Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Lipid Res ; 62: 100074, 2021.
Article in English | MEDLINE | ID: mdl-33872606

ABSTRACT

Cytosolic sulfotransferases (SULTs) catalyze the transfer of a sulfonate group from the cofactor 3'-phosphoadenosine 5'-phosphosulfate to a hydroxyl (OH) containing substrate and play a critical role in the homeostasis of endogenous compounds, including hormones, neurotransmitters, and bile acids. In human, SULT2A1 sulfonates the 3-OH of bile acids; however, bile acid metabolism in mouse is dependent on a 7α-OH sulfonating SULT2A8 via unknown molecular mechanisms. In this study, the crystal structure of SULT2A8 in complex with adenosine 3',5'-diphosphate and cholic acid was resolved at a resolution of 2.5 Å. Structural comparison with human SULT2A1 reveals different conformations of substrate binding loops. In addition, SULT2A8 possesses a unique substrate binding mode that positions the target 7α-OH of the bile acid close to the catalytic site. Furthermore, mapping of the critical residues by mutagenesis and enzyme activity assays further highlighted the importance of Lys44 and His48 for enzyme catalysis and Glu237 in loop 3 on substrate binding and stabilization. In addition, limited proteolysis and thermal shift assays suggested that the cofactor and substrates have protective roles in stabilizing SULT2A8 protein. Together, the findings unveil the structural basis of bile acid sulfonation targeting 7α-OH and shed light on the functional diversity of bile acid metabolism across species.


Subject(s)
Bile Acids and Salts
2.
Transgenic Res ; 29(5-6): 499-510, 2020 12.
Article in English | MEDLINE | ID: mdl-33029720

ABSTRACT

SULT2A8 is a male-predominant and liver-specific mouse cytosolic sulfotransferase (SULT) that sulfonates 7α-hydroxyl (7α-OH) bile acids in vitro. Sulfonation regulates bile acid homeostasis, which in turn regulates cholesterol and energy metabolism. Using the Sult2a8-heterozygous (HT) mouse model created earlier in our laboratory, we aimed to investigate the physiological role of SULT2A8 in sulfonating 7α-OH bile acids and its impact on energy metabolism in vivo under both fed and energy-deprivation conditions. Disruption of one allele of the Sult2a8 gene in male HT mice resulted in losing ~ 50% of the 7α-OH sulfonating activity compared to wild-type (WT) control, but no significant change in female HT mice. Under the fed condition comparing the levels of hepatic and biliary bile acids as well as plasma/serum energy metabolites, HT mice displayed a profile similar to that of WT mice, suggesting that the Sult2a8-haplodeficient mice conducted normal energy metabolism. However, after 48-h fasting, a significant decrease in plasma cholesterol level was found in male HT mice but without any significant reduction in female HT mice. Of interest, in male Sult2a8-haplodeficient mice, an increase of the hepatic taurine-conjugated cholic acid level was noted but no noticeable change in other tested bile acids after fasting. Taken together, SULT2A8 is a male-specific and key hepatic SULT in metabolizing 7α-OH primary bile acids. During energy deprivation, SULT2A8 is required to maintain the bile acid and cholesterol metabolism, suggesting SULT is a potential therapeutic target for controlling metabolic diseases.


Subject(s)
Cholesterol/blood , Liver/metabolism , Sulfotransferases/metabolism , Taurocholic Acid/metabolism , Animals , Bile Acids and Salts/metabolism , Energy Metabolism , Fasting , Haploinsufficiency/genetics , Heterozygote , Male , Mice, Mutant Strains , Sulfotransferases/genetics
3.
J Lipid Res ; 58(6): 1114-1131, 2017 06.
Article in English | MEDLINE | ID: mdl-28442498

ABSTRACT

PPARα has been known to play a pivotal role in orchestrating lipid, glucose, and amino acid metabolism via transcriptional regulation of its target gene expression during energy deprivation. Recent evidence has also suggested that PPARα is involved in bile acid metabolism, but how PPARα modulates the homeostasis of bile acids during fasting is still not clear. In a mechanistic study aiming to dissect the spectrum of PPARα target genes involved in metabolic response to fasting, we identified a novel mouse gene (herein named mL-STL for mouse liver-sulfotransferase-like) that shared extensive homology with the Sult2a subfamily of a superfamily of cytosolic sulfotransferases, implying its potential function in sulfonation. The mL-STL gene expressed predominantly in liver in fed state, but PPARα was required to sustain its expression during fasting, suggesting a critical role of PPARα in regulating the mL-STL-mediated sulfonation during fasting. Functional studies using recombinant His-tagged mL-STL protein revealed its narrow sulfonating activities toward 7α-hydroxyl primary bile acids, including cholic acid, chenodeoxycholic acid, and α-muricholic acid, and thus suggesting that mL-STL may be the major hepatic bile acid sulfonating enzyme in mice. Together, these studies identified a novel PPARα-dependent gene and uncovered a new role of PPARα as being an essential regulator in bile acid biotransformation via sulfonation during fasting.


Subject(s)
Bile Acids and Salts/metabolism , Cytosol/enzymology , PPAR alpha/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biocatalysis , Biotransformation , Cloning, Molecular , DNA, Complementary/genetics , Down-Regulation , Fasting/metabolism , Liver/cytology , Male , Mice , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity , Sulfotransferases/chemistry
4.
Arterioscler Thromb Vasc Biol ; 34(4): 830-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24482374

ABSTRACT

OBJECTIVE: 5' Adenosine monophosphate-activated protein kinase (AMPK) interacts with peroxisome proliferator-activated receptor δ (PPARδ) to induce gene expression synergistically, whereas the activation of AMPK inhibits endoplasmic reticulum (ER) stress. Whether the vascular benefits of antidiabetic drug metformin (AMPK activator) in diabetes mellitus and obesity is mediated by PPARδ remains unknown. We aim to investigate whether PPARδ is crucial for metformin in ameliorating ER stress and endothelial dysfunction induced by high-fat diet. APPROACH AND RESULTS: Acetylcholine-induced endothelium-dependent relaxation in aortae was measured on wire myograph. ER stress markers were determined by Western blotting. Superoxide production in mouse aortae and NO generation in mouse aortic endothelial cells were assessed by fluorescence imaging. Endothelium-dependent relaxation was impaired and ER stress markers and superoxide level were elevated in aortae from high-fat diet-induced obese mice compared with lean mice. These effects of high-fat diet were reversed by oral treatment with metformin in diet-induced obese PPARδ wild-type mice but not in diet-induced obese PPARδ knockout littermates. Metformin and PPARδ agonist GW1516 reversed tunicamycin (ER stress inducer)-induced ER stress, oxidative stress, and impairment of endothelium-dependent relaxation in mouse aortae as well as NO production in mouse aortic endothelial cells. Effects of metformin were abolished by cotreatment of GSK0660 (PPARδ antagonist), whereas effects of GW1516 were unaffected by compound C (AMPK inhibitor). CONCLUSIONS: Metformin restores endothelial function through inhibiting ER stress and oxidative stress and increasing NO bioavailability on activation of AMPK/PPARδ pathway in obese diabetic mice.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus/drug therapy , Diet, High-Fat , Endoplasmic Reticulum Stress/drug effects , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Obesity/drug therapy , PPAR gamma/agonists , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Diabetes Mellitus/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Enzyme Activation , Mice , Mice, Knockout , Nitric Oxide/metabolism , Obesity/enzymology , Obesity/genetics , Obesity/physiopathology , Oxidative Stress/drug effects , PPAR gamma/deficiency , PPAR gamma/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Superoxides/metabolism , Time Factors , Vasodilation/drug effects , Vasodilator Agents/pharmacology
5.
Diabetes ; 61(12): 3285-93, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22933110

ABSTRACT

Recent evidence highlights the therapeutic potential of peroxisome proliferator-activated receptor-δ (PPARδ) agonists to increase insulin sensitivity in diabetes. However, the role of PPARδ in regulating vascular function is incompletely characterized. We investigate whether PPARδ activation improves endothelial function in diabetic and obese mice. PPARδ knockout (KO) and wild-type (WT) mice fed with high-fat diet and db/db mice were used as diabetic mouse models, compared with PPARδ KO and WT mice on normal diet and db/m(+) mice. Endothelium-dependent relaxation (EDR) was measured by wire myograph. Flow-mediated vasodilatation (FMD) was measured by pressure myograph. Nitric oxide (NO) production was examined in primary endothelial cells from mouse aortae. PPARδ agonist GW1516 restored EDRs in mouse aortae under high-glucose conditions or in db/db mouse aortae ex vivo. After oral treatment with GW1516, EDRs in aortae and FMDs in mesenteric resistance arteries were improved in obese mice in a PPARδ-specific manner. The effects of GW1516 on endothelial function were mediated through phosphatidylinositol 3-kinase (PI3K) and Akt with a subsequent increase of endothelial nitric oxide synthase (eNOS) activity and NO production. The current study demonstrates an endothelial-protective effect of PPARδ agonists in diabetic mice through PI3K/Akt/eNOS signaling, suggesting the therapeutic potential of PPARδ agonists for diabetic vasculopathy.


Subject(s)
Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , PPAR delta/metabolism , Thiazoles/therapeutic use , Animals , Diabetes Mellitus/genetics , Diet, High-Fat/adverse effects , Endothelium/drug effects , Endothelium/metabolism , Endothelium/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , PPAR delta/agonists , PPAR delta/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
6.
Protein Expr Purif ; 68(2): 190-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19683057

ABSTRACT

Millions of candidate clones are commonly obtained following rounds of phage-displayed antibody library panning, and expression of those selected single-chain variable fragment (scFv) is required for secondary functional screening to identify positive clones. Large scale functional screening is often hampered by the time-consuming and labor-intensive subcloning of those candidate scFv clones into a bacterial expression vector carrying an affinity tag for scFv purification and detection. To overcome the limitations and to develop a multiplex approach, an improved hexahistidine tag phagemid vector was constructed for one-step scFv expression and purification. By using hexahistidine as an affinity tag, soluble scFvs can be rapidly and cost-effectively captured from Escherichia coli periplasmic extracts. For proof-of-concept, feasibility of the improved phagemid vector was examined against two scFvs, L17E4d targeting a cell surface antigen and L18Hh5 recognizing a monoclonal antibody (mAb). Using 1 ml of Ni-NTA agarose, 0.2-0.5 mg of soluble scFv was obtained from 1 L of bacteria culture, and the purified scFvs bound specifically to their target antigens with high affinity. Moreover, using two randomly selected hapten-specific scFv phage clones, it was demonstrated that the display of scFvs on phage surface was not affected by the hexahistidine affinity tag. These results suggest the improved phagemid vector allows the shuttle of phage-displayed antibody library panning and functional scFv production. Importantly, the improved phagemid vector can be easily adapted for multiplex screening.


Subject(s)
Genetic Vectors/chemistry , Histidine/metabolism , Oligopeptides/metabolism , Recombinant Fusion Proteins/isolation & purification , Single-Chain Antibodies/isolation & purification , Base Sequence , Chromatography, Affinity/methods , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Flow Cytometry , Histidine/chemistry , Histidine/genetics , Humans , Molecular Sequence Data , Oligopeptides/chemistry , Oligopeptides/genetics , Peptide Library , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
7.
Cancer Invest ; 26(7): 708-17, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18608213

ABSTRACT

Farnesol (FOH) and geranylgeraniol (GGOH) possess anti-tumor potential, while peroxisome proliferator-activated receptor gamma (PPARgamma) has exhibited modulating effects in colorectal cancers. We investigated the anti-carcinogenic effects of these isoprenols in HT-29 and HCT116 colon cancer cells and PPARgamma involvement. Results indicate that the FOH- and GGOH-induced apoptosis involve caspase 3 activation, PARP cleavage, nuclear chromatin condensation, down-regulation of Bcl-x(L) and survivin expression, with increased PPARgamma promoter activity. Pretreatment of the PPARgamma antagonist GW9662 reduces FOH-induced growth inhibition and the associated PARP cleavage. We conclude that PPARgamma activation is essential to elicit the anti-carcinogenic action of herbal isoprenols in colonic cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/metabolism , Diterpenes/pharmacology , Farnesol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , PPAR gamma/agonists , Transcriptional Activation/drug effects , Anilides/pharmacology , Caspase 3/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Enzyme Activation , HCT116 Cells , HT29 Cells , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Promoter Regions, Genetic/drug effects , Survivin , Time Factors , bcl-X Protein/metabolism
8.
Int J Biochem Cell Biol ; 40(9): 1775-91, 2008.
Article in English | MEDLINE | ID: mdl-18289917

ABSTRACT

The peroxisome proliferator-activated receptor alpha (PPARalpha) has been known to play a pivotal role in maintaining the energy balance during fasting; however, the battery of PPARalpha target genes involved in this metabolic response is still not fully characterized. Here, we report the identification and characterization of Ppsig (for PPARalpha-regulated and starvation-induced gene) with unknown biological function from mouse liver. Multiple Ppsig cDNAs which differed in the 3'-untranslated regions were identified. The open reading frame of Ppsig cDNA is 1830 bp which encodes a protein of 67.33 kDa. Ppsig contains 11 exons spanning at least 10 kb. Although the exact biological function of Ppsig is still not known, we found that Ppsig mRNA transcript was dramatically up-regulated during 72 h fasting and following treatment with a potent PPARalpha agonist, in a tissue-specific and PPARalpha-dependent manner. A functional peroxisome proliferator-response element was found in the intron 1 of Ppsig, thus confirming that Ppsig is a novel direct mouse PPARalpha target gene. This finding might help in elucidating the transcriptional regulatory mechanism of Ppsig in the cellular response to fasting.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , PPAR alpha/metabolism , Starvation/genetics , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Fasting/metabolism , Genomics , Humans , Introns/genetics , Lipid Metabolism , Male , Mice , Mice, Knockout , Molecular Sequence Data , Oxidoreductases Acting on CH-CH Group Donors/biosynthesis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Up-Regulation
9.
Toxicology ; 228(2-3): 310-22, 2006 Dec 07.
Article in English | MEDLINE | ID: mdl-17084009

ABSTRACT

Previous study using Cyp2e1-null mice showed that Cyp2e1 is required in CCl(4)-induced liver injury at 24h, what remains unclear are the temporal changes in liver damage and the spectrum of genes involved in this process. We investigated the time-dependent liver changes that occurred at morphological, histopathological, biochemical and molecular levels in both Cyp2e1(+/+) and Cyp2e1(-/-) mice after treating with either corn oil or CCl(4) (1 ml/kg) for 2, 6, 12, 24 and 48 h. A pale orange colored liver, indicative of fatty infiltration, was observed in Cyp2e1(+/+) mice treated with CCl(4) for 24 and 48 h, while the Cyp2e1(+/+) mice treated with corn oil and Cyp2e1(-/-) mice treated with either corn oil or CCl(4) showed normal reddish brown colored liver. Ballooned hepatocytes with multiple vacuoles in their cytoplasm were observed in the livers of Cyp2e1(+/+) mice 24 and 48 h after treating with CCl(4). The levels of serum alanine aminotransferase and aspartate aminotransferase, markers for liver injury, were significantly higher at 12h, peaked at 24h and gradually decreased at 48 h after CCl(4) intoxication. In contrast, this kind of damage was not apparent in the Cyp2e1(-/-) mice treated with CCl(4). Altered expressions of genes related to liver cirrhosis, apoptosis, oxidative stress, xenobiotic detoxification, lipid metabolism, chemsensory signaling or tumorigenesis, structural organization, regeneration and inflammatory response were identified, and the time-dependent changes in expression of these genes were varied. Overall, the present study provides insights into the mechanism of CCl(4)-induced hepatotoxicity in animal models.


Subject(s)
Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/physiology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Blotting, Northern , Carbon Tetrachloride Poisoning/enzymology , Chemical and Drug Induced Liver Injury/enzymology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Gene Expression/drug effects , Liver/enzymology , Liver/pathology , Mice , Mice, Knockout , Oxidative Stress/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Biochem Pharmacol ; 71(3): 319-37, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16336942

ABSTRACT

In the present study, a phage-displayed random peptide library was used to identify surrogate peptide ligands for orphan GPCR mas. Sequence analysis of the isolated phage clones indicated a selective enrichment of some peptide sequences. Moreover, multiple alignments of the isolated phage clones gave two conserved peptide motifs from which we synthesized peptide MBP7 for further evaluation. Characterization of the representative phage clones and the synthetic peptide MBP7 by immunocytochemistry revealed a strong punctate cell surface staining in CHO cells expressing mas-GFP fusion protein. The isolated phage clones and synthetic peptide MBP7 induced mas internalization in a stable CHO cell clone (MC0M80) over-expressing mas. In addition, MBP7-stimulated phospholipase C activity and intracellular calcium mobilization in these same cells. In summary, we have demonstrated a systematic approach to derive surrogate peptide ligands for orphan GPCRs. With this technique, we have identified two conserved peptide motifs which allow us to identify potential protein partners for mas, and have generated a peptide agonist MBP7 which will be invaluable for functional characterization of the mas oncogene.


Subject(s)
Membrane Proteins/metabolism , Peptide Library , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , CHO Cells , Calcium/metabolism , Cloning, Molecular , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay , Inositol Phosphates/metabolism , Ligands , Microscopy, Confocal , Molecular Sequence Data , Protein Binding , Proto-Oncogene Mas , Transfection
11.
Methods Enzymol ; 357: 214-40, 2002.
Article in English | MEDLINE | ID: mdl-12424913

ABSTRACT

In conclusion, we have applied the fluorescent differential method and the PPAR alpha-null mouse model for the rapid isolation of expression tags of PPAR alpha target genes that are involved in the action of peroxisome proliferators and in the regulation of lipid homeostasis under energy deprivation. Identification of a wide spectrum of PPAR alpha target genes will provide new insights into the diverse cellular pathways regulated by these receptor, and this information will be critical for understanding the complicated biological interactions among members of the PPAR alpha target genes. With the recent technological advancement, a newer method, such as DNA microarray, has emerged in the identification of differential gene expressions. This new DNA microarray method, in conjunction with the differential display method, is the first important step toward understanding the molecular mechanisms of gene interactions in any biological systems and can speed up the search for differential gene expressions.


Subject(s)
Fluorescence , Gene Expression Profiling/methods , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Animals , DNA/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Fasting , Genotype , Lipid Metabolism , Liver/drug effects , Liver/physiology , Male , Mice , Mice, Knockout , Peroxisome Proliferators/pharmacology , Polymerase Chain Reaction/methods , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...