Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Can J Gastroenterol Hepatol ; 2021: 8854432, 2021.
Article in English | MEDLINE | ID: mdl-33604316

ABSTRACT

Cytochrome P450 2E1 (CYP2E1) is a key target protein in the development of alcoholic and nonalcoholic fatty liver disease (FLD). The pathophysiological correlate is the massive production of reactive oxygen species. The role of CYP2E1 in the development of hepatocellular carcinoma (HCC), the final complication of FLD, remains controversial. Specifically, CYP2E1 has not yet been defined as a molecular target for HCC therapy. In addition, a CYP2E1-specific drug has not been developed. We have already shown that our newly developed CYP2E1 inhibitor 12-imidazolyl-1-dodecanol (I-ol) was therapeutically effective against alcoholic and nonalcoholic steatohepatitis. In this study, we investigated the effect of I-ol on HCC tumorigenesis and whether I-ol could serve as a possible treatment option for terminal-stage FLD. I-ol exerted a very highly significant antitumour effect against hepatocellular HepG2 cells. Cell viability was reduced in a dose-dependent manner, with only the highest doses causing a cytotoxic effect associated with caspase 3/7 activation. Comparable results were obtained for the model colorectal adenocarcinoma cell line, DLD-1, whose tumorigenesis is also associated with CYP2E1. Transcriptome analyses showed a clear effect of I-ol on apoptosis and cell-cycle regulation, with the increased expression of p27Kip1 being particularly noticeable. These observations were confirmed at the protein level for HepG2 and DLD-1 cells grafted on a chorioallantoic membrane. Cell-cycle analysis showed a complete loss of proliferating cells with a simultaneous increase in S-phase arrest beginning at a threshold dose of 30 µM. I-ol also reduced xenograft tumour growth in nude mice. This antitumour effect was not associated with tumour cachexia. I-ol was not toxic to healthy tissues or organs. This study demonstrates for the first time the therapeutic effect of the specific CYP2E1 inhibitor I-ol on the tumorigenesis of HCC. Our findings imply that I-ol can potentially be applied therapeutically on patients at the final stage of FLD.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/drug therapy , Cytochrome P-450 CYP2E1/metabolism , Dodecanol , Humans , Liver Neoplasms/drug therapy , Mice , Mice, Nude , Oxidative Stress
2.
Molecules ; 26(2)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445710

ABSTRACT

Boswellic acids, and particularly 11-keto-boswellic acids, triterpenoids derived from the genus Boswellia (Burseraceae), are known for their anti-inflammatory and potential antitumor efficacy. Although boswellic acids generally occur as α-isomers (oleanane type) and ß-isomers (ursane type), 11-keto-boswellic acid (KBA) was found only as the ß-isomer, ß-KBA. Here, the existence and natural occurrence of the respective α-isomer, 11-keto-α-boswellic acid (α-KBA), is demonstrated for the first time. Initially, α-KBA was synthesized and characterized by high-resolution mass spectrometry (HR-MS) and nuclear magnetic resonance (NMR) spectroscopy, and a highly selective, sensitive, and accurate high-performance liquid chromatography coupled with tandem mass spectrometry (HPLC-MS/MS) method was developed by Design of Experiments (DoE) using a pentafluorophenyl stationary phase. This method allowed the selective quantification of individual 11-keto-boswellic acids and provided evidence for α-KBA in Boswellia spp. oleogum resins. The contents of α-KBA as well as further boswellic acids and the composition of essential oils were used to chemotaxonomically classify 41 Boswellia oleogum resins from 9 different species. Moreover, α-KBA exhibited cytotoxicity against three treatment-resistant triple-negative breast cancer (TNBC) cell lines in vitro and also induced apoptosis in MDA-MB-231 xenografts in vivo. The respective ß-isomer and the acetylated form demonstrate higher cytotoxic efficacies against TNBC cells. This provides further insights into the structure-activity relationship of boswellic acids and could support future developments of potential anti-inflammatory and antitumor drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Boswellia/chemistry , Triple Negative Breast Neoplasms/pathology , Triterpenes/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Chickens , Humans , Isomerism , Triterpenes/chemical synthesis , Triterpenes/chemistry , Triterpenes/isolation & purification
3.
PLoS One ; 15(7): e0235990, 2020.
Article in English | MEDLINE | ID: mdl-32701948

ABSTRACT

BACKGROUND AND AIMS: Alcoholic steatohepatitis (ASH)-the inflammation of fatty liver-is caused by chronic alcohol consumption and represents one of the leading chronic liver diseases in Western Countries. ASH can lead to organ dysfunction or progress to hepatocellular carcinoma (HCC). Long-term alcohol abstinence reduces this probability and is the prerequisite for liver transplantation-the only effective therapy option at present. Elevated enzymatic activity of cytochrome P450 2E1 (CYP2E1) is known to be critically responsible for the development of ASH due to excessively high levels of reactive oxygen species (ROS) during metabolization of ethanol. Up to now, no rational drug discovery process was successfully initiated to target CYP2E1 for the treatment of ASH. METHODS: In this study, we applied a rational drug design concept to develop drug candidates (NCE) including preclinical studies. RESULTS: A new class of drug candidates was generated successfully. Two of the most promising small compounds named 12-Imidazolyl-1-dodecanol (abbr.: I-ol) and 1-Imidazolyldodecane (abbr.: I-an) were selected at the end of this process of drug discovery and developability. These new ω-imidazolyl-alkyl derivatives act as strong chimeric CYP2E1 inhibitors at a nanomolar range. They restore redox balance, reduce inflammation process as well as the fat content in the liver and rescue the physiological liver architecture of rats consuming continuously a high amount of alcohol. CONCLUSIONS: Due to its oral application and therapeutic superiority over an off-label use of the hepatoprotector ursodeoxycholic acid (UDCA), this new class of inhibitors marks the first rational, pharmaceutical concept in long-term treatment of ASH.


Subject(s)
Cytochrome P-450 CYP2E1 Inhibitors/therapeutic use , Cytochrome P-450 CYP2E1/metabolism , Fatty Liver, Alcoholic/drug therapy , Alkanes/chemical synthesis , Alkanes/pharmacology , Alkanes/therapeutic use , Animals , Cytochrome P-450 CYP2E1/chemistry , Cytochrome P-450 CYP2E1 Inhibitors/chemical synthesis , Cytochrome P-450 CYP2E1 Inhibitors/pharmacology , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Female , Hep G2 Cells , Humans , Lipoproteins, VLDL/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Oxidative Stress/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Severity of Illness Index , Triglycerides/blood , Ursodeoxycholic Acid/analysis
4.
J Pharmacol Exp Ther ; 357(3): 487-94, 2016 06.
Article in English | MEDLINE | ID: mdl-27044804

ABSTRACT

Intraperitoneal injection of arglabin (2.5 ng/g of body weight, twice daily, 13 weeks) into female human apolipoprotein E2 gene knock-in (ApoE2Ki) mice fed a high-fat Western-type diet (HFD) reduced plasma levels of glucose and insulin by ∼20.0% ± 3.5% and by 50.0% ± 2.0%, respectively, in comparison with vehicle-treated mice. Immunohistochemical analysis revealed the absence of active caspase-3 in islet sections from ApoE2Ki mice fed a HFD and treated with arglabin. In addition, arglabin reduced interleukin-1ß (IL-1ß) production in a concentration-dependent manner in Langerhans islets isolated from ApoE2Ki mice treated with lipopolysaccharide (LPS) and with cholesterol crystals. This inhibitory effect is specific for the inflammasome NOD-like receptor family, pyrin domain-containing 3 (NLRP3) because IL-1ß production was abolished in Langerhans islets isolated from Nlrp3(-/-) mice. In the insulin-secreting INS-1 cells, arglabin inhibited, in a concentration-dependent manner, the maturation of pro-IL-1ß into biologically active IL-1ß probably through the inhibition of the maturation of procaspase-1 into active capsase-1. Moreover, arglabin reduced the susceptibility of INS-1 cells to apoptosis by increasing Bcl-2 levels. Similarly, autophagy activation by rapamycin decreased apoptosis susceptibility while autophagy inhibition by 3-methyladenin treatment promoted apoptosis. Arglabin further increased the expression of the autophagic markers Bcl2-interacting protein (Beclin-1) and microtubule-associated protein 1 light chain 3 II (LC3-II) in a concentration-dependent manner. Thus, arglabin reduces NLRP3-dependent inflammation as well as apoptosis in pancreatic ß-cells in vivo and in the INS-1 cell line in vitro, whereas it increases autophagy in cultured INS-1 cells, indicating survival-promoting properties of the compound in these cells. Hence, arglabin may represent a new promising compound to treat inflammation and type 2 diabetes mellitus development.


Subject(s)
Apolipoprotein E2/genetics , Apoptosis/drug effects , Diabetes Mellitus, Type 2/prevention & control , Diet, High-Fat/adverse effects , Inflammasomes/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Sesquiterpenes/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Caspase 1/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Enzyme Activation/drug effects , Female , Gene Expression Regulation/drug effects , Gene Knock-In Techniques , Humans , Inflammation/drug therapy , Insulin/blood , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Interleukin-1beta/biosynthesis , Mice , Rats , Sesquiterpenes/therapeutic use , Sesquiterpenes, Guaiane , bcl-2-Associated X Protein/metabolism
6.
Circulation ; 131(12): 1061-70, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25613820

ABSTRACT

BACKGROUND: This study was designed to evaluate the effect of arglabin on the NLRP3 inflammasome inhibition and atherosclerotic lesion in ApoE2Ki mice fed a high-fat Western-type diet. METHODS AND RESULTS: Arglabin was purified, and its chemical identity was confirmed by mass spectrometry. It inhibited, in a concentration-dependent manner, interleukin (IL)-1ß and IL-18, but not IL-6 and IL-12, production in lipopolysaccharide and cholesterol crystal-activated cultured mouse peritoneal macrophages, with a maximum effect at ≈50 nmol/L and EC50 values for both cytokines of ≈ 10 nmol/L. Lipopolysaccharide and cholesterol crystals did not induce IL-1ß and IL-18 production in Nlrp3(-/-) macrophages. In addition, arglabin activated autophagy as evidenced by the increase in LC3-II protein. Intraperitoneal injection of arglabin (2.5 ng/g body weight twice daily for 13 weeks) into female ApoE2.Ki mice fed a high-fat diet resulted in a decreased IL-1ß plasma level compared with vehicle-treated mice (5.2±1.0 versus 11.7±1.1 pg/mL). Surprisingly, arglabin also reduced plasma levels of total cholesterol and triglycerides to 41% and 42%, respectively. Moreover, arglabin oriented the proinflammatory M1 macrophages into the anti-inflammatory M2 phenotype in spleen and arterial lesions. Finally, arglabin treatment markedly reduced the median lesion areas in the sinus and whole aorta to 54% (P=0.02) and 41% (P=0.02), respectively. CONCLUSIONS: Arglabin reduces inflammation and plasma lipids, increases autophagy, and orients tissue macrophages into an anti-inflammatory phenotype in ApoE2.Ki mice fed a high-fat diet. Consequently, a marked reduction in atherosclerotic lesions was observed. Thus, arglabin may represent a promising new drug to treat inflammation and atherosclerosis.


Subject(s)
Apolipoprotein E2/deficiency , Atherosclerosis/drug therapy , Carrier Proteins/antagonists & inhibitors , Diet, High-Fat/adverse effects , Inflammasomes/antagonists & inhibitors , Sesquiterpenes/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/blood , Atherosclerosis/etiology , Female , Inflammasomes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Sesquiterpenes/pharmacology , Sesquiterpenes, Guaiane , Treatment Outcome
7.
J Pharmacol Exp Ther ; 352(1): 33-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25316122

ABSTRACT

Here we provide evidence that αATA(8,24) (3α-acetyloxy-tir-8,24-dien-21-oic acid) inhibits Akt/mammalian target of rapamycin (mTOR) signaling. αATA(8,24) and other tirucallic acids were isolated from the acetylated extract of the oleo gum resin of Boswellia serrata to chemical homogeneity. Compared with related tirucallic acids, αATA(8,24) was the most potent inhibitor of the proliferation of androgen-insensitive prostate cancer cells in vitro and in vivo, in prostate cancer xenografted onto chick chorioallantoic membranes. αATA(8,24) induced loss of cell membrane asymmetry, caspase-3 activation, and DNA fragmentation in vitro and in vivo. These effects were selective for cancer cells, because αATA(8,24) exerted no overt toxic effects on peripheral blood mononuclear cells or the chick embryo. At the molecular level, αATA(8,24) inhibited the Akt1 kinase activity. Prior to all biochemical signs of cellular dysfunction, αATA(8,24) induced inhibition of the Akt downstream target mTOR as indicated by dephosphorylation of S6K1. This event was followed by decreased expression of cell cycle regulators, such as cyclin D1, cyclin E, and cyclin B1, as well as cyclin-dependent kinases CDK4 and CDK2 and phosphoretinoblastoma protein, which led to inhibition of the cell-cycle progression. In agreement with the mTOR inhibition, αATA(8,24) and rapamycin increased the volume of acidic vesicular organelles. In contrast to rapamycin, αATA(8,24) destabilized lysosomal and mitochondrial membranes and induced reactive oxygen species production in cancer cells. The ability of αATA(8,24) to inhibit Akt/mTOR signaling and to induce simultaneously oxidative stress could be exploited for the development of novel antitumor therapeutics with a lower profile of toxic side effects.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Oxidative Stress/drug effects , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Triterpenes/chemistry , Triterpenes/pharmacology , Animals , Antineoplastic Agents/isolation & purification , Apoptosis/drug effects , Boswellia/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chick Embryo , Chorioallantoic Membrane/cytology , Chorioallantoic Membrane/drug effects , Humans , Isomerism , Male , Membrane Potential, Mitochondrial/drug effects , Models, Molecular , Molecular Conformation , Organelles/drug effects , Organelles/metabolism , Reactive Oxygen Species/metabolism , Triterpenes/isolation & purification
8.
ACS Nano ; 8(11): 11042-52, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25337989

ABSTRACT

Amyloid oligomers are nonfibrillar polypeptide aggregates linked to diseases, such as Alzheimer's and Parkinson's. Here we show that these aggregates possess a compact, quasi-crystalline architecture that presents significant nanoscale regularity. The amyloid oligomers are dynamic assemblies and are able to release their individual subunits. The small oligomeric size and spheroid shape confer diffusible characteristics, electrophoretic mobility, and the ability to enter hydrated gel matrices or cells. We finally showed that the amyloid oligomers can be labeled with both fluorescence agents and iron oxide nanoparticles and can target macrophage cells. Oligomer amyloids may provide a new biological nanomaterial for improved targeting, drug release, and medical imaging.


Subject(s)
Amyloid/chemistry , Biopolymers/chemistry , Nanoparticles , Microscopy, Electron, Transmission , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation
9.
PLoS One ; 9(6): e100129, 2014.
Article in English | MEDLINE | ID: mdl-24941000

ABSTRACT

Boswellic acid (BA)-containing extracts such as BSE have anti-inflammatory and immunomodulatory activity. In chronic schistosomiasis, the hepatic granuloma and fibrosis induced by egg deposition in the liver is the most serious pathological manifestations. However, little is known regarding the role of BAs in Schistosoma japonicum (S. japonicum) egg-induced liver granuloma and fibrosis. In order to investigate the effect of a water-soluble complex preparation of BSE, BSE-CD, on S. japonicum egg-induced liver pathology, liver granuloma and fibrosis were induced by infecting C57BL/6 mice with 18-22 cercariae of S. japonicum. S. japonicum cercariae infected mice were injected with BSE-CD at the onset of egg granuloma formation (early phase BSE-CD treatment after 4 weeks infection) or after the formation of liver fibrosis (late phase BSE-CD treatment after 7 weeks infection). Our data show that treatment of infected mice with BSE-CD significantly reduced both the extent of hepatic granuloma and fibrosis. Consistent with an inhibition of NF-κB signaling as evidenced by reduced IκB kinase (IKK) activation, the mRNA expression of VEGF (vascular endothelial growth factor, VEGF), TNF-α (tumor necrosis factor-alpha TNF-α) and MCP-1 (monocyte chemotactic protein 1, MCP-1) was decreased. Moreover, immunohistochemical analysis (IHC) revealed that the content of α-SMA in liver tissue of BSE-CD treated mice was dramatically decreased. Our findings suggest that BSE-CD treatment attenuates S. japonicum egg-induced hepatic granulomas and fibrosis, at least partly due to reduced NF-κB signaling and the subsequently decreased expression of VEGF, TNF-α, and MCP-1. Suppression of the activation of hepatic stellate cells (HSC) may also be involved in the therapeutic efficacy of BSE-CD.


Subject(s)
Anthelmintics/pharmacology , Granuloma/drug therapy , Liver Cirrhosis/drug therapy , NF-kappa B/antagonists & inhibitors , Plant Extracts/pharmacology , Schistosomiasis japonica/drug therapy , Triterpenes/pharmacology , Actins/genetics , Actins/metabolism , Animals , Anthelmintics/chemistry , Cercaria/drug effects , Cercaria/physiology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression Regulation , Granuloma/genetics , Granuloma/parasitology , Granuloma/pathology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/parasitology , Liver/drug effects , Liver/metabolism , Liver/parasitology , Liver Cirrhosis/genetics , Liver Cirrhosis/parasitology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Parasite Egg Count , Plant Extracts/chemistry , Schistosoma japonicum/drug effects , Schistosoma japonicum/physiology , Schistosomiasis japonica/genetics , Schistosomiasis japonica/parasitology , Schistosomiasis japonica/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
10.
J Biol Chem ; 288(44): 31635-45, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24003215

ABSTRACT

Monocyte infiltration and macrophage formation are pivotal steps in atherosclerosis and plaque vulnerability. Gremlin-1/Drm is crucial in embryo-/organogenesis and has been shown to be expressed in the adult organism at sites of arterial injury and to inhibit monocyte migration. The purpose of the present study was to evaluate and characterize the role of Gremlin-1 in atherosclerosis. Here we report that Gremlin-1 is highly expressed primarily by monocytes/macrophages in aortic atherosclerotic lesions of ApoE(-/-) mice and is secreted from activated monocytes and during macrophage development in vitro. Gremlin-1 reduces macrophage formation by inhibiting macrophage migration inhibitory factor (MIF), a cytokine critically involved in atherosclerotic plaque progression and vulnerability. Gremlin-1 binds with high affinity to MIF (KD = 54 nm), as evidenced by surface plasmon resonance analysis and co-immunoprecipitation, and reduces MIF-induced release of TNF-α from macrophages. Treatment of ApoE(-/-) mice with a dimeric recombinant fusion protein, mGremlin1-Fc, but not with equimolar control Fc or inactivated mGremlin1-Fc, reduced TNF-α expression, the content of monocytes/macrophages of atherosclerotic lesions, and attenuated atheroprogression. The present data disclose that Gremlin-1 is an endogenous antagonist of MIF and define a role for Gremlin-1/MIF interaction in atherosclerosis.


Subject(s)
Apolipoproteins E , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/biosynthesis , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/pathology , Mice , Mice, Knockout , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/genetics
11.
Parasitol Res ; 112(3): 1105-11, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23271565

ABSTRACT

Granuloma formation has been shown to be induced and elicited by schistosome egg antigens, and it finally develops into fibrosis in intestine and the liver. Hepatic fibrosis is the main cause of increased morbidity and mortality in humans infected with schistosomes. Boswellic acid (BA)-containing extracts such as extracts of the oleogum resin from Boswellia serrata (BSE) have anti-inflammatory and immunomodulatory activity. However, little is known about the role of such extracts in schistosome egg-induced granulomatous inflammation. In order to investigate the effect of a watersoluble cyclodextrin complex preparation of BSE (BSE-CD) on Schistosoma japonicum (S. japonicum) egg-induced liver granuloma, mice infected with S. japonicum cercariae were injected with BSE-CD during egg granuloma formation. The data showed that BSE-CD significantly reduced the size of liver granuloma and levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST); however, BSE-CD treatment had no effect on worm load and egg burden. The data also showed that BSE-CD significantly decreased the expression of leukotriene B(4) (LTB(4)) and prostaglandin E(2) (PGE(2)), as well as the expression of matrix metallopeptidase 9 (MMP-9) in liver both on the mRNA and protein level. Thus, BSE-CD can significantly attenuate S. japonicum egg-induced hepatic granuloma, which may be partly dependent on the downregulation of some biochemical mediators.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Plant Extracts/administration & dosage , Schistosoma japonicum/drug effects , Schistosomiasis japonica/drug therapy , Triterpenes/administration & dosage , Alanine Transaminase/blood , Animals , Anti-Inflammatory Agents/isolation & purification , Boswellia/chemistry , Disease Models, Animal , Female , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/parasitology , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Parasite Egg Count , Parasite Load , Plant Extracts/isolation & purification , Schistosomiasis japonica/parasitology , Schistosomiasis japonica/pathology , Treatment Outcome , Triterpenes/isolation & purification
12.
Biochem Pharmacol ; 85(1): 38-45, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23085438

ABSTRACT

Malaria causes millions of death cases per year. Since Plasmodium falciparum rapidly develops drug resistance, it is of high importance to investigate potential drug targets which may lead to novel rational therapy approaches. Here we report on the interaction of translationally controlled tumor protein of P. falciparum (PfTCTP) with the anti-malarial drug artemisinin. Furthermore, we investigated the crystal structure of PfTCTP. Using mass spectrometry, bioinformatic approaches and surface plasmon resonance spectroscopy, we identified novel binding sites of artemisinin which are in direct neighborhood to amino acids 19-46, 108-134 and 140-163. The regions covered by these residues are known to be functionally important for TCTP function. We conclude that interaction of artemisinin with TCTP may be at least in part explain the antimalarial activity of artemisinin.


Subject(s)
Antimalarials/chemistry , Artemisinins/chemistry , Biomarkers, Tumor/chemistry , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Binding Sites , Biomarkers, Tumor/metabolism , Computer Simulation , Crystallography, X-Ray , Humans , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protozoan Proteins/metabolism , Recombinant Proteins/chemistry , Surface Plasmon Resonance , Tumor Protein, Translationally-Controlled 1
13.
Mol Pharmacol ; 83(2): 531-41, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23208958

ABSTRACT

The mammalian target of rapamycin (mTOR) is a key regulator of cell growth and its uncontrolled activation is a hallmark of cancer. Moreover, mTOR activation has been implicated in the resistance of cancer cells to many anticancer drugs, rendering this pathway a promising pharmacotherapeutic target. Here we explored the capability of a semisynthetic compound to intercept mTOR signaling. We synthesized and chemically characterized a novel, semisynthetic triterpenoid derivative, 3-cinnamoyl-11-keto-ß-boswellic acid (C-KßBA). Its pharmacodynamic effects on mTOR and several other signaling pathways were assessed in a number of prostate and breast cancer cell lines as well as in normal prostate epithelial cells. C-KßBA exhibits specific antiproliferative and proapoptotic effects in cancer cell lines in vitro as well as in PC-3 prostate cancer xenografts in vivo. Mechanistically, the compound significantly inhibits the cap-dependent transition machinery, decreases expression of eukaryotic translation initiation factor 4E and cyclin D1, and induces G(1) cell-cycle arrest. In contrast to conventional mTOR inhibitors, C-KßBA downregulates the phosphorylation of p70 ribosomal S6 kinase, the major downstream target of mTOR complex 1, without concomitant activation of mTOR complex 2/Akt and extracellular signal-regulated kinase pathways, and independently of protein phosphatase 2A, liver kinase B1/AMP-activated protein kinase/tuberous sclerosis complex, and F12-protein binding. At the molecular level, the compound binds to the FKBP12-rapamycin-binding domain of mTOR with high affinity, thereby competing with the endogenous mTOR activator phosphatidic acid. C-KßBA represents a new type of proapoptotic mTOR inhibitor that, due to its special mechanistic profile, might overcome the therapeutic drawbacks of conventional mTOR inhibitors.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triterpenes/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Down-Regulation/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , G1 Phase/drug effects , Humans , Male , Phosphorylation/drug effects , Prostatic Neoplasms/pathology , Protein Interaction Domains and Motifs , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
14.
J Nat Prod ; 74(8): 1731-6, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21800858

ABSTRACT

Mastic, a resinous exudate from Pistacia lentiscus, has been reported to exhibit selective cytotoxicity against different cancer cell lines. There are, however, no data published correlating distinct mastic-derived compounds with the postulated cytotoxic activity. A polypodane-type bicyclic triterpenoid, (8R)-3ß,8-dihydroxypolypoda-13E,17E,21-triene (1), was isolated from P. lentiscus oleogum resin. In androgen-independent PC-3 prostate cancer cells, 1 potently inhibited the expression of cyclins D1 and E, but had no effect on the expression of the cyclin kinase inhibitor p21(Waf1/Cip1). Inhibition of the expression of cell cycle-regulating cyclins resulted in cell cycle arrest in the G0/G1 phase, reduction in the number of cells in the S phase, and the triggering of apoptosis, as detected by increased expression of phosphatidylserine on the cell surface and by formation of DNA laddering. In addition, 1 suppressed the formation of prostate cancer colonies in soft agar and inhibited proliferation, angiogenesis, and the growth of prostate tumors xenografted onto chick chorioallantoic membranes without overt systemic toxicity. Taken together, these data show that 1 triggers apoptosis in chemoresistant, androgen-independent human prostate cancer cells in vitro and in vivo. Thus, 1 may serve as a lead compound for targeting so far incurable androgen-insensitive prostate cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Terpenes/isolation & purification , Terpenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Drug Resistance, Neoplasm/drug effects , Humans , Male , Mastic Resin , Molecular Structure , Prostatic Neoplasms/metabolism , Resins, Plant/chemistry , Resins, Plant/pharmacology , Stereoisomerism , Terpenes/chemistry
15.
J Mol Med (Berl) ; 88(9): 909-20, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20454888

ABSTRACT

The appearance of lipid-rich foam cells is a major feature of vulnerable atherosclerotic plaque formation. The transformation of macrophages into foam cells results from excessive uptake of cholesterol-rich particles by scavenger receptors such as CD68. We cloned a CD68-Fc immunoadhesin, a fusion protein consisting of the extracellular domain of the human CD68 and a human Fc domain, and investigated the function in vitro. Specific binding of CD68-Fc to OxLDL with an affinity of 10 nmol/L was determined by surface plasmon resonance and increased binding to lipid-rich human and ApoE(-/-) mice plaque tissue. This was confirmed both by immunohistochemical staining of CD68-Fc-treated paraffin sections from human plaques and by ELISA-based quantification of CD68-Fc binding to human atherosclerotic plaque extracts. In an in vitro model of macrophage/foam cell formation, CD68-Fc reduced foam cell formation significantly. This was caused both by interference of CD68-Fc with OxLDL uptake into macrophages and platelets and by the inhibition of platelet/OxLDL phagocytosis. Finally, expression of metalloproteinases by macrophages/foam cells was inhibited by CD68-Fc. In conclusion, CD68-Fc seems to be a promising new tool for preventing macrophage/foam cell formation. Thus, CD68-Fc might offer a novel therapeutic strategy for patients with acute coronary syndrome by modulating the generation of vulnerable plaques.


Subject(s)
Antigens, CD/chemistry , Antigens, Differentiation, Myelomonocytic/chemistry , Foam Cells/cytology , Immunoglobulin Fc Fragments/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Apolipoproteins E/metabolism , CHO Cells , Cricetinae , Cricetulus , Foam Cells/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Mice , Plaque, Atherosclerotic/metabolism , Receptors, Scavenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Surface Plasmon Resonance , Transfection
16.
Biomaterials ; 31(19): 5063-71, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20381862

ABSTRACT

Superparamagnetic iron oxide nanoparticles are frequently used for cell labeling or as diagnostic contrast media, yet studies analyzing their effects on immune cells remain scarce. Here we investigated how nanosized carboxydextran-coated superparamagnetic iron oxide (SPIO) and ultrasmall superparamagnetic iron oxide (USPIO) might affect human macrophages. Within 1 h, both SPIO and USPIO were rapidly taken up by macrophages. Confocal microscopy revealed that after 24 h the particles were almost exclusively localized within the lysosomal compartment. Continued cultivation of the macrophages for several days was associated with apoptosis induction caused by a long-lasting activation of the c-Jun N-terminal kinase (JNK) pathway. JNK activation was due to significantly elevated levels of reactive oxygen species, whereas no TNF-alpha was produced by the macrophages treated with nanoparticles. Compared to SPIO, USPIO induced more pronounced biochemical alterations and cytotoxicity, which could be antagonized by the JNK inhibitor V. Alternatively, treatment of macrophages with Trolox or N-acetyl-L-cysteine, two functionally different scavengers of reactive oxygen species, abolished both the JNK activation and the subsequent cytotoxic effects. These data indicate that nanosized superparamagnetic iron oxide-based contrast media exert cytotoxicity in human macrophages that can be functionally antagonized with radical scavengers.


Subject(s)
Apoptosis/physiology , Coated Materials, Biocompatible/administration & dosage , Dextrans/administration & dosage , Ferrosoferric Oxide/administration & dosage , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/cytology , Macrophages/enzymology , Nanoparticles/administration & dosage , Apoptosis/drug effects , Cells, Cultured , Humans , Macrophages/drug effects , Magnetite Nanoparticles
17.
Anal Biochem ; 401(1): 30-7, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20175984

ABSTRACT

Sesquiterpene lactones such as helenalin have generally been considered as highly promising compounds for the treatment of inflammatory disorders. Although sesquiterpene lactones are known to inhibit signaling through transcription factor nuclear factor-kappaB (NF-kappaB), the nature of their molecular targets remains controversial. To characterize the interactions of helenalin with putative target proteins, a surface plasmon resonance-based method was developed and validated to analyze the interactions of helenalin with the NF-kappaB protein p65/RelA, with recombinant IkappaB kinases (IKKs) alpha and beta, and with the intracellular antioxidant glutathione, all immobilized on sensor chips. At pH 7.4, helenalin is interacting with RelA (K(D)=4.8microM), yet it failed to bind either IKKalpha or IKKbeta. When DNA with NF-kappaB binding sites was immobilized on sensor chips, the binding of RelA was inhibited by helenalin with an IC(50) of 5.0microM. At pH 8.0, helenalin was also able to interact with reduced, but not oxidized, glutathione with a K(D) of 24microM, but no significant interaction was observed at pH 7.4. Thus, with this optimized method, we showed that the sesquiterpene lactone helenalin interacts with the NF-kappaB protein RelA but not with IKKalpha or IKKbeta. Moreover, at physiological pH, helenalin does not interact with glutathione to any significant extent.


Subject(s)
Sesquiterpenes/metabolism , Surface Plasmon Resonance/methods , Transcription Factor RelA/metabolism , Binding Sites , Biosensing Techniques , Glutathione/metabolism , Hydrogen-Ion Concentration , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Immobilized Proteins/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sesquiterpenes, Guaiane , Transcription Factor RelA/genetics
18.
Mol Pharmacol ; 77(3): 378-87, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20018812

ABSTRACT

Activation of the serine/threonine kinase Akt is associated with aggressive clinical behavior of prostate cancer. We found that the human prostate cancer cell lines LNCaP and PC-3 express predominantly Akt1 and Akt2. Selective down-regulation of Akt1, but not Akt2, by short-hairpin RNA reduced the viability of prostate cancer cells. In addition, structurally different Akt inhibitors were cytotoxic for the prostate cancer cells, confirming that the Akt pathway is indispensable for their viability. We have purified the tetracyclic triterpenoids 3-oxo-tirucallic acid, 3-alpha-acetoxy-tirucallic acid, and 3-beta-acetoxy-tirucallic acid from the oleogum resin of Boswellia carterii to chemical homogeneity. The acetoxy-derivatives in particular potently inhibited the activities of human recombinant Akt1 and Akt2 and of constitutively active Akt immunoprecipitated from PC-3 cells, whereas inhibitor of nuclear factor-kappaB kinases remained unaffected. Docking data indicated that these tetracyclic triterpenoids form hydrogen bonds within the phosphatidylinositol binding pocket of the Akt pleckstrin homology domain. Accordingly, 3-beta-acetoxy-tirucallic acid did not inhibit the activity of Akt1 lacking the pleckstrin homology domain. In the prostate cancer cell lines investigated, these compounds inhibited the phosphorylation of cellular Akt and the Akt signaling pathways, including glycogen synthase kinase-3beta and BAD phosphorylation, nuclear accumulation of p65, the androgen receptor, beta-catenin, and c-Myc. These events culminated in the induction of apoptosis in prostate cancer, but not in nontumorigenic cells. The tirucallic acid derivatives inhibited proliferation and induced apoptosis in tumors xenografted onto chick chorioallantoic membranes and decreased the growth of pre-established prostate tumors in nude mice without overt systemic toxicity. Thus, tirucallic acid derivatives represent a new class of Akt inhibitors with antitumor properties.


Subject(s)
Apoptosis/drug effects , Blood Proteins/chemistry , Phosphoproteins/chemistry , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Triterpenes/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Binding Sites/drug effects , Binding Sites/physiology , Blood Proteins/pharmacology , Blood Proteins/therapeutic use , Boswellia , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Nude , Phosphoproteins/pharmacology , Phosphoproteins/therapeutic use , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prostatic Neoplasms/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/physiology , Triterpenes/pharmacology , Triterpenes/therapeutic use , Xenograft Model Antitumor Assays/methods
19.
J Immunol ; 183(7): 4755-63, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19752240

ABSTRACT

Psoriasis vulgaris is a common chronic inflammatory skin disease involving cytokines and an activated cellular immune system. At variance to skin from patients with atopic dermatitis or from healthy subjects, human psoriatic skin lesions exhibit strong activation of transcription factor NF-kappaB that is mainly confined to dermal macrophages, whereas only a few dendritic cells but no CD3+ lymphocytes show activated NF-kappaB. Since NF-kappaB signaling is required for the induction and/or function of many cytokines and aberrant cytokine expression has been proposed as an underlying cause of psoriasis, we investigated whether NF-kappaB targeting would affect the course of the disease in the CD18 hypomorphic (CD18(hypo)) mouse model of psoriasis. When mice with severe psoriasiform lesions were treated systemically or locally with the IkappaB kinase inhibitor acetyl-11-keto-beta-boswellic acid (AKbetaBA), NF-kappaB signaling and the subsequent NF-kappaB-dependent cytokine production as shown by the TNF-alpha production of macrophages were profoundly suppressed. Additionally, application of the compound counteracted the intradermal MCP-1, IL-12, and IL-23 expression in previously lesional skin areas, led to resolution of the abundant immune cell infiltrates, and significantly reduced the increased proliferation of the keratinocytes. Overall, the AKbetaBA treatment was accompanied by a profound improvement of the psoriasis disease activity score in the CD18(hypo) mice with reconstitution of a nearly normal phenotype within the chosen observation period. Our data demonstrate that NF-kappaB signaling is pivotal for the pathogenesis in the CD18(hypo) mouse model of psoriasis. Therefore, targeting NF-kappaB might provide an effective strategy for the treatment of psoriasis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Drug Delivery Systems , Inflammation Mediators/therapeutic use , NF-kappa B/metabolism , Psoriasis/drug therapy , Psoriasis/immunology , Triterpenes/therapeutic use , Animals , Disease Models, Animal , Drug Delivery Systems/methods , Humans , Mice , Mice, Mutant Strains , Psoriasis/metabolism
20.
Thromb Haemost ; 101(4): 682-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19350111

ABSTRACT

The Extracellular Matrix Metalloproteinase Inducer (EMMPRIN, CD147, basigin) is an immunoglobulin-like receptor expressed in various cell types. During cellular interactions homotypic EMMPRIN-EMMPRIN interactions are known to induce the synthesis of matrix metalloproteinases. Recently, we have identified EMMPRIN as a novel receptor on platelets. To our knowledge EMMPRIN has not been shown to serve as adhesion receptor, yet. Here we characterise platelet glycoprotein VI (GPVI) as a novel adhesion receptor for EMMPRIN. Human platelets were prestimulated with ADP and perfused over immobilised recombinant EMMPRIN-Fc or Fc-fragments under arterial shear conditions. ADP-stimulated platelets showed significantly enhanced rolling (but not enhanced firm adhesion) on immobilised EMMPRIN-Fc compared to Fc. Pretreatment of platelets with blocking mAbs anti-EMMPRIN or anti-GPVI leads to a significant reduction of rolling platelets on immobilised EMMPRIN-Fc, whereas pretreatment with blocking mAbs anti-p-selectin, anti-alpha4-integrin or anti-GPIIb/IIIa complex (20 microg/ml each) had no effect. Consistently, chinese hamster ovary (CHO) cells stably transfected with GPVI showed enhanced rolling (but not adhesion) on immobilised EMMPRIN-Fc in comparison to non-transfected CHO cells. Similarly, CHO cells stably transfected with EMMPRIN showed enhanced rolling on immobilised GPVI-Fc (or EMMPRIN-Fc) compared to non transfected CHO-cells. Finally, specific binding of EMMPRIN to GPVI was demonstrated by a modified ELISA and surface plasmon resonance technology with a dissociation constant of 88 nM. Platelet GPVI is a novel receptor for EMMPRIN and can mediate platelet rolling via GPVI-EMMPRIN interaction.


Subject(s)
Basigin/metabolism , Blood Platelets/metabolism , CD36 Antigens/metabolism , Cell Movement , Platelet Activation , Adenosine Diphosphate/metabolism , Animals , Antibodies, Monoclonal , Basigin/genetics , Basigin/immunology , CD36 Antigens/genetics , CD36 Antigens/immunology , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , Humans , Mice , P-Selectin/metabolism , Platelet Adhesiveness , Protein Binding , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...