Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Res ; 61(4): 1486-92, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245455

ABSTRACT

A high-throughput screening assay was designed to select compounds that inhibit the growth of cultured mammalian cells. After screening more than 60,000 compounds, A-105972 was identified and selected for further testing. A-105972 is a small molecule that inhibits the growth of breast, central nervous system, colon, liver, lung, and prostate cancer cell lines, including multidrug-resistant cells. The cytotoxic IC50 values of A-105972 were between 20 and 200 nM, depending on the specific cell type. The potency of A-105972 is similar in cells expressing wild-type or mutant p53. A majority of cells treated with A-105972 were trapped in the G2-M phases, suggesting that A-105972 inhibits the progression of the cell cycle. Using [3H]A-105972, we found that A-105972 bound to purified tubulin. Unlabeled A-105972 competed with [3H]A-105972 binding with an IC50 value of 3.6 microL. Colchicine partially inhibited [3H]A-105972 binding with an IC50 value of approximately 90 microM, whereas paclitaxel and vinblastine had no significant effect. Tumor cells treated with A-105972 were observed to contain abnormal microtubule arrangement and apoptotic bodies. DNA ladder studies also indicated that A-105972 induced apoptosis. A-105972 caused a mobility shift of bcl-2 on SDS-PAGE, suggesting that A-105972 induced bcl-2 phosphorylation. A-105972 treatment increased the life span of mice inoculated with B16 melanoma, P388 leukemia, and Adriamycin-resistant P388. These results suggest that A-105972 is a small molecule that interacts with microtubules, arrests cells in G2-M phases, and induces apoptosis in both multidrug resistance-negative and multidrug resistance-positive cancer cells. A-105972 and its analogues may be useful for treating cell proliferative disorders such as cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Oxadiazoles/pharmacology , Animals , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Inhibitory Concentration 50 , Leukemia P388/drug therapy , Leukemia P388/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Microtubules/drug effects , Microtubules/metabolism , Oxadiazoles/metabolism , Phosphorylation/drug effects , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin/metabolism , Tumor Cells, Cultured/drug effects
2.
Life Sci ; 66(10): 937-45, 2000.
Article in English | MEDLINE | ID: mdl-10714894

ABSTRACT

An assay using scintillation proximity bead technology has been developed suitable for the quantitation of endothelin (ET) receptor antagonists in preclinical and clinical samples of plasma. The assay measures the competitive inhibition of radiolabelled ET-1 binding to ET(A) receptor membranes bound to wheat germ agglutinin (WGA)-coated scintillation proximity assay (SPA) beads in the presence of plasma containing A-127722, a potent orally active, ET(A) selective ET antagonist. The assay requires as little as 50 microl plasma and no extraction procedure is needed. The SPA methodology eliminates the need for the separation of bound from free ligand. Using this method, A-127722 could be directly quantified in rat plasma with a detection limit of 1 ng/ml.


Subject(s)
Endothelin Receptor Antagonists , Pyrrolidines/blood , Animals , Atrasentan , CHO Cells , Chromatography, High Pressure Liquid , Cricetinae , Endothelin-1/metabolism , Humans , Radioligand Assay , Rats , Receptors, Endothelin/metabolism , Recombinant Proteins/antagonists & inhibitors , Scintillation Counting , Spectrometry, Fluorescence
3.
Eur J Pharmacol ; 388(1): 29-35, 2000 Jan 24.
Article in English | MEDLINE | ID: mdl-10657544

ABSTRACT

Tactile allodynia, the enhanced perception of pain in response to normally non-painful stimulation, represents a common complication of diabetic neuropathy. The activation of endothelin ET(A) receptors has been implicated in diabetes-induced reductions in peripheral neurovascularization and concomitant endoneurial hypoxia. Endothelin receptor activation has also been shown to alter the peripheral and central processing of nociceptive information. The present study was conducted to evaluate the antinociceptive effects of the novel endothelin ET(A) receptor-selective antagonist, 2R-(4-methoxyphenyl)-4S-(1,3-benzodioxol-5-yl)-1-(N, N-di(n-butyl)aminocarbonyl-methyl)-pyrrolidine-3R-carboxylic acid (ABT-627), in the streptozotocin-induced diabetic rat model of neuropathic pain. Rats were injected with 75 mg/kg streptozotocin (i. p.), and drug effects were assessed 8-12 weeks following streptozotocin treatment to allow for stabilization of blood glucose levels (>/=240 mg/dl) and tactile allodynia thresholds (

Subject(s)
Analgesics, Non-Narcotic/pharmacology , Diabetic Neuropathies/complications , Endothelin Receptor Antagonists , Pain/drug therapy , Pyrrolidines/pharmacology , Analgesics, Non-Narcotic/blood , Animals , Atrasentan , Diabetes Mellitus, Experimental/physiopathology , Humans , Male , Pain/etiology , Pain Measurement/drug effects , Physical Stimulation , Pyrrolidines/blood , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptor, Endothelin B , Stereoisomerism , Vasoconstrictor Agents/pharmacology
4.
Am J Physiol Renal Physiol ; 278(2): F279-86, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10662732

ABSTRACT

Experiments were designed to elucidate the role of endothelin B receptors (ET(B)) on arterial pressure and renal function in deoxycorticosterone acetate (DOCA)-salt hypertensive rats. Male Sprague-Dawley rats underwent uninephrectomy and were treated with either DOCA and salt (0.9% NaCl to drink) or placebo. DOCA-salt rats given the ET(B)-selective antagonist, A-192621, for 1 wk (10 mg. kg(-1). day(-1) in the food) had significantly greater systolic arterial pressure compared with untreated DOCA-salt rats (208 +/- 7 vs. 182 +/- 4 mmHg) whereas pressure in placebo rats was unchanged. In DOCA-salt, but not placebo rats, A-192621 significantly decreased sodium and water excretion along with parallel decreases in food and water intake. To determine whether the response in DOCA-salt rats was due to increased expression of ET(B) receptors, endothelin receptor binding was performed by using membranes from renal medulla. Maximum binding (B(max)) of [(125)I]ET-1, [(125)I]ET-3, and [(125)I]IRL-1620 increased from 227 +/- 42, 146 +/- 28, and 21 +/- 1 fmol/mg protein, respectively, in placebo rats to 335 +/- 27, 300 +/- 38, and 61 +/- 6 fmol/mg protein, respectively, in DOCA-salt hypertensive rats. The fraction of receptors that are the ET(B) subtype was significantly increased in DOCA-salt (0.88 +/- 0.07) compared with placebo (0.64 +/- 0.01). The difference between [(125)I]ET-3 and [(125)I]IRL-1620 binding is consistent with possible ET(B) receptor subtypes in the kidney. These results indicate that ET(B) receptors in the renal medulla are up-regulated in the DOCA-salt hypertensive rat and may serve to maintain a lower arterial pressure by promoting salt and water excretion.


Subject(s)
Hypertension/metabolism , Kidney Medulla/metabolism , Receptors, Endothelin/drug effects , Animals , Blood Pressure/drug effects , Desoxycorticosterone , Endothelin Receptor Antagonists , Hypertension/chemically induced , Kidney Medulla/drug effects , Male , Pyrrolidines/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Endothelin B , Receptors, Endothelin/metabolism , Up-Regulation , Urine
5.
Eur J Pharmacol ; 366(2-3): 189-201, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-10082200

ABSTRACT

Endothelins, 21-amino acid peptides involved in the pathogenesis of various diseases, bind to endothelin ET(A) and ET(B) receptors to initiate their effects. Here, we characterize the pharmacology of A-216546 ([2S-(2,2-dimethylpentyl)-4S-(7-methoxy-1,3-benzodioxol-5-yl )-1-(N,N-di(n-butyl) aminocarbonylmethyl)-pyrrolidine-3R-carboxylic acid), a potent antagonist with > 25,000-fold selectivity for the endothelin ET(A) receptor. A-216546 inhibited [125I]endothelin-1 binding to cloned human endothelin ET(A) and ET(B) receptors competitively with Ki of 0.46 and 13,000 nM, and blocked endothelin-1-induced arachidonic acid release and phosphatidylinositol hydrolysis with IC50 of 0.59 and 3 nM, respectively. In isolated vessels, A-216546 inhibited endothelin ET(A) receptor-mediated endothelin-1-induced vasoconstriction, and endothelin ET(B) receptor-mediated sarafotoxin 6c-induced vasoconstriction with pA2 of 8.29 and 4.57, respectively. A-216546 was orally available in rat, dog and monkey. In vivo, A-216546 dose-dependently blocked endothelin-1-induced pressor response in conscious rats. Maximal inhibition remained constant for at least 8 h after dosing. In conclusion, A-216546 is a potent, highly endothelin ET(A) receptor-selective and orally available antagonist, and will be useful for treating endothelin-1-mediated diseases.


Subject(s)
Endothelin Receptor Antagonists , Pyrrolidines/pharmacology , Administration, Oral , Animals , Area Under Curve , Binding, Competitive/drug effects , Blood Pressure/drug effects , Blood Vessels/drug effects , Blood Vessels/physiology , CHO Cells , Cell Line , Cricetinae , Dogs , Dose-Response Relationship, Drug , Endothelin-1/metabolism , Endothelin-1/pharmacology , Endothelin-3/metabolism , Endothelin-3/pharmacology , Female , Humans , In Vitro Techniques , Macaca fascicularis , Male , Membranes/drug effects , Membranes/metabolism , Muscle Contraction/drug effects , Pyrrolidines/chemistry , Pyrrolidines/pharmacokinetics , Rabbits , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptors, Endothelin/metabolism , Sensitivity and Specificity , Swine , Treatment Outcome , Vasoconstriction/drug effects
6.
J Cardiovasc Pharmacol ; 30(1): 33-41, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9268219

ABSTRACT

Balloon angioplasty has become an important intervention in clinical cardiology; however, the technique is associated with a high incidence of restenosis, requiring repeated procedures. Endothelin-1 (ET-1), specifically through its action on ET(A) receptors, has been implicated in the cell proliferation and subsequent neointimal formation that leads to restenosis. Therefore we examined a potent antagonist of the ET(A) receptor, A127722.5, in a pig model of balloon angioplasty in iliac and carotid arteries. Ten pigs received A-127722.5 (7.5 mg/kg b.i.d.) orally, starting 3 days before angioplasty and continuing for 4 weeks; 10 additional pigs were treated with the same dosing regimen of the angiotensin-converting enzyme (ACE) inhibitor captopril (3.0 mg/kg b.i.d.), while a third group of 10 animals received placebo. At 2 and 4 weeks after the start of treatment, these doses of the ET(A) receptor antagonist and ACE inhibitor blocked the presser responses induced by big ET-1 and angiotensin I, respectively. In the iliac arteries, neointimal formation, neointimal/medial ratio, and maximal neointimal thickness were all significantly reduced, and the residual lumen area was significantly increased in pigs treated with the ET(A) receptor antagonist compared with placebo and captopril-treated groups. Medial collagen content, collagen deposition, and medial growth also were significantly reduced relative to the placebo group. Beneficial effects also were observed in the carotid arteries, although the results were less striking. Captopril was ineffective in protecting against the effects of balloon angioplasty in both vessels. Our results indicate that an orally active and potent antagonist of the ET(A) receptor inhibits cell proliferation and synthesis of extracellular matrix in pigs and may provide an important therapeutic approach to the prevention of restenosis.


Subject(s)
Angioplasty, Balloon/adverse effects , Endothelin Receptor Antagonists , Muscle, Smooth, Vascular/drug effects , Pyrrolidines/pharmacology , Angiotensin I/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Atrasentan , Blood Pressure/drug effects , Captopril/pharmacology , Carotid Artery Injuries , Carotid Artery, Common/drug effects , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Collagen/biosynthesis , Endothelin-1/blood , Endothelin-1/pharmacology , Hyperplasia , Iliac Artery/drug effects , Iliac Artery/injuries , Iliac Artery/metabolism , Iliac Artery/pathology , Male , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Pyrrolidines/blood , Receptor, Endothelin A , Swine , Swine, Miniature , Tunica Intima/drug effects , Tunica Intima/pathology
7.
J Med Chem ; 40(3): 322-30, 1997 Jan 31.
Article in English | MEDLINE | ID: mdl-9022798

ABSTRACT

The benzodioxole ((methylenedioxy)benzene) group is present in a number of endothelin (ET) receptor antagonists thus far reported. As part of our own endothelin antagonist program we have developed (2R*,3R*,4S*)-1-(N,N-dibutylacetamido)-4-(1,3-benzodioxol-5- yl)-2-(4-methoxyphenyl)pyrrolidine-3-carboxylic acid (A-127722). This is a potent antagonist, binding to the ETA and ETB receptor subtypes with affinities (IC50) of 0.4 and 520 nM, respectively, and also contains the aforementioned benzodioxole. While this compound was seemingly optimized at its N-terminus, no effort had been directed toward understanding the contributions to binding affinity or receptor subtype selectivity conferred by the benzodioxole. Substitution by 1- or 2-naphthyl yielded weak antagonists. Oxygenated benzenes, such as p-anisyl, were potent compounds with IC50s in the low-nanomolar range. Simple deletion of either of the two oxygen atoms (dihydrobenzofurans) yielded extremely potent agents, possessing subnanomolar affinity for the ETA receptor. Additionally, the compounds showed enhanced selectivity, binding to the ETB receptor subtype in the micromolar range. This paper describes the development of this novel class of compounds.


Subject(s)
Acetamides/pharmacology , Endothelin Receptor Antagonists , Proline/analogs & derivatives , Acetamides/chemical synthesis , Acetamides/chemistry , Acetamides/pharmacokinetics , Animals , Cells, Cultured , Dioxoles/metabolism , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Molecular Conformation , Molecular Structure , Proline/chemical synthesis , Proline/chemistry , Proline/pharmacokinetics , Proline/pharmacology , Protein Binding , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Structure-Activity Relationship
8.
Recept Signal Transduct ; 7(3): 165-75, 1997.
Article in English | MEDLINE | ID: mdl-9440503

ABSTRACT

Endothelins (ETs) are 21-amino acid peptides that bind to membrane receptors to initiate pathophysiological effects. This report characterizes ET receptors in benign prostatic hyperplasia-1 (BPH-1) cells, a prostate cell line isolated from a specimen of a 60-yr-old man with benign prostatic hyperplasia. [(125)I]ET-1 or -3 binding was of high affinity, with B(max) and K(d) values of 48 fmol/1 x 10(6) cells and 0.16 nM for ET-1, and 2.9 fmol/1 x 10(6) cells and 0.033 nM for ET-3, respectively. ET-1, ET-3, FR139317, Ro 46-2005, and IRL1620 inhibited [(125)I]ET-1 binding to these cells with IC50 values of 0.22, 186, 0.20, 52.8, and 772.3 nM, respectively. Reverse transcription-polymerase chain reaction confirmed that BPH-1 cells expressed more ET(A) than ET(B) receptors. ET-1 did not have any effect on arachidonic acid release, but caused a modest stimulation of phosphatidylinositol hydrolysis, and induced a prominent, sustained elevation in intracellular Ca2+ concentrations. The functional effects of ET-1 were completely inhibited by the ET(A)-selective antagonists FR139317 and A-127722, suggesting that the effects were mediated by the ET(A) receptor. These results suggest that ET may play functional roles in benign prostatic hyperplasia.


Subject(s)
Prostatic Hyperplasia/metabolism , Receptors, Endothelin/metabolism , Arachidonic Acid/metabolism , Calcium/metabolism , Endothelin-1/metabolism , Humans , Hydrolysis , Male , Middle Aged , Phosphatidylinositols/metabolism , Polymerase Chain Reaction , Prostatic Hyperplasia/pathology , Receptor, Endothelin A , Receptor, Endothelin B , Receptors, Endothelin/physiology , Tumor Cells, Cultured
9.
Am J Physiol ; 271(3 Pt 1): C869-77, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8843717

ABSTRACT

Endothelins (ET) are potent vasoconstricting peptides with 21 amino acid residues. Endothelin-1 (ET-1) stimulates arachidonic acid (AA) release in human pericardial smooth muscle cells (HPSMC), which is primarily mediated through the ETA receptor. Manoalide, an inhibitor for phospholipase A2, inhibited the ET-1-evoked response by 50% at 1 microM. RHC-80267, an inhibitor for diacylglycerol lipase, did not have a significant effect. The Ca2+ ionophore A-23187 at 2 microM greatly stimulated AA release in the presence of extracellular Ca2+. ET-1 (10 nM) evoked a robust Ca2+ response. The intracellular Ca2+ concentration reached a peak after 10 s and gradually decreased to a new plateau level in the presence of extracellular Ca2+. ET-1-evoked AA release closely followed the change in the intracellular Ca2+ concentration. Removal of extracellular Ca2+ or treating cells with 250 microM bis(2-amino-5-methylphenoxy)ethane-N,N,N',N'-tetraacetic acid tetraacetoxymethyl ester (MAPTAM; an intracellular Ca2+ chelator) greatly reduced ET-1-stimulated AA release. The protein kinase C (PKC) inhibitors, staurosporine (1 microM) and chelerythrine chloride (2.5 microM), inhibited ET-1-evoked AA release by 70%. Phorbol 12-myristate 13-acetate, a PKC activator, potentiated the effect of ET on AA release. The data suggest that the effect of ET on AA release in HPSMC is via phospholipase A2, which is modulated by Ca2+ and PKC.


Subject(s)
Arachidonic Acid/metabolism , Calcium/metabolism , Endothelin-1/pharmacology , Muscle, Smooth/metabolism , Signal Transduction/drug effects , Cells, Cultured , Endothelin-1/metabolism , Humans , Pericardium/cytology , Pericardium/metabolism , Receptors, Endothelin/agonists , Receptors, Endothelin/metabolism
10.
J Pharmacol Exp Ther ; 276(2): 473-81, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8632312

ABSTRACT

Endothelins (ET) are potent vasoactive peptides implicated in the pathogenesis of a number of vascular diseases. The effects of ET on mammalian organs and cells are initiated by binding to ETA or ETB receptors. In this report, we document the pharmacology of A-127722, a novel ETA-selective receptor antagonist. A-127722 inhibits [125I]ET-1 binding to cloned human ETA and ETB receptors competitively with Ki values of 69 pM and 139 nM, respectively. A-127722 exhibits a dose-dependent inhibition of ET-1-induced arachidonic acid release in human pericardium smooth muscle cells with a pA2 value of 10.5 and inhibits ET-1-induced vasoconstriction in isolated rat aorta with a pA2 value of 9.2. In vivo, A-127722 dose-dependently blocks the pressor response to ET-1 (0.3 nmol/kg i.v.) in conscious rats. Statistically significant (P < .05) antagonism is seen at doses greater than 0.1 mg/kg p.o. Maximal inhibition, at 10 mg/kg, remains constant for at least 8 hr after dosing. No effect is seen on the ETB-mediated transient vasodepressor effect of exogenous ET-1. In conclusion, A-127722 is ETA-selective, orally bioavailable and efficacious for inhibiting the effects of ET in the rat, and A-127722 is the most potent ET receptor antagonist yet reported.


Subject(s)
Endothelin Receptor Antagonists , Pyrrolidines/pharmacology , Administration, Oral , Animals , Arachidonic Acid/metabolism , Atrasentan , CHO Cells , Cricetinae , Dogs , Female , Humans , Macaca fascicularis , Male , Phosphatidylinositols/metabolism , Pyrrolidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Stereoisomerism , Vasoconstriction/drug effects
11.
J Med Chem ; 39(4): 982-91, 1996 Feb 16.
Article in English | MEDLINE | ID: mdl-8632421

ABSTRACT

The oral absorption profile of a family of azole-based ET(A)-selective antagonists has been improved through a rational series of structural modifications which were suggested by analysis of the physicochemical parameter delta log P. Comparison of urea 2 with a series of well-absorbed compounds using delta log P analysis suggested that 2 has an excess capacity for forming hydrogen bonds with solvent. A series of urea modifications were explored as a means of reducing H-bonding capacity while maintaining affinity for the ET(A)-receptor. The correlation between delta log P values and absorption in an intraduodenal (id) bioavailability model was good; this strategy uncovered replacements for each of the urea NH groups which simultaneously improve both potency and drug absorption. A combination of these optimized modifications produces carbamate 16h, a highly-selective ET(A) antagonist with a potency/bioavailability profile consistent with an oral route of administration.


Subject(s)
Azoles/chemical synthesis , Azoles/pharmacokinetics , Endothelin Receptor Antagonists , Intestinal Absorption , Administration, Oral , Animals , Azepines/pharmacokinetics , Azoles/chemistry , Drug Design , Hydrogen Bonding , Indicators and Reagents , Indoles/pharmacokinetics , Injections, Intravenous , Kinetics , Male , Metabolic Clearance Rate , Models, Biological , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptors, Endothelin/metabolism , Structure-Activity Relationship
12.
Clin Chem ; 38(11): 2239-43, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1330373

ABSTRACT

To establish whether the use of proteinase inhibitors in the routine determination of in vitro plasma renin activity overestimates the potency of renin inhibitors in vivo, we examined the effects of phenylmethylsulfonyl fluoride and 8-hydroxyquinoline sulfate on the binding to plasma proteins and the respective IC50 values (50% inhibiting concentrations) of three renin inhibitors. All three renin inhibitors, A-64662, A-65317, and A-74273, bound (> 60%) to plasma proteins at both pH 6.0 and 7.4, with slightly greater binding at pH 7.4. Phenylmethylsulfonyl fluoride (1.45 mmol/L) had no significant effect on the protein binding at either pH 6.0 or 7.4; 8-hydroxyquinoline sulfate (3.4 mmol/L) caused a modest dissociation (10-30%) of the renin inhibitors from plasma proteins at both pH values; and the effects of both proteinase inhibitors together were similar to those of 8-hydroxyquinoline alone. At pH 7.4, phenylmethylsulfonyl fluoride increased the potencies of the three renin inhibitors slightly (< or = 43%), whereas IC50 values determined in the presence of 8-hydroxyquinoline decreased by 1.5- to 3.7-fold. The greatest increase in potency occurred with the most hydrophilic compound, and with both angiotensinase inhibitors the effect was no greater than that of 8-hydroxyquinoline alone. The results show that any dissociation of the hypotensive activity measured in vivo from the plasma renin activity measured in vitro is not simply an artifact in the plasma renin activity assay stemming from the use of these angiotensinase inhibitors, especially if only phenylmethylsulfonyl fluoride is used.


Subject(s)
Blood Proteins/metabolism , Oxazolidinones , Oxyquinoline/pharmacology , Phenylmethylsulfonyl Fluoride/pharmacology , Protease Inhibitors , Renin/antagonists & inhibitors , Amides/metabolism , Amides/pharmacology , Dipeptides/metabolism , Dipeptides/pharmacology , Histidine/analogs & derivatives , Histidine/metabolism , Histidine/pharmacology , Humans , Hydrogen-Ion Concentration , Morpholines/metabolism , Morpholines/pharmacology , Oxazoles/metabolism , Oxazoles/pharmacology , Protein Binding/drug effects
13.
FEBS Lett ; 300(3): 301-4, 1992 Apr 06.
Article in English | MEDLINE | ID: mdl-1555659

ABSTRACT

A series of nonpeptidic human renin inhibitors with a 4-methoxymethoxypiperidinylamide at the P4 position of the molecule exhibited slow tight binding to the enzyme. Replacement of the methoxymethoxy moiety on the piperidine ring with H, OH, methoxyethyl, propyloxy or n-butyl eliminated the effect. The inhibition was partially reversed by prolonged dialysis at 4 degrees C, arguing against formation of a covalent bond in the tightened complex.


Subject(s)
Piperidines/chemistry , Renin/antagonists & inhibitors , Binding Sites , Enzyme Activation , Humans , Protein Binding , Renin/blood , Renin/chemistry , Structure-Activity Relationship
14.
Anal Biochem ; 188(2): 408-15, 1990 Aug 01.
Article in English | MEDLINE | ID: mdl-2221392

ABSTRACT

A rapid, high-throughput radiometric assay for HIV-1 protease has been developed using ion-exchange chromatography performed in 96-well filtration plates. The assay monitors the activity of the HIV-1 protease on the radiolabeled form of a heptapeptide substrate, [tyrosyl-3,5-3H]Ac-Ser-Gln-Asn-Tyr-Pro-Val-Val-NH2, which is based on the p17-p24 cleavage site found in the viral polyprotein substrate Pr55gag. Specific cleavage of this uncharged heptapeptide substrate by HIV-1 protease releases the anionic product [tyrosyl-3,5-3H]Ac-Ser-Gln-Asn-Tyr, which is retained upon minicolumns of the anion-exchange resin AG1-X8. Protease activity is determined from the recovery of this radiolabeled product following elution with formic acid. This facile and highly sensitive assay may be utilized for steady-state kinetic analysis of the protease, for measurements of enzyme activity during its purification, and as a routine assay for the evaluation of protease inhibitors from natural product or synthetic sources.


Subject(s)
Chromatography, Ion Exchange , HIV Protease/metabolism , Amino Acid Sequence , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Reproducibility of Results , Substrate Specificity , Tritium
15.
Proc Natl Acad Sci U S A ; 86(6): 1841-5, 1989 Mar.
Article in English | MEDLINE | ID: mdl-2648384

ABSTRACT

Recombinant human immunodeficiency virus 1 (HIV-1) protease, purified from a bacterial expression system, processed a recombinant form of its natural substrate, Pr55gag, into protein fragments that possess molecular weights commensurate with those of the virion gag proteins. Molecular weights of the protease obtained under denaturing and nondenaturing conditions (11,000 and 22,000, respectively) and chemical crosslinking studies were consistent with a dimeric structure for the active enzyme. The protease appropriately cleaved the nonapeptide Ac-Arg-Ala-Ser-Gln-Asn-Tyr-Pro-Val-Val-NH2 between the tyrosine and proline residues. HIV-1 protease was sensitive to inactivators of the aspartic proteases. The aspartic protease inactivator 1,2-epoxy-3-(4-nitrophenoxy)propane produced irreversible, time-dependent inactivation of the protease. The pH-dependent kinetics of this inactivator were consistent with the requirement of an unprotonated carboxyl group in the active site of the enzyme, suggesting that HIV-1 protease is also an aspartic protease.


Subject(s)
Endopeptidases/metabolism , Escherichia coli/enzymology , HIV/enzymology , Recombinant Proteins/metabolism , Aspartic Acid Endopeptidases , Binding Sites , Centrifugation, Density Gradient , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Endopeptidases/genetics , Epoxy Compounds/pharmacology , Escherichia coli/genetics , Gene Products, gag , HIV/genetics , Hydrogen-Ion Concentration , Kinetics , Macromolecular Substances , Molecular Weight , Nitrophenols/pharmacology , Oligopeptides/metabolism , Peptide Fragments/metabolism , Protease Inhibitors , Retroviridae Proteins , Substrate Specificity
16.
Biochem Biophys Res Commun ; 159(2): 420-5, 1989 Mar 15.
Article in English | MEDLINE | ID: mdl-2649094

ABSTRACT

Oligopeptides containing the consensus retroviral protease cleavage sequence Ser/Thr-X-Y-Tyr/Phe-Pro are substrates for purified recombinant HIV-1 protease with Km's in the millimolar range. The minimum sequence containing the consensus pentapeptide which serves as a good substrate is a heptapeptide spanning the P4-P3' residues. Substitution of reduced Phe-Pro or Tyr-Pro dipeptide isosteres or the statine analog 3-hydroxy-4-amino-5-phenylpentanoic acid for the scissile dipeptide afforded inhibitors of HIV-1 protease with Ki values in the micromolar range, three orders of magnitude better in affinity than the corresponding substrates. Inhibitors of HIV-1 protease may provide a novel and potentially useful therapeutic approach to the treatment of acquired immune deficiency syndrome (AIDS).


Subject(s)
Oligopeptides/metabolism , Protease Inhibitors , Amino Acid Sequence , Endopeptidases/metabolism , HIV Protease , Hydrolysis , Kinetics , Molecular Sequence Data , Oligopeptides/chemical synthesis , Recombinant Proteins/metabolism , Retroviridae Proteins/metabolism , Substrate Specificity
17.
Cancer Res ; 42(7): 2821-8, 1982 Jul.
Article in English | MEDLINE | ID: mdl-7083173

ABSTRACT

CC-1065 (NSC 298223), a potent new antitumor antibiotic produced by Streptomyces zelensis, interacts strongly with double-stranded DNA and appears to exert its cytotoxic effects through disruption of DNA synthesis. We undertook this study to elucidate the sites and mechanisms of CC-1065 interaction with DNA. The binding of CC-1065 to synthetic and native DNA was examined by differential circular dichroism or by Sephadex chromatography with photometric detection. The binding of CC-1065 with calf thymus DNA was rapid, being complete within 2 hr, and saturated at 1 drug per 7 to 11 base pairs. The interaction of CC-1065 with synthetic DNA polymers indicated a specificity for adenine- and thymine-rich sites. Agarose gel electrophoresis of CC-1065-treated supercoiled DNA showed that CC-1065 did not intercalate. Site exclusion studies using substitutions in the DNA grooves showed CC-1065 to bind primarily in the minor groove. CC-1065 did not cause DNA breaks; it inhibited susceptibility of DNA to nuclease S1 digestion. It raised the thermal melting temperature of DNA, and it inhibited the ethidium-induced unwinding of DNA. Thus, in contrast to many antitumor agents, CC-1065 stabilized the DNA helix. DNA helix overstabilization may be relevant to the mechanism of action of CC-1065.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA , Indoles , Leucomycins/pharmacology , Animals , Anthramycin/metabolism , Cattle , Circular Dichroism , DNA/metabolism , Deoxyribonucleases/pharmacology , Duocarmycins , Electrophoresis, Agar Gel , Hot Temperature , Leucomycins/metabolism , Thymus Gland/metabolism
18.
Cancer Res ; 42(3): 999-1004, 1982 Mar.
Article in English | MEDLINE | ID: mdl-6174220

ABSTRACT

CC-1065, a novel antibiotic produced by Streptomyces zelensis, was active against several experimental tumors in vivo and a broad spectrum of human tumor cells in vitro. This report describes its biological and biochemical effects of L1210 leukemia cells. CC-1065 is one of the most cytotoxic agents known. The concentrations required for a 50 and 90% inhibition of cell growth are 0.02 and 0.05 ng/ml, respectively. It is about 400 times more cytotoxic than was Adriamycin. The action of CC-1065 is rapid and is dose and time dependent. CC-1065 inhibits DNA synthesis much more than it inhibits RNA and protein synthesis. The concentrations required for a 50% inhibition of DNA synthesis and RNA synthesis are 4 to 6 and 45 to 60 ng/ml, respectively. Although the drug inhibition of DNA synthesis cannot completely account for its cytotoxic effects on L1210 cells, these results, along with those generated by other investigators, suggest that the inhibition of DNA synthesis represents a major mode of action of CC-1065. CC-1065 inhibited both thymidine kinase and DNA polymerase alpha activities, but the effect on highly purified DNA polymerase alpha was more pronounced. At 1 microgram/ml, CC-1065 inhibited more than 70% of the enzyme activity. In order to elucidate the mechanism of inhibition of DNA polymerase alpha, the interaction between CC-1065 and DNA was investigated. The studies with thermal melting of DNA and difference circular dichroism measurement indicate that CC-1065 is one of the strongest DNA-binding agents. It induced an increase in thermal melting temperature of calf thymus DNA by at least 31 degrees. The circular dichroism studies also reveal that CC-1065 binds only to double-stranded DNA but not to heat-denatured DNA or yeast RNA. These observations were supported by those obtained with two other experimental approaches. CC-1065 also appeared to interact with proteins, but the interaction was weak and reversible.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA/biosynthesis , Indoles , Leucomycins/pharmacology , Leukemia L1210/metabolism , Animals , Cattle , Cell Division/drug effects , Cell Line , Circular Dichroism , DNA Polymerase II/metabolism , Dose-Response Relationship, Drug , Duocarmycins , Leucomycins/metabolism , Leukemia L1210/enzymology , Mice , RNA/biosynthesis , Templates, Genetic , Thymidine Kinase/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...