Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
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
2.
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
3.
Int J Mol Sci ; 23(19)2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36232834

ABSTRACT

Particulate matter 2.5 (PM2.5), an atmospheric pollutant with an aerodynamic diameter of <2.5 µm, can cause serious human health problems, including skin damage. Since sebocytes are involved in the regulation of skin homeostasis, it is necessary to study the effects of PM2.5 on sebocytes. We examined the role of PM2.5 via the identification of differentially expressed genes, functional enrichment and canonical pathway analysis, upstream regulator analysis, and disease and biological function analysis through mRNA sequencing. Xenobiotic and lipid metabolism, inflammation, oxidative stress, and cell barrier damage-related pathways were enriched; additionally, PM2.5 altered steroid hormone biosynthesis and retinol metabolism-related pathways. Consequently, PM2.5 increased lipid synthesis, lipid peroxidation, inflammatory cytokine expression, and oxidative stress and altered the lipid composition and expression of factors that affect cell barriers. Furthermore, PM2.5 altered the activity of sterol regulatory element binding proteins, mitogen-activated protein kinases, transforming growth factor beta-SMAD, and forkhead box O3-mediated pathways. We also suggest that the alterations in retinol and estrogen metabolism by PM2.5 are related to the damage. These results were validated using the HairSkin® model. Thus, our results provide evidence of the harmful effects of PM2.5 on sebocytes as well as new targets for alleviating the skin damage it causes.


Subject(s)
Environmental Pollutants , Particulate Matter , Cytokines/genetics , Estrogens , Gene Expression Profiling , Humans , Lipids , Mitogen-Activated Protein Kinases/metabolism , Particulate Matter/chemistry , Particulate Matter/toxicity , RNA, Messenger , Steroids , Sterol Regulatory Element Binding Proteins/genetics , Transforming Growth Factor beta/genetics , Vitamin A , Xenobiotics
4.
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
5.
Fungal Genet Biol ; 163: 103739, 2022 11.
Article in English | MEDLINE | ID: mdl-36089227

ABSTRACT

Fungal biofilm founder cells experience self-generated hypoxia leading to dramatic changes in their cell biology. For example, during Aspergillus nidulans biofilm formation microtubule (MT) disassembly is triggered causing dispersal of EB1 from MT tips. This process is dependent on SrbA, a sterol regulatory element-binding transcription factor required for adaptation to hypoxia. We show that SrbA, an ER resident protein prior to activation, is proteolytically activated during early stages of biofilm formation and that, like SrbA itself, its activating proteases are also required for normal biofilm MT disassembly. In addition to SrbA, the AtrR transcription factor is also found to be required to modulate cellular responses to gaseous signaling during biofilm development. Using co-cultures, we further show that cells lacking srbA or atrR are capable of responding to biofilm generated gaseous microenvironments but are actually more sensitive to this signal than wild type cells. SrbA is a regulator of ergosterol biosynthetic genes and we find that the levels of seven GFP-tagged Erg proteins differentially accumulate during biofilm formation with various dependencies on SrbA for their accumulation. This uncovers a complex pattern of regulation with biofilm accumulation of only some Erg proteins being dependent on SrbA with others accumulating to higher levels in its absence. Because different membrane sterols are known to influence cell permeability to gaseous molecules, including oxygen, we propose that differential regulation of ergosterol biosynthetic proteins by SrbA potentially calibrates the cell's responsiveness to gaseous signaling which in turn modifies the cell biology of developing biofilm cells.


Subject(s)
Aspergillus nidulans , Aspergillus nidulans/genetics , Aspergillus nidulans/metabolism , Aspergillus fumigatus/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Sterols/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gases/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Hypoxia , Biofilms , Ergosterol/metabolism
6.
J Integr Neurosci ; 21(3): 95, 2022 May 16.
Article in English | MEDLINE | ID: mdl-35633176

ABSTRACT

BACKGROUND: The central nervous system (CNS) is enriched in lipids; despite this, studies exploring the functional roles of lipids in the brain are still limited. Sterol regulatory element binding protein (SREBP) signaling is a transcriptomic pathway that predominantly participates in the maintenance of lipid homeostasis; however, its involvement in the CNS dysfunction is not well-established. In this study, we aimed to characterize and pinpoint specific genes of the SREBP pathway which may be implicated in neurodegenerative, neurological, and neuropsychiatric diseases. METHODS: In silico bioinformatic analysis was performed using the open-source databases DisGeNET and MSigDB. Protein-protein interaction data were visualized and analyzed using STRING, after which GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analyses were conducted via DAVID (Database for Annotation, Visualization and Integrated Discovery). RESULTS: Several common genes were identified between the SREBP pathway and CNS disorders. In GO enrichment analysis, the most enriched biological processes included lipid, cholesterol, and steroid biosynthetic processes; the most enriched molecular functions were transcription factor-related; and the most enriched subcellular compartments revealed that the genes involved in CNS disorders were mainly associated with the enzyme complexes of acetyl-CoA carboxylase (ACC) and fatty acid synthase (FASN). In KEGG enrichment analysis, the most enriched pathway was the AMP-activated protein kinase (AMPK) signaling pathway, and the top-ranked genes significantly enriched under this pathway were ACACA, ACACB, FASN, HMGCR, MTOR, PPARGC1A, PRKAA1, SCD, SIRT1, and SREBF1. CONCLUSIONS: The findings of this study strengthen the evidence linking the involvement of lipid homeostasis in CNS functions. We suggest herein the roles of downstream ACC and FASN enzymes and upstream AMPK signaling in the SREBP pathway as mechanisms underlying neurodegenerative, neurological, and neuropsychiatric CNS disorders.


Subject(s)
Central Nervous System Diseases , Sterol Regulatory Element Binding Proteins , AMP-Activated Protein Kinases/metabolism , Humans , Lipids , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Proteins/genetics
7.
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
8.
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
9.
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
10.
Endocrinology ; 162(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34431998

ABSTRACT

In the liver, the sterol response element binding protein (SREBP) and the SREBP cleavage-activated protein (SCAP) complex upregulate cholesterol biosynthesis by gene induction of de novo cholesterol synthetic enzymes (Hmgcr, Cyp51, and Dhcr7). Insulin induced gene 1 (INSIG1) negatively regulates cholesterol biosynthesis by the inhibition of de novo cholesterol biosynthetic gene expression. In the ovary, cholesterol is de novo synthesized; however, the roles of SREBP and its regulators (SCAP and INSIG1) are not well understood. In this study, when immature mice were treated with gonadotropins (eCG followed by hCG), eCG induced and hCG maintained the expression of SREBP-1a, -2, and SCAP granulosa cells, whereas INSIG1 expression was dramatically downregulated after hCG injection. Downregulation of INSIG1 led to generate the SREBPs active form and translocate the SREBPs active form to nuclei. Inhibition of generation of the SREBPs active form by fatostatin or Scap siRNA in both in vivo and in vitro significantly decreased the expressions of de novo cholesterol biosynthetic enzymes, cholesterol accumulation, and progesterone (P4) production compared with the control group. Fatostatin treatment inhibited the ovulation and increased the formation of abnormal corpus luteum which trapped the matured oocyte in the corpus luteum; however, the phenomenon was abolished by P4 administration. The results showed that decreasing INSIG1 level after hCG stimulation activated SREBP-induced de novo cholesterol biosynthesis in granulosa cells of preovulatory follicles, which is essential for P4 production and the rupture of matured oocyte during ovulation process.


Subject(s)
Cholesterol/biosynthesis , Granulosa Cells/drug effects , Luteinizing Hormone/pharmacology , Ovulation/drug effects , Animals , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Granulosa Cells/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Ovulation/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
11.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34204880

ABSTRACT

Dysregulated mammalian target of rapamycin (mTOR) activity is associated with various neurodevelopmental disorders ranging from idiopathic autism spectrum disorders (ASD) to syndromes caused by single gene defects. This suggests that maintaining mTOR activity levels in a physiological range is essential for brain development and functioning. Upon activation, mTOR regulates a variety of cellular processes such as cell growth, autophagy, and metabolism. On a molecular level, however, the consequences of mTOR activation in the brain are not well understood. Low levels of cholesterol are associated with a wide variety of neurodevelopmental disorders. We here describe numerous genes of the sterol/cholesterol biosynthesis pathway to be transcriptionally regulated by mTOR complex 1 (mTORC1) signaling in vitro in primary neurons and in vivo in the developing cerebral cortex of the mouse. We find that these genes are shared targets of the transcription factors SREBP, SP1, and NF-Y. Prenatal as well as postnatal mTORC1 inhibition downregulated expression of these genes which directly translated into reduced cholesterol levels, pointing towards a substantial metabolic function of the mTORC1 signaling cascade. Altogether, our results indicate that mTORC1 is an essential transcriptional regulator of the expression of sterol/cholesterol biosynthesis genes in the developing brain. Altered expression of these genes may be an important factor contributing to the pathogenesis of neurodevelopmental disorders associated with dysregulated mTOR signaling.


Subject(s)
Cholesterol/genetics , Neurons/metabolism , Protein Kinases/genetics , Sterol Regulatory Element Binding Proteins/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Autophagy/genetics , CCAAT-Binding Factor/genetics , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cholesterol/biosynthesis , Gene Expression Regulation, Developmental/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Neurogenesis/genetics , Primary Cell Culture , Signal Transduction/genetics , Transcription, Genetic/genetics
12.
J Med Chem ; 64(9): 5689-5709, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33899473

ABSTRACT

Vitamin D3 metabolites inhibit the expression of lipogenic genes by impairing sterol regulatory element-binding protein (SREBP), a master transcription factor of lipogenesis, independent of their canonical activity through a vitamin D receptor (VDR). Herein, we designed and synthesized a series of vitamin D derivatives to search for a drug-like small molecule that suppresses the SREBP-induced lipogenesis without affecting the VDR-controlled calcium homeostasis in vivo. Evaluation of the derivatives in cultured cells and mice led to the discovery of VDR-silent SREBP inhibitors and to the development of KK-052 (50), the first vitamin D-based SREBP inhibitor that has been demonstrated to mitigate hepatic lipid accumulation without calcemic action in mice. KK-052 maintained the ability of 25-hydroxyvitamin D3 to induce the degradation of SREBP but lacked in the VDR-mediated activity. KK-052 serves as a valuable compound for interrogating SREBP/SCAP in vivo and may represent an unprecedented translational opportunity of synthetic vitamin D analogues.


Subject(s)
Drug Design , Sterol Regulatory Element Binding Proteins/metabolism , Vitamin D/analogs & derivatives , Animals , Body Weight/drug effects , CHO Cells , Cricetinae , Cricetulus , Cycloaddition Reaction , Disease Models, Animal , Drug Evaluation, Preclinical , Fatty Liver/drug therapy , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipogenesis/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/metabolism , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors , Sterol Regulatory Element Binding Proteins/genetics , Vitamin D/metabolism , Vitamin D/pharmacology , Vitamin D/therapeutic use
13.
Protein Cell ; 12(4): 279-296, 2021 04.
Article in English | MEDLINE | ID: mdl-32666500

ABSTRACT

Sterol-regulatory element binding proteins (SREBPs) are the key transcriptional regulators of lipid metabolism. The activation of SREBP requires translocation of the SREBP precursor from the endoplasmic reticulum to the Golgi, where it is sequentially cleaved by site-1 protease (S1P) and site-2 protease and releases a nuclear form to modulate gene expression. To search for new genes regulating cholesterol metabolism, we perform a genome-wide CRISPR/Cas9 knockout screen and find that partner of site-1 protease (POST1), encoded by C12ORF49, is critically involved in the SREBP signaling. Ablation of POST1 decreases the generation of nuclear SREBP and reduces the expression of SREBP target genes. POST1 binds S1P, which is synthesized as an inactive protease (form A) and becomes fully mature via a two-step autocatalytic process involving forms B'/B and C'/C. POST1 promotes the generation of the functional S1P-C'/C from S1P-B'/B (canonical cleavage) and, notably, from S1P-A directly (non-canonical cleavage) as well. This POST1-mediated S1P activation is also essential for the cleavages of other S1P substrates including ATF6, CREB3 family members and the α/ß-subunit precursor of N-acetylglucosamine-1-phosphotransferase. Together, we demonstrate that POST1 is a cofactor controlling S1P maturation and plays important roles in lipid homeostasis, unfolded protein response, lipoprotein metabolism and lysosome biogenesis.


Subject(s)
Membrane Proteins/metabolism , Signal Transduction , Sterol Regulatory Element Binding Proteins/metabolism , CRISPR-Cas Systems , HeLa Cells , Humans , Lipoproteins/biosynthesis , Lipoproteins/genetics , Lysosomes/genetics , Lysosomes/metabolism , Membrane Proteins/genetics , Sterol Regulatory Element Binding Proteins/genetics
14.
Cell Chem Biol ; 28(2): 169-179.e7, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33096051

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) are master transcriptional regulators of the mevalonate pathway and lipid metabolism and represent an attractive therapeutic target for lipid metabolic disorders. SREBPs are maintained in the endoplasmic reticulum (ER) in a tripartite complex with SREBP cleavage-activating protein (SCAP) and insulin-induced gene protein (INSIG). When new lipid synthesis is required, the SCAP-SREBP complex dissociates from INSIG and undergoes ER-to-Golgi transport where the N-terminal transcription factor domain is released by proteolysis. The mature transcription factor translocates to the nucleus and stimulates expression of the SREBP gene program. Previous studies showed that dipyridamole, a clinically prescribed phosphodiesterase (PDE) inhibitor, potentiated statin-induced tumor growth inhibition. Dipyridamole limited nuclear accumulation of SREBP, but the mechanism was not well resolved. In this study, we show that dipyridamole selectively blocks ER-to-Golgi movement of the SCAP-SREBP complex and that this is independent of its PDE inhibitory activity.


Subject(s)
Dipyridamole/pharmacology , Endoplasmic Reticulum/drug effects , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Lipogenesis/drug effects , Membrane Proteins/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Proteins/genetics
15.
Cell Mol Gastroenterol Hepatol ; 11(4): 949-971, 2021.
Article in English | MEDLINE | ID: mdl-33246135

ABSTRACT

BACKGROUND & AIMS: cAMP responsive element-binding protein 3 like 3 (CREB3L3) is a membrane-bound transcription factor involved in the maintenance of lipid metabolism in the liver and small intestine. CREB3L3 controls hepatic triglyceride and glucose metabolism by activating plasma fibroblast growth factor 21 (FGF21) and lipoprotein lipase. In this study, we intended to clarify its effect on atherosclerosis. METHODS: CREB3L3-deficifient, liver-specific CREB3L3 knockout, intestine-specific CREB3L3 knockout, both liver- and intestine-specific CREB3L3 knockout, and liver CREB3L3 transgenic mice were crossed with LDLR-/- mice. These mice were fed with a Western diet to develop atherosclerosis. RESULTS: CREB3L3 ablation in LDLR-/- mice exacerbated hyperlipidemia with accumulation of remnant APOB-containing lipoprotein. This led to the development of enhanced aortic atheroma formation, the extent of which was additive between liver- and intestine-specific deletion. Conversely, hepatic nuclear CREB3L3 overexpression markedly suppressed atherosclerosis with amelioration of hyperlipidemia. CREB3L3 directly up-regulates anti-atherogenic FGF21 and APOA4. In contrast, it antagonizes hepatic sterol regulatory element-binding protein (SREBP)-mediated lipogenic and cholesterogenic genes and regulates intestinal liver X receptor-regulated genes involved in the transport of cholesterol. CREB3L3 deficiency results in the accumulation of nuclear SREBP proteins. Because both transcriptional factors share the cleavage system for nuclear transactivation, full-length CREB3L3 and SREBPs in the endoplasmic reticulum (ER) functionally inhibit each other. CREB3L3 promotes the formation of the SREBP-insulin induced gene 1 complex to suppress SREBPs for ER-Golgi transport, resulting in ER retention and inhibition of proteolytic activation at the Golgi and vice versa. CONCLUSIONS: CREB3L3 has multi-potent protective effects against atherosclerosis owing to new mechanistic interaction between CREB3L3 and SREBPs under atherogenic conditions.


Subject(s)
Atherosclerosis/prevention & control , Cyclic AMP Response Element-Binding Protein/physiology , Gene Expression Regulation , Hyperlipidemias/prevention & control , Lipid Metabolism , Receptors, LDL/physiology , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Female , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sterol Regulatory Element Binding Proteins/genetics
16.
Sci Rep ; 10(1): 22244, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33335263

ABSTRACT

Effective drugs are needed for lung cancer, as this disease remains the leading cause of cancer-related deaths. Rexinoids are promising drug candidates for cancer therapy because of their ability to modulate genes involved in inflammation, cell proliferation or differentiation, and apoptosis through activation of the retinoid X receptor (RXR). The only currently FDA-approved rexinoid, bexarotene, is ineffective as a single agent for treating epithelial cancers and induces hypertriglyceridemia. Here, we used a previously validated screening paradigm to evaluate 23 novel rexinoids for biomarkers related to efficacy and safety. These biomarkers include suppression of inducible nitric oxide synthase (iNOS) and induction of sterol regulatory element-binding protein (SREBP). Because of its potent iNOS suppression, low SREBP induction, and activation of RXR, MSU-42011 was selected as our lead compound. We next used MSU-42011 to treat established tumors in a clinically relevant Kras-driven mouse model of lung cancer. KRAS is one of the most common driver mutations in human lung cancer and correlates with aggressive disease progression and poor patient prognosis. Ultrasound imaging was used to detect and monitor tumor development and growth over time in the lungs of the A/J mice. MSU-42011 markedly decreased the tumor number, size, and histopathology of lung tumors compared to the control and bexarotene groups. Histological sections of lung tumors in mice treated with MSU-42011 exhibited reduced cell density and fewer actively proliferating cells compared to the control and bexarotene-treated tumors. Although bexarotene significantly (p < 0.01) elevated plasma triglycerides and cholesterol, treatment with MSU-42011 did not increase these biomarkers, demonstrating a more favorable toxicity profile in vivo. The combination of MSU-42011 and carboplatin and paclitaxel reduced macrophages in the lung and increased activation markers of CD8+T cells compared to the control groups. Our results validate our screening paradigm for in vitro testing of novel rexinoids and demonstrate the potential for MSU-42011 to be developed for the treatment of KRAS-driven lung cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinogens , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Retinoid X Receptors/agonists , Tetrahydronaphthalenes/pharmacology , Animals , Anticarcinogenic Agents/chemistry , Apoptosis/drug effects , Bexarotene/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Immunohistochemistry , Immunomodulation/drug effects , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Mice , Molecular Structure , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Tetrahydronaphthalenes/chemistry , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
17.
BMC Genomics ; 21(1): 655, 2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32967608

ABSTRACT

BACKGROUND: The AMP-activated protein kinase (AMPK) is an intracellular fuel sensor for lipid and glucose metabolism. In addition to the short-term regulation of metabolic enzymes by phosphorylation, AMPK may also exert long-term effects on the transcription of downstream genes through the regulation of transcription factors and coactivators. In this study, RNA interference (RNAi) was conducted to investigate the effects of knockdown of TcAMPKα on lipid and carbohydrate metabolism in the red flour beetle, Tribolium castaneum, and the transcriptome profiles of dsTcAMPKα-injected and dsEGFP-injected beetles under normal conditions were compared by RNA-sequencing. RESULTS: RNAi-mediated suppression of TcAMPKα increased whole-body triglyceride (TG) level and the ratio between glucose and trehalose, as was confirmed by in vivo treatment with the AMPK-activating compound, 5-Aminoimidazole-4-carboxamide1-ß-D-ribofuranoside (AICAR). A total of 1184 differentially expressed genes (DEGs) were identified between dsTcAMPKα-injected and dsEGFP-injected beetles. These include genes involved in lipid and carbohydrate metabolism as well as insulin/insulin-like growth factor signaling (IIS). Real-time quantitative polymerase chain reaction analysis confirmed the differential expression of selected genes. Interestingly, metabolism-related transcription factors such as sterol regulatory element-binding protein 1 (SREBP1) and carbohydrate response element-binding protein (ChREBP) were also significantly upregulated in dsTcAMPKα-injected beetles. CONCLUSIONS: AMPK plays a critical role in the regulation of beetle metabolism. The findings of DEGs involved in lipid and carbohydrate metabolism provide valuable insight into the role of AMPK signaling in the transcriptional regulation of insect metabolism.


Subject(s)
Insect Proteins/genetics , Metabolome , Protein Kinases/genetics , Transcriptome , Tribolium/genetics , AMP-Activated Protein Kinase Kinases , Animals , Glucose/metabolism , Insect Proteins/metabolism , Insulin/metabolism , Protein Kinases/metabolism , Somatomedins/genetics , Somatomedins/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Trehalose/metabolism , Tribolium/metabolism , Triglycerides/metabolism
18.
Life Sci ; 258: 118030, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32739470

ABSTRACT

The risk of atherosclerosis (AS) ascends among post-menopausal women, while current hormone replacement therapy exerts several adverse effects. Alisol B 23-acetate (AB23A), a tetracyclic triterpenoid isolated from the rhizome of Alisma orientale, was reported to show multiple physiological activities, including regulating lipid metabolism. According to molecular docking analysis, it was predicted to bind with estrogen receptor α (ERα). In this study, we aimed to observe the effect of AB23A on preventing post-menopausal AS and explore whether the mechanism was mediated by ERα. In vitro, free fatty acid (FFA) was applied to induce the abnormal lipid metabolism of L02 cells. In vivo, the ApoE-/- mice were ovariectomized to mimic the cessation of estrogen. The high-fat diet was also given to induce post-menopausal AS. We demonstrated AB23A attenuated the accumulation of total cholesterol and triglyceride induced by free fatty acids in hepatocytes. In high-fat diet-ovariectomy-treated ApoE-/- mice, AB23A eliminated lipids in blood and liver. AB23A not only reduced the synthesis of proprotein convertase subtilisin/kexin type 9 (PCSK9) through sterol-regulatory element binding proteins (SREBPs) but also suppressed the secretion of PCSK9 through silent information regulator 1 (SIRT1). Notably, AB23A promoted the expression of ERα in vivo and in vitro. The both ERα inhibitor and ERα siRNA were also applied in confirming whether the hepatic protective effect of AB23A was mediated by ERα. We found that AB23A significantly promoted the expression of ERα. AB23A could inhibit the synthesis and secretion of PCSK9 through ERα, lower the accumulation of triglyceride and cholesterol, and prevent post-menopausal AS.


Subject(s)
Atherosclerosis/pathology , Cholestenones/pharmacology , Estrogen Receptor alpha/metabolism , Lipid Metabolism/drug effects , Postmenopause/drug effects , Animals , Atherosclerosis/genetics , Cell Line , Cell Survival/drug effects , Cholestenones/chemistry , Diet, High-Fat , Fatty Acids/metabolism , Female , Lipoproteins, LDL/metabolism , Mice , Ovariectomy , Promoter Regions, Genetic/genetics , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sirtuin 1/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Up-Regulation/drug effects
19.
Biofactors ; 46(5): 777-787, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32663368

ABSTRACT

Butein, a flavonoid found in annatto seeds and lacquer trees, may be used for many health benefits, including the prevention of obesity. However, its anti-obesity effects are not completely understood; in particular, the effects of butein on the regulation of lipid metabolism have not been explained. Thus, the goal of the current study was to determine the effects of butein on lipid metabolism in Caenorhabditis elegans, which is a multi-organ nematode used as an animal model in obesity research. Butein at 70 µM reduced triglyceride content by 27% compared to the control without altering food intake and energy expenditure. The reduced triglyceride content by butein was associated with the downregulation of sbp-1, fasn-1, and fat-7, the lipogenesis-related homologs of sterol regulatory element-binding proteins, fatty acid synthase and stearoyl-CoA desaturase, respectively. Furthermore, fat-7 and skn-1, a homolog of nuclear respiratory factors, were identified as genetic requirements for butein's effects on triglyceride content in C. elegans. The effects of butein on sbp-1 and fasn-1 were dependent on skn-1, but the downregulation of fat-7 was independent of skn-1. These results suggest that the inhibitory effects of butein on lipogenesis are via SKN-1- and FAT-7-dependent pathways in C. elegans.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Chalcones/pharmacology , DNA-Binding Proteins/genetics , Obesity/drug therapy , Stearoyl-CoA Desaturase/genetics , Transcription Factors/genetics , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Obesity/genetics , Sterol Regulatory Element Binding Proteins/genetics , Triglycerides/metabolism
20.
Article in English | MEDLINE | ID: mdl-32544537

ABSTRACT

The sterol regulatory element binding proteins (SREBPs) transcription factors family, which regulate the expression of genes involved in cellular lipid metabolism and homeostasis, have recently been implicated in various physiological and pathophysiological processes such as immune regulation and inflammation in vertebrates. Consistent with other invertebrates, we identified a single SREBP ortholog in Penaeus vannamei (designated PvSREBP) with transcripts ubiquitously expressed in tissues and induced by lipopolysaccharide (LPS), Vibrio parahaemolyticus and Streptococcus iniae. In vivo RNA interference (RNAi) of PvSREBP attenuated the expression of several fatty acid metabolism-related genes (i.e., cyclooxygenase (PvCOX), lipoxygenase (PvLOX), fatty acid binding protein (PvFABP) and fatty acid synthase (PvFASN)), which consequently decreased the levels of total polyunsaturated fatty acids (ΣPUFAs). In addition, PvSREBP silencing decreased transcript levels of several immune-related genes such as hemocyanin (PvHMC) and trypsin (PvTrypsin), as well as genes encoding for heat-shock proteins (i.e., PvHSP60, PvHSP70 and PvHSP90). Moreover, in silico analysis revealed the presence of SREBP binding motifs on the promoters of most of the dysregulated genes, while shrimp depleted of PvSREBP were more susceptible to V. parahaemolyticus infection. Collectively, we demonstrated the involvement of shrimp SREBP in fatty acids metabolism and immune response, and propose that PvSREBP and PvHMC modulate each other through a feedback mechanism to establish homeostasis. The current study is the first to show the dual role of SREBP in fatty acid metabolism and immune response in invertebrates and crustaceans.


Subject(s)
Fatty Acids/metabolism , Penaeidae , Sterol Regulatory Element Binding Proteins , Animals , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Arthropod Proteins/metabolism , Hemocyanins/genetics , Hemocytes/immunology , Hepatopancreas/immunology , Lipopolysaccharides/pharmacology , Penaeidae/immunology , Penaeidae/metabolism , Penaeidae/microbiology , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/immunology , Sterol Regulatory Element Binding Proteins/metabolism , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Streptococcal Infections/veterinary , Streptococcus iniae , Trypsin/genetics , Vibrio Infections/immunology , Vibrio Infections/metabolism , Vibrio Infections/veterinary , Vibrio parahaemolyticus
SELECTION OF CITATIONS
SEARCH DETAIL
...