Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Biol Chem ; 299(4): 103050, 2023 04.
Article in English | MEDLINE | ID: mdl-36813233

ABSTRACT

Consecutive oxygenation of arachidonic acid by 5-lipoxygenase and cyclooxygenase-2 yields the hemiketal eicosanoids, HKE2 and HKD2. Hemiketals stimulate angiogenesis by inducing endothelial cell tubulogenesis in culture; however, how this process is regulated has not been determined. Here, we identify vascular endothelial growth factor receptor 2 (VEGFR2) as a mediator of HKE2-induced angiogenesis in vitro and in vivo. We found that HKE2 treatment of human umbilical vein endothelial cells dose-dependently increased the phosphorylation of VEGFR2 and the downstream kinases ERK and Akt that mediated endothelial cell tubulogenesis. In vivo, HKE2 induced the growth of blood vessels into polyacetal sponges implanted in mice. HKE2-mediated effects in vitro and in vivo were blocked by the VEGFR2 inhibitor vatalanib, indicating that the pro-angiogenic effect of HKE2 was mediated by VEGFR2. HKE2 covalently bound and inhibited PTP1B, a protein tyrosine phosphatase that dephosphorylates VEGFR2, thereby providing a possible molecular mechanism for how HKE2 induced pro-angiogenic signaling. In summary, our studies indicate that biosynthetic cross-over of the 5-lipoxygenase and cyclooxygenase-2 pathways gives rise to a potent lipid autacoid that regulates endothelial cell function in vitro and in vivo. These findings suggest that common drugs targeting the arachidonic acid pathway could prove useful in antiangiogenic therapy.


Subject(s)
Arachidonate 5-Lipoxygenase , Vascular Endothelial Growth Factor Receptor-2 , Mice , Humans , Animals , Cyclooxygenase 2/metabolism , Arachidonic Acid , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor A/metabolism , Neovascularization, Physiologic , Human Umbilical Vein Endothelial Cells/metabolism , Angiogenesis Inhibitors/pharmacology , Cell Movement , Cell Proliferation
2.
J Immunol ; 209(4): 796-805, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35896340

ABSTRACT

Colonization by Helicobacter pylori is associated with gastric diseases, ranging from superficial gastritis to more severe pathologies, including intestinal metaplasia and adenocarcinoma. The interplay of the host response and the pathogen affect the outcome of disease. One major component of the mucosal response to H. pylori is the activation of a strong but inefficient immune response that fails to control the infection and frequently causes tissue damage. We have shown that polyamines can regulate H. pylori-induced inflammation. Chemical inhibition of ornithine decarboxylase (ODC), which generates the polyamine putrescine from l-ornithine, reduces gastritis in mice and adenocarcinoma incidence in gerbils infected with H. pylori However, we have also demonstrated that Odc deletion in myeloid cells enhances M1 macrophage activation and gastritis. Here we used a genetic approach to assess the specific role of gastric epithelial ODC during H. pylori infection. Specific deletion of the gene encoding for ODC in gastric epithelial cells reduces gastritis, attenuates epithelial proliferation, alters the metabolome, and downregulates the expression of immune mediators induced by H. pylori Inhibition of ODC activity or ODC knockdown in human gastric epithelial cells dampens H. pylori-induced NF-κB activation, CXCL8 mRNA expression, and IL-8 production. Chronic inflammation is a major risk factor for the progression to more severe pathologies associated with H. pylori infection, and we now show that epithelial ODC plays an important role in mediating this inflammatory response.


Subject(s)
Adenocarcinoma , Gastritis , Helicobacter Infections , Helicobacter pylori , Adenocarcinoma/metabolism , Animals , Epithelial Cells/metabolism , Gastric Mucosa/pathology , Helicobacter pylori/metabolism , Humans , Inflammation/metabolism , Mice , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism
3.
Gastroenterology ; 162(3): 813-827.e8, 2022 03.
Article in English | MEDLINE | ID: mdl-34767785

ABSTRACT

BACKGROUND & AIMS: Because inflammatory bowel disease is increasing worldwide and can lead to colitis-associated carcinoma (CAC), new interventions are needed. We have shown that spermine oxidase (SMOX), which generates spermidine (Spd), regulates colitis. Here we determined whether Spd treatment reduces colitis and carcinogenesis. METHODS: SMOX was quantified in human colitis and associated dysplasia using quantitative reverse-transcription polymerase chain reaction and immunohistochemistry. We used wild-type (WT) and Smox-/- C57BL/6 mice treated with dextran sulfate sodium (DSS) or azoxymethane (AOM)-DSS as models of colitis and CAC, respectively. Mice with epithelial-specific deletion of Apc were used as a model of sporadic colon cancer. Animals were supplemented or not with Spd in the drinking water. Colonic polyamines, inflammation, tumorigenesis, transcriptomes, and microbiomes were assessed. RESULTS: SMOX messenger RNA levels were decreased in human ulcerative colitis tissues and inversely correlated with disease activity, and SMOX protein was reduced in colitis-associated dysplasia. DSS colitis and AOM-DSS-induced dysplasia and tumorigenesis were worsened in Smox-/- vs WT mice and improved in both genotypes with Spd. Tumor development caused by Apc deletion was also reduced by Spd. Smox deletion and AOM-DSS treatment were both strongly associated with increased expression of α-defensins, which was reduced by Spd. A shift in the microbiome, with reduced abundance of Prevotella and increased Proteobacteria and Deferribacteres, occurred in Smox-/- mice and was reversed with Spd. CONCLUSIONS: Loss of SMOX is associated with exacerbated colitis and CAC, increased α-defensin expression, and dysbiosis of the microbiome. Spd supplementation reverses these phenotypes, indicating that it has potential as an adjunctive treatment for colitis and chemopreventive for colon carcinogenesis.


Subject(s)
Carcinogenesis/drug effects , Carcinogenesis/genetics , Colitis/genetics , Colonic Neoplasms/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Spermidine/therapeutic use , Adenomatous Polyposis Coli Protein/genetics , Animals , Azoxymethane , Colitis/chemically induced , Colitis/enzymology , Colitis/prevention & control , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Colon/enzymology , Colon/pathology , Colonic Neoplasms/prevention & control , Dextran Sulfate , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Male , Mice , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Precancerous Conditions/enzymology , Protective Factors , RNA, Messenger/metabolism , Severity of Illness Index , Spermidine/metabolism , Spermidine/pharmacology , Weight Loss/drug effects , alpha-Defensins/genetics , alpha-Defensins/metabolism , Polyamine Oxidase
4.
Biochem Biophys Res Commun ; 595: 41-46, 2022 03 05.
Article in English | MEDLINE | ID: mdl-35093639

ABSTRACT

Curcumin is a yellow pigment in turmeric (Curcuma longa) with various physiological effects in the body. To elucidate the molecular mechanisms by which bioactive compounds exert their function, identification of their molecular targets is crucial. In this study, we show that curcumin activates G protein-coupled receptor 97 (GPR97). Curcumin dose-dependently activated serum-response element-, but not serum-response factor-response element-, nuclear factor of activated T-cell-response element-, or cAMP-response element-, mediated transcription in cells overexpressed with GPR97. The structure-activity relationship indicated that (i) the double-bonds of the central 7-carbon chain were essential for activation; (ii) a methoxy group on the aromatic ring was required for maximal activity; (iii) the addition of glucuronic acid moiety or a methoxy group to the aromatic ring, but not the methylation of the aromatic p-hydroxy group, eliminated the activity; (iv) the stability of curcumin would be related to receptor activation. Both mutant GPR97(T250A) lacking the cleavage at GPCR proteolysis site and mutant GPR97(ΔN) lacking the N-terminal extracellular region were activated by curcumin and its related compounds similar to wild-type GPR97. In contrast, the synthetic glucocorticoid beclomethasone dipropionate and l-Phe activated wild-type GPR97 and GPR97(T250A), but not GPR97(ΔN). Moreover, curcumin exerted an additive effect on the activation of wild-type GPR97 with beclomethasone dipropionate, but not with l-Phe. Taken together, these results indicate that curcumin activates GPR97 coupled to Gi/Go subunit, and suggest that curcumin and glucocorticoid activate GPR97 in a different manner.


Subject(s)
Beclomethasone/pharmacology , Curcumin/pharmacology , Gene Expression Regulation/drug effects , Receptors, G-Protein-Coupled/genetics , Beclomethasone/chemistry , Curcuma/chemistry , Curcumin/chemistry , Curcumin/metabolism , Glucocorticoids/chemistry , Glucocorticoids/pharmacology , HEK293 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Molecular Structure , Mutation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Receptors, G-Protein-Coupled/metabolism , Response Elements/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Structure-Activity Relationship
5.
Proc Natl Acad Sci U S A ; 116(11): 5077-5085, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30804204

ABSTRACT

Infection by Helicobacter pylori is the primary cause of gastric adenocarcinoma. The most potent H. pylori virulence factor is cytotoxin-associated gene A (CagA), which is translocated by a type 4 secretion system (T4SS) into gastric epithelial cells and activates oncogenic signaling pathways. The gene cagY encodes for a key component of the T4SS and can undergo gene rearrangements. We have shown that the cancer chemopreventive agent α-difluoromethylornithine (DFMO), known to inhibit the enzyme ornithine decarboxylase, reduces H. pylori-mediated gastric cancer incidence in Mongolian gerbils. In the present study, we questioned whether DFMO might directly affect H. pylori pathogenicity. We show that H. pylori output strains isolated from gerbils treated with DFMO exhibit reduced ability to translocate CagA in gastric epithelial cells. Further, we frequently detected genomic modifications in the middle repeat region of the cagY gene of output strains from DFMO-treated animals, which were associated with alterations in the CagY protein. Gerbils did not develop carcinoma when infected with a DFMO output strain containing rearranged cagY or the parental strain in which the wild-type cagY was replaced by cagY with DFMO-induced rearrangements. Lastly, we demonstrate that in vitro treatment of H. pylori by DFMO induces oxidative DNA damage, expression of the DNA repair enzyme MutS2, and mutations in cagY, demonstrating that DFMO directly affects genomic stability. Deletion of mutS2 abrogated the ability of DFMO to induce cagY rearrangements directly. In conclusion, DFMO-induced oxidative stress in H. pylori leads to genomic alterations and attenuates virulence.


Subject(s)
Bacterial Proteins/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Eflornithine/pharmacology , Helicobacter pylori/genetics , Mutation/genetics , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Animals , DNA Damage , Gene Deletion , Gene Rearrangement , Gerbillinae , Helicobacter pylori/drug effects , Helicobacter pylori/pathogenicity , Male , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Virulence
6.
Proc Natl Acad Sci U S A ; 114(5): E751-E760, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096401

ABSTRACT

Macrophage activation is a critical step in host responses during bacterial infections. Ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine metabolism, has been well studied in epithelial cells and is known to have essential roles in many different cellular functions. However, its role in regulating macrophage function during bacterial infections is not well characterized. We demonstrate that macrophage-derived ODC is a critical regulator of M1 macrophage activation during both Helicobacter pylori and Citrobacter rodentium infection. Myeloid-specific Odc deletion significantly increased gastric and colonic inflammation, respectively, and enhanced M1 activation. Add-back of putrescine, the product of ODC, reversed the increased macrophage activation, indicating that ODC and putrescine are regulators of macrophage function. Odc-deficient macrophages had increased histone 3, lysine 4 (H3K4) monomethylation, and H3K9 acetylation, accompanied by decreased H3K9 di/trimethylation both in vivo and ex vivo in primary macrophages. These alterations in chromatin structure directly resulted in up-regulated gene transcription, especially M1 gene expression. Thus, ODC in macrophages tempers antimicrobial, M1 macrophage responses during bacterial infections through histone modifications and altered euchromatin formation, leading to the persistence and pathogenesis of these organisms.


Subject(s)
Enterobacteriaceae Infections/immunology , Helicobacter Infections/immunology , Histones/metabolism , Macrophages/immunology , Ornithine Decarboxylase/immunology , Animals , Cell Line , Citrobacter rodentium , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/pathology , Cytokines/immunology , Enterobacteriaceae Infections/pathology , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastritis/immunology , Gastritis/pathology , Helicobacter Infections/pathology , Helicobacter pylori , Humans , Macrophage Activation , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Ornithine Decarboxylase/genetics , Putrescine/metabolism
7.
J Nat Prod ; 82(3): 500-509, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30794412

ABSTRACT

The biological basis for documented in vivo bone-protective effects of turmeric-derived curcumin is unclear since curcumin is barely detectable in serum, being rapidly conjugated to form what is thought to be an inactive glucuronide. Studies were therefore undertaken to test the postulate that antiresorptive effects of curcumin require deconjugation within bone to form the bioactive aglycone and that ß-glucuronidase (GUSB), a deconjugating enzyme expressed by hematopoietic marrow cells, facilitates this site-specific transformation. Consistent with this postulate, aglycone, but not glucuronidated, curcumin inhibited RANKL-stimulated osteoclastogenesis, a key curcumin target in bone. Aglycone curcumin, expressed relative to total curcumin, was higher in bone marrow than in serum of curcumin-treated C57BL/6J mice, while remaining a minor component. Ex vivo, under conditions preventing further metabolism of the unstable aglycone, the majority of curcumin-glucuronide delivered to marrow in vivo was hydrolyzed to the aglycone, a process that was inhibited by treatment with saccharolactone, a GUSB inhibitor, or in mice having reduced (C3H/HeJ) or absent (mps/mps) GUSB activity. These findings suggest that curcumin, despite low systemic bioavailability, may be enzymatically activated (deconjugated) within GUSB-enriched bone to exert protective effects, a metabolic process that could also contribute to bone-protective effects of other highly glucuronidated dietary polyphenols.


Subject(s)
Bone and Bones/metabolism , Curcumin/metabolism , Glucuronidase/metabolism , Glucuronides/metabolism , Administration, Oral , Animals , Area Under Curve , Biological Availability , Catalysis , Curcumin/administration & dosage , Curcumin/pharmacokinetics , Female , Half-Life , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Osteoclasts/cytology , RANK Ligand/metabolism
8.
J Biol Chem ; 292(52): 21243-21252, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29097552

ABSTRACT

The spice turmeric, with its active polyphenol curcumin, has been used as anti-inflammatory remedy in traditional Asian medicine for centuries. Many cellular targets of curcumin have been identified, but how such a wide range of targets can be affected by a single compound is unclear. Here, we identified curcumin as a pro-drug that requires oxidative activation into reactive metabolites to exert anti-inflammatory activities. Synthetic curcumin analogs that undergo oxidative transformation potently inhibited the pro-inflammatory transcription factor nuclear factor κB (NF-κB), whereas stable, non-oxidizable analogs were less active, with a correlation coefficient (R2) of IC50versus log of autoxidation rate of 0.75. Inhibition of glutathione biosynthesis, which protects cells from reactive metabolites, increased the potency of curcumin and decreased the amount of curcumin-glutathione adducts in cells. Oxidative metabolites of curcumin adducted to and inhibited the inhibitor of NF-κB kinase subunit ß (IKKß), an activating kinase upstream of NF-κB. An unstable, alkynyl-tagged curcumin analog yielded abundant adducts with cellular protein that were decreased by pretreatment with curcumin or an unstable analog but not by a stable analog. Bioactivation of curcumin occurred readily in vitro, which may explain the wide range of cellular targets, but if bioactivation is insufficient in vivo, it may also help explain the inconclusive results in human studies with curcumin so far. We conclude that the paradigm of metabolic bioactivation uncovered here should be considered for the evaluation and design of clinical trials of curcumin and other polyphenols of medicinal interest.


Subject(s)
Curcumin/metabolism , Oxidation-Reduction/drug effects , Animals , Anti-Inflammatory Agents/metabolism , Curcumin/pharmacology , Glutathione/drug effects , Glutathione/metabolism , HeLa Cells , Humans , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , Mice , NF-kappa B/drug effects , NF-kappa B/metabolism , Oxidative Stress/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
9.
Chem Res Toxicol ; 31(4): 269-276, 2018 04 16.
Article in English | MEDLINE | ID: mdl-29569909

ABSTRACT

The polypharmacological effects of the turmeric compound curcumin may be partly mediated by covalent adduction to cellular protein. Covalent binding to small molecule and protein thiols is thought to occur through a Michael-type addition at the enone moiety of the heptadienedione chain connecting the two methoxyphenol rings of curcumin. Here we show that curcumin forms the predicted thiol-Michael adducts with three model thiols, glutathione, N-acetylcysteine, and ß-mercaptoethanol. More abundant, however, are respective thiol adducts of the dioxygenated spiroepoxide intermediate of curcumin autoxidation. Two electrophilic sites at the quinone-like ring of the spiroepoxide are identified. Addition of ß-mercaptoethanol at the 5'-position of the ring gives a 1,7-dihydroxycyclopentadione-5' thioether, and addition at the 1'-position results in cleavage of the aromatic ring from the molecule, forming methoxyphenol-thioether and a tentatively identified cyclopentadione aldehyde. The curcuminoids demethoxy- and bisdemethoxycurcumin do not form all of the possible thioether adducts, corresponding with their increased stability toward autoxidation. RAW264.7 macrophage-like cells activated with phorbol ester form curcumin-glutathionyl and the 1,7-dihydroxycyclopentadione-5'-glutathionyl adducts. These studies indicate that the enone of the parent compound is not the only functional electrophile in curcumin, and that its oxidation products provide additional electrophilic sites. This suggests that protein binding by curcumin may involve oxidative activation into reactive quinone methide and spiroepoxide electrophiles.


Subject(s)
Curcumin/chemistry , Sulfhydryl Compounds/chemistry , Animals , Curcumin/chemical synthesis , Curcumin/metabolism , Macrophages/chemistry , Macrophages/metabolism , Mice , Molecular Structure , Oxidation-Reduction , RAW 264.7 Cells , Sulfhydryl Compounds/metabolism
10.
Org Biomol Chem ; 16(17): 3273-3281, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29664496

ABSTRACT

The efficacy of the curry spice compound curcumin as a natural anti-inflammatory agent is limited by its rapid reductive metabolism in vivo. A recent report described a novel synthetic derivative, 2,6-dimethyl-curcumin, with increased stability against reduction in vitro and in vivo. It is also known that curcumin is unstable at physiological pH in vitro and undergoes rapid autoxidative transformation. Since the oxidation products may contribute to the biological effects of curcumin, we tested oxidative stability of 2,6-dimethyl-curcumin in buffer (pH 7.5). The rate of degradation was similar to curcumin. The degradation products were identified as a one-carbon chain-shortened alcohol, vanillin, and two isomeric epoxides that underwent cleavage to vanillin and a corresponding hydroxylated cleavage product. 2,6-Dimethyl-curcumin was more potent than curcumin in inhibiting NF-κB activity but less potent in inhibiting expression of cyclooxygenase-2 in LPS-activated RAW264.7 cells. 2,6-Dimethyl-curcumin and some of its degradation products covalently bound to a peptide that contains the redox-sensitive cysteine of IKKß kinase, the activating kinase upstream of NF-κB, providing a mechanism for the anti-inflammatory activity. In RAW264.7 cells vanillin, the chain-shortened alcohol, and reduced 2,6-dimethyl-curcumin were detected as major metabolites. These studies provide new insight into the oxidative transformation mechanism of curcumin and related compounds. The products resulting from oxidative transformation contribute to the anti-inflammatory activity of 2,6-dimethyl-curcumin in addition to its enhanced resistance against enzymatic reduction.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Curcumin/analogs & derivatives , Curcumin/pharmacology , Macrophages/drug effects , Animals , Drug Stability , Macrophages/immunology , Methylation , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Oxidation-Reduction , RAW 264.7 Cells
11.
J Nat Prod ; 81(12): 2756-2762, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30560664

ABSTRACT

Evidence that anti-inflammatory and other biological effects of curcumin may at least in part be mediated by its metabolites underscores the importance of identifying novel transformation products. Spontaneous degradation of curcumin in buffer pH 7.5 results mainly in dioxygenated products with a characteristic cyclopentadione ring composed of carbons 2 through 6 of the former heptadienedione chain. When analyzing degradation reactions of 4'- O-methylcurcumin, a product was identified missing one of the terminal carbons of the heptadienedione moiety while containing a cyclopentadione ring and adjacent hydroxy group typical of curcumin degradation products. Analysis of curcumin autoxidation reactions showed formation of an analogous compound, 7-norcyclopentadione, a degradation product exhibiting net loss of a carbon and gain of an oxygen atom. Removal of the carbon is proposed to occur via a peroxide-linked curcumin dimer in conjunction with radical-mediated 1,2-aryl migration of a guaiacol moiety. Oxidation reactions of demethoxycurcumin gave demethoxy-7-norcyclopentadione, whereas an analogous product was not observed from bis-demethoxycurcumin. Incubation of RAW264.7 macrophage-like cells with curcumin showed the presence of 7-norcyclopentadione, the formation of which was not increased upon activation of the cells with 12- O-tetradecanoylphorbol-13-acetate . 7-Norcyclopentadione is a novel type of degradation product that is most likely formed via autoxidative processes when cells are incubated with curcumin.


Subject(s)
Curcumin/chemistry , Cyclopentanes/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Carbon/chemistry , Magnetic Resonance Spectroscopy , Mice , Molecular Structure , RAW 264.7 Cells
12.
J Biol Chem ; 290(8): 4817-4828, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25564617

ABSTRACT

Curcumin is a dietary anti-inflammatory and chemopreventive agent consisting of two methoxyphenol rings connected by a conjugated heptadienedione chain. Curcumin is unstable at physiological pH and rapidly degrades in an autoxidation reaction to a major bicyclopentadione product in which the 7-carbon chain has undergone oxygenation and double cyclization. Early degradation products (but not the final bicyclopentadione) mediate topoisomerase poisoning and possibly many other activities of curcumin, but it is not known how many and what autoxidation products are formed, nor their mechanism of formation. Here, using [(14)C2]curcumin as a tracer, seven novel autoxidation products, including two reaction intermediates, were isolated and identified using one- and two-dimensional NMR and mass spectrometry. The unusual spiroepoxide and vinylether reaction intermediates are precursors to the final bicyclopentadione product. A mechanism for the autoxidation of curcumin is proposed that accounts for the addition and exchange of oxygen that have been determined using (18)O2 and H2(18)O. Several of the by-products are formed from an endoperoxide intermediate via reactions that are well precedented in lipid peroxidation. The electrophilic spiroepoxide intermediate formed a stable adduct with N-acetylcysteine, suggesting that oxidative transformation is required for biological effects mediated by covalent adduction to protein thiols. The spontaneous autoxidation distinguishes curcumin among natural polyphenolic compounds of therapeutic interest; the formation of chemically diverse reactive and electrophilic products provides a novel paradigm for understanding the polypharmacological effects of curcumin.


Subject(s)
Curcumin/analogs & derivatives , Curcumin/chemistry , Spiro Compounds/chemistry , Curcumin/pharmacokinetics , Magnetic Resonance Spectroscopy , Oxidation-Reduction , Oxygen Isotopes/chemistry
13.
Chem Res Toxicol ; 28(5): 989-96, 2015 May 18.
Article in English | MEDLINE | ID: mdl-25806475

ABSTRACT

Extracts from the rhizome of the turmeric plant are widely consumed as anti-inflammatory dietary supplements. Turmeric extract contains the three curcuminoids, curcumin (≈80% relative abundance), demethoxycurcumin (DMC; ≈15%), and bisdemethoxycurcumin (BDMC; ≈5%). A distinct feature of pure curcumin is its instability at physiological pH, resulting in rapid autoxidation to a bicyclopentadione within 10-15 min. Here, we describe oxidative transformation of turmeric extract, DMC, and BDMC and the identification of their oxidation products using LC-MS and NMR analyses. DMC autoxidized over the course of 24 h to the expected bicyclopentadione diastereomers. BDMC was resistant to autoxidation, and oxidative transformation required catalysis by horseradish peroxidase and H2O2 or potassium ferricyanide. The product of BDMC oxidation was a stable spiroepoxide that was equivalent to a reaction intermediate in the autoxidation of curcumin. The ability of DMC and BDMC to poison recombinant human topoisomerase IIα was significantly increased in the presence of potassium ferricyanide, indicating that oxidative transformation was required to achieve full DNA cleavage activity. DMC and BDMC are less prone to autoxidation than curcumin and contribute to the enhanced stability of turmeric extract at physiological pH. Their oxidative metabolites may contribute to the biological effects of turmeric extract.


Subject(s)
Antigens, Neoplasm/metabolism , Curcuma/toxicity , Curcumin/analogs & derivatives , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Plant Extracts/toxicity , Antigens, Neoplasm/chemistry , Curcuma/chemistry , Curcuma/metabolism , Curcumin/chemistry , Curcumin/metabolism , Curcumin/toxicity , DNA Cleavage/drug effects , DNA Topoisomerases, Type II/chemistry , DNA-Binding Proteins/chemistry , Diarylheptanoids , Epoxy Compounds/chemistry , Epoxy Compounds/metabolism , Epoxy Compounds/toxicity , Humans , Oxidation-Reduction , Plant Extracts/chemistry , Plant Extracts/metabolism
14.
J Inherit Metab Dis ; 37(3): 353-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24154984

ABSTRACT

BACKGROUND: Valproic acid (VPA) is an effective antiepileptic drug that may induce progressive microvesicular steatosis. The impairment of mitochondrial function may be an important metabolic effect of VPA treatment with potential adverse consequences. OBJECTIVE: To investigate the influence of VPA on the activity of GTP- and ATP-specific succinate:CoA ligases (G-SUCL and A-SUCL). METHODS: The GTP- and ATP-specific SUCL activities were measured in human fibroblasts in the reverse direction, i.e. the formation of succinyl-CoA. These were assessed at different concentrations of succinate in the presence of VPA, valproyl-CoA and zinc chloride, an established inhibitor of the enzymes. Activities were measured using an optimized HPLC procedure. RESULTS: Valproyl-CoA (1 mM) inhibited the activity of A-SUCL and G-SUCL by 45-55% and 25-50%, respectively. VPA (1 mM) had no influence on the activity of the two enzymes. DISCUSSION: Valproyl-CoA appears to affect the activity of SUCL, especially with the ATP-specific enzyme. Considering the key role of SUCL in the Krebs cycle, interference with its activity might impair the cellular energy status. Moreover, A-SUCL is bound to the nucleoside diphosphate kinase (NDPK), which is responsible for the mitochondrial (deoxy)nucleotide synthesis. An inhibition of A-SUCL might influence the activity of NDPK inducing an imbalance of nucleotides in the mitochondria and eventually mitochondrial DNA depletion. This may account for the potential liver failure associated with valproate therapy, reported in patients with deficiencies within the mitochondrial DNA replicase system such as polymerase gamma 1.


Subject(s)
Acyl Coenzyme A/pharmacology , Adenosine Triphosphate/physiology , Guanosine Triphosphate/physiology , Succinate-CoA Ligases/antagonists & inhibitors , DNA, Mitochondrial/metabolism , Humans , Liver Failure/chemically induced , Nucleoside-Diphosphate Kinase/physiology , Valproic Acid/adverse effects , Valproic Acid/pharmacology
15.
ACS Omega ; 9(35): 37025-37034, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39246485

ABSTRACT

Curcumin exerts some of its biological effects via degradation products formed by spontaneous oxidation at physiological, i.e., weakly basic, pH. Here, we analyzed products formed by dry heating of curcumin in the presence of a basic salt (sodium bicarbonate and others). Under the dry heating conditions employed, curcumin was completely consumed, yielding products entirely different from those obtained by autoxidative degradation in buffer. Bioassay-guided fractionation of the reaction mixture was used to identify and isolate compounds with anti-inflammatory activity in a cell-based assay. This provided two dimers of curcumin, dicurmins A and B, featuring a partly saturated naphthalene core that inhibited lipopolysaccharide-induced activation of NF-κB in RAW264.7 cells. Dicurmin A and B are unusual derivatives of curcumin lacking key functional moieties yet exhibit increased anti-inflammatory activity. The process of dry heating of polyphenols in the presence of a basic salt can serve as a novel approach to generating bioactive compounds.

16.
J Inherit Metab Dis ; 35(3): 443-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22189597

ABSTRACT

BACKGROUND: Valproic acid (VPA) is a widely used anticonvulsant drug which affects mitochondrial metabolism including the catabolism of fatty acids and branched-chain amino acids. AIMS: To elucidate the effect of valproate on the leucine pathway through a targeted metabolomics approach and the evaluation of the effects of valproate on the activity of biotinidase and 3-methylcrotonyl-CoA carboxylase (3MCC). METHODS: Urine organic acid analysis was performed in patients under VPA therapy and healthy controls using gas-chromatography/mass spectrometry (GC-MS). Biotinidase activity was determined in plasma samples of both groups using an optimized spectrophotometric assay. After immunoprecipitation of short-chain enoyl-CoA hydratase (crotonase, ECHS1), 3MCC activity was measured in human liver homogenate using high-performance liquid chromatography (HPLC), in the absence and presence of valproyl-CoA. RESULTS: The levels of 3-hydroxyisovaleric acid (3OH-IVA), one secondary metabolite of the leucine pathway, were significantly elevated in human urine after VPA treatment. Biotinidase activity in plasma samples ranged from very low to normal levels in treated patients as compared with controls. Enzyme activity measurements revealed inhibition of 3-methylcrotonyl-CoA carboxylase by valproyl-CoA (IC(50) = 1.36 mM). Furthermore, we show that after complete immunoprecipitation of crotonase in a human liver homogenate, 3-hydroxyisovaleryl-CoA is not formed. DISCUSSION: Our results suggest the interference of VPA with the activity of 3MCC through a potential cumulative effect: direct inhibition of the enzyme activity by the drug metabolite valproyl-CoA and the inhibition of biotinidase by valproate and/or its metabolites. These interactions may be associated with the skin rash and hair loss which are side effects often reported in VPA-treated patients.


Subject(s)
Carbon-Carbon Ligases/antagonists & inhibitors , Carbon-Carbon Ligases/chemistry , Enzyme Inhibitors/pharmacology , Valerates/metabolism , Biotinidase/metabolism , Case-Control Studies , Chromatography, High Pressure Liquid/methods , Gas Chromatography-Mass Spectrometry/methods , Humans , Inhibitory Concentration 50 , Kinetics , Leucine/metabolism , Liver/enzymology , Metabolomics/methods , Models, Chemical , Valproic Acid/pharmacology
17.
J Nutr Biochem ; 99: 108842, 2022 01.
Article in English | MEDLINE | ID: mdl-34407450

ABSTRACT

TGFß signaling promotes progression of bone-metastatic (BMET) breast cancer (BCa) cells by driving tumor-associated osteolysis, a hallmark of BCa BMETs, thus allowing for tumor expansion within bone. Turmeric-derived bioactive curcumin, enriched in bone via local enzymatic deconjugation of inactive circulating curcumin-glucuronides, inhibits osteolysis and BMET progression in human xenograft BCa BMET models by blocking tumoral TGFß signaling pathways mediating osteolysis. This is a unique antiosteolytic mechanism in contrast to current osteoclast-targeting therapeutics. Therefore, experiments were undertaken to elucidate the mechanism for curcumin inhibition of BCa TGFß signaling and the application of this finding across multiple BCa cell lines forming TGFß-dependent BMETs, including a possible role for bioactive curcumin metabolites in mediating these effects. Immunoblot analysis of TGFß signaling proteins in bone tropic human (MDA-SA, MDA-1833, MDA-2287) and murine (4T1) BCa cells revealed uniform curcumin blockade of TGFß-induced Smad activation due to down-regulation of plasma membrane associated TGFßR2 and cellular receptor Smad proteins that propagate Smad-mediated gene expression, resulting in downregulation of PTHrP expression, the osteolytic factor driving in vivo BMET progression. With the exception of early decreases in TGFßR2, inhibitory effects appeared to be mediated by oxidative metabolites of curcumin and involved inhibition of gene expression. Interestingly, while not contributing to changes in Smad-mediated TGFß signaling, curcumin caused early activation of MAPK signaling in all cell lines, including JNK, an effect possibly involving interactions with TGFßR2 within lipid rafts. Treatment with curcumin or oxidizable analogs of curcumin may have clinical relevancy in the management of TGFß-dependent BCa BMETs.


Subject(s)
Bone Neoplasms/prevention & control , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Curcumin/administration & dosage , Transforming Growth Factor beta1/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Curcumin/chemistry , Female , Humans , Mice , Oxidation-Reduction , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction/drug effects , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/genetics
18.
NPJ Sci Food ; 6(1): 4, 2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35031622

ABSTRACT

The identification of molecular targets of bioactive food components is important to understand the mechanistic aspect of their physiological functions. Here, we have developed a screening system that enables us to determine the activation of G protein-coupled receptors (GPCRs) by food components and have identified GPR55 as a target for curcumin. Curcumin activated GPR55 and induced serum-response element- and serum-response factor-mediated transcription, which were inhibited by Rho kinase and GPR55 antagonists. Both the methoxy group and the heptadienone moiety of curcumin were required for GPR55 activation. The F1905.47 residue of GPR55 was important for the interaction with curcumin. The curcumin-induced secretion of glucagon-like peptide-1 in GLUTag cells was inhibited by a GPR55 antagonist. These results indicate that expression screening is a useful system to identify GPCRs as targets of food components and strongly suggest that curcumin activates GPR55 as an agonist, which is involved in the physiological function of curcumin.

19.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35579952

ABSTRACT

Macrophages play a crucial role in the inflammatory response to the human stomach pathogen Helicobacter pylori, which infects half of the world's population and causes gastric cancer. Recent studies have highlighted the importance of macrophage immunometabolism in their activation state and function. We have demonstrated that the cysteine-producing enzyme cystathionine γ-lyase (CTH) is upregulated in humans and mice with H. pylori infection. Here, we show that induction of CTH in macrophages by H. pylori promoted persistent inflammation. Cth-/- mice had reduced macrophage and T cell activation in H. pylori-infected tissues, an altered metabolome, and decreased enrichment of immune-associated gene networks, culminating in decreased H. pylori-induced gastritis. CTH is downstream of the proposed antiinflammatory molecule, S-adenosylmethionine (SAM). Whereas Cth-/- mice exhibited gastric SAM accumulation, WT mice treated with SAM did not display protection against H. pylori-induced inflammation. Instead, we demonstrated that Cth-deficient macrophages exhibited alterations in the proteome, decreased NF-κB activation, diminished expression of macrophage activation markers, and impaired oxidative phosphorylation and glycolysis. Thus, through altering cellular respiration, CTH is a key enhancer of macrophage activation, contributing to a pathogenic inflammatory response that is the universal precursor for the development of H. pylori-induced gastric disease.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Animals , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Inflammation/metabolism , Macrophages/metabolism , Mice
20.
Drug Metab Dispos ; 39(7): 1155-60, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21430231

ABSTRACT

Many biological systems including the oxidative catabolic pathway for branched-chain amino acids (BCAAs) are affected in vivo by valproate therapy. In this study, we investigated the potential effect of valproic acid (VPA) and some of its metabolites on the metabolism of BCAAs. In vitro studies were performed using isovaleryl-CoA dehydrogenase (IVD), isobutyryl-CoA dehydrogenase (IBD), and short branched-chain acyl-CoA dehydrogenase (SBCAD), enzymes involved in the degradation pathway of leucine, valine, and isoleucine. The enzymatic activities of the three purified human enzymes were measured using optimized high-performance liquid chromatography procedures, and the respective kinetic parameters were determined in the absence and presence of VPA and the corresponding CoA and dephosphoCoA conjugates. Valproyl-CoA and valproyl-dephosphoCoA inhibited IVD activity significantly by a purely competitive mechanism with K(i) values of 74 ± 4 and 170 ± 12 µM, respectively. IBD activity was not affected by any of the tested VPA esters. However, valproyl-CoA did inhibit SBCAD activity by a purely competitive mechanism with a K(i) of 249 ± 29 µM. In addition, valproyl-dephosphoCoA inhibited SBCAD activity via a distinct mechanism (K(i) = 511 ± 96 µM) that appeared to be of the mixed type. Furthermore, we show that both SBCAD and IVD are active, using valproyl-CoA as a substrate. The catalytic efficiency of SBCAD turned out to be much higher than that of IVD, demonstrating that SBCAD is the most probable candidate for the first dehydrogenation step of VPA ß-oxidation. Our data explain some of the effects of valproate on the branched-chain amino acid metabolism and shed new light on the biotransformation pathway of valproate.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Butyryl-CoA Dehydrogenase/metabolism , Isovaleryl-CoA Dehydrogenase/metabolism , Valproic Acid/metabolism , Chromatography, High Pressure Liquid , Oxidation-Reduction , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL