Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
J Hand Surg Am ; 2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36175248

ABSTRACT

PURPOSE: Olecranon osteotomy is commonly used to obtain access to the distal humerus for fracture fixation. The goal of this study was to accurately describe the anatomy of the bare area to minimize articular cartilage damage while performing olecranon osteotomies. METHODS: Twenty cadaveric ulnae were denuded to expose the bare area. Laser surface mapping was used to create 3-dimensional models, and the nonarticular portions of the ulnae were digitally measured. RESULTS: The morphology of the bare area from all aspects of the proximal ulna was defined. The central bare area was consistent in its location, 4.9 ± 1.5 mm distal to the deepest portion of the trochlear notch and 23.2 ± 2.3 mm distal to the olecranon tip. The maximum chevron osteotomy apical angle to stay within the bare area averaged 110° ± 11.8°. However, there was little tolerance for error without the risk of violating the articular cartilage. With transverse osteotomy, averaging 18° ± 10.6° in the coronal plane, there is less risk of damaging the articular cartilage. CONCLUSIONS: Transverse osteotomy perpendicular to the posterior surface of the ulna aiming at the visible bare area on the medial and lateral sides of the greater sigmoid notch may reduce the chances of violating the nonvisible articular cartilage of the proximal ulna. Based on the findings of this study, if chevron osteotomy is used, a shallow apex distal angle of more than 110° is recommended. CLINICAL RELEVANCE: This study provides intraoperative landmarks to guide surgeons performing olecranon osteotomies to stay within the bare area.

2.
J Shoulder Elbow Surg ; 30(1): 172-177, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32788041

ABSTRACT

INTRODUCTION: There is limited research on the surgical management techniques for scapular spine fractures after reverse shoulder arthroplasty (RTSA). As such, the purpose of this in vitro biomechanical study was to compare 4 fixation constructs to stabilize scapular spine insufficiency fractures. METHODS: Twelve paired fresh-frozen cadaveric scapulae (N = 24) were randomized into 4 fixation groups: subcutaneous border plating (± hook) and supraspinatus fossa plating (± hook). A Levy type II fracture was simulated. Each specimen was cyclically loaded incrementally up to 700 N in 50 N steps or until failure. Between 50 and 200 N construct stiffness was measured, and stability failure was defined as displacement greater than 2.5 mm. RESULTS: Seventy-nine percent (19 of 24) of the specimens failed before the maximum load of 700 N. The average survival force with subcutaneous border plating was 480 ± 80 N compared with 380 ± 30 N for supraspinatus fossa plating (P = .3). Fixation construct failure was significantly more likely with fossa plating over subcutaneous plating (P = .012). The presence of the lateral plate hook was beneficial in preventing failure of the lateral acromion (P = .016). CONCLUSION: When appropriately surgically indicated, a dorsally applied plate to the subcutaneous border of the scapular spine with a lateral inferior supporting hook may be advantageous for internal fixation of type II scapular spine insufficiency fractures after RTSA.


Subject(s)
Arthroplasty, Replacement, Shoulder , Fracture Fixation, Internal , Fractures, Stress , Biomechanical Phenomena , Bone Plates , Humans , Scapula/surgery
3.
Int J Impot Res ; 20(4): 333-42, 2008.
Article in English | MEDLINE | ID: mdl-18418391

ABSTRACT

Cyclic guanosine monophosphate (cGMP) in penile vascular smooth muscle cells (VSMC) plays a key role in promoting penile erection. Phosphodiesterase-5 (PDE5) in VSMC breaks down cGMP to counter this effect. Sildenafil (Viagra), vardenafil (Levitra) and tadalafil (Cialis), treatments for erectile dysfunction, inhibit PDE5 action. Many men with erectile dysfunction have improved erectile function after plasma inhibitor concentration falls below therapeutic levels. Maximum effect plus onset and duration of action of inhibitor determines its efficacy. The rate and extent of cellular drug accumulation and efflux of drug from smooth muscle cells plus persistence of drug effects in these cell impact these parameters. We propose possible molecular mechanisms that could account for prolonged action of PDE5 inhibitors including (1) persistence of biochemical effects after inhibitor is cleared from cells, and (2) retention of drug in VSMC beyond plasma clearance.


Subject(s)
Erectile Dysfunction/drug therapy , Erectile Dysfunction/enzymology , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/therapeutic use , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Erectile Dysfunction/blood , Humans , Ligands , Male , Muscle, Smooth/drug effects , Phosphodiesterase Inhibitors/blood , Time Factors
4.
Int J Impot Res ; 17(5): 467-8, 2005.
Article in English | MEDLINE | ID: mdl-16079899

ABSTRACT

PDE11, the newest member of the PDE family of phosphodiesterases, has become the center of controversy. Four splice variants were recently identified, PDE11A1-4. Historical data have suggested that PDE11A3 is found in the testis while PDE11A4 is found in the prostate. The controversial issue is the inhibition of PDE11 by tadalafil. In the light of tadalafil's commercial success, its inhibition of PDE11A has been the subject of heated debate. A variety of published reports addressed this issue, suggesting that the target organ of tadalafil's inhibition, the testis, is not adversely affected. Daily tadalafil given to healthy volunteers did not alter semen analysis parameters or blood hormonal parameters, seemingly mitigating the clinical effect of the PDE11 inhibition. However, two recent papers published in this journal have added fuel to this proverbial fire. In this perspective, Sharron Francis, a noted PDE expert, and a co-author of one of the recent papers mentioned above, sheds further light on this contested topic.


Subject(s)
Phosphoric Diester Hydrolases/physiology , Testis/physiology , 3',5'-Cyclic-GMP Phosphodiesterases , Animals , Carbolines/pharmacology , Humans , Male , Mice , Mice, Knockout , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/drug effects , Phosphoric Diester Hydrolases/genetics , Species Specificity , Tadalafil
5.
Int J Impot Res ; 17(1): 5-9, 2005.
Article in English | MEDLINE | ID: mdl-15538396

ABSTRACT

The physiological role of phosphodiesterase (PDE)11 is unknown and its biochemical characteristics are poorly understood. We have expressed human His-tagged PDE11A4 and purified the enzyme to apparent homogeneity. PDE11A4 displays K(m) values of 0.97 microM for cGMP and 2.4 microM for cAMP, and maximal velocities were 4- to 10-fold higher for cAMP than for cGMP. Given the homology between PDE11 and PDE5, we have compared the biochemical potencies of tadalafil (Cialis, Lilly-ICOS), vardenafil (Levitra, Bayer-GSK), and sildenafil (Viagra, Pfizer Inc.) for PDE11A4 and PDE5A1. PDE5A1/PDE11A4 selectivities are 40-, 9300-, and 1000-fold for tadalafil, vardenafil, and sildenafil, respectively. This suggests that none of these three compounds is likely to crossreact with PDE11A4 in patients.


Subject(s)
Carbolines/pharmacology , Imidazoles/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Piperazines/pharmacology , Sulfones/pharmacology , Triazines/pharmacology , 3',5'-Cyclic-GMP Phosphodiesterases , Cross Reactions , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5 , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , Kinetics , Purines , Recombinant Proteins , Sildenafil Citrate , Substrate Specificity , Tadalafil , Vardenafil Dihydrochloride
6.
Int J Impot Res ; 15(5): 369-72, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14562139

ABSTRACT

Sildenafil, the active ingredient in Viagra, has been purified from commercially available tablets. The purification, using Sephadex G25 chromatography under conditions of low ionic strength, is simple and inexpensive. Sildenafil purified according to this protocol has been characterized with respect to its IC50 for PDE5, its ultraviolet absorption profile, and by collision-induced dissociation fingerprinting, positive ion nanospray, and MALDI mass spectrometry. Tritated sildenafil (6 Ci/mmol) was prepared commercially using the sildenafil purified by this protocol and was verified to retain the potency of unlabeled sildenafil. This protocol and similar procedures will allow investigators to easily isolate sufficient amounts of sildenafil or other PDE5 inhibitors for conducting biochemical and in vitro studies of drug action.


Subject(s)
Phosphodiesterase Inhibitors/isolation & purification , Piperazines/chemistry , Piperazines/isolation & purification , Dextrans , Hydrophobic and Hydrophilic Interactions , Phosphodiesterase Inhibitors/chemistry , Purines , Sildenafil Citrate , Sulfones , Tritium
7.
Int J Clin Pract ; 56(6): 453-9, 2002.
Article in English | MEDLINE | ID: mdl-12166544

ABSTRACT

The clinical properties (efficacy and safety profile) of a medicine are related not only to its mode of action, but also to its selectivity for its target (usually a receptor or enzyme) and are also influenced by its pharmacokinetic properties (absorption, distribution, metabolism and elimination). The growing number of phosphodiesterase inhibitors that are selective for phosphodiesterase-5 (PDE5) represent a promising new class of compounds that are useful for the treatment of erectile dysfunction and perhaps other disorders. Some of the basic pharmacodynamic and pharmacokinetic parameters that describe drug action are discussed with regard to the new PDE5 inhibitors. Central topics reviewed are the concentration that produces a given in vitro response, or potency (IC50), maximum plasma concentration (Cmax), time to Cmax (Tmax), half-life (t 1/2), area under the curve (AUC), bioavailability, onset and duration of action, and the balance to achieve optimum safety and efficacy. To illustrate these concepts, a group of inhibitors with varying selectivities and potencies for PDE5 (theophylline, IBMX, zaprinast, sildenafil, tadalafil and vardenafil) are discussed. Each drug has its own set of unique pharmacological characteristics based on its specific molecular structure, enzyme inhibition profile and pharmacokinetic properties. Each PDE5 inhibitor has a distinct selectivity that contributes to its safety profile. As with all new drugs, and especially those in a new class, careful evaluation will be necessary to ensure the optimal use of the PDE5 inhibitors.


Subject(s)
Phosphodiesterase Inhibitors/pharmacokinetics , Phosphoric Diester Hydrolases , 1-Methyl-3-isobutylxanthine/pharmacokinetics , 3',5'-Cyclic-GMP Phosphodiesterases , Area Under Curve , Carbolines , Cyclic Nucleotide Phosphodiesterases, Type 5 , Erectile Dysfunction/drug therapy , Humans , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Male , Muscle, Smooth, Vascular/drug effects , Phosphodiesterase Inhibitors/therapeutic use , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Purines , Purinones/pharmacokinetics , Purinones/therapeutic use , Sildenafil Citrate , Sulfones , Tadalafil , Theophylline/pharmacokinetics , Triazines , Vardenafil Dihydrochloride
8.
Eur J Biochem ; 268(11): 3304-12, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11389733

ABSTRACT

To date, relative cellular levels of cGMP and cGMP-binding proteins have not been considered important in the regulation of smooth muscle or any other tissue. In rabbit penile corpus cavernosum, intracellular cGMP was determined to be 18 +/- 4 nM, whereas the cGMP-binding sites of types Ialpha and Ibeta cGMP-dependent protein kinase (PKG) and cGMP-binding cGMP-specific phosphodiesterase (PDE5) were 58 +/- 14 nM and 188 +/- 6 nM, respectively, as estimated by two different methods for each protein. Thus, total cGMP-binding sites (246 nM) greatly exceed total cGMP. Given this excess of cGMP-binding sites and the high affinities of PKG and PDE5 for cGMP, it is likely that a large portion of intracellular cGMP is associated with these proteins, which could provide a dynamic reservoir for cGMP. Phosphorylation of PDE5 by PKG is known to increase the affinity of PDE5 allosteric sites for cGMP, suggesting the potential for regulation of a reservoir of cGMP bound to this protein. Enhanced binding of cGMP by phosphorylated PDE5 could reduce the amount of cGMP available for activation of PKG, contributing to feedback inhibition of smooth muscle relaxation or other processes. This introduces a new concept for cyclic nucleotide signaling.


Subject(s)
Cyclic GMP/metabolism , Intracellular Signaling Peptides and Proteins , Penis/enzymology , Phosphoric Diester Hydrolases/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases , Allosteric Site , Animals , Carrier Proteins/analysis , Chromatography, DEAE-Cellulose , Cyclic AMP/analysis , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/analysis , Cyclic GMP/analysis , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/analysis , Cyclic Nucleotide Phosphodiesterases, Type 5 , Enzyme Activation , Feedback , Male , Penis/chemistry , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/chemistry , Piperazines/pharmacology , Purines , Rabbits , Signal Transduction , Sildenafil Citrate , Sulfones , Tissue Extracts/chemistry
9.
Article in English | MEDLINE | ID: mdl-11008484

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDEs) comprise a superfamily of metallophosphohydrolases that specifically cleave the 3',5'-cyclic phosphate moiety of cAMP and/or cGMP to produce the corresponding 5'-nucleotide. PDEs are critical determinants for modulation of cellular levels of cAMP and/or cGMP by many stimuli. Eleven families of PDEs with varying selectivities for cAMP or cGMP have been identified in mammalian tissues. Within these families, multiple isoforms are expressed either as products of different genes or as products of the same gene through alternative splicing. Regulation of PDEs is important for controlling myriad physiological functions, including the visual response, smooth muscle relaxation, platelet aggregation, fluid homeostasis, immune responses, and cardiac contractility. PDEs are critically involved in feedback control of cellular cAMP and cGMP levels. Activities of the various PDEs are highly regulated by a panoply of processes, including phosphorylation events, interaction with small molecules such as cGMP or phosphatidic acid, subcellular localization, and association with specific protein partners. The PDE superfamily continues to be a major target for pharmacological intervention in a number of medically important maladies.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/chemistry , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Amino Acid Sequence , Animals , Catalysis , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , Structure-Activity Relationship
11.
Biochemistry ; 39(31): 9591-6, 2000 Aug 08.
Article in English | MEDLINE | ID: mdl-10924156

ABSTRACT

Class I cyclic nucleotide phosphodiesterases (PDEs) share a catalytic domain containing 18 invariant residues. In cGMP-binding cGMP-specific PDE (PDE5), we showed previously that point mutation of nine of these profoundly decreases k(cat) when the assay is conducted in the presence of Mg(2+); seven of these are in the prototypical metal-binding motifs A and B (HX(3)HX(n)()E) that we identified earlier. Tandem arrangement of two of these metal-binding motifs in PDEs is novel, and whether residues within these motifs are involved in metal support of catalytic activity is a fundamental question in this field. This report shows that mutation of either His-607 (A motif) or His-643 (B motif) to alanine profoundly diminishes support of PDE catalysis by Mn(2+) or Mg(2+), but mutation of His-647 in B motif or of Glu in either motif does not. H607A and H643A mutants have much greater maximum catalytic rates supported by Mn(2+) than that by Mg(2+); catalytic activity of H603A mutant is supported weakly by either. In H607A and H643A, K(a)s for Mn(2+) and Mg(2+) are increased, but the effect of Mn(2+) is 2-fold greater than that of Mg(2+) in each. Mutation of any of the other conserved residues (Asn-604, Asp-644, His-675, Asp-714, and Asp-754) causes unremarkable changes in Mn(2+) or Mg(2+) support of catalysis. This study identifies specific residues in PDE5 that contribute to interactions with catalytically relevant metals. The combined data suggest that despite a high degree of sequence similarity between each HX(3)HX(n)()E motif in PDEs and certain metallo-endopeptidases, PDEs employ a distinct complement of residues for interacting with metals involved in catalysis.


Subject(s)
Catalytic Domain , Histidine/chemistry , Metals/chemistry , Phosphoric Diester Hydrolases/chemistry , 3',5'-Cyclic-GMP Phosphodiesterases , Amino Acid Sequence , Animals , Binding Sites/genetics , Catalysis , Catalytic Domain/genetics , Cations, Divalent/chemistry , Cattle , Cyclic Nucleotide Phosphodiesterases, Type 5 , Histidine/genetics , Magnesium/chemistry , Manganese/chemistry , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphoric Diester Hydrolases/genetics , Point Mutation
12.
Eur J Biochem ; 267(9): 2760-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10785399

ABSTRACT

In addition to its cGMP-selective catalytic site, cGMP-binding cGMP-specific phosphodiesterase (PDE5) contains two allosteric cGMP-binding sites and at least one phosphorylation site (Ser92) on each subunit [Thomas, M.K., Francis, S.H. & Corbin, J.D. (1990) J. Biol. Chem. 265, 14971-14978]. In the present study, prior incubation of recombinant bovine PDE5 with a phosphorylation reaction mixture [cGMP-dependent protein kinase (PKG) or catalytic subunit of cAMP-dependent protein kinase (PKA), MgATP, cGMP, 3-isobutyl-1-methylxanthine], shown earlier to produce Ser92 phosphorylation, caused a 50-70% increase in enzyme activity and also increased the affinity of cGMP binding to the allosteric cGMP-binding sites. Both effects were associated with increases in its phosphate content up to 0.6 mol per PDE5 subunit. Omission of any one of the preincubation components caused loss of stimulation of catalytic activity. Addition of the phosphorylation reaction mixture to a crude bovine lung extract, which contains PDE5, also produced a significant increase in cGMP PDE catalytic activity. The increase in recombinant PDE5 catalytic activity brought about by phosphorylation was time-dependent and was obtained with 0.2-0.5 microM PKG subunit, which is approximately the cellular level of this enzyme in vascular smooth muscle. Significantly greater stimulation was observed using cGMP substrate concentrations below the Km value for PDE5, although stimulation was also seen at high cGMP concentrations. Considerably higher concentration of the catalytic subunit of PKA than of PKG was required for activation. There was no detectable difference between phosphorylated and unphosphorylated PDE5 in median inhibitory concentration for the PDE5 inhibitors, sildenafil, or zaprinast 3-isobutyl-1-methylxanthine. Phosphorylation reduced the cGMP concentration required for half-maximum binding to the allosteric cGMP-binding sites from 0.13 to 0.03 microM. The mechanism by which phosphorylation of PDE5 by PKG could be involved in physiological negative-feedback regulation of cGMP levels is discussed.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Phosphoric Diester Hydrolases/metabolism , Protein Kinases/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases , Allosteric Regulation , Animals , Catalysis , Cattle , Cyclic GMP-Dependent Protein Kinases , Cyclic Nucleotide Phosphodiesterases, Type 5 , Phosphorylation , Protein Binding , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism
13.
J Biol Chem ; 275(1): 154-8, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10617599

ABSTRACT

The role of each of the two different cGMP-binding sites (referred to as slow and fast sites) of type I cGMP-dependent protein kinase (PKG) in altering the rate of catalysis of phosphorylation of exogenous substrates (heterophosphorylation) or the rate of autophosphorylation has not been resolved. In the present study, the cGMP concentration required for half-maximal activation (A(50)) of wild-type PKG type Ibeta (WT) was 5-fold higher for heterophosphorylation than for autophosphorylation. cGMP occupation of the slow site was associated with an increase in the autophosphorylation rate, whereas occupation of the fast and slow site together was associated with a decrease in the autophosphorylation rate compared with the rate observed with occupation of the slow site alone. The contributions of each cGMP-binding site were investigated using PKG mutants containing substitutions of an invariant threonine residue that is critical for high affinity cGMP-binding in each site. Site-directed mutagenesis of Thr-317 of the fast site (T317A) increased the cGMP A(50) for heterophosphorylation 4-fold at 30 degrees C, with nominal effect on cGMP A(50) for autophosphorylation compared with WT. The analogous slow site mutation (T193A) increased the cGMP A(50) for heterophosphorylation and autophosphorylation 32- and 64-fold, respectively. Compared with WT, the cGMP A(50) of the double mutant (T193A/T317A) for heterophosphorylation was increased 300-fold, whereas the cGMP A(50) for autophosphorylation was similar to that of T193A. Thus, occupation of both cGMP-binding sites of PKG is required for maximal stimulation of heterophosphorylation, whereas occupation of the slow site alone is sufficient for stimulation of the rate of autophosphorylation, and additional occupation of the fast site reduces this rate.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Animals , Binding Sites , Cattle , Cyclic GMP-Dependent Protein Kinases/genetics , Humans , Kinetics , Mutation , Phosphorylation
14.
J Biol Chem ; 274(49): 34613-20, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10574925

ABSTRACT

Phosphodiesterases (PDEs) comprise a superfamily of phosphohydrolases that degrade 3',5'-cyclic nucleotides. All known mammalian PDEs are dimeric, but the functional significance of dimerization is unknown. A deletion mutant of cGMP-binding cGMP-specific PDE (PDE5), encoding the 357 carboxyl-terminal amino acids including the catalytic domain, has been generated, expressed, and purified. The K(m) of the catalytic fragment for cGMP (5.5 +/- 0. 51 microM) compares well with those of the native bovine lung PDE5 (5.6 microM) and full-length wild type recombinant PDE5 (2 +/- 0.4 microM). The catalytic fragment and full-length PDE5 have similar IC(50) values for the inhibitors 3-isobutyl-1-methylxanthine (20 microM) and sildenafil (Viagra(TM))(4 nM). Based on measured values for Stokes radius (29 A) and sedimentation coefficient (2.9 S), the PDE5 catalytic fragment has a calculated molecular mass of 35 kDa, which agrees well with that predicted by amino acid content (43.3 kDa) and with that estimated using SDS-polyacrylamide gel electrophoresis (39 kDa). The combined data indicate that the recombinant PDE5 catalytic fragment is monomeric, and retains the essential catalytic features of the dimeric, full-length enzyme. Therefore, the catalytic activity of PDE5 holoenzyme requires neither interaction between the catalytic and regulatory domains nor interactions between subunits of the dimer.


Subject(s)
Cyclic GMP/chemistry , Cyclic GMP/genetics , Gene Expression Regulation, Enzymologic , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , 1-Methyl-3-isobutylxanthine/pharmacology , 3',5'-Cyclic-GMP Phosphodiesterases , Animals , Cattle , Centrifugation, Density Gradient , Cyclic Nucleotide Phosphodiesterases, Type 5 , Dimerization , Dose-Response Relationship, Drug , Inhibitory Concentration 50 , Kinetics , Leucine Zippers , Mutagenesis , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/biosynthesis , Piperazines/pharmacology , Purines , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sildenafil Citrate , Sulfones
15.
J Biol Chem ; 274(41): 29038-41, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10506154

ABSTRACT

The regulatory domain of the cGMP-binding cGMP-specific 3':5'-cyclic nucleotide phosphodiesterase (PDE5) contains two homologous segments of amino acid sequence that encode allosteric cyclic nucleotide-binding sites, referred to as site a and site b, which are highly selective for cGMP over cAMP. The possibility that the state of protonation in these sites contributes to cyclic nucleotide selectivity was investigated. The binding of cGMP or cAMP was determined using saturation and competition kinetics at pH values between 5.2 and 9.5. The total cGMP binding by PDE5 was unchanged by variation in pH, but the relative affinity for cGMP versus cAMP progressively decreased as the pH was lowered. Using site-directed mutagenesis, a conserved residue, Asp-289, in site a of PDE5 has been identified as being important for cyclic nucleotide discrimination in this site. It is proposed that deprotonation of Asp-289 enhances the number and strength of bonds formed with cGMP, while concomitantly decreasing the interactions with cAMP.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/chemistry , Cyclic AMP/chemistry , Cyclic GMP/chemistry , 3',5'-Cyclic-GMP Phosphodiesterases/genetics , Allosteric Regulation , Animals , Binding Sites , Cattle , Cyclic Nucleotide Phosphodiesterases, Type 5 , Hydrogen-Ion Concentration , Kinetics , Lung , Molecular Structure , Mutagenesis, Site-Directed , Protein Binding
16.
Crit Rev Clin Lab Sci ; 36(4): 275-328, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10486703

ABSTRACT

Intracellular cAMP and cGMP levels are increased in response to a variety of hormonal and chemical stimuli; these nucleotides play key roles as second messenger signals in modulating myriad physiological processes. The cAMP-dependent protein kinase and cGMP-dependent protein kinase are major intracellular receptors for these nucleotides, and the actions of these enzymes account for much of the cellular responses to increased levels of cAMP or cGMP. This review summarizes many studies that have contributed significantly to an improved understanding of the catalytic, regulatory, and structural properties of these protein kinases. These accumulated findings provide insights into the mechanisms by which these enzymes produce their specific physiological effects and are helpful in considering the actions of other protein kinases as well.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Animals , Binding Sites , Humans
17.
Mol Pharmacol ; 56(1): 124-30, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385692

ABSTRACT

The cGMP-binding cGMP-specific phosphodiesterase (PDE5) degrades cGMP and regulates the intracellular level of cGMP in many tissues, including the smooth muscle of the corpus cavernosum of the penis. Sildenafil (Viagra), a specific PDE5 inhibitor, promotes penile erection by blocking the activity of PDE5, which causes cGMP to accumulate in the corpus cavernosum. In the present study, sildenafil, like other PDE5 inhibitors, stimulates cGMP binding to the allosteric sites of PDE5 by interacting at the catalytic site of this enzyme, but the drug does not compete with cGMP for binding at the allosteric sites. Both sildenafil and zaprinast are competitive inhibitors of PDE5, and double-inhibition analysis shows that these two inhibitors added together interact with the catalytic site of PDE5 in a mutually exclusive manner. After site-directed mutagenesis of each of 23 conserved amino acid residues in the catalytic domain of PDE5, the pattern of changes in the IC50 values for sildenafil or UK-122764 is similar to that found for zaprinast. However, among the three inhibitors, sildenafil exhibits the most similar pattern of changes in the IC50 to that found for the affinity of cGMP, implying similar interactions with the catalytic domain. This may explain in part the stronger inhibitory potency of sildenafil for wild-type PDE5 compared with the other inhibitors [sildenafil (Ki = 1 nM) > UK-122764 (Ki = 5 nM) > zaprinast (Ki = 130 nM)]. The affinity of each of these inhibitors for PDE5 is much higher than that of cGMP itself (Km = 2000 nM). It is concluded that residues such as Tyr602, His607, His643, and Asp754 may form important interactions for sildenafil in PDE5, but because these amino acids are conserved in all mammalian PDEs, the selectivity and potency of sildenafil is likely to be provided by a nonconserved residue or residues in the PDE5 catalytic domain.


Subject(s)
Cyclic GMP/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Piperazines/pharmacology , 3',5'-Cyclic-GMP Phosphodiesterases , Allosteric Regulation , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cattle , Cyclic Nucleotide Phosphodiesterases, Type 5 , Gene Expression , Inhibitory Concentration 50 , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/isolation & purification , Purines , Sildenafil Citrate , Sulfones
19.
Am J Cardiol ; 83(5A): 3C-12C, 1999 Mar 04.
Article in English | MEDLINE | ID: mdl-10078537

ABSTRACT

Sildenafil is a selective inhibitor of phosphodiesterase type 5 (PDE5), which has been shown to be a clinically effective treatment for erectile dysfunction. Its action results from increased levels of cyclic guanosine monophosphate (cGMP), which is normally degraded by PDE5. This cyclic nucleotide is a second messenger for nitric oxide, which is involved in the regulation of numerous functions, including vascular smooth muscle tone. In an attempt to better predict the effects of sildenafil on cardiovascular function, the distribution of PDE activity was determined with anti-PDE1 and anti-PDE5 antibodies in the human cardiac ventricle and saphenous vein, and in vitro studies were performed on the isolated human cardiac ventricle, corpus cavernosum, saphenous vein, and mesenteric artery as well as on rabbit aorta, dog coronary artery, dog trabecular tissue, and rabbit and human platelets. The major PDE activity in the human cardiac ventricle was shown to be calcium/calmodulin-dependent PDE1, but there was no detectable level of PDE5. In contrast, the human saphenous vein contained PDEs 1, 4, and 5, and the human mesenteric artery contained PDEs 1, 2, 3, 4, and 5. The distribution of PDE5 in the cardiovascular system is consistent with the observed pharmacodynamic and clinical effects of sildenafil. Sildenafil, unlike milrinone, a selective PDE3 inhibitor, had no effect on the isolated trabeculae carneae; this is consistent with the lack of PDE5 expression in cardiac myocytes. Sildenafil selectively increased cGMP levels in coronary vascular smooth muscle tissue but produced no change in cyclic adenosine monophosphate (cAMP) levels, which is consistent with the drug's selectivity for PDE5. In phenylephrine-contracted isolated rabbit aortic rings, sildenafil enhanced the relaxation induced by the nitric oxide donor glyceryl trinitrate, suggesting that sildenafil may potentiate the hypotensive effects of nitric oxide donor agents on the vasculature, an effect that has been observed clinically. Human platelets were found to contain PDE5, which was inhibited by 50% (IC50) by sildenafil at a concentration of 6.3 nM, consistent with the IC50 value in the corpus cavernosum. Sildenafil alone had no direct effect on platelet function, but it potentiated the in vitro antiaggregatory activity of sodium nitroprusside on rabbit and human platelets. The pharmacodynamic and adverse event profiles observed in clinical trials with sildenafil are consistent with the in vitro profile of the tissue distribution of PDE5 and its known mechanism of action as a selective inhibitor of PDE5.


Subject(s)
Blood Platelets/drug effects , Blood Vessels/enzymology , Heart Ventricles/enzymology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Myocardial Contraction/drug effects , Nucleotides, Cyclic/metabolism , Penis/enzymology , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Animals , Aorta/enzymology , Blood Vessels/drug effects , Cadaver , Coronary Vessels/enzymology , Dogs , Heart Ventricles/drug effects , Humans , In Vitro Techniques , Isoenzymes/analysis , Male , Mesenteric Arteries/enzymology , Penis/drug effects , Phosphoric Diester Hydrolases/analysis , Purines , Rabbits , Saphenous Vein/enzymology , Sildenafil Citrate , Sulfones , Tissue Distribution
20.
J Biol Chem ; 273(38): 24485-90, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9733741

ABSTRACT

Chimeric cGMP phosphodiesterases (PDEs) have been constructed using components of the cGMP-binding PDE (PDE5) and cone photoreceptor phosphodiesterase (PDE6alpha') in order to study structure and function of the photoreceptor enzyme. A fully functional chimeric PDE6alpha'/PDE5 enzyme containing the PDE6alpha' noncatalytic cGMP-binding sites, and the PDE5 catalytic domain has been efficiently expressed in the baculovirus/High Five cell system. The catalytic properties of this chimera were practically indistinguishable from those of PDE5, whereas the noncatalytic cGMP binding was similar to that of native purified PDE6alpha'. The inhibitory gamma subunit of PDE6 (Pgamma) enhanced the affinity of cGMP binding at noncatalytic sites of native PDE6alpha' by approximately 6-fold. The polycationic region of Pgamma, Pgamma-24-45, was mainly responsible for this effect, while the inhibitory domain of Pgamma, Pgamma-63-87, was ineffective. On the contrary, Pgamma failed to inhibit catalytic activity of the chimeric PDE6alpha'/PDE5 or to modulate its noncatalytic cGMP binding. Substitutions of Ala residues for the conserved Asn, Asn193 or Asn402, in the two N(K/R)XD-like motifs of the chimeric PDE noncatalytic cGMP-binding sites, each led to a loss of the noncatalytic cGMP binding. Our data suggest that both putative noncatalytic sites of PDE6alpha' are important for binding of cGMP, and that the two binding sites are coupled. Furthermore, mutation Asn402 --> Ala resulted in an approximately 10-fold increase of the Km value for cGMP, indicating that occupation of the noncatalytic cGMP- binding sites of PDE6alpha' may regulate catalytic properties of the enzyme.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Retinal Cone Photoreceptor Cells/enzymology , Rod Cell Outer Segment/enzymology , 3',5'-Cyclic-GMP Phosphodiesterases/chemistry , Alanine , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Catalysis , Cattle , Cell Line , Cloning, Molecular , Conserved Sequence , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5 , Kinetics , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Spodoptera , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...