Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Physiol ; 235(3): 2441-2451, 2020 03.
Article in English | MEDLINE | ID: mdl-31556103

ABSTRACT

Acupuncture has many advantages in the treatment of certain diseases as opposed to drug therapy. Besides, adenosine has been revealed to affect cellular progression including proliferation. Therefore, this study aimed at exploring the mechanism involving acupuncture stress and adenosine in fibroblast proliferation. The fibroblasts from fascia tissues of the acupoint area (Zusanli) were stimulated by different levels of stress, different concentrations of adenosine, and agonist or antagonist of A3 receptor (A3 R) to investigate the effect of stress stimulation, adenosine, and adenosine-A3 R inhibition on fibroblasts. Then, the fibroblasts were treated with stress stimulation of 200 kPa or/and mitogen-activated protein kinase (MAPK) blocker. We revealed that stress stimulation and the binding of adenosine and A3 R promoted fibroblast proliferation in the fascial tissue, increased the expression of immune-related factors, adenosine and A3 R, and activated the MAPK signaling pathway. MAPK signaling pathway also directly affected the expression of adenosine, A3 R, and immune-related factors. Stress stimulation and adenosine treatment upregulated A3 R expression, and then activated the MAPK signaling pathway, which could in turn upregulate expression of adenosine, A3 R and immune-related factors, and promote cell proliferation. Adenosine is shown to form a positive feedback loop with the MAPK signaling pathway. Collectively, stress stimulation in vitro induces the increase of adenosine in fibroblasts through the energy metabolism and activation of the MAPK signaling pathway through A3 R, ultimately promoting fibroblast proliferation.


Subject(s)
Acupuncture/methods , Adenosine/genetics , Energy Metabolism/genetics , Receptor, Adenosine A3/genetics , Acupuncture Points , Adenosine A3 Receptor Agonists/pharmacology , Animals , Cell Proliferation/genetics , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System/genetics , Microscopy, Confocal , Primary Cell Culture , Rats , Signal Transduction/drug effects
2.
FASEB J ; 26(12): 5106-14, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22982377

ABSTRACT

In nonhuman primates, we previously demonstrated that a maternal high-fat diet (MHFD) induces fetal nonalcoholic fatty liver disease (NAFLD) and alters the fetal metabolome. These changes are accompanied by altered acetylation of histone H3 (H3K14ac). However, the mechanism behind this alteration in acetylation remains unknown. As SIRT1 is both a lysine deacetylase and a crucial sensor of cellular metabolism, we hypothesized that SIRT1 may be involved in fetal epigenomic alterations. Here we show that in utero exposure to a MHFD, but not maternal obesity per se, increases fetal H3K14ac with concomitant decreased SIRT1 expression and diminished in vitro protein and histone deacetylase activity. MHFD increased H3K14ac and DBC1-SIRT1 complex formation in fetal livers, both of which were abrogated with diet reversal despite persistent maternal obesity. Moreover, MHFD was associated with altered expression of known downstream effectors deregulated in NAFLD and modulated by SIRT1 (e.g., PPARΑ, PPARG, SREBF1, CYP7A1, FASN, and SCD). Finally, ex vivo purified SIRT1 retains deacetylase activity on an H3K14ac peptide substrate with preferential activity toward acetylated histone H3; mutagenesis of the catalytic domain of SIRT1 (H363Y) abrogates H3K14ac deacetylation. Our data implicate SIRT1 as a likely molecular mediator of the fetal epigenome and metabolome under MHFD conditions.


Subject(s)
Diet, High-Fat , Histones/metabolism , Macaca/metabolism , Sirtuin 1/metabolism , Acetylation , Animals , Blotting, Western , COS Cells , Catalytic Domain/genetics , Female , Fetus/enzymology , Gene Expression Regulation, Developmental , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Liver/embryology , Liver/metabolism , Lysine/metabolism , Macaca/embryology , Macaca/genetics , Male , Mass Spectrometry , Mutation , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1/genetics , Substrate Specificity , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
3.
PLoS One ; 6(11): e27586, 2011.
Article in English | MEDLINE | ID: mdl-22110680

ABSTRACT

Endothelium-leukocyte interaction is critical for inflammatory responses. Whereas the tissue microenvironments are often acidic at inflammatory sites, the mechanisms by which cells respond to acidosis are not well understood. Using molecular, cellular and biochemical approaches, we demonstrate that activation of GPR4, a proton-sensing G protein-coupled receptor, by isocapnic acidosis increases the adhesiveness of human umbilical vein endothelial cells (HUVECs) that express GPR4 endogenously. Acidosis in combination with GPR4 overexpression further augments HUVEC adhesion with U937 monocytes. In contrast, overexpression of a G protein signaling-defective DRY motif mutant (R115A) of GPR4 does not elicit any increase of HUVEC adhesion, indicating the requirement of G protein signaling. Downregulation of GPR4 expression by RNA interference reduces the acidosis-induced HUVEC adhesion. To delineate downstream pathways, we show that inhibition of adenylate cyclase by inhibitors, 2',5'-dideoxyadenosine (DDA) or SQ 22536, attenuates acidosis/GPR4-induced HUVEC adhesion. Consistently, treatment with a cAMP analog or a G(i) signaling inhibitor increases HUVEC adhesiveness, suggesting a role of the G(s)/cAMP signaling in this process. We further show that the cAMP downstream effector Epac is important for acidosis/GPR4-induced cell adhesion. Moreover, activation of GPR4 by acidosis increases the expression of vascular adhesion molecules E-selectin, VCAM-1 and ICAM-1, which are functionally involved in acidosis/GPR4-mediated HUVEC adhesion. Similarly, hypercapnic acidosis can also activate GPR4 to stimulate HUVEC adhesion molecule expression and adhesiveness. These results suggest that acidosis/GPR4 signaling regulates endothelial cell adhesion mainly through the G(s)/cAMP/Epac pathway and may play a role in the inflammatory response of vascular endothelial cells.


Subject(s)
Acidosis/metabolism , Acidosis/pathology , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Acidosis/genetics , Blood Vessels/metabolism , Blood Vessels/physiology , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen-Ion Concentration , Receptors, G-Protein-Coupled/genetics , Up-Regulation
4.
J Med Chem ; 46(23): 4847-59, 2003 Nov 06.
Article in English | MEDLINE | ID: mdl-14584936

ABSTRACT

A three-dimensional model of the human A(2A) adenosine receptor (AR) and its docked ligands was built by homology to rhodopsin and validated with site-directed mutagenesis and the synthesis of chemically complementary agonists. Different binding modes of A(2A)AR antagonists and agonists were compared by using the FlexiDock automated docking procedure, with manual adjustment. Putative binding regions for the 9H-purine ring in agonist NECA 3 and the 1H-[1,2,4]triazolo[1,5-c]quinazoline ring in antagonist CGS15943 1 overlapped, and the exocyclic amino groups of each were H-bonded to the side chain of N(6.55). For bound agonist, H-bonds formed between the ribose 3'- and 5'-substituents and the hydrophilic amino acids T(3.36), S(7.42), and H(7.43), and the terminal methyl group of the 5'-uronamide interacted with the hydrophobic side chain of F(6.44). Formation of the agonist complex destabilized the ground-state structure of the A(2A)AR, which was stabilized through a network of H-bonding and hydrophobic interactions in the transmembrane helical domain (TM) regions, facilitating a conformational change upon activation. Both flexibility of the ribose moiety, required for the movement of TM6, and its H-bonding to the receptor were important for agonism. Two sets of interhelical H-bonds involved residues conserved among ARs but not in rhodopsin: (1) E13(1.39) and H278(7.43) and (2) D52(2.50), with the highly conserved amino acids N280(7.45) and S281(7.46), and N284(7.49) with S91(3.39). Most of the amino acid residues lining the putative binding site(s) were conserved among the four AR subtypes. The A(2A)AR/3 complex showed a preference for an intermediate conformation about the glycosidic bond, unlike in the A(3)AR/3 complex, which featured an anti-conformation. Hydrophilic amino acids of TMs 3 and 7 (ribose-binding region) were replaced with anionic residues for enhanced binding to amine-derivatized agonists. We identified new neoceptor (T88D)-neoligand pairs that were consistent with the model.


Subject(s)
Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Adenosine-5'-(N-ethylcarboxamide)/chemistry , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Binding Sites , COS Cells , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Quinazolines/chemistry , Quinazolines/pharmacology , Radioligand Assay , Receptors, Adenosine A2/genetics , Triazoles/chemistry , Triazoles/pharmacology
5.
Mol Pharmacol ; 63(5): 1021-31, 2003 May.
Article in English | MEDLINE | ID: mdl-12695530

ABSTRACT

We examined the effects on allosteric modulation and ligand binding of the mutation of amino acid residues of the human A(3) adenosine receptor (A(3)AR) that are hypothesized to be near one of three loci: the putative sodium binding site, the putative ligand binding site, and the DRY motif in transmembrane helical domain 3. The effects of three heterocyclic allosteric modulators [the imidazoquinoline 2-cyclopentyl-4-phenylamino-1H-imidazo[4,5-c]quinoline (DU124183), the pyridinylisoquinoline 4-methoxy-N-[7-methyl-3-(2-pyridinyl)-1-isoquinolinyl]benzamide (VUF5455), and the amiloride analog 5-(N,N-hexamethylene)-amiloride] on the dissociation of the agonist radioligand, N(6)- (4-amino-3-[(125)I]iodobenzyl)-5'-N-methylcarboxamidoadenosine, were compared at wild-type (WT) and mutant A(3)ARs. The F182A(5.43) and N274A(7.45) mutations eliminated the allosteric effects of all three modulators but had little effect on agonist binding. The N30A(1.50) and D58N(2.50) mutations abolished the allosteric effects of DU124183 and VUF5455, but not HMA, whereas the D107N(3.49) mutation abolished the effects of DU124183, but not HMA or VUF5455. The T94A(3.36), H95A(3.37), K152A(EL2), W243A(6.48), L244A(6.49), and S247A(6.52) mutations did not influence allosteric effects of the modulators. Sodium ions (100 mM), which modulate agonist binding at a variety of receptors, caused an approximately 80% inhibition of agonist binding in WT A(3)ARs but did not show any effect on D58N(2.50), D107N(3.49), and F182A(5.43) mutant receptors. In contrast, NaCl induced a modest increase of agonist binding in N30A(1.50) and N274A(7.45) mutant receptors. NaCl decreased the dissociation rate of the antagonist radioligand [(3)H]8-ethyl-4-methyl-2-phenyl-(8R)-4,5,7,8-tetrahydro-1H-imidazo[2.1-i]purin-5-one (PSB-11) at the WT A(3)ARs, but not the D58N(2.50) mutant receptor. The results were interpreted using a rhodopsin-based molecular model of the A(3)AR to suggest multiple binding modes of the allosteric modulators.


Subject(s)
Adenosine/analogs & derivatives , Receptors, Purinergic P1/metabolism , Adenosine/pharmacology , Allosteric Regulation , Amino Acid Motifs , Animals , COS Cells , Humans , Imidazoles/pharmacology , Iodine Radioisotopes , Models, Molecular , Mutation , Protein Structure, Tertiary , Purines/pharmacology , Radioligand Assay , Receptor, Adenosine A3 , Receptors, Purinergic P1/chemistry , Sodium/pharmacology , Tritium
6.
Drug Dev Res ; 58(4): 330-339, 2003 Apr.
Article in English | MEDLINE | ID: mdl-35799977

ABSTRACT

Modification of the ribose moiety of nucleotides and nucleosides has provided new insights into structural and conformational requirements for ligands at P2Y nucleotide receptors and at adenosine receptors (ARs). Methanocarba derivatives (containing a rigid bicyclic ring system in place of ribose) of adenosine, ATP, ADP, UTP, UDP, and other receptor agonist analogs were synthesized. Biological evaluation led to the conclusion that in general the Northern (N)-conformation was favored over the Southern (S)-conformation of the pseudoribose moiety at A1 and A3 ARs and at P2Y1, P2Y2, P2Y4, or P2Y11 receptors, but not P2Y6 receptors. At the hA3 AR a new full agonist, MRS1898, the (N)-methanocarba equivalent of CI-IB-MECA (2-chloro-N 6-(3-iodobenzyl)-5'-N-methylcarbamoyladenosine), had a Ki value of 1.9 nM in binding to the hA3 AR expressed in CHO cells. Functional assays confirmed the selectivity of MRS1898, although CI-IB-MECA was even more functionally selective for human A3 vs. hA1 and hA2A ARs. Thirty µM MRS1898 did not induce apoptosis in HL-60 cells, suggesting that some of the proapoptotic effects of CI-IB-MECA may be nonreceptor-mediated. Manipulation of the sequence of A3 ARs through site-directed mutagenesis has led to pharmacologically unique constructs: constitutively active receptors and "neoceptors." Such engineered receptors may later prove to have potential for cardioprotection through gene transfer. Effects of single amino acid replacement were interpreted using a rhodopsin-based model of ligand-A3 receptor interactions, leading to the proposal that a movement of the conserved W243 in TM6 may be involved in AR activation.

7.
J Med Chem ; 45(26): 5694-709, 2002 Dec 19.
Article in English | MEDLINE | ID: mdl-12477353

ABSTRACT

Activation by ADP of both P2Y(1) and P2Y(12) receptors in platelets contributes to platelet aggregation, and antagonists at these receptor subtypes have antithrombotic properties. In an earlier publication, we have characterized the SAR as P2Y(1) receptor antagonists of acyclic analogues of adenine nucleotides, containing two phosphate groups on a symmetrically branched aliphatic chain, attached at the 9-position of adenine. In this study, we have focused on antiaggregatory effects of P2Y antagonists related to a 2-chloro-N(6)-methyladenine-9-(2-methylpropyl) scaffold, containing uncharged substitutions of the phosphate groups. For the known nucleotide (cyclic and acyclic) bisphosphate antagonists of P2Y(1) receptors, there was a significant correlation between inhibition of aggregation induced by 3.3 microM ADP in rat platelets and inhibition of P2Y(1) receptor-induced phospholipase C (PLC) activity previously determined in turkey erythrocytes. Substitution of the phosphate groups with nonhydrolyzable phosphonate groups preserved platelet antiaggregatory activity. Substitution of one of the phosphate groups with O-acyl greatly reduced the inhibitory potency, which tended to increase upon replacement of both phosphate moieties of the acyclic derivatives with uncharged (e.g., ester) groups. In the series of nonsymmetrically substituted monophosphates, the optimal antagonist potency occurred with the phenylcarbamate group. Among symmetrical diester derivatives, the optimal antagonist potency occurred with the di(phenylacetyl) group. A dipivaloyl derivative, a representative uncharged diester, inhibited ADP-induced aggregation in both rat (K(I) 3.6 microM) and human platelets. It antagonized the ADP-induced inhibition of the cyclic AMP pathway in rat platelets (IC(50) 7 microM) but did not affect hP2Y(1) receptor-induced PLC activity measured in transfected astrocytoma cells. We propose that the uncharged derivatives are acting as antagonists of a parallel pro-aggregatory receptor present on platelets, that is, the P2Y(12) receptor. Thus, different substitution of the same nucleoside scaffold can target either of two P2Y receptors in platelets.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemical synthesis , Platelet Aggregation Inhibitors/chemical synthesis , Purinergic P2 Receptor Antagonists , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Line , Cyclic AMP/metabolism , Humans , In Vitro Techniques , Inositol Phosphates/metabolism , Nucleosides/chemical synthesis , Nucleosides/chemistry , Nucleosides/pharmacology , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y1 , Structure-Activity Relationship , Type C Phospholipases/metabolism
8.
J Biol Chem ; 277(21): 19056-63, 2002 May 24.
Article in English | MEDLINE | ID: mdl-11891221

ABSTRACT

Ligand recognition has been extensively explored in G protein-coupled A(1), A(2A), and A(2B) adenosine receptors but not in the A(3) receptor, which is cerebroprotective and cardioprotective. We mutated several residues of the human A(3) adenosine receptor within transmembrane domains 3 and 6 and the second extracellular loop, which have been predicted by previous molecular modeling to be involved in the ligand recognition, including His(95), Trp(243), Leu(244), Ser(247), Asn(250), and Lys(152). The N250A mutant receptor lost the ability to bind both radiolabeled agonist and antagonist. The H95A mutation significantly reduced affinity of both agonists and antagonists. In contrast, the K152A (EL2), W243A (6.48), and W243F (6.48) mutations did not significantly affect the agonist binding but decreased antagonist affinity by approximately 3-38-fold, suggesting that these residues were critical for the high affinity of A(3) adenosine receptor antagonists. Activation of phospholipase C by wild type (WT) and mutant receptors was measured. The A(3) agonist 2-chloro-N(6)-(3-iodobenzyl)-5'-N-methylcarbamoyladenosine stimulated phosphoinositide turnover in the WT but failed to evoke a response in cells expressing W243A and W243F mutant receptors, in which agonist binding was less sensitive to guanosine 5'-gamma-thiotriphosphate than in WT. Thus, although not important for agonist binding, Trp(243) was critical for receptor activation. The results were interpreted using a rhodopsin-based model of ligand-A(3) receptor interactions.


Subject(s)
Receptors, Purinergic P1/metabolism , Amino Acid Sequence , Animals , COS Cells , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphatidylinositols/metabolism , Purinergic P1 Receptor Agonists , Receptor, Adenosine A3 , Receptors, Purinergic P1/chemistry , Receptors, Purinergic P1/genetics , Sequence Homology, Amino Acid
9.
Biochem Pharmacol ; 63(5): 871-80, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11911839

ABSTRACT

A(3) adenosine receptor (A(3)AR) agonists have been reported to influence cell death and survival. The effects of an A(3)AR agonist, 1-[2-chloro-6-[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-1-deoxy-N-methyl-beta-D-ribofuranonamide (Cl-IB-MECA), on apoptosis in two human leukemia cell lines, HL-60 and MOLT-4, were investigated. Cl-IB-MECA (> or =30 microM) increased the apoptotic fractions, as determined using fluorescence-activated cell sorting (FACS) analysis, and activated caspase 3 and poly-ADP-ribose-polymerase. Known messengers coupled to A(3)AR (phospholipase C and intracellular calcium) did not seem to play a role in the induction of apoptosis. Neither dantrolene nor BAPTA-AM affected the Cl-IB-MECA-induced apoptosis. Cl-IB-MECA failed to activate phospholipase C in HL-60 cells, while UTP activated it through endogenous P2Y(2) receptors. Induction of apoptosis during a 48hr exposure to Cl-IB-MECA was not prevented by the A(3)AR antagonists [5-propyl-2-ethyl-4-propyl-3-(ethylsulfanylcarbonyl)-6-phenylpyridine-5-carboxylate] (MRS 1220) or N-[9-chloro-2-(2-furanyl)[1,2,4]triazolo[1,5-c]quinazolin-5-yl]benzeneacetamide (MRS 1523). Furthermore, higher concentrations of MRS 1220, which would also antagonize A(1) and A(2A) receptors, were ineffective in preventing the apoptosis. Although Cl-IB-MECA has been shown in other systems to cause apoptosis through an A(3)AR-mediated mechanism, in these cells it appeared to be an adenosine receptor-independent effect, which required prolonged incubation. In both HL-60 and MOLT-4 cells, Cl-IB-MECA induced the expression of Fas, a death receptor. This induction of Fas was not dependent upon p53, because p53 is not expressed in an active form in either HL-60 or MOLT-4 cells. Cl-IB-MECA-induced apoptosis in HL-60 cells was augmented by an agonistic Fas antibody, CH-11, and this increase was suppressed by the antagonistic anti-Fas antibody ZB-4. Therefore, Cl-IB-MECA induced apoptosis via a novel, p53-independent up-regulation of Fas.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/pharmacology , Apoptosis , Tumor Suppressor Protein p53/metabolism , Type C Phospholipases/metabolism , fas Receptor/biosynthesis , Antibodies/pharmacology , Blotting, Western , Calcium/metabolism , Drug Interactions , HL-60 Cells , Humans , Leukemia/pathology , Purinergic P1 Receptor Antagonists , Receptor, Adenosine A3 , Tumor Cells, Cultured , fas Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...