Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Science ; 374(6572): 1221-1227, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34855475

ABSTRACT

Increased blood levels of low-density lipoprotein cholesterol (LDL-C) and fibrinogen are independent risk factors for cardiovascular disease. We identified associations between an Amish-enriched missense variant (p.Asn352Ser) in a functional domain of beta-1,4-galactosyltransferase 1 (B4GALT1) and 13.9 milligrams per deciliter lower LDL-C (P = 4.1 × 10­19) and 29 milligrams per deciliter lower plasma fibrinogen (P = 1.3 × 10­5). B4GALT1 gene­based analysis in 544,955 subjects showed an association with decreased coronary artery disease (odds ratio = 0.64, P = 0.006). The mutant protein had 50% lower galactosyltransferase activity compared with the wild-type protein. N-linked glycan profiling of human serum found serine 352 allele to be associated with decreased galactosylation and sialylation of apolipoprotein B100, fibrinogen, immunoglobulin G, and transferrin. B4galt1 353Ser knock-in mice showed decreases in LDL-C and fibrinogen. Our findings suggest that targeted modulation of protein galactosylation may represent a therapeutic approach to decreasing cardiovascular disease.


Subject(s)
Cholesterol, LDL/blood , Fibrinogen/analysis , Galactosyltransferases/genetics , Mutation, Missense , Animals , Coronary Artery Disease/genetics , Coronary Artery Disease/prevention & control , Female , Galactose/metabolism , Galactosyltransferases/metabolism , Gene Knock-In Techniques , Gene Knockdown Techniques , Glycoproteins/blood , Glycosylation , Humans , Liver/enzymology , Male , Mice , N-Acetylneuraminic Acid/metabolism , Polysaccharides/blood , Whole Genome Sequencing
2.
Proteomics Clin Appl ; 7(3-4): 217-24, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23339088

ABSTRACT

PURPOSE: Evaluate combination of heat and elevated pressure to enhance protein extraction and quality of formalin-fixed (FF), and FF paraffin-embedded (FFPE) aorta for proteomics. EXPERIMENT DESIGN: Proteins were extracted from fresh frozen aorta at room temperature (RT). FF and FFPE aortas (3 months and 15 years) were extracted at RT, heat alone, or a combination of heat and high pressure. Protein yields were compared, and digested peptides from the extracts were analyzed with MS. RESULTS: Combined heat and elevated pressure increased protein yield from human FF or FFPE aorta compared to matched tissues with heat alone (1.5-fold) or at RT (8.3-fold), resulting in more proteins identified and with more sequence coverage. The length of storage did adversely affect the quality of proteins from FF tissue. For long-term storage, aorta was preserved better with FFPE than FF alone. Periostin and MGF-E8 were demonstrated suitable for MRM assays from FFPE aorta. CONCLUSIONS AND CLINICAL RELEVANCE: Combination of heat and high pressure is an effective method to extract proteins from FFPE aorta for downstream proteomics. This method opens the possibility for use of archival and often rare FFPE aortas and possibly other tissues available to proteomics for biomarker discovery and quantification.


Subject(s)
Aorta/metabolism , Fixatives/chemistry , Formaldehyde/chemistry , Paraffin Embedding/methods , Paraffin/chemistry , Proteins/isolation & purification , Aorta/pathology , Biomarkers/chemistry , Humans , Proteomics , Tissue Fixation/methods
3.
Eur J Pharmacol ; 698(1-3): 228-34, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23183107

ABSTRACT

The effect of the (R,S)-ketamine metabolites (R,S)-norketamine, (R,S)-dehydronorketamine, (2S,6S)-hydroxynorketamine and (2R,6R)-hydroxynorketamine on the activity of α7 and α3ß4 neuronal nicotinic acetylcholine receptors was investigated using patch-clamp techniques. The data indicated that (R,S)-dehydronorketamine inhibited acetylcholine-evoked currents in α7-nicotinic acetylcholine receptor, IC(50) = 55 ± 6 nM, and that (2S,6S)-hydroxynorketamine, (2R,6R)-hydroxynorketamine and (R,S)-norketamine also inhibited α7-nicotinic acetylcholine receptor function at concentrations ≤ 1 µM, while (R,S)-ketamine was inactive at these concentrations. The inhibitory effect of (R,S)-dehydronorketamine was voltage-independent and the compound did not competitively displace selective α7-nicotinic acetylcholine receptor ligands [(125)I]-α-bungarotoxin and [(3)H]-epibatidine indicating that (R,S)-dehydronorketamine is a negative allosteric modulator of the α7-nicotinic acetylcholine receptor. (R,S)-Ketamine and (R,S)-norketamine inhibited (S)-nicotine-induced whole-cell currents in cells expressing α3ß4-nicotinic acetylcholine receptor, IC(50) 3.1 and 9.1 µM, respectively, while (R,S)-dehydronorketamine, (2S,6S)-hydroxynorketamine and (2R,6R)-hydroxynorketamine were weak inhibitors, IC(50) >100 µM. The binding affinities of (R,S)-dehydronorketamine, (2S,6S)-hydroxynorketamine and (2R,6R)-hydroxynorketamine at the NMDA receptor were also determined using rat brain membranes and the selective NMDA receptor antagonist [(3)H]-MK-801. The calculated K(i) values were 38.95 µM for (S)-dehydronorketamine, 21.19 µM for (2S,6S)-hydroxynorketamine and>100 µM for (2R,6R)-hydroxynorketamine. The results suggest that the inhibitory activity of ketamine metabolites at the α7-nicotinic acetylcholine receptor may contribute to the clinical effect of the drug.


Subject(s)
Acetylcholine/antagonists & inhibitors , Acetylcholine/pharmacology , Electrophysiological Phenomena/drug effects , Ketamine/metabolism , Ketamine/pharmacology , Receptors, Nicotinic/metabolism , Anesthetics/chemistry , Anesthetics/metabolism , Anesthetics/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Ketamine/chemistry , Nicotinic Agonists/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , alpha7 Nicotinic Acetylcholine Receptor
4.
ChemMedChem ; 7(5): 920-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22383251

ABSTRACT

Three heterocyclic systems were selected as potential bioisosteres of the amide linker for a series of 1,6-disubstituted-4-quinolone-3-carboxamides, which are potent and selective CB2 ligands that exhibit poor water solubility, with the aim of improving their physicochemical profile and also of clarifying properties of importance for amide bond mimicry. Among the newly synthesized compounds, a 1,2,3-triazole derivative (1-(adamantan-1-yl)-4-[6-(furan-2-yl)-1,4-dihydro-4-oxo-1-pentylquinolin-3-yl]-1H-1,2,3-triazole) emerged as the most promising in terms of both physicochemical and pharmacodynamic properties. When assayed in vitro, this derivative exhibited inverse agonist activity, whereas, in the formalin test in mice, it produced analgesic effects antagonized by a well-established inverse agonist. Metabolic studies allowed the identification of a side chain hydroxylated derivative as its only metabolite, which, in its racemic form, still showed appreciable CB2 selectivity, but was 150-fold less potent than the parent compound.


Subject(s)
4-Quinolones/chemistry , Carboxylic Acids/chemistry , Computer Simulation , Receptor, Cannabinoid, CB2/agonists , Animals , CHO Cells , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Humans , Ligands , Lipids/chemistry , Mice , Models, Molecular , Molecular Structure , Protein Structure, Tertiary , Solubility , Structure-Activity Relationship
5.
Int J Biochem Cell Biol ; 42(9): 1525-35, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20684041

ABSTRACT

The interaction of ibogaine and phencyclidine (PCP) with human (h) alpha3beta4-nicotinic acetylcholine receptors (AChRs) in different conformational states was determined by functional and structural approaches including, radioligand binding assays, Ca2+ influx detections, and thermodynamic and kinetics measurements. The results established that (a) ibogaine inhibits (+/-)-epibatidine-induced Ca2+ influx in h(alpha)3beta4 AChRs with approximately 9-fold higher potency than that for PCP, (b) [3H]ibogaine binds to a single site in the h(alpha)3beta4 AChR ion channel with relatively high affinity (Kd = 0.46 +/- 0.06 microM), and ibogaine inhibits [3H]ibogaine binding to the desensitized h(alpha)3beta4 AChR with slightly higher affinity compared to the resting AChR. This is explained by a slower dissociation rate from the desensitized ion channel compared to the resting ion channel, and (c) PCP inhibits [3H]ibogaine binding to the h(alpha)3beta4 AChR, suggesting overlapping sites. The experimental results correlate with the docking simulations suggesting that ibogaine and PCP interact with a binding domain located between the serine (position 6') and valine/phenylalanine (position 13') rings. This interaction is mediated mainly by van der Waals contacts, which is in agreement with the observed enthalpic contribution determined by non-linear chromatography. However, the calculated entropic contribution also indicates local conformational changes. Collectively our data suggest that ibogaine and PCP bind to overlapping sites located between the serine and valine/phenylalanine rings, to finally block the AChR ion channel, and in the case of ibogaine, to probably maintain the AChR in the desensitized state for longer time.


Subject(s)
Ibogaine/metabolism , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/metabolism , Biological Transport/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcium/metabolism , Cell Line , Humans , Kinetics , Phencyclidine/pharmacology , Protein Binding , Protein Structure, Secondary , Pyridines/pharmacology , Thermodynamics
6.
Biochim Biophys Acta ; 1798(6): 1153-63, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20303928

ABSTRACT

The interaction of 18-methoxycoronaridine (18-MC) with nicotinic acetylcholine receptors (AChRs) was compared with that for ibogaine and phencyclidine (PCP). The results established that 18-MC: (a) is more potent than ibogaine and PCP inhibiting (+/-)-epibatidine-induced AChR Ca(2+) influx. The potency of 18-MC is increased after longer pre-incubation periods, which is in agreement with the enhancement of [(3)H]cytisine binding to resting but activatable Torpedo AChRs, (b) binds to a single site in the Torpedo AChR with high affinity and inhibits [(3)H]TCP binding to desensitized AChRs in a steric fashion, suggesting the existence of overlapping sites. This is supported by our docking results indicating that 18-MC interacts with a domain located between the serine (position 6') and valine (position 13') rings, and (c) inhibits [(3)H]TCP, [(3)H]ibogaine, and [(3)H]18-MC binding to desensitized AChRs with higher affinity compared to resting AChRs. This can be partially attributed to a slower dissociation rate from the desensitized AChR compared to that from the resting AChR. The enthalpic contribution is more important than the entropic contribution when 18-MC binds to the desensitized AChR compared to that for the resting AChR, and vice versa. Ibogaine analogs inhibit the AChR by interacting with a luminal domain that is shared with PCP, and by inducing desensitization.


Subject(s)
Cholinergic Antagonists/chemistry , Electric Organ/chemistry , Fish Proteins/chemistry , Ibogaine/analogs & derivatives , Receptors, Cholinergic/chemistry , Torpedo , Animals , Binding Sites , Ibogaine/chemistry , Protein Binding , Protein Structure, Tertiary
7.
Int J Biochem Cell Biol ; 42(6): 1007-18, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20223294

ABSTRACT

The interaction of tricyclic antidepressants with the human (h) alpha4beta2 nicotinic acetylcholine receptor in different conformational states was compared with that for the noncompetitive antagonist mecamylamine by using functional and structural approaches. The results established that: (a) [(3)H]imipramine binds to halpha4beta2 receptors with relatively high affinity (K(d)=0.83+/-0.08 microM), but imipramine does not differentiate between the desensitized and resting states, (b) although tricyclic antidepressants inhibit (+/-)-epibatidine-induced Ca(2+) influx in HEK293-halpha4beta2 cells with potencies that are in the same concentration range as that for (+/-)-mecamylamine, tricyclic antidepressants inhibit [(3)H]imipramine binding to halpha4beta2 receptors with affinities >100-fold higher than that for (+/-)-mecamylamine. This can be explained by our docking results where imipramine interacts with the leucine (position 9') and valine (position 13') rings by van der Waals contacts, whereas mecamylamine interacts electrostatically with the outer ring (position 20'), (c) van der Waals interactions are in agreement with the thermodynamic results, indicating that imipramine interacts with the desensitized and resting receptors by a combination of enthalpic and entropic components. However, the entropic component is more important in the desensitized state, suggesting local conformational changes. In conclusion, our data indicate that tricyclic antidepressants and mecamylamine efficiently inhibit the ion channel by interacting at different luminal sites. The high proportion of protonated mecamylamine calculated at physiological pH suggests that this drug can be attracted to the channel mouth before binding deeper within the receptor ion channel finally blocking ion flux.


Subject(s)
Antidepressive Agents, Tricyclic/metabolism , Imipramine/metabolism , Ion Channels/metabolism , Mecamylamine/metabolism , Nicotinic Antagonists/metabolism , Receptors, Nicotinic/metabolism , Antidepressive Agents, Tricyclic/chemistry , Antidepressive Agents, Tricyclic/pharmacology , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcium Signaling/drug effects , Cell Line , Humans , Imipramine/chemistry , Imipramine/pharmacology , Ion Channels/chemistry , Ion Channels/genetics , Mecamylamine/chemistry , Mecamylamine/pharmacology , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/pharmacology , Protein Binding , Protein Conformation/drug effects , Pyridines/pharmacology , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics
8.
Biochemistry ; 48(21): 4506-18, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19334677

ABSTRACT

To characterize the binding sites and the mechanisms of inhibition of bupropion on muscle-type nicotinic acetylcholine receptors (AChRs), structural and functional approaches were used. The results established that bupropion (a) inhibits epibatidine-induced Ca(2+) influx in embryonic muscle AChRs, (b) inhibits adult muscle AChR macroscopic currents in the resting/activatable state with approximately 100-fold higher potency compared to that in the open state, (c) increases the desensitization rate of adult muscle AChRs from the open state and impairs channel opening from the resting state, (d) inhibits binding of [(3)H]TCP and [(3)H]imipramine to the desensitized/carbamylcholine-bound Torpedo AChR with higher affinity compared to the resting/alpha-bungarotoxin-bound AChR, (e) binds to the Torpedo AChR in either state mainly by an entropy-driven process, and (f) interacts with a binding domain located between the serine (position 6') and valine (position 13') rings, by a network of van der Waals, hydrogen bond, and polar interactions. Collectively, our data indicate that bupropion first binds to the resting AChR, decreasing the probability of ion channel opening. The remnant fraction of open ion channels is subsequently decreased by accelerating the desensitization process. Bupropion interacts with a luminal binding domain shared with PCP that is located between the serine and valine rings, and this interaction is mediated mainly by an entropy-driven process.


Subject(s)
Bupropion/metabolism , Muscles/metabolism , Nicotinic Antagonists/metabolism , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/metabolism , Animals , Binding, Competitive , Biological Transport/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bupropion/pharmacology , Calcium/metabolism , Cell Line , Drug Discovery , Electric Conductivity , Humans , Imipramine/metabolism , Immobilized Proteins/antagonists & inhibitors , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Ion Channel Gating , Kinetics , Mice , Models, Molecular , Nicotinic Antagonists/pharmacology , Piperidines/chemistry , Piperidines/metabolism , Protein Binding , Protein Conformation , Pyridines/pharmacology , Thermodynamics , Torpedo
SELECTION OF CITATIONS
SEARCH DETAIL
...