from reaching lysosomes. This results in a reduction of cholesterol in the ER and activation of SREBP-2. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
40. Hexacosanol reduces plasma and hepatic cholesterol by activation of AMP-activated protein kinase and suppression of sterol regulatory element-binding protein-2 in HepG2 and C57BL/6J mice.
- Author
-
Lee, Ji Hae, Jia, Yaoyao, Thach, Trung Thanh, Han, Yuri, Kim, Bobae, Wu, Chunyan, Kim, Yeonji, Seo, Woo Duck, and Lee, Sung-Joon
- Abstract
Policosanols have hypocholesterolemic activity; however, the molecular mechanism of the policosanol effects is currently poorly characterized. We hypothesized that hexacosanol, a policosanol compound derived from barley sprout, may decrease cellular and plasma cholesterol levels; we thus investigated the hypocholesterolemic activity and mechanism of hexacosanol on both hepatocytes and high-fat–induced obese C57BL/6J mice. The reduction of total cholesterol, free cholesterol, and cholesteryl ester concentrations was confirmed in hexacosanol-stimulated hepatocytes (−38%, −33%, and −53%, respectively). Plasma, hepatic cholesterol concentrations, and hepatic steatosis were significantly reduced in high-fat–fed mice orally administered with hexacosanol (0.7 mg/kg body weight a day) for 8 weeks compared with those of vehicle-fed control mice (−15% and −40%, respectively). Hexacosanol in fact bound to the allosteric regulation site of AMP-activated protein kinase (AMPK)- β subunit and thus activated AMPK that inhibited the activity of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase by inhibitory phosphorylation. In addition, activation of AMPK by hexacosanol induced hepatic autophagy activity, which could further reduce hepatic lipid accumulation. Alternatively, hexacosanol suppressed the nuclear translocation and activation of sterol regulatory element-binding protein-2 (SREBP-2), a key transcription factor in cholesterol biosynthesis. These results collectively suggest that hexacosanol is a major hypocholesterolemic compound in barley sprouts with regulation of AMPK activation and SREBP-2 suppression. These suppress 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase at both mRNA expression and protein activity levels. In conclusion, hexacosanol activates AMPK and hepatic autophagy and inhibits SREBP2, resulting in hypocholesterolemic activities and improvement of hepatic steatosis. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
41. New insights into cellular cholesterol acquisition: promoter analysis of human HMGCR and SQLE, two key control enzymes in cholesterol synthesis.
- Author
-
Howe, Vicky, Sharpe, Laura J., Prabhu, Anika V., and Brown, Andrew J.
- Subjects
- *
CHOLESTEROL , *COENZYME A , *TRANSCRIPTION factors , *LUCIFERASES , *ELECTROPHORETIC displays - Abstract
Background The two control points of cholesterol synthesis, 3-hydroxy-3-methylglutaryl-coenzyme A reductase ( HMGCR ) and squalene monooxygenase ( SQLE ) are known targets of the transcription factor sterol-regulatory element binding protein-2 (SREBP-2). Yet the location of the sterol-regulatory elements (SREs) and cofactor binding sites, nuclear factor-Y (NF-Y) and specificity protein 1 (Sp1), have not been satisfactorily mapped in the human SQLE promoter, or at all in the human HMGCR promoter. Methods We used luciferase reporter assays to screen the sterol-responsiveness of a library of predicted SRE, Sp1 and NF-Y site mutants and hence identify bone fide binding sites. We confirmed SREs via an electrophoretic mobility shift assay (EMSA) and ChIP-PCR. Results We identified two SREs in close proximity in both the human HMGCR and SQLE promoters, as well as one NF-Y site in HMGCR and two in SQLE . In addition, we found that HMGCR expression is highly activated only when SREBP-2 levels are very high, in contrast to the low density lipoprotein receptor ( LDLR ), a result reflected in mouse models used in other studies. Conclusions Both HMGCR and SQLE promoters have two SREs that may act as a homing region to attract a single SREBP-2 homodimer, with HMGCR being activated only when there is absolute need for cholesterol synthesis. This ensures preferential uptake of exogenous cholesterol via LDLR, thereby conserving energy. General Significance We provide the first comprehensive investigation of SREs and NF-Ys in the human HMGCR and SQLE promoters, increasing our fundamental understanding of the transcriptional regulation of cholesterol synthesis. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
42. Circulating PCSK9 affects serum LDL and cholesterol levels more than SREBP-2 expression.
- Author
-
Mohammadi, Asghar, Shabani, Mohamad, Naseri, Faezeh, Hosseni, Bita, Soltanmohammadi, Elham, Piran, Sadegh, and Najafi, Mohammad
- Subjects
PHYSIOLOGICAL effects of cholesterol ,HOMEOSTASIS ,STEROL regulatory element-binding proteins ,LOW density lipoproteins ,PROPROTEIN convertases ,GENE expression - Abstract
Background. Cholesterol homeostasis is dependent upon the sterol regulatory element binding protein 2 (SREBP-2) regulatory system and the functioning of plasma proprotein convertase subtilisin/kexin type 9 (PCSK9). Many studies have also reported that low density lipoprotein receptor (LDLR) levels in cellular membranes are related to the functioning of these proteins. Objectives. The aim of this study was to investigate the association of lipid profiles with circulating PCSK9 protein values and SREBP-2 expression levels in normal subjects. Material and methods. The study involved 120 randomly chosen healthy subjects. Their lipid profiles were measured using routine laboratory techniques, and the plasma PCSK9 protein and SREBP-2 expression levels were determined by ELISA and real time quantitative PCR methods, respectively. A statistical analysis was carried out using a statistical software package. Results. Linear regression analyses showed a significant correlation between total cholesterol and PCSK9 (3.54 ° 1.31 ng/mL), as well as between total cholesterol and SREBP-2 (0.1-35.38) (p = 0.002 and p = 0.02, respectively). Furthermore, multiple regression analyses showed strict correlations between PCSK9 and cholesterol-related parameters especially the total cholesterol/HDL-C ratio (β = 3.53, p = 0.001). There was no significant correlation between circulating PCSK9 and SREBP-2 expression levels (r = 1.2, p = 0.3). Conclusions. The study results revealed that serum cholesterol-related parameters are strictly associated with plasma PCSK9 values, suggesting that PCSK9 function has a greater effect on serum total cholesterol levels than SREBP-2 expression does. Furthermore, the total cholesterol/HDL-C ratio was a better indicator for evaluating PCSK9 level than total cholesterol. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
43. The moderate essential amino acid restriction entailed by low-protein vegan diets may promote vascular health by stimulating FGF21 secretion.
- Author
-
McCarty, Mark F.
- Subjects
- *
ADIPONECTIN , *LOW density lipoproteins , *CHOLESTEROL , *VEGANISM , *FIBROBLAST growth factors , *LOW-protein diet - Abstract
The serum total and LDL cholesterol levels of long-term vegans tend to be very low. The characteristically low ratio of saturated to unsaturated fat in vegan diets, and the absence of cholesterol in such diets, clearly contribute to this effect. But there is reason to suspect that the quantity and composition of dietary protein also play a role in this regard. Vegan diets of moderate protein intake tend to be relatively low in certain essential amino acids, and as a result may increase hepatic activity of the kinase GCN2, which functions as a gauge of amino acid status. GCN2 activation boosts the liver's production of fibroblast growth factor 21 (FGF21), a factor which favorably affects serum lipids and metabolic syndrome. The ability of FGF21 to decrease LDL cholesterol has now been traced to at least two mechanisms: a suppression of hepatocyte expression of sterol response element-binding protein-2 (SREBP-2), which in turn leads to a reduction in cholesterol synthesis; and up-regulated expression of hepatocyte LDL receptors, reflecting inhibition of a mechanism that promotes proteasomal degradation of these receptors. In mice, the vascular benefits of FGF21 are also mediated by favorable effects on adipocyte function - most notably, increased adipocyte secretion of adiponectin, which directly exerts anti-inflammatory effects on the vasculature which complement the concurrent reduction in LDL particles in preventing or reversing atherosclerosis. If, as has been proposed, plant proteins preferentially stimulate glucagon secretion owing to their amino acid composition, this would represent an additional mechanism whereby plant protein promotes FGF21 activity, as glucagon acts on the liver to boost transcription of the FGF21 gene. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
44. Dietary flavones counteract phorbol 12-myristate 13-acetate-induced SREBP-2 processing in hepatic cells.
- Author
-
Tan, Yan, Wong, Tsz, Lin, Shu-Mei, and Leung, Lai
- Abstract
Consumption of fruits and vegetables is generally regarded as beneficial to plasma lipid profile. The mechanism by which the plant foods induce desirable lipid changes remains unclear. SREBP-2 is crucial in cholesterol metabolism, and it is a major regulator of the cholesterol biosynthesis enzyme HMGCR. Our lab has previously illustrated that apigenin and luteolin could attenuate the nuclear translocation of SREBP-2 through an AMPK-dependent pathway. In the present study, these two flavones were studied for their ability to deter the same in an AMPK-independent signaling route. The processing of SREBP-2 protein was promoted by phorbol 12-myristate 13-acetate (PMA) in the hepatic cells WRL and HepG2, and the increased processing was reversed by apigenin or luteolin co-administration. EMSA results demonstrated that the PMA-induced DNA-binding activity was weakened by the flavones. The increased amount of nuclear SREBP-2 in cells was attenuated by the flavonoid as shown by immunocytochemical imaging. Quantitative reverse transcriptase-polymerase chain reaction assay demonstrated that the transcription of HMGCR under both flavone treatments was reduced. However, apigenin appeared to be stronger than luteolin in restraining PMA-induced HMGCR mRNA expression. Since PMA is a diacylglycerol analog, these findings might have some physiological implications. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
45. Ginsenoside Rg3 Reduces Lipid Accumulation with AMP-Activated Protein Kinase (AMPK) Activation in HepG2 Cells
- Author
-
Yangha Kim, In-Hwan Kim, Mak-Soon Lee, Chong-Tai Kim, and Seohyun Lee
- Subjects
Cardiovascular disease (CVD) ,ginsenoside Rg3 ,cholesterol ,triglyceride ,SREBP-2 ,HMGCR ,AMPK ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Cardiovascular disease (CVD) is one of the main causes of mortality worldwide, and dyslipidemia is a major risk factor for CVD. Ginseng has been widely used in the clinic to treat CVD. Ginsenoside Rg3, one of the major active components of ginseng, has been reported to exhibit antiobesity, antidiabetic, and cardioprotective effects. However, the effect of ginsenoside Rg3 on hepatic lipid metabolism remains unclear. Therefore, we investigated whether ginsenoside Rg3 would regulate hepatic lipid metabolism with AMP-activated protein kinase (AMPK) activation in HepG2 cells. Ginsenoside Rg3 significantly reduced hepatic cholesterol and triglyceride levels. Furthermore, ginsenoside Rg3 inhibited expression of sterol regulatory element binding protein-2 (SREBP-2) and 3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMGCR). Ginsenoside Rg3 increased activity of AMPK, a major regulator of energy metabolism. These results suggest that ginsenoside Rg3 reduces hepatic lipid accumulation with inhibition of SREBP-2 and HMGCR expression and stimulation of AMPK activity in HepG2 cells. Therefore, ginsenoside Rg3 may be beneficial as a food ingredient to lower the risk of CVD by regulating dyslipidemia.
- Published
- 2012
- Full Text
- View/download PDF
46. Possible involvement of PCSK9 overproduction in hyperlipoproteinemia associated with hepatocellular carcinoma: A case report.
- Author
-
Nagashima, Shuichi, Morishima, Kazue, Okamoto, Hiroaki, and Ishibashi, Shun
- Subjects
CELL receptors ,GENE expression ,HEPATITIS viruses ,HEPATOCELLULAR carcinoma ,HYPERLIPOPROTEINEMIA ,PROTEOLYTIC enzymes ,DISEASE complications ,SYMPTOMS ,GENETICS - Abstract
Herein, we describe a 69-year-old Japanese man with massive type III hyperlipoproteinemia (total cholesterol, 855 mg/dL; triglyceride, 753 mg/dL) presenting as a paraneoplastic manifestation of hepatitis B virus–associated hepatocellular carcinoma. The messenger RNA expression of sterol regulatory element–binding protein-2 and proprotein convertase subtilisin/kexin 9 in the tumor tissue was increased by 13-fold and 4-fold, respectively, compared with the non-tumor tissue. Serum level of active form of PCSK9 was 382 ng/mL (reference range: 253 ± 79 ng/mL). The non-tumor tissue had extremely low expression of low-density lipoprotein receptor and low-density lipoprotein receptor–related protein 1. Together, we speculate that marked overexpression of sterol regulatory element–binding protein-2 in the tumor may stimulate the secretion of PCSK9, which inhibits the lipoprotein receptors in the non-tumor tissue, thereby causing paraneoplastic hyperlipoproteinemia. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
47. Phorbol 12-myristate 13-acetate promotes nuclear translocation of hepatic steroid response element binding protein-2.
- Author
-
Wong, Tsz Yan, Tan, Yan Qin, Lin, Shu-mei, and Leung, Lai K.
- Subjects
- *
PHORBOLS , *STEROL regulatory element-binding proteins , *CHROMOSOMAL translocation , *CHOLESTEROL metabolism , *PROTEIN kinase C , *LIVER cells , *POST-translational modification - Abstract
Sterol regulatory element-binding protein (SREBP)-2 is a pivotal transcriptional factor in cholesterol metabolism. Factors interfering with the proper functioning of SREBP-2 potentially alter plasma lipid profiles. Phorbol 12-myristate 13-acetate (PMA), which is a common protein kinase C (PKC) activator, was shown to promote the post-translational processing and nuclear translocation of SREBP-2 in hepatic cells in the current study. Following SREBP-2 translocation, the transcripts of its target genes HMGCR and LDLR were upregulated as demonstrated by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) assay. Electrophoretic mobility shift assays (EMSA) also demonstrated an induced DNA-binding activity on the sterol response element (SRE) domain under PMA treatment. The increase of activated Srebp-2 without the concurrent induced mRNA expression was also observed in an animal model. As the expression of SREBP-2 was not increased by PMA, the activation of PKC was the focus of investigation. Specific PKC isozyme inhibition and overexpression supported that PKCβ was responsible for the promoting effect. Further studies showed that the mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinases (ERK) and c-Jun N-terminal kinases (JNK), but not 5′ adenosine monophosphate-activated protein kinase (AMPK), were the possible downstream signaling proteins of PKCβ. In conclusion, this study illustrated that PKCβ increased SREBP-2 nuclear translocation in a pathway mediated by MEK/ERK and JNK, rather than the one dictated by AMPK. These results revealed a novel signaling target of PKCβ in the liver cells. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
48. Tanshinone IIA affects the HDL subfractions distribution not serum lipid levels: Involving in intake and efflux of cholesterol.
- Author
-
Jia, Lian-qun, Zhang, Ni, Xu, Ying, Chen, Wen-na, Zhu, Mei-lin, Song, Nan, Ren, Lu, Cao, Hui-min, Wang, Jun-yan, and Yang, Guan-lin
- Subjects
- *
HIGH density lipoproteins , *BLOOD serum analysis , *CHOLESTEROL , *LIPID analysis , *CHINESE medicine , *HYPERLIPIDEMIA - Abstract
Aim of study Tanshinone IIA is an active component of the traditional Chinese medicine. This study aimed at investigating the mechanism of tanshinone IIA on anti-atherosclerosis, which may be because of that Tanshinone IIA can affect the HDL subfractions distribution and then regulate reverse cholesterol transport. Materials and methods A model of hyperlipidaemia in rats was used. Tanshinone IIA was given daily after hyperlipidaemia. In vivo, lipid deposition and morphological changes in liver were analyzed; HDL subfractions and lipid level in serum as well as in liver were measured; the expression of genes related to cholesterol intake in liver and peritoneal macrophage cholesterol efflux were evaluated. In vitro, HepG2 cells and THP-1 cells were pretreated with tanshinone IIA and subsequently with ox-LDL to evaluate the total cholesterol and the expression of related genes. Results Tanshinone IIA reduced the lipid deposition in liver. Moreover, it did not affect the serum lipid levels but reduced the levels of HDL middle subfractions and increased the levels of HDL large subfractions. Furthermore, tanshinone IIA could regulate the expressions of CYP7A1, LDL-R, SREBP2 and LCAT in the liver as well as the ABCA1 and CD36 in macrophage cells which is involving in the cholesterol intake and efflux respectively. It could reduce lipid accumulation caused by ox-LDL induction, and that also regulate the expressions of LDL-R, HMGCR and SREBP2 in HepG2 and ABCA1, CD36 in THP-1 cells. Conclusion A novel finding that tanshinone IIA was not reduce the serum lipid level but affects the HDL subfractions distribution and thereby regulating the intake and efflux of cholesterol. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
49. Genistein upregulates LDLR levels via JNK-mediated activation of SREBP-2.
- Author
-
Kartawijaya, Medicia, Han, Hye Won, Kim, Yunhye, and Lee, Seung-Min
- Subjects
- *
ATHEROSCLEROSIS prevention , *HYPERCHOLESTEREMIA prevention , *GENISTEIN , *CELL culture , *GENE expression , *WESTERN immunoblotting , *THERAPEUTICS - Abstract
Background: Genistein has been proved in vitro and in vivo to lower LDLR level. It is also widely consumed and implicated for its anti-atherogenic effects. However, the molecular mechanism by which genistein lowers the LDL level is still unknown. Objective: To understand the anti-atherogenic molecular mechanism of action, genistein was investigated for its impact on the expression of LDLR, the receptor for LDL cholesterol, and related signaling pathways in a human hepatoma cell line. Design: HepG2 cell was used for the experiments. Genistein with different concentrations was diluted in media and was incubated for 24 h or more as indicated. Protein levels were measured by western blotting, and mRNA expression was detected by RT-qPCR. Chromatin immunoprecipitation assay (CHIP) assay was used to determine protein binding levels, and luciferase assay was used to measure promoter activity. Result: Genistein increased the mRNA and protein levels of LDLR in a time-dependent manner. Genistein increased the transcriptional activity of the LDLR promoter containing the reporter gene (pLDLR-luc, -805 to +50). But the sterol regulatory element deletion mutant construct failed to be activated by genistein. Genistein increased the nuclear fraction of SREBP-2 and the DNA-binding activity of SREBP-2 to LDLR promoter, as assessed by CHIP. The genistein-phosphorylated JNK inhibitor (SP600126) abolished the genistein-stimulated levels of LDLR and the nuclear SREBP-2. The addition of cholesterol up to 5 µg/mL for 24 h did not affect the effect of genistein on LDLR protein expression. Even the addition of 40 mM genistein increased the cholesterol uptake by more than 10% in the human hepatoma cell line. Conclusion: Our data support the idea that genistein may have anti-atherogenic effects by activating JNK signals and SREBP-2 processing, which is followed by the upregulation of LDLR. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
50. Cholesterol metabolism is a potential therapeutic target in Duchenne muscular dystrophy
- Author
-
Guillaume Corre, Laurence Suel, David Israeli, Ai Vu Hong, Laurent Servais, Thomas Voit, Isabelle Richard, Fatima Amor, Mathilde Sanson, Stephanie Blaie, Généthon, Approches génétiques intégrées et nouvelles thérapies pour les maladies rares (INTEGRARE), Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Généthon, University of Oxford [Oxford], University College of London [London] (UCL), University of Oxford, and Richard, Isabelle
- Subjects
0301 basic medicine ,Male ,mdx mouse ,Simvastatin ,Duchenne muscular dystrophy ,Diseases of the musculoskeletal system ,Extracellular matrix ,Mice ,0302 clinical medicine ,Glucocorticoid ,Myocyte ,Orthopedics and Sports Medicine ,DLK1‐DIO3 ,[SDV.MHEP.EM] Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,0303 health sciences ,Host gene ,[SDV.MHEP.EM]Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,3. Good health ,Cell biology ,Cholesterol ,030220 oncology & carcinogenesis ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Original Article ,Mevalonate pathway ,musculoskeletal diseases ,SREBP-2 ,congenital, hereditary, and neonatal diseases and abnormalities ,Biology ,Biological interpretation of miRNA dysregulation ,03 medical and health sciences ,Physiology (medical) ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,medicine ,Animals ,Humans ,SREBP‐1 ,SREBP‐2 ,DLK1-DIO3 ,Muscle, Skeletal ,030304 developmental biology ,SREBP-1 ,Sarcolemma ,business.industry ,QM1-695 ,Lipid metabolism ,Original Articles ,medicine.disease ,Actin cytoskeleton ,Sterol regulatory element-binding protein ,Muscular Dystrophy, Duchenne ,030104 developmental biology ,RC925-935 ,Human anatomy ,Mice, Inbred mdx ,business ,Homeostasis ,030217 neurology & neurosurgery - Abstract
BackgroundDuchenne Muscular Dystrophy (DMD) is a lethal muscle disease detected in approximately 1:5000 male births. DMD is caused by mutations in the DMD gene, encoding a critical protein that link the cytoskeleton and the extracellular matrix in skeletal and cardiac muscles. The primary consequence of the disrupted link between the extracellular matrix and the myofiber actin cytoskeleton is thought to involve sarcolemma destabilization, perturbation of Ca+2 homeostasis, activation of proteases, mitochondrial damage and tissue degeneration. A recently emphasized secondary aspect of the dystrophic process is a progressive metabolic change of the dystrophic tissue; however, the mechanism and nature of the metabolic dysregulation is yet poorly understood. In this study, we characterized a molecular mechanism of metabolic perturbation in DMD.MethodsWe sequenced plasma miRNA in a DMD cohort, comprising of 54 DMD patients treated or not by glucocorticoid, compared to 27 healthy controls, in three age groups. We developed an original approach for the biological interpretation of miRNA dysregulation, and produced a novel hypothesis concerning metabolic perturbation in DMD. We then used the mdx mouse model for DMD for the investigation of this hypothesis.ResultsWe identified 96 dysregulated miRNAs, of which 74 were up- and 22 down-regulated in DMD. We confirmed the dysregulation in DMD of Dystro-miRs, Cardio-miRs and a large number of the DLK1-DIO3 miRNAs. We also identified numerous dysregulated miRNAs, yet unreported in DMD. Bioinformatics analysis of both target and host genes for dysregulated miRNAs predicted that lipid metabolism might be a critical metabolic perturbation in DMD. Investigation of skeletal muscles of the mdx mouse uncovered dysregulation of transcription factors of cholesterol and fatty acid metabolism (SREBP1 and SREBP2), perturbation of the mevalonate pathway, and accumulation of cholesterol. Elevated cholesterol level was also found in muscle biopsies of DMD patients. Treatment of mdx mice with Simvastatin, a cholesterol-reducing agent, normalized these perturbations and partially restored the dystrophic parameters.ConclusionThis investigation supports that cholesterol metabolism and the mevalonate pathway are potential therapeutic targets in DMD.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.