Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 408
Filter
1.
Biomolecules ; 14(7)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39062491

ABSTRACT

The sterol regulatory element-binding protein (SREBP) pathway is an integral cellular mechanism that regulates lipid homeostasis, in which transcriptional activator SREBPs regulate the expression of various genes. In the carotenogenic yeast Xanthophyllomyces dendrorhous, Sre1 (the yeast SREBP homolog) regulates lipid biosynthesis and carotenogenesis, among other processes. Despite the characterization of several components of the SREBP pathway across various eukaryotes, the specific elements of this pathway in X. dendrorhous remain largely unknown. This study aimed to explore the potential regulatory mechanisms of the SREBP pathway in X. dendrorhous using the strain CBS.cyp61- as a model, which is known to have Sre1 in its active state under standard culture conditions, resulting in a carotenoid-overproducing phenotype. This strain was subjected to random mutagenesis with N-methyl-N'-nitro-N-nitrosoguanidine (NTG), followed by a screening methodology that focused on identifying mutants with altered Sre1 activation phenotypes. Single-nucleotide polymorphism (SNP) analysis of 20 selected mutants detected 5439 single-nucleotide variants (SNVs), narrowing them down to 1327 SNPs of interest after a series of filters. Classification based on SNP impact identified 116 candidate genes, including 49 genes with high impact and 68 genes with deleterious moderate-impact mutations. BLAST, InterProScan, and gene ontology enrichment analyses highlighted 25 genes as potential participants in regulating Sre1 in X. dendrorhous. The key findings of this study include the identification of genes potentially encoding proteins involved in protein import/export to the nucleus, sterol biosynthesis, the ubiquitin-proteasome system, protein regulatory activities such as deacetylases, a subset of kinases and proteases, as well as transcription factors that could be influential in SREBP regulation. These findings are expected to significantly contribute to the current understanding of the intricate regulation of the transcription factor Sre1 in X. dendrorhous, providing valuable groundwork for future research and potential biotechnological applications.


Subject(s)
Basidiomycota , Sterol Regulatory Element Binding Proteins , Basidiomycota/genetics , Basidiomycota/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Polymorphism, Single Nucleotide , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Carotenoids/metabolism , Mutation
2.
Mol Metab ; 85: 101964, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823776

ABSTRACT

OBJECTIVE: Cancer cells must maintain lipid supplies for their proliferation and do so by upregulating lipogenic gene programs. The sterol regulatory element-binding proteins (SREBPs) act as modulators of lipid homeostasis by acting as transcriptional activators of genes required for fatty acid and cholesterol synthesis and uptake. SREBPs have been recognized as chemotherapeutic targets in multiple cancers, however it is not well understood which SREBP target genes are essential for tumorigenesis. In this study, we examined the requirement of SREBP target genes for pancreatic ductal adenocarcinoma (PDAC) tumor growth. METHODS: Here we constructed a custom CRISPR knockout library containing known SREBP target genes and performed in vitro 2D culture and in vivo orthotopic xenograft CRISPR screens using a patient-derived PDAC cell line. In vitro, we grew cells in medium supplemented with 10% fetal bovine serum (FBS) or 10% lipoprotein-deficient serum (LPDS) to examine differences in gene essentiality in different lipid environments. In vivo, we injected cells into the pancreata of nude mice and collected tumors after 4 weeks. RESULTS: We identified terpenoid backbone biosynthesis genes as essential for PDAC tumor development. Specifically, we identified the non-sterol isoprenoid product of the mevalonate pathway, geranylgeranyl diphosphate (GGPP), as an essential lipid for tumor growth. Mechanistically, we observed that restricting mevalonate pathway activity using statins and SREBP inhibitors synergistically induced apoptosis and caused disruptions in small G protein prenylation that have pleiotropic effects on cellular signaling pathways. Finally, we demonstrated that geranylgeranyl diphosphate synthase 1 (GGPS1) knockdown significantly reduces tumor burden in an orthotopic xenograft mouse model. CONCLUSIONS: These findings indicate that PDAC tumors selectively require GGPP over other lipids such as cholesterol and fatty acids and that this is a targetable vulnerability of pancreatic cancer cells.


Subject(s)
Cell Proliferation , Mice, Nude , Pancreatic Neoplasms , Polyisoprenyl Phosphates , Humans , Animals , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/pharmacology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics
3.
Mol Cell Biol ; 44(4): 123-137, 2024.
Article in English | MEDLINE | ID: mdl-38747374

ABSTRACT

SREBP transcription factors are central regulators of lipid metabolism. Their proteolytic activation requires ER to the Golgi translocation and subsequent cleavage by site-1-protease (S1P). Produced as a proprotein, S1P undergoes autocatalytic cleavage from its precursor S1PA to mature S1PC form. Here, we report that SPRING (previously C12ORF29) and S1P interact through their ectodomains, and that this facilitates the autocatalytic cleavage of S1PA into its mature S1PC form. Reciprocally, we identified a S1P recognition-motif in SPRING and demonstrate that S1P-mediated cleavage leads to secretion of the SPRING ectodomain in cells, and in liver-specific Spring knockout (LKO) mice transduced with AAV-mSpring. By reconstituting SPRING variants into SPRINGKO cells we show that the SPRING ectodomain supports proteolytic maturation of S1P and SREBP signaling, but that S1P-mediated SPRING cleavage is not essential for these processes. Absence of SPRING modestly diminishes proteolytic maturation of S1PA→C and trafficking of S1PC to the Golgi. However, despite reaching the Golgi in SPRINGKO cells, S1PC fails to rescue SREBP signaling. Remarkably, whereas SREBP signaling was severely attenuated in SPRINGKO cells and LKO mice, that of ATF6, another S1P substrate, was unaffected in these models. Collectively, our study positions SPRING as a dedicated licensing factor for SREBP-specific activation by S1P.


Subject(s)
Proprotein Convertases , Serine Endopeptidases , Animals , Humans , Mice , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , Liver/metabolism , Mice, Knockout , Proprotein Convertases/metabolism , Proprotein Convertases/genetics , Proteolysis , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Signal Transduction , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Proteins/genetics
4.
Tissue Cell ; 86: 102276, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37979395

ABSTRACT

The sterol regulatory element-binding protein (SREBP) activation and cytokine level were significantly increased in coronavirus disease-19. The NLRP3 inflammasome is an amplifier for cellular inflammation. This study aimed to elucidate the modulatory effect of SARS-CoV-2 nucleocapsid protein (SARS-CoV-2 NP) on trimethylamine N-oxide (TMAO)-induced lipogenesis and NLRP3 inflammasome activation and the underlying mechanisms in vascular smooth muscle cells (VSMCs). Our data indicated that SARS-CoV-2 NP activates the dissociation of the SREBP cleavage activating protein (SCAP) from the endoplasmic reticulum, resulting in SREBP activation, increased lipogenic gene expression, and NLRP3 inflammasome activation. TMAO was applied to VSMC-induced NLRP3 inflammasome by promoting the SCAP-SREBP complex endoplasmic reticulum-to-Golgi translocation, which facilitates directly binding of SARS-CoV-2 NP to the NLRP3 protein for NLRP3 inflammasome assembly. SARS-CoV-2 NP amplified the TMAO-induced lipogenic gene expression and NLRP3 inflammasome. Knockdown of SCAP-SREBP2 can effectively reduce lipogenic gene expression and alleviate NLRP3 inflammasome-mediated systemic inflammation in VSMCs stimulated with TMAO and SARS-CoV-2 NP. These results reveal that SARS-CoV-2 NP amplified TMAO-induced lipogenesis and NLRP3 inflammasome activation via priming the SCAP-SREBP signaling pathway.


Subject(s)
COVID-19 , Methylamines , Sterol Regulatory Element Binding Proteins , Humans , Sterol Regulatory Element Binding Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , SARS-CoV-2 , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Inflammation , Nucleocapsid Proteins
5.
J Nutr ; 154(2): 626-637, 2024 02.
Article in English | MEDLINE | ID: mdl-38110182

ABSTRACT

BACKGROUND: Artificially fermented dark loose tea is a type of novel dark tea prepared via fermentation by Eurotium cristatum. The effects of artificially fermented dark loose tea on lipid metabolism are still unclear. OBJECTIVES: This study aimed to explore if artificially fermented dark loose tea has the same effects as naturally fermented dark loose tea in regulating hepatic lipid metabolism. METHODS: Thirty-six 8-wk-old male C57BL/6 mice were randomly divided into 6 treatment groups, including normal control (NC), high-fat diet (HFD), positive control (PC), Wuniuzao dark raw tea (WDT), Wuniuzao naturally fermented dark loose tea (NFLT), and Wuniuzao artificially fermented dark loose tea (AFLT) groups. The HFD, PC, WDT, NFLT, and AFLT groups were fed a HFD. The PC group was supplemented with atorvastatin (10 mg/kg). The WDT group was supplemented with WDT (300 mg/kg), the NFLT group with NFLT (300 mg/kg), and the AFLT group with AFLT (300 mg/kg). RESULTS: The study compared the effect of WDT, NFLT, and AFLT on liver steatosis and gut microbiota disorder in obese mice. All 3 tea extracts reduced body weight, glucose tolerance, and serum lipid concentrations. Via sterol-regulatory element binding protein (SREBP)-mediated lipid metabolism, all 3 tea extracts alleviated hepatic steatosis in mice with obesity. Furthermore, NFLT and AFLT intervened in the abundance of Firmicutes, Bacteroidetes, Clostridia, Muribaculaceae, and Lachnospiraceae. CONCLUSION: In mice with obesity induced by a HFD, WDT, NFLT, and AFLT may improve hepatic steatosis through an SREBP-mediated lipid metabolism. Moreover, NFLT and AFLT improved the composition of gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Tea , Male , Mice , Animals , Tea/chemistry , Mice, Obese , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Proteins/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/pharmacology , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Lipid Metabolism , Sterols/pharmacology , Diet, High-Fat
6.
Sci Rep ; 13(1): 19438, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37945738

ABSTRACT

To provide a theoretical basis for the prevention and treatment of atherosclerosis (As), the current study aimed to investigate the mechanism underlying the effect of homocysteine (Hcy) on inducing the lipid deposition and foam cell formation of the vascular smooth muscle cell (VSMC) via C1q/Tumor necrosis factor-related protein9 (CTRP9) promoter region Hypermethylation negative regulating endoplasmic reticulum stress (ERs). Therefore, apolipoprotein E deficient (ApoE-/-) mice were randomly divided into the control [ApoE-/- + normal diet (NC)] and high methionine [ApoE-/- + (normal diet supplemented with 1.7% methionine (HMD)] groups (n = 6 mice/group). Following feeding for 15 weeks, the serum levels of Homocysteine (Hcy), total cholesterol (TC), and triglyceride (TG) were measured using an automatic biochemical analyzer. HE and oil red O staining were performed on the aorta roots to observe the pathological changes. Additionally, immunofluorescence staining was performed to detect the protein expression levels of CTRP9, glucose-regulated protein 78 kD (GRP78), phosphorylated protein kinase RNA-like ER kinase (p-PERK), activating transcription factor 6a (ATF6a), phosphorylated inositol-requiring enzyme-1α (p-IRE1α), sterol regulatory element binding proteins-1c (SREBP1c) and sterol regulatory element binding proteins-2 (SREBP2) in VSMC derived from murine aortic roots. In vitro, VSMC was stimulated with 100 µmol/l Hcy. After transfection of plasmids with overexpression and interference of CTRP9, ERs agonist (TM) and inhibitor (4-PBA) were given to stimulate VSMC cells. HE staining and oil red O staining were used to observe the effect of Hcy stimulation on lipid deposition in VSMC. Additionally, The mRNA and protein expression levels of CTRP9, GRP78, PERK, ATF6a, IRE1α, SREBP1c, and SREBP2 in VSMC were detected by RT-qPCR and western blot analysis, respectively. Finally, The methylation modification of the CTRP9 promoter region has been studied. The NCBI database was used to search the promoter region of the CTRP9 gene, and CpG Island was used to predict the methylation site. After Hcy stimulation of VSMC, overexpression of DNMT1, and intervention with 5-Azc, assess the methylation level of the CTRP9 promoter through bisulfite sequencing PCR (BSP). The results showed that the serum levels of Hcy, TC, and TG in the ApoE-/- + HMD group were significantly increased compared with the ApoE-/- + NC group. In addition, HE staining and oil red O staining showed obvious AS plaque formation in the vessel wall, and a large amount of fat deposition in VSMC, thus indicating that the hyperhomocysteinemia As an animal model was successfully established. Furthermore, CTRP9 were downregulated, while GRP78, p-PERK, ATF6a, p-IRE1α, SREBP1c, SREBP2 was upregulated in aortic VSMC in the ApoE-/- + HMD group. Consistent with the in vivo results, Hcy can inhibit the expression of CTRP9 in VSMC and induce ERs and lipid deposition in VSMC. Meanwhile, the increased expression of CTRP9 can reduce ERs and protect the lipid deposition in Hcy induced VSMC. Furthermore, ERs can promote Hcy induced VSMC lipid deposition, inhibition of ERs can reduce Hcy induced VSMC lipid deposition, and CTRP9 may play a protective role in Hcy induced VSMC lipid deposition and foam cell transformation through negative regulation of ERs. In addition, The CTRP9 promoter in the Hcy group showed hypermethylation. At the same time as Hcy intervention, overexpression of DNMT1 increases the methylation level of the CTRP9 promoter, while 5-Azc can reduce the methylation level of the CTRP9 promoter. Finally, Hcy can up-regulate the expression of DNMT1 and down-regulate the expression of CTRP9. After overexpression of DNMT1, the expression of CTRP9 is further decreased. After 5-Azc inhibition of DNMT1, the expression of DNMT1 decreases, while the expression of CTRP9 increases. It is suggested that the molecular mechanism of Hcy inhibiting the expression of CTRP9 is related to the hypermethylation of the CTRP9 promoter induced by Hcy and regulated by DNMT1. 5-Azc can inhibit the expression of DNMT1 and reverse the regulatory effect of DNMT1 on CTRP9. Overall, the results of the present study suggested that Hcy induces DNA hypermethylation in the CTRP9 promoter region by up-regulating DNMT1 expression, and negatively regulates ERs mediated VSMC lipid deposition and foam cell formation. CTRP9 may potentially be a therapeutic target in the treatment of hyperhomocysteinemia and As.


Subject(s)
Atherosclerosis , Hyperhomocysteinemia , Mice , Animals , Endoribonucleases/metabolism , Endoplasmic Reticulum Chaperone BiP , Muscle, Smooth, Vascular/metabolism , Foam Cells/metabolism , Hyperhomocysteinemia/pathology , Protein Serine-Threonine Kinases/metabolism , Atherosclerosis/metabolism , Promoter Regions, Genetic , Methionine/metabolism , Apolipoproteins E/metabolism , Lipids/pharmacology , Homocysteine/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Endoplasmic Reticulum Stress
7.
Mol Cell ; 83(16): 3010-3026.e8, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37595559

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth that stimulates macromolecule synthesis through transcription, RNA processing, and post-translational modification of metabolic enzymes. However, the mechanisms of how mTORC1 orchestrates multiple steps of gene expression programs remain unclear. Here, we identify family with sequence similarity 120A (FAM120A) as a transcription co-activator that couples transcription and splicing of de novo lipid synthesis enzymes downstream of mTORC1-serine/arginine-rich protein kinase 2 (SRPK2) signaling. The mTORC1-activated SRPK2 phosphorylates splicing factor serine/arginine-rich splicing factor 1 (SRSF1), enhancing its binding to FAM120A. FAM120A directly interacts with a lipogenic transcription factor SREBP1 at active promoters, thereby bridging the newly transcribed lipogenic genes from RNA polymerase II to the SRSF1 and U1-70K-containing RNA-splicing machinery. This mTORC1-regulated, multi-protein complex promotes efficient splicing and stability of lipogenic transcripts, resulting in fatty acid synthesis and cancer cell proliferation. These results elucidate FAM120A as a critical transcription co-factor that connects mTORC1-dependent gene regulation programs for anabolic cell growth.


Subject(s)
Arginine , Lipogenesis , Sterol Regulatory Element Binding Protein 1 , Lipogenesis/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , RNA Splicing Factors , Sterol Regulatory Element Binding Protein 1/metabolism , Humans , Sterol Regulatory Element Binding Proteins/metabolism
8.
J Agric Food Chem ; 71(32): 12250-12263, 2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37493643

ABSTRACT

Sterol regulatory element-binding protein, Sre1, regulates sterol biosynthesis, lipid metabolism, hypoxia adaptation, and virulence in some fungi, even though its roles are varied in fungal species. However, few studies report its other functions in fungi. Here, we report novel roles of Sre1 homolog, BbSre1, in the insect fungal pathogen, Beauveria bassiana, that regulates oxidative stress response, peroxisome division, and redox homeostasis. The gene disruption stain showed increased sensitivity to oxidative stress, which was in line with oxidative stress-induced-BbSre1 nuclear import and control of antioxidant and detoxification-involved genes. The gene mutation also inhibited peroxisome division, affected redox homeostasis, and impaired lipid/fatty acid metabolism and sterol biosynthesis, which was verified by downregulation of their associated genes. These data broaden our understanding of role of Sre1, which regulates peroxisome division, antioxidant, and detoxification-involved genes for control of redox homeostasis and oxidative stress response that links to lipid/fatty acid metabolism and sterol biosynthesis.


Subject(s)
Antioxidants , Sterol Regulatory Element Binding Proteins , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Antioxidants/metabolism , Peroxisomes/genetics , Peroxisomes/metabolism , Homeostasis , Oxidative Stress , Oxidation-Reduction , Sterols/metabolism , Fatty Acids/metabolism , Lipids
9.
Nat Commun ; 14(1): 763, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36808152

ABSTRACT

Sleep behavior is conserved throughout evolution, and sleep disturbances are a frequent comorbidity of neuropsychiatric disorders. However, the molecular basis underlying sleep dysfunctions in neurological diseases remains elusive. Using a model for neurodevelopmental disorders (NDDs), the Drosophila Cytoplasmic FMR1 interacting protein haploinsufficiency (Cyfip85.1/+), we identify a mechanism modulating sleep homeostasis. We show that increased activity of the sterol regulatory element-binding protein (SREBP) in Cyfip85.1/+ flies induces an increase in the transcription of wakefulness-associated genes, such as the malic enzyme (Men), causing a disturbance in the daily NADP+/NADPH ratio oscillations and reducing sleep pressure at the night-time onset. Reduction in SREBP or Men activity in Cyfip85.1/+ flies enhances the NADP+/NADPH ratio and rescues the sleep deficits, indicating that SREBP and Men are causative for the sleep deficits in Cyfip heterozygous flies. This work suggests modulation of the SREBP metabolic axis as a new avenue worth exploring for its therapeutic potential in sleep disorders.


Subject(s)
Drosophila Proteins , Sterol Regulatory Element Binding Proteins , Animals , Sterol Regulatory Element Binding Proteins/metabolism , NADP/metabolism , Drosophila/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sleep , Sterol Regulatory Element Binding Protein 2/metabolism , Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein
10.
Commun Biol ; 6(1): 1, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596887

ABSTRACT

Ganoderic acids (GAs) are well recognized as important pharmacological components of the medicinal species belonging to the basidiomycete genus Ganoderma. However, transcription factors directly regulating the expression of GA biosynthesis genes remain poorly understood. Here, the genome of Ganoderma lingzhi is de novo sequenced. Using DNA affinity purification sequencing, we identify putative targets of the transcription factor sterol regulatory element-binding protein (SREBP), including the genes of triterpenoid synthesis and lipid metabolism. Interactions between SREBP and the targets are verified by electrophoretic mobility gel shift assay. RNA-seq shows that SREBP targets, mevalonate kinase and 3-hydroxy-3-methylglutaryl coenzyme A synthetase in mevalonate pathway, sterol isomerase and lanosterol 14-demethylase in ergosterol biosynthesis, are significantly upregulated in the SREBP overexpression (OE::SREBP) strain. In addition, 3 targets involved in glycerophospholipid/glycerolipid metabolism are upregulated. Then, the contents of mevalonic acid, lanosterol, ergosterol and 13 different GAs as well as a variety of lipids are significantly increased in this strain. Furthermore, the effects of SREBP overexpression on triterpenoid and lipid metabolisms are recovered when OE::SREBP strain are treated with exogenous fatostatin, a specific inhibitor of SREBP. Taken together, our genome-wide study clarify the role of SREBP in triterpenoid and lipid metabolisms of G. lingzhi.


Subject(s)
Ganoderma , Triterpenes , Lanosterol/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Lipid Metabolism , Genome-Wide Association Study , Triterpenes/pharmacology , Triterpenes/metabolism , Ganoderma/genetics , Ganoderma/chemistry , Ganoderma/metabolism , Sterols/metabolism , Ergosterol/metabolism
11.
Food Funct ; 13(21): 10923-10936, 2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36205648

ABSTRACT

In the present study, mice with high-fat-diet-induced obesity were used in investigating the anti-obesity effects of an aqueous extract and isoquercitrin from Apocynum venetum L. The aqueous extract and the signal molecule isoquercitrin significantly reduced the body weight gain, food intake, water consumption, and fasting blood glucose, plasma triglyceride and total cholesterol levels of the obese mice. Furthermore, the mechanism of action of isoquercitrin was explored through RT-PCR analyses and uptake experiments of adenosine 5'-monophosphate-activated protein kinase (AMPK) and sterol regulatory-element binding protein (SREBP-1c) inhibitors and glucose. The indexes of SREBP-1c, fatty acid synthase (FAS), stearoyl-CoA desaturase-1 (SCD), and cluster of differentiation 36 (CD36) in obese mice significantly increased but returned to normal levels after the administration of isoquercitrin. Meanwhile, the anti-obesity effect of isoquercitrin was diminished by the inhibitors of AMPK and SREBP-1c. In addition, intestinal glucose uptake in normal mice was significantly inhibited after the oral administration of isoquercitrin. Moreover, 2D gel electrophoresis based proteome-wide cellular thermal shift assay (CETSA) showed that the potential target proteins of isoquercitrin were C-1-tetrahydrofolate synthase, carbonyl reductase, and glutathione S-transferase P. These results suggested that isoquercitrin produces an anti-obesity effect by targeting the above-mentioned proteins and regulating the AMPK/SREBP-1c signaling pathway and potentially prevents obesity and obesity-related metabolic disorders.


Subject(s)
Apocynum , Sterol Regulatory Element Binding Proteins , Mice , Animals , Mice, Obese , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Proteins/pharmacology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Apocynum/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Liver/metabolism , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Obesity/drug therapy , Obesity/metabolism , Signal Transduction , Tetrahydrofolates/metabolism , Tetrahydrofolates/pharmacology , Mice, Inbred C57BL , Lipid Metabolism
12.
Chemosphere ; 308(Pt 3): 136424, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36116629

ABSTRACT

Bisphenol AF (BPAF) is one of the substitutes for bisphenol A (BPA), which has endocrine-disrupting, reproductive and neurological toxicity. BPAF has frequently been detected in the aquatic environment, which has been a long-term threat to the health of aquatic organisms. In this study, female marine medaka (Oryzias melastigma) were exposed to 6.7 µg/L, 73.4 µg/L, and 367.0 µg/L BPAF for 120 d. The effects of BPAF on behavior, growth, liver and ovarian histology, gene transcriptional profiles, and reproduction of marine medaka were determined. The results showed that with the increase of BPAF concentration, the swimming speed of female marine medaka showed an increasing trend and then decreasing trend. BPAF (367.0 µg/L) significantly increased body weight and condition factors in females. BPAF (73.4 µg/L and 367.0 µg/L) significantly delayed oocyte maturation. Exposure to 367.0 µg/L BPAF showed an increasing trend in the transcript levels of lipid synthesis and transport-related genes such as fatty acid synthase (fasn), sterol regulatory element binding protein (srebf), diacylglycerol acyltransferase (dgat), solute carrier family 27 member 4 (slc27a4), fatty acid-binding protein (fabp), and peroxisome proliferator-activated receptor gamma (pparγ) in the liver. In addition, 6.7 µg/L BPAF significantly down-regulated the expression levels of antioxidant-related genes [superoxide dismutase (sod), glutathione peroxidase (gpx), and catalase (cat)], and complement system-related genes [complement component 5 (c5), complement component 7a (c7a), mannan-binding lectin serine peptidase 1 (masp1), and tumor necrosis factor (tnf)] were significantly up-regulated in the 73.4 and 367.0 µg/L groups, which implies the effect of BPAF on the immune system in the liver. In the hypothalamic-pituitary-ovarian axis (HPG) results, the transcription levels of estrogen receptor α (erα), estrogen receptor ß (erß), androgen receptor (arα), gonadotropin-releasing hormone 2 (gnrh2), cytochrome P450 19b (cyp19b), aromatase (cyp19a), and luteinizing hormone receptor (lhr) in the brain and ovary, and vitellogenin (vtg) and choriogenin (chg) in the liver of 367.0 µg/L BPAF group showed a downward trend. In addition, exposure to 367.0 µg/L BPAF for 120 d inhibited the spawning behavior of marine medaka. Our results showed that long-term BPAF treatment influenced growth (body weight and condition factors), lipid metabolism, and ovarian maturation, and significantly altered the immune response and the transcriptional expression levels of HPG axis-related genes.


Subject(s)
Mannose-Binding Lectin , Oryzias , Water Pollutants, Chemical , Animals , Antioxidants/metabolism , Aromatase/metabolism , Benzhydryl Compounds , Body Weight , Catalase/metabolism , Complement C5/genetics , Complement C5/metabolism , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Fatty Acid-Binding Proteins/genetics , Female , Fluorocarbons , Gene Expression , Glutathione Peroxidase/metabolism , Gonadotropin-Releasing Hormone/metabolism , Lipids , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/metabolism , Oryzias/physiology , PPAR gamma/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Receptors, Androgen/metabolism , Receptors, LH/genetics , Serine/genetics , Serine/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Superoxide Dismutase/metabolism , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism , Vitellogenins/metabolism , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/toxicity
13.
Environ Pollut ; 307: 119579, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35671893

ABSTRACT

DEHP is commonly found in the environment, biota, food, and humans, raising significant health concerns. Whether developmental stage and exposure duration modify the obesogenic effects of DEHP is unclear, especially the underlying mechanisms by which chronic exposure to DEHP as well as its metabolites remain largely unknown. This study investigated the obesogenic effects of chronic DEHP exposure, with levels below environmentally-relevant amounts and provide the mechanism in Caenorhabditis elegans. We show that early-life DEHP exposure resulted in an increased lipid and triglyceride (TG) accumulation mainly attributed to DEHP itself, not its metabolite mono-2-ethylhexyl phthalate (MEHP). In addition, developmental stage and exposure timing influence DEHP-induced TG accumulation and chronic DEHP exposure resulted in the most significant effect. Analysis of fatty acid composition shows that chronic DEHP exposure altered fatty acid composition and TG, resulting in an increased ω-6/ω-3 ratio. The increased TG content by chronic DEHP exposure required lipogenic genes fat-6, fat-7, pod-2, fasn-1, and sbp-1. Moreover, chronic DEHP exposure induced XBP-1-mediated endoplasmic reticulum (ER) stress which might lead to up-regulation of sbp-1. This study suggests the possible involvement of ER stress and SBP-1/SREBP-mediated lipogenesis in chronic DEHP-induced obesogenic effects. Results from this study implies that chronic exposure to DEHP disrupts lipid metabolism, which is likely conserved across species due to evolutionary conservation of molecular mechanisms, raising concerns in ecological and human health.


Subject(s)
Caenorhabditis elegans Proteins , Diethylhexyl Phthalate , Endoplasmic Reticulum Stress , Sterol Regulatory Element Binding Proteins , Transcription Factors , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Diethylhexyl Phthalate/toxicity , Endoplasmic Reticulum Stress/drug effects , Fatty Acids/metabolism , Humans , Lipid Metabolism/drug effects , Sterol Regulatory Element Binding Proteins/metabolism , Transcription Factors/metabolism
14.
Biosci Biotechnol Biochem ; 86(9): 1220-1230, 2022 Aug 24.
Article in English | MEDLINE | ID: mdl-35723236

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) are transcription factors that have important roles in the genes involved in lipid biosynthesis. In this study, it was found that the flavonoid pectolinarigenin, reduced the activity of SRE-containing fatty acid synthase (FAS) promoter and the mRNA expressions of SREBP target genes in human hepatoma (Huh-7) cells. Moreover, compared with other flavonoids, pectolinarigenin reduced the mature forms of SREBPs in a dose-dependent manner. The insulin-induced gene (INSIG) and proteasome were not involved in the pectolinarigenin-mediated reduction of mature forms of SREBPs. Pectolinarigenin also reduced the lipid contents in vitro. These results suggest that pectolinarigenin may inhibit lipogenesis through suppressing SREBP activity, at least partially, via the formation of SREBPs mature forms, thereby reducing the expression of their downstream genes related to lipogenesis. To the best of our knowledge, this is the first work that shows how pectolinarigenin affects cellular lipid levels by affecting SREBPs.


Subject(s)
Flavonoids , Sterol Regulatory Element Binding Proteins , CCAAT-Enhancer-Binding Proteins , Chromones , Flavonoids/pharmacology , Humans , Lipids , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 2 , Sterol Regulatory Element Binding Proteins/metabolism , Sterols
15.
Sci Rep ; 12(1): 8715, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35610278

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) are transcription factors that regulate various genes involved in cholesterol and fatty acid synthesis. In this study, we describe that naturally occurring isothiocyanate sulforaphane (SFaN) impairs fatty acid synthase promoter activity and reduces SREBP target gene (e.g., fatty acid synthase and acetyl-CoA carboxylase 1) expression in human hepatoma Huh-7 cells. SFaN reduced SREBP proteins by promoting the degradation of the SREBP precursor. Amino acids 595-784 of SREBP-1a were essential for SFaN-mediated SREBP-1a degradation. We also found that such SREBP-1 degradation occurs independently of the SREBP cleavage-activating protein and the Keap1-Nrf2 pathway. This study identifies SFaN as an SREBP inhibitor and provides evidence that SFaN could have major potential as a pharmaceutical preparation against hepatic steatosis and obesity.


Subject(s)
NF-E2-Related Factor 2 , Sterol Regulatory Element Binding Proteins , CCAAT-Enhancer-Binding Proteins/metabolism , Cholesterol/metabolism , Fatty Acid Synthases/metabolism , Humans , Isothiocyanates/pharmacology , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Sulfoxides
16.
Dev Cell ; 57(6): 719-731.e8, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35216681

ABSTRACT

The coordinated regulation of growth control and metabolic pathways is required to meet the energetic and biosynthetic demands associated with proliferation. Emerging evidence suggests that the Hippo pathway effector Yes-associated protein 1 (YAP) reprograms cellular metabolism to meet the anabolic demands of growth, although the mechanisms involved are poorly understood. Here, we demonstrate that YAP co-opts the sterol regulatory element-binding protein (SREBP)-dependent lipogenic program to facilitate proliferation and tissue growth. Mechanistically, YAP stimulates de novo lipogenesis via mechanistic target of rapamcyin (mTOR) complex 1 (mTORC1) signaling and subsequent activation of SREBP. Importantly, YAP-dependent regulation of serum- and glucocorticoid-regulated kinase 1 (SGK1) is required to activate mTORC1/SREBP and stimulate de novo lipogenesis. We also find that the SREBP target genes fatty acid synthase (FASN) and stearoyl-CoA desaturase (SCD) are conditionally required to support YAP-dependent proliferation and tissue growth. These studies reveal that de novo lipogenesis is a metabolic vulnerability that can be targeted to disrupt YAP-dependent proliferation and tissue growth.


Subject(s)
Lipogenesis , Sterol Regulatory Element Binding Proteins , Cell Proliferation , Lipogenesis/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
17.
Mol Med ; 28(1): 3, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35030992

ABSTRACT

The mechanisms of chronic intermittent hypoxia (CIH)-induced cognitive deficits remain unclear. Here, our study found that about 3 months CIH treatment induced lipid droplets (LDs) accumulation in hippocampal nerve and glia cells of C57BL/6 mice, and caused severe neuro damage including neuron lesions, neuroblast (NB) apoptosis and abnormal glial activation. Studies have shown that the neuronal metabolism disorders might contribute to the CIH induced-hippocampal impairment. Mechanistically, the results showed that pyruvate dehydrogenase complex E1ɑ subunit (PDHA1) and the pyruvate dehydrogenase complex (PDC) activator pyruvate dehydrogenase phosphatase 1 (PDP1) did not noticeable change after intermittent hypoxia. Consistent with those results, the level of Acetyl-CoA in hippocampus did not significantly change after CIH exposure. Interestingly, we found that CIH produced large quantities of ROS, which activated the JNK/SREBP/ACC pathway in nerve and glia cells. ACC catalyzed the carboxylation of Acetyl-CoA to malonyl-CoA and then more lipid acids were synthesized, which finally caused aberrant LDs accumulation. Therefore, the JNK/SREBP/ACC pathway played a crucial role in the cognitive deficits caused by LDs accumulation after CIH exposure. Additionally, LDs were peroxidized by the high level of ROS under CIH conditions. Together, lipid metabolic disorders contributed to nerve and glia cells damage, which ultimately caused behavioral dysfunction. An active component of Salvia miltiorrhiza, SMND-309, dramatically alleviated these injuries and improved cognitive deficits of CIH mice.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Cognition , Eye Proteins/metabolism , GTP-Binding Protein Regulators/metabolism , Lipid Droplets/metabolism , Phosphoproteins/metabolism , Protein Phosphatase 2C/metabolism , Sleep Apnea, Obstructive/etiology , Sleep Apnea, Obstructive/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Caffeic Acids/pharmacology , Cognitive Dysfunction , Disease Models, Animal , Hippocampus/metabolism , Hypoxia/metabolism , Learning , Lipid Peroxidation , MAP Kinase Signaling System , Memory , Mice , Neurons , Reactive Oxygen Species/metabolism , Signal Transduction , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/drug therapy
18.
Acta Pharmacol Sin ; 43(3): 505-519, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33990764

ABSTRACT

Renal fibrosis contributes to progressive damage to renal structure and function. It is a common pathological process as chronic kidney disease develops into kidney failure, irrespective of diverse etiologies, and eventually leads to death. However, there are no effective drugs for renal fibrosis treatment at present. Lipid aggregation in the kidney and consequent lipotoxicity always accompany chronic kidney disease and fibrosis. Numerous studies have revealed that restoring the defective fatty acid oxidation in the kidney cells can mitigate renal fibrosis. Thus, it is an important strategy to reverse the dysfunctional lipid metabolism in the kidney, by targeting critical regulators of lipid metabolism. In this review, we highlight the potential "druggability" of lipid metabolism to ameliorate renal fibrosis and provide current pre-clinical evidence, exemplified by some representative druggable targets and several other metabolic regulators with anti-renal fibrosis roles. Then, we introduce the preliminary progress of noncoding RNAs as promising anti-renal fibrosis drug targets from the perspective of lipid metabolism. Finally, we discuss the prospects and deficiencies of drug targeting lipid reprogramming in the kidney.


Subject(s)
Fibrosis/pathology , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Renal Insufficiency, Chronic/pathology , Animals , CD36 Antigens/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Epithelial Cells/pathology , Fatty Acids/metabolism , Inflammation/pathology , Kidney/physiology , MicroRNAs/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , RNA, Untranslated/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism
19.
Arch Physiol Biochem ; 128(5): 1383-1390, 2022 Oct.
Article in English | MEDLINE | ID: mdl-32643419

ABSTRACT

The study aimed to assess the effect of simvastatin on gene expression of LDLR, SREBPs, and SCD1 in rat hepatic tissues fed with high-fat diets (HFD) and its association with some biochemical parameters. Thirty-two male Wister albino rats were divided into four equal groups (three test and one control groups). The biochemical parameters were determined by using spectrophotometer techniques and the Elisa method. Low-density lipoprotein receptor, sterol regulatory element-binding proteins, stearoyl-CoA desaturase1, Beta-actin were analysed by real-time quantitative polymerase chain reaction (RT-PCR) method. At the end of study, the livers of the rats were separated and changes of hepatic tissue were determined. LDLR, SREBP2, and SCD1 expression increased significantly when compared G1 versus G4 and G2 versus G4. The expression of LDLR, SREBP2, and SCD1 also increased significantly when compared G2 versus G3, G1versus G3 and G1 versus G3 and G2 versus G3. The serum level of cholesterol, triglyceride, glucose, LDL, and HDL increased significantly when compared G1 versus G3. LDL showed significantly decreased when compared G1 versus G2. Cholesterol, glucose and HDL and triglyceride levels were increased significantly when compared G1 versus G4 and G2. Treatment of rats with HFD and simvastatin 20 mg/kg, triglyceride and LDL were almost the same as a control group and LDLR expression increased 98% in liver tissue. Gene expressions may be up-regulated in liver tissue and they showed different effects on biochemical parameters.


Subject(s)
Stearoyl-CoA Desaturase , Sterol Regulatory Element Binding Proteins , Actins/genetics , Actins/metabolism , Actins/pharmacology , Animals , Cholesterol , Gene Expression , Glucose/metabolism , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Liver/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Simvastatin/pharmacology , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/pharmacology , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Proteins/pharmacology , Triglycerides
20.
Microbiologyopen ; 10(6): e1249, 2021 11.
Article in English | MEDLINE | ID: mdl-34964293

ABSTRACT

In fungal pathogens, the transcription factor SrbA (a sterol regulatory element-binding protein, SREBP) and CBC (CCAAT binding complex) have been reported to regulate azole resistance by competitively binding the TR34 region (34 mer) in the promoter of the drug target gene, erg11A. However, current knowledge about how the SrbA and CBC coordinately mediate erg11A expression remains limited. In this study, we uncovered a novel relationship between HapB (a subunit of CBC) and SrbA in which deletion of hapB significantly prolongs the nuclear retention of SrbA by increasing its expression and cleavage under azole treatment conditions, thereby enhancing Erg11A expression for drug resistance. Furthermore, we verified that loss of HapB significantly induces the expression of the rhomboid protease RbdB, Dsc ubiquitin E3 ligase complex, and signal peptide peptidase SppA, which are required for the cleavage of SrbA, suggesting that HapB acts as a repressor for these genes which contribute to the activation of SrbA by proteolytic cleavage. Together, our study reveals that CBC functions not only to compete with SrbA for binding to erg11A promoter region but also to affect SrbA expression, cleavage, and translocation to nuclei for the function, which ultimately regulate Erg11A expression and azole resistance.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Azoles/pharmacology , CCAAT-Binding Factor/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Aspergillus fumigatus/genetics , Aspergillus fumigatus/metabolism , CCAAT-Binding Factor/genetics , Cytochrome P450 Family 51/genetics , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Microbial Sensitivity Tests , Mutation , Proteolysis , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL