Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Biochemistry ; 36(10): 2939-50, 1997 Mar 11.
Article in English | MEDLINE | ID: mdl-9062124

ABSTRACT

Comparative molecular field analysis (CoMFA) was used to analyze the relationship between the structure of a group of ryanoids and the modulation of the calcium channel function of the ryanodine receptor. The conductance properties of ryanodine receptors purified from sheep heart were measured using the planar, lipid bilayer technique. The magnitude of the ryanoid-induced fractional conductance was strongly correlated to specific structural loci on the ligand. Briefly, electrostatic effects were more prominent than steric effects. The 10-position of the ryanoid had the greatest influence on fractional conductance. Different regions of the ligand have opposing effects on fractional conductance. For example, steric bulk at the 10-position is correlated with decreased fractional conductance, whereas steric bulk at the 2-position (isopropyl position) is correlated with increased fractional conductance. In contrast to fractional conductance, the 3-position (the pyrrole locus) had the greatest influence on ligand binding, whereas the 10-position had comparatively little influence on binding. Two possible models of ryanodine action, a direct (or channel plug) mechanism and an allosteric mechanism, were examined in light of the CoMFA. Taken together, the data do not appear to be consistent with direct interaction between ryanodine and the translocating ion. The data appear to be more consistent with an allosteric mechanism. It is suggested the ryanoids act by inducing or stabilizing a conformational change in the ryanodine receptor that results in the observed alterations in cation conductance.


Subject(s)
Calcium Channels/metabolism , Muscle Proteins/metabolism , Ryanodine/pharmacology , Sarcoplasmic Reticulum/chemistry , Allosteric Regulation , Animals , Electric Conductivity , Guanidines/pharmacology , Lipid Bilayers/metabolism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Molecular Structure , Myocardium/metabolism , Protein Binding , Protein Conformation , Pyrroles/pharmacology , Ryanodine/analogs & derivatives , Ryanodine/chemistry , Ryanodine Receptor Calcium Release Channel , Sheep
2.
Biophys J ; 70(5): 2110-9, 1996 May.
Article in English | MEDLINE | ID: mdl-9172735

ABSTRACT

We have examined the effects of a number of derivatives of ryanodine on K+ conduction in the Ca2+ release channel purified from sheep cardiac sarcoplasmic reticulum (SR). In a fashion comparable to that of ryanodine, the addition of nanomolar to micromolar quantities to the cytoplasmic face (the exact amount depending on the derivative) causes the channel to enter a state of reduced conductance that has a high open probability. However, the amplitude of that reduced conductance state varies between the different derivatives. In symmetrical 210 mM K+, ryanodine leads to a conductance state with an amplitude of 56.8 +/- 0.5% of control, ryanodol leads to a level of 69.4 +/- 0.6%, ester A ryanodine modifies to one of 61.5 +/- 1.4%, 9,21-dehydroryanodine to one of 58.3 +/- 0.3%, 9 beta,21beta-epoxyryanodine to one of 56.8 +/- 0.8%, 9-hydroxy-21-azidoryanodine to one of 56.3 +/- 0.4%, 10-pyrroleryanodol to one of 52.2 +/- 1.0%, 3-epiryanodine to one of 42.9 +/- 0.7%, CBZ glycyl ryanodine to one of 29.4 +/- 1.0%, 21-p-nitrobenzoyl-amino-9-hydroxyryanodine to one of 26.1 +/- 0.5%, beta-alanyl ryanodine to one of 14.3 +/- 0.5%, and guanidino-propionyl ryanodine to one of 5.8 +/- 0.1% (chord conductance at +60 mV, +/- SEM). For the majority of the derivatives the effect is irreversible within the lifetime of a single-channel experiment (up to 1 h). However, for four of the derivatives, typified by ryanodol, the effect is reversible, with dwell times in the substate lasting tens of seconds to minutes. The effect caused by ryanodol is dependent on transmembrane voltage, with modification more likely to occur and lasting longer at +60 than at -60 mV holding potential. The addition of concentrations of ryanodol insufficient to cause modification does not lead to an increase in single-channel open probability, such as has been reported for ryanodine. At concentrations of > or = 500 mu M, ryanodine after initial rapid modification of the channel leads to irreversible closure, generally within a minute. In contrast, comparable concentrations of beta-alanyl ryanodine do not cause such a phenomenon after modification, even after prolonged periods of recording (>5 min). The implications of these results for the site(s) of interaction with the channel protein and mechanism of the action of ryanodine are discussed. Changes in the structure of ryanodine can lead to specific changes in the electrophysiological consequences of the interaction of the alkaloid with the sheep cardiac SR Ca2+ release channel.


Subject(s)
Calcium Channels/physiology , Heart/physiology , Muscle Proteins/physiology , Ryanodine/analogs & derivatives , Ryanodine/pharmacology , Sarcoplasmic Reticulum/physiology , Animals , Calcium Channels/drug effects , Calcium Channels/isolation & purification , Cell Fractionation , Lipid Bilayers , Membrane Potentials/drug effects , Molecular Structure , Muscle Proteins/drug effects , Muscle Proteins/isolation & purification , Ryanodine/chemistry , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum/ultrastructure , Sheep , Structure-Activity Relationship
3.
Mol Cell Biochem ; 149-150: 145-60, 1995.
Article in English | MEDLINE | ID: mdl-8569724

ABSTRACT

The plant alkaloids ryanodine and dehydroryanodine are high affinity, biphasic modulators of the intracellularly located, calcium-regulated calcium release channels of a variety of cell types. To date, little is certain about the molecular basis of the interactions that prompt low concentrations of ryanodine (nanomolar to low micromolar) to activate (open) the channels and higher concentrations to deactivate (functionally close) the sarcoplasmic reticulum calcium release channel. In the present study, we approached this question using novel, semi-synthetic C10-Oeq ester derivatives of ryanodine and dehydroryanodine as molecular probes of the ryanodine binding sites on the calcium release channel. Binding affinities of these C10-Oeq ester derivatives of ryanodine and dehydroryanodine with acidic, basic and neutral side chains (Kd values > 53.9 nM, Kd values 0.3-0.7 nM and Kd values 1.3-20.4 nM, compared with 2.3 and 2.8 nM for ryanodine and dehydroryanodine, respectively) were evaluated for their ability to modulate the patency of the sarcoplasmic reticulum calcium release channel. With the exception of only two derivatives tested to date, all the semi-synthetic C10-Oeq esters selectively activate the Ca2+ release channel. That is, they produce no functional closure of the sarcoplasmic reticulum calcium release channels at the highest concentration that could be tested. Half-maximal concentrations for activation (EC50act values) ranged from 0.87-4.2 microM, compared with an EC50act of 1.3 microM for ryanodine. Using a low concentration (0.5 nM) of a high specific activity, radioiodinated derivative of ryanodine, C10-Oeq N-(4-azido-5-125iodo salicyloyl) glycyl ryanodine (1400 Ci/mmol) as the radioligand in displacement binding affinity assays, two distinct, sequential ryanodine binding isotherms were demonstrated within the normal 0-300 nM ryanodine sigmoidal displacement curve. A high affinity site had an IC50 of 0.5 nM (Kd = 0.26 +/- 0.02 nM). Above this concentration, an apparent plateau occurred between 3 and 6 nM ryanodine, and at higher concentrations a lower affinity site was revealed that demonstrated an IC50 of about 25 nM (Kd = 11.7 +/- 1.2 nM). Scatchard analysis from direct binding of C10-Oeq N-(4-azido-5-125iodo salicyloyl) glycyl ryanodine to junctional sarcoplasmic reticulum vesicles also suggests the presence of more than one class of binding sites within the nanomolar concentration range. The high affinity site demonstrated a Bmax of 3 pmol/mg protein. We were unable to saturate the lower affinity binding sites with this ligand. To evaluate the functional effects occurring among sarcoplasmic reticulum calcium release channel monomers as a consequence of ryanodine's binding, we utilized a photo-activatable derivative of ryanodine, C10-Oeq N-(4-azido salicyloyl) glycyl ryanodine that demonstrates channel modulating characteristics similar to ryanodine. Covalently labeling the sarcoplasmic reticulum calcium-release channels with this ligand, followed by measurements of rates of calcium efflux and SDS-PAGE of the labeled protein, revealed that deactivation of the sarcoplasmic reticulum calcium release channels of skeletal muscle by this ryanoid occurred at concentrations which apparently produce virtually irreversibly interactions between receptor monomers. This 'polymerization' was indicated by the progressive appearance of two higher molecular weight protein bands on SDS-PAGE, concomitant with progressive decreases in the ryanodine receptor monomer band that runs at an apparent molecular mass of 365 kDa. In summary, we have prepared and utilized novel C10-Oeq ester derivatives of ryanodine and dehydroryanodine in studies aimed at better understanding the molecular basis for the complex biphasic actions of ryanodine on the sarcoplasmic reticulum calcium release channels from rabbit skeletal muscle cells. The described studies presage correlations that may be useful in furthering our understa


Subject(s)
Calcium Channels/physiology , Calcium/physiology , Muscle Proteins/physiology , Ryanodine/analogs & derivatives , Sarcoplasmic Reticulum/physiology , Animals , Calmodulin-Binding Proteins/physiology , Ion Channel Gating , Ion Channels/physiology , Muscle Contraction , Muscles/physiology , Photochemistry , Rabbits , Radioligand Assay , Ryanodine/chemistry , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel , Structure-Activity Relationship
4.
J Biol Chem ; 269(48): 30243-53, 1994 Dec 02.
Article in English | MEDLINE | ID: mdl-7982934

ABSTRACT

The plant alkaloids ryanodine and dehydroryanodine are specific and potent modulators of the sarcoplasmic reticulum calcium release channel. In the present study, acidic, basic, and neutral side chains esters of these diterpene compounds were prepared and their pharmacologic activities were assessed. Binding affinities of the novel C10-Oeq ester derivatives for the sarcoplasmic reticulum Ca2+ release channel were evaluated with sarcoplasmic reticular vesicles prepared from rabbit skeletal muscle. Kd values of the derivatives varied 500-fold, ranging from 0.5 to 244 nM. In comparison, Kd values for ryanodine and dehydroryanodine were 4.4 nM and 5.4 nM, respectively. Basic substituents at the C10-Oeq side chain terminus produced the highest affinity derivatives (Kd values from 0.5 to 1.3 nM). Neutral and/or hydrophobic side chain derivatives exhibited intermediate affinities for the high affinity ryanodine receptor site (Kd values from 2.5 to 39 nM), whereas a derivative with a terminal acidic group had the lowest affinity (Kd value > 100 nM). Certain of the higher affinity C10-Oeq derivatives were evaluated more extensively for their pharmacologic activity on the sarcoplasmic reticular Ca2+ release channel. Both channel activating (opening) and deactivating (closing) actions were assessed from the ability of the ryanoids to alter Ca2+ efflux rates from skeletal junctional sarcoplasmic reticular vesicles that had been passively loaded with Ca2+. The natural Ryania secondary metabolites ryanodine, dehydroryanodine and esters E and F, all exhibit antithetical concentration-effect curves, indicating both activator and deactivator actions. In contrast, the semi-synthetic C10-Oeq esters selectively activate the Ca2+ release channel. Half-maximal concentrations for such activation (EC50 act) ranged from 0.87 microM to 4.2 microM, compared with an EC50 act of 1.3 microM for ryanodine. These derivatives were also evaluated for their ability to augment ATP-dependent CA2+ accumulation by cardiac junctional sarcoplasmic reticular vesicles, an effect that results from deactivation of the Ca2+ release channels. None of the derivatives tested was able to significantly augment Ca2+ accumulation, further substantiating their inability to deactivate the sarcoplasmic reticular Ca2+ release channel. Additionally, these derivatives functionally antagonized the action of ryanodine to close the Ca2+ release channel. The results presented demonstrate that these C10-Oeq ester derivatives of ryanodine and dehydroryanodine bind specifically to the SR Ca2+ release channel, selectively activate the channel, and, although they fail to effect channel closure, they nevertheless functionally compete with ryanodine at its low affinity (deactivator) site(s).


Subject(s)
Calcium Channel Agonists/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Muscle Proteins/metabolism , Ryanodine/analogs & derivatives , Ryanodine/pharmacology , Sarcoplasmic Reticulum/metabolism , Adenosine Triphosphate/pharmacology , Animals , Binding, Competitive , Cell Fractionation , Muscles/metabolism , Myocardium/metabolism , Rabbits , Ryanodine/chemical synthesis , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel , Structure-Activity Relationship
5.
Eur J Pharmacol ; 268(3): 365-73, 1994 Aug 16.
Article in English | MEDLINE | ID: mdl-7805760

ABSTRACT

An 125I-iodinated ryanodine analog, modified by attaching an iodo-Cbz-beta-alanyl group to the C10eq hydroxy of ryanodine (iodo-carbobenzyloxy-beta-alanyl-ryanodine), binds to cardiac sarcoplasmic reticulum Ca2+ release channels with equal affinity as [3H]ryanodine. In the present study, both iodo-Cbz-beta-alanyl-ryanodine and ryanodine bound to canine cardiac microsomal membrane preparations in a Ca2+ dependent manner. At 10 microM free Ca2+ doxorubicin increased specific binding of both ligands, with doxorubicin concentrations of 4.06 +/- 0.44 and 6.22 +/- 1.31 microM inducing 50% maximal enhancement of binding for ryanodine and iodo-Cbz-beta-alanyl-ryanodine, respectively. Effects of ryanodine and iodo-Cbz-beta-alanyl-ryanodine +/- doxorubicin in vitro on cardiac sarcoplasmic reticulum Ca2+ release were compared indirectly by determining Ca2+ accumulation in cardiac microsomal vesicles loaded with 45Ca2+. In the absence of oxalate, neither ryanodine nor iodo-Cbz-beta-alanyl-ryanodine (10 microM) decreased net Ca2+ uptake, whereas doxorubicin reduced Ca2+ accumulation 20 +/- 2%. In the presence of oxalate and 0.4 microM free Ca2+ ("low"), both ryanodine and iodo-Cbz-beta-alanyl-ryanodine modestly decreased (by 19% and 17% at 10 nM, respectively) maximum Ca2+ accumulation. Increasing concentrations of ryanodine (100 nM-100 microM) and iodo-Cbz-beta-alanyl-ryanodine (100 nM-30 microM) had no greater effect, but 100 microM iodo-Cbz-beta-alanyl- ryanodine decreased net Ca2+ uptake 57 +/- 3%. Doxorubicin (30 microM) alone reduced Ca2+ uptake 36%; its effects with 1 nM-10 microM ryanodine or 1 nM-100 microM iodo-Cbz-beta-alanyl-ryanodine were additive.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Calcium/metabolism , Doxorubicin/pharmacology , Myocardium/metabolism , Ryanodine/analogs & derivatives , Ryanodine/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Calcium Radioisotopes , Dogs , Heart/drug effects , In Vitro Techniques , Iodine Radioisotopes , Ligands , Microsomes/drug effects , Microsomes/metabolism , Radioligand Assay , Sarcoplasmic Reticulum/drug effects
6.
Biochemistry ; 33(20): 6074-85, 1994 May 24.
Article in English | MEDLINE | ID: mdl-8193121

ABSTRACT

Ryanodine binds to specific membrane proteins, altering the calcium permeability of intracellular membranes. In this study 19 ryanoids were isolated or synthesized and the structures correlated to the strength of binding to vertebrate skeletal muscle ryanodine receptors. Global minima were determined by employment of molecular mechanics and dynamics augmented by systematic searching of conformational space. Overall, steric and electrostatic factors contribute about equally to the differences in the experimentally determined dissociation constants. The dominant electrostatic interaction is localized to a hydroxyl group in an apolar region of the molecule. The pyrrole and isopropyl groups located together at one pole of the molecule have the greatest effect on steric interactions between ligand and receptor. We suggest ryanodine binds to the receptor with the pyrrole and isopropyl groups buried deep inside a cleft in the protein. This arrangement places special importance on the conformation of the pyrrole and isopropyl groups. In contrast, the opposite pole appears to be positioned at the entrance of the binding pocket because bulky adducts placed in the 9 position of ryanodine alter binding minimally. For example, a fluorescent ryanodine adduct was synthesized which has a dissociation constant close to that of ryanodine. Detailed examination reveals subtle interactions between ryanoid and receptor. In many cases, the major factors altering the strength of binding were found to be conformational alterations in the molecule remote from the site of covalent modification.


Subject(s)
Calcium Channels/metabolism , Muscle Proteins/metabolism , Muscles/metabolism , Ryanodine/analogs & derivatives , Animals , Chemical Phenomena , Chemistry, Physical , Chickens , Electrochemistry , Models, Molecular , Molecular Conformation , Molecular Structure , Rabbits , Ryanodine/chemistry , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel , Structure-Activity Relationship , Thermodynamics
7.
Mol Pharmacol ; 44(2): 412-21, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8394996

ABSTRACT

Two novel natural ryanoids from extracts of the wood of Ryania speciosa Vahl were evaluated with sarcoplasmic reticulum (SR) vesicles for their binding affinities and their activating and deactivating effects on Ca2+ release channels. The new ryanoids, which are more polar than the known Ryania constituents ryanodine and didehydro-(9,21)-ryanodine, were purified using silica gel column chromatography and reverse phase high performance liquid chromatography. The new ryanoids were designated ester E and ester F, in keeping with nomenclature previously used in the literature. These compounds were identified by NMR spectroscopy and mass spectroscopy as C9ax-hydroxyryanodine and C8ax-hydroxy-C10-epi-dehydroryanodine, respectively. Binding of esters E and F to the high affinity (nanomolar Kd) site on SR Ca2+ release channels was determined from relative binding affinity assays using 6.7 nM [3H]ryanodine. Apparent Kd values of ryanodine, ester E, and ester F for binding to this domain on the skeletal muscle ryanodine receptor/SR Ca2+ release channel were 4.4 +/- 0.8, 65 +/- 10, and 257 +/- 53 nM, respectively (mean +/- standard deviation, four or more experiments). Apparent Kd values for cardiac muscle receptors were 0.51 +/- 0.01, 12 +/- 0.4, and 57 nM, respectively. As a functional indication of the effects of the ryanoids, channel-opening (activator) and channel-closing (deactivator) actions were assessed from the ability of the ryanoids to alter the rate of Ca2+ efflux from passively loaded skeletal muscle junctional sarcoplasmic reticular vesicles (JSRV). Activator actions among the ryanoids were similar, in that they exhibited apparently parallel concentration-effect curves, having a slope of 40% Ca2+ loss/decade increment in ryanoid concentration. Half-maximal values for activation (EC50 values) were 2.5, 63, and 43 microM for ryanodine, ester E, and ester F, respectively. Maximal channel opening by ester E was significantly less than that produced by the other ryanoids. The deactivator actions of the compounds on skeletal JSRV were dissimilar, in that their concentration-effect curves appeared not to be parallel. The quotient of the EC50 for deactivation and that for activation was taken as the concentration-coupling ratio (CCR). The CCR for ryanodine was 114 and that for ester F was 72, but the CCR for ester E was only 21. ATP-dependent Ca2+ accumulation by cardiac JSRV provided a second means to evaluate deactivator actions of the ryanoids. Results from cardiac JSRV assays were in general similar to those from skeletal JSRV assays.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Ryanodine/analogs & derivatives , Ryanodine/pharmacology , Sarcoplasmic Reticulum/drug effects , Animals , Calcium Channels/drug effects , Muscles/drug effects , Muscles/ultrastructure , Myocardium/metabolism , Myocardium/ultrastructure , Plants/chemistry , Rabbits , Ryanodine/isolation & purification , Ryanodine/metabolism , Sarcoplasmic Reticulum/metabolism
8.
J Med Chem ; 36(10): 1319-23, 1993 May 14.
Article in English | MEDLINE | ID: mdl-8388466

ABSTRACT

Amino- and guanidinoacyl esters of ryanodine were prepared to evaluate the effect of basicity on the binding affinity of these derivatives for the sarcoplasmic reticulum Ca(2+)-release channel (SR CRC). In the presence of DCC and DMAP Cbz-beta-alanine reacts with ryanodine in CH2Cl2 to give O10eq-Cbz-beta-alanylryanodine (3a), which on hydrogenolysis yields the beta-alanyl ester (4a). N,N'-bis-Cbz-S-methylthiourea reacts with 4a to yield beta-N,N'-bis-Cbz-guanidinopropionylryanodine (5a). O10eq-beta-guanidinopropionylryanodine (6a) is obtained on hydrogenolytic deprotection of 5a. The binding affinity of beta-alanine ester (4a) and its glycyl congener (4b) is 2-3-fold greater, and that of the beta-guanidinopropionyl ester (6a) and its acetyl congener (6b) 3-6-fold greater, than that of ryanodine. The effect of ryanodine on SR Ca2+ flux is of a biphasic nature: nanomolar levels open (activate) the channel, while micromolar levels close (deactivate) it. The base-substituted esters 4a and 6a both display a unidirectional effect: they only open the channel. An understanding of ryanodine's mode of action and the design of effective SR CRC activating and deactivating ryanoids for possible therapeutic application are major research objectives.


Subject(s)
Amino Acids/chemical synthesis , Calcium Channels/drug effects , Guanidines/chemical synthesis , Ryanodine/analogs & derivatives , Sarcoplasmic Reticulum/drug effects , Amino Acids/metabolism , Amino Acids/pharmacology , Animals , Binding Sites , Esters/chemical synthesis , Esters/metabolism , Esters/pharmacology , Guanidines/metabolism , Guanidines/pharmacology , Rabbits , Sarcoplasmic Reticulum/metabolism , Structure-Activity Relationship
9.
Steroids ; 48(3-4): 223-31, 1986.
Article in English | MEDLINE | ID: mdl-3445280

ABSTRACT

Propargyl amine was protected by condensing it with 2,5-hexane-dione to give 2,5-dimethyl-N-(2'-propyn-1'-yl)pyrrole (2). The latter was converted to the corresponding Grignard reagent with ethylmagnesium bromide, and then condensed with estrone tetrahydropyranyl ether to give 17 alpha-[3'-(2'',5''-dimethyl-1''-pyrryl)-1'-propyn-1'-yl)-1,3 ,5( 10)- estratriene-3,17 beta-diol 3-tetrahydropyranyl ether (3), in 85% yield. Acetic acid and methanol cleaved the tetrahydropyranyl ether group, and hydroxylamine and sodium bicarbonate cleaved the pyrrole ring to give 17 alpha-(3'-amino-1'-propyn-1'-yl)-1,3,5(10)-estratriene-3,17 beta-diol (1), estrynamine. Several derivatives and analogs of 1 were also synthesized. Estrynamine binds to estrogen receptor with an RBA of 0.0045 (estradiol = 1.0). Several of the compounds, including estrynamine, are weak estrogens (stimulation of prolactin synthesis).


Subject(s)
Estradiol Congeners , Estradiol/analogs & derivatives , Animals , Antineoplastic Agents , Cells, Cultured , Chemical Phenomena , Chemistry , Estradiol/chemical synthesis , Estradiol/metabolism , Estradiol/pharmacology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Prolactin/biosynthesis , Rats , Receptors, Estrogen/metabolism , Steroids
10.
Steroids ; 46(4-5): 889-902, 1985.
Article in English | MEDLINE | ID: mdl-3837957

ABSTRACT

As part of a search for estradiol derivatives designed for conjugation to carboxyl or amine functions of anti-cancer agents or suitable derivatives thereof, estradiol analogs with side chains at the C-16 or -17 position were prepared for biological assay. These analogs include several which have a substituted nitrogenous function at C-17. The avidity of some of these analogs for binding to estrogen receptor was found to be of a low order.


Subject(s)
Estradiol/analogs & derivatives , Vinblastine/analogs & derivatives , Estradiol/chemical synthesis , Estradiol/metabolism , Humans , Receptors, Estrogen/metabolism
11.
J Med Chem ; 22(4): 391-400, 1979 Apr.
Article in English | MEDLINE | ID: mdl-430477

ABSTRACT

While structure-activity relationships for vinblastine (VLB), vincristine, deacetyl-VLB, and deacetyl-VLB amide (vindesine, VDS) in several tumor and leukemia models have been reported previously, the present study explores these relationships for a series of N-substituted vindesine analogues. These compounds were prepared from the reaction of deacetyl-VLB acid azide with the appropriate amines and were characterized by mass spectral analysis, 1H and 13C NMR spectra, electrometric titration, and infrared spectra. N-Alkylvindesines have reduced activity compared to that of VDS against the Gardner lymphosarcoma (GLS). N-beta-Hydroxyethyl-VDS surpasses vindesine in its activity against the Ridgway osteogenic sarcoma and the GLS, whereas against the B16 melanoma it is less active than VDS. N-beta-(4-Hydroxyphenethyl)-VDS, envisaged as a substrate for the enzyme tryosinase, was shown to be more active than VDS against the B16 melanoma but has only marginal activity against the GLS. In terms of collective antitumor activity against the model systems used, vindesine emerges as the congener with optimum qualities. Bis(N-ethylidenevindesine) disulfide, the first example of a bridged bisvindesine and comparable to VDS in its antitumor profile, shows evidence of activity against a P388/VCR leukemia strain known to be resistant to maytansine as well as to vincristine.


Subject(s)
Antineoplastic Agents/chemical synthesis , Vinblastine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Lethal Dose 50 , Male , Mice , Neoplasms, Experimental/drug therapy , Structure-Activity Relationship , Vinblastine/chemical synthesis , Vinblastine/pharmacology , Vinblastine/therapeutic use , Vinblastine/toxicity
13.
J Med Chem ; 21(1): 88-96, 1978 Jan.
Article in English | MEDLINE | ID: mdl-412968

ABSTRACT

Exploration of the effects of "minor" structural differences on the antitumor activity and toxicity of dimeric Catharanthus alkaloids resulted in the preparation of deacetylvinblastine amide (vindesine, VDS) from either vinblastine (VLB) or deacetylvinblastine. Adequate amounts of vindesine for biological testing were prepared by preferential hydrazinolysis of the C23-ester in the vindoline moiety of VLB, followed by hydrogenolysis of the resulting deacetylvinblastine hydrazide. Vindesine in its activity spectrum against rodent tumor systems resembles vincristine (VCR) rather than its parent VLB, while its neurotoxic potential appears to be less than that of VCR. The experimental models developed to estimate this potential include in vitro measurements of axoplasmic transport effects in the cat sciatic nerve and the estimation of neuromuscular disturbances in chickens and monkeys by vindesine in comparison with VCR. A radioimmunoassay for VLB, VCR, and VDS, developed by means of deacetylvinblastine acid azide, has been used to study the pharmacokinetics of vindesine in man. The clinical investigation of vindesine is in progress. Deacetylvinblastine, in contrast to earlier reports, showed activity against several murine tumor systems.


Subject(s)
Vinblastine/analogs & derivatives , Animals , Blood Pressure/drug effects , Cats , Chickens , Haplorhini , Heart Rate/drug effects , Humans , In Vitro Techniques , Kinetics , Lethal Dose 50 , Leukemia, Experimental/drug therapy , Male , Mice , Mice, Inbred Strains , Neoplasms, Experimental/drug therapy , Rats , Respiration/drug effects , Structure-Activity Relationship , Vinblastine/chemical synthesis , Vinblastine/metabolism , Vinblastine/pharmacology
19.
Appl Microbiol ; 18(1): 14-20, 1969 Jul.
Article in English | MEDLINE | ID: mdl-4308535

ABSTRACT

With the agar diffusion test and BS-C-1 cells, mycophenolic acid was found to give a straight-line dose-response activity in inhibiting the cytopathic effects of vaccinia, herpes simplex, and measles viruses. Plaque tests have shown 100% reduction of virus plaques by mycophenolic acid over drug ranges of 10 to 50 mug/ml and virus input as high as 6,000 plaque-forming units (PFU) per flask. Back titration studies with measles virus inhibited by mycophenolic acid have indicated that extracellular virus titers were reduced by approximately 3 logs(10) and total virus was reduced by 1 log(10). The agar diffusion test system lends itself readily to drug reversal studies. Mycophenolic acid incorporated into agar at 10 mug/ml gave 100% protection to virus-infected cells. Filter paper discs impregnated with selected chemical agents at concentrations of 1,000 mug/ml (20 mug per filter paper disc) were placed on the agar surface. Reversal of the antiviral activity of mycophenolic acid was indicated by virus breakthrough in those cells in close proximity to the filter paper disc. Chemicals showing the best reversal of the antiviral activity of mycophenolic acid were guanine, guanosine, guanylic acid, deoxyguanylic acid, and 2,6-diaminopurine. The reversal of antiviral activity was confirmed by titrations of virus produced with various amounts of both mycophenolic acid and guanine present and by isotope tracer methods with uptakes of labeled uridine, guanine, leucine, and thymidine in treated and nontreated, infected and noninfected cells as parameters. All antiviral effects of mycophenolic acid at 10 mug/ml could be reversed to the range shown by untreated controls by the addition of 10 mug/ml of those chemicals exhibiting reversal activity.


Subject(s)
Caproates/pharmacology , Measles virus/drug effects , Simplexvirus/drug effects , Vaccinia virus/drug effects , Animals , Caproates/antagonists & inhibitors , Caproates/blood , Cell Line , Cytopathogenic Effect, Viral , Guanine/pharmacology , Guanine Nucleotides/pharmacology , Haplorhini , Kidney , Methods , Mice , Nucleosides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...