Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 279(2): H601-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10924059

ABSTRACT

Severe ischemic injury or infarction of myocardium may cause activation of matrix metalloproteinases (MMPs) and damage the interstitial matrix. However, it is unknown whether MMP activation and matrix damage occur after moderate ischemia and reperfusion that result in myocardial stunning without infarction, and if so whether such changes contribute to postischemic myocardial expansion and contractile dysfunction. To address these questions, open-chest anesthetized pigs underwent 90 min of regional ischemia (subendocardial blood flow 0.4 +/- 0.1 ml. g(-1). min(-1)) and 90 min of reperfusion. After ischemia plus reperfusion, histological and ultrastructural examination revealed no myocardial infarction or inflammatory cell infiltration. Myocardial MMP-9 content increased threefold with a fourfold increase in the active form (P < 0.001). Myocardial collagenase content doubled (P < 0.01) but remained in latent form. MMP-2 and tissue inhibitors of metalloproteinases were unaffected. Despite increases in MMPs, collagen ultrastructure (assessed by cell maceration scanning electron microscopy) was unaltered. Intracoronary administration of the MMP inhibitor GM-2487 did not prevent or attenuate myocardial expansion (assessed by regional diastolic dimensions at near-zero left ventricular pressure) or contractile dysfunction. We conclude that although moderate ischemia and reperfusion alter myocardial MMP content and activity, these effects do not result in damage to interstitial collagen, nor do they contribute to myocardial expansion or contractile dysfunction.


Subject(s)
Collagen/ultrastructure , Hemodynamics , Matrix Metalloproteinases/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion , Myocardium/metabolism , Myocardium/ultrastructure , Animals , Blood Pressure , Collagenases/metabolism , Coronary Circulation , Female , Heart/physiopathology , Heart Rate , Matrix Metalloproteinase 9/metabolism , Myocardial Ischemia/metabolism , Myocardial Stunning/metabolism , Myocardial Stunning/pathology , Myocardial Stunning/physiopathology , Myocardium/pathology , Swine , Tissue Inhibitor of Metalloproteinases/metabolism , Ventricular Function, Left
2.
Diabetes ; 47(8): 1311-7, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9703333

ABSTRACT

The degree of hyperglycemia correlates with the development of diabetic retinopathy. We investigated the effect of glucose on the expression of matrix metalloproteinase (MMP)-2 and MMP-9 (72-kDa and 92-kDa type IV collagenases, respectively) by human retinal microvascular endothelial cells (HRECs). Cultured HRECs from nondiabetic and diabetic donors were exposed to 5 or 30 mmol/l glucose. Using gelatin zymography, conditioned medium (CM) from all cultures revealed a gelatinolytic band migrating at 65 kDa (representing the proform of MMP-2 that runs at 72 kDa under reducing conditions). This band was unchanged by glucose exposure or the disease state of the donors. CM from nondiabetic HREC cultures demonstrated an additional proteolytic activity migrating at 90 kDa when cells were exposed to 30 mmol/l glucose, but not when they were exposed to 5 mmol/l glucose. This same activity was seen in CM from HREC cultures of diabetic origin in the presence of both 5 and 30 mmol/l glucose. Western analysis confirmed the identity of the 65-kDa band as MMP-2. The anomalous activity at 90 kDa was identified as MMP-2 associated and co-migrating with a fibronectin fragment. Competition-based reverse transcription-polymerase chain reaction revealed that nondiabetic and diabetic HRECs expressed constitutively mRNA for MMP-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-2, and fibronectin. After exposure to 5 or 30 mmol/l glucose, no changes were detected in mRNA levels in MMP-2 or MMP-9, their inhibitors TIMP-1 and TIMP-2, or fibronectin in either nondiabetic or diabetic HREC cultures. These results support the notion that modulation of MMP function by extracellular matrix components occurs in response to glucose and may be relevant to the development of diabetic retinopathy.


Subject(s)
Collagenases/metabolism , Gelatinases/metabolism , Metalloendopeptidases/metabolism , Retinal Vessels/enzymology , Blotting, Western , Cells, Cultured , Collagenases/genetics , Diabetes Mellitus/enzymology , Diabetes Mellitus/pathology , Gelatinases/genetics , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Metalloendopeptidases/genetics , Microcirculation/physiology , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/metabolism , Retinal Vessels/pathology , Transcription, Genetic
3.
J Med Chem ; 41(2): 199-223, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9457244

ABSTRACT

Modifications around the dipeptide-mimetic core of a hydroxamic acid based matrix metalloproteinase inhibitor were studied. These variations incorporated a variety of natural, unnatural, and synthetic amino acids in addition to modifications of the P1' and P3' substituents. The results of this study indicate the following structural requirements: (1) Two key hydrogen bonds must be present between the enzyme and potent substrates. (2) Potent inhibitors must possess strong zinc-binding functionalities. (3) The potential importance of the hydrophobic group at position R3 as illustrated by its ability to impart greater relative potency against stromelysin when larger hydrophobic groups are used. (4) Requirements surrounding the nature of the amino acid appear to be more restrictive for stromelysin than for neutrophil collagenase, 72 kDa gelatinase, and 92 kDa gelatinase. These requirements may involve planar fused-ring aryl systems and possibly hydrogen-bonding capabilities.


Subject(s)
Dipeptides/chemistry , Dipeptides/pharmacology , Gelatinases/antagonists & inhibitors , Matrix Metalloproteinase Inhibitors , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Chromatography, High Pressure Liquid , Kinetics , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 8 , Matrix Metalloproteinase 9 , Models, Chemical , Structure-Activity Relationship
4.
Shock ; 7(6): 427-31, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9185243

ABSTRACT

Excessive tumor necrosis factor alpha (TNF alpha) production in response to Gram-negative bacteremia or endotoxemia can often lead to hypotension, shock, and increased mortality. Current approaches used to block the deleterious effects of exaggerated TNF alpha production rely on monoclonal antibodies or immunoadhesins that bind TNF alpha and thus prevent the interaction with its cellular receptors. This report examines whether a previously described inhibitor of matrix metalloproteinases, GM-6001, can inhibit TNF alpha processing and release and attenuate endotoxin-induced mortality. In human peripheral blood mononuclear cells stimulated in vitro with 1 microgram/mL endotoxin, GM-6001 at concentrations > 5 micrograms/mL blocked release of TNF alpha, but did not affect the release of either IL-1 beta or IL-6. GM-6001 also inhibited the release of soluble TNF receptor (p75) from peripheral blood mononuclear cells stimulated with endotoxin and/or TNF alpha. To confirm the role of secreted TNF alpha in endotoxic shock-induced mortality, C57BL/6 mice were challenged with either endotoxin alone (500 micrograms/mouse) or endotoxin (100 ng/mouse) plus D-galactosamine (8 mg/mouse). GM-6001 pretreatment (100 mg/kg) significantly attenuated the 90-minute plasma TNF alpha response in both models and improved survival in mice treated with low-dose endotoxin plus D-galactosamine. However, plasma IL-1 beta and IL-6 concentrations at 90 min after endotoxin treatment were unaffected by GM-6001 following lethal endotoxin challenge, confirming the in vivo specificity of this matrix metalloproteinase inhibitor for TNF alpha processing. These findings demonstrate that a novel inhibitor of matrix metalloproteinases can prevent the release of TNF alpha both in vitro and in vivo, and can abrogate the harmful sequelae of endotoxemic shock.


Subject(s)
Dipeptides/administration & dosage , Endotoxins , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/administration & dosage , Shock, Septic/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Shock, Septic/metabolism , Shock, Septic/mortality
5.
Arch Dermatol Res ; 289(3): 138-44, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9128761

ABSTRACT

Matrix metalloproteinases (MMPs) are a family of zinc-dependent proteases which play key roles in extracellular matrix remodeling, connective tissue damage, inflammation and cell proliferation in a variety of tissues. Since MMP inhibitors have been recently shown to decrease proliferation of vascular smooth-muscle cells, and to prevent neutrophil infiltration in response to alkali burns, we sought to determine whether MMPs play a role in the pathogenesis of inflammatory or hyperproliferative skin disorders. The effects of a specific MMP inhibitor and its analogues on phorbol dibutyrate (PdiBu)-induced inflammation and epidermal hyperplasia in murine skin were assessed. Topical GM 6001, a hydroxamic acid analog with potent inhibitory activity against several MMPs, markedly inhibited PdiBu-induced increases in both ear thickness and ear punch-biopsy weight in a dose-dependent manner 30 h after topical application of PdiBu. Maximal inhibition (75%) was obtained at a dose of 100 micrograms/cm2 (P < 0.01). Moreover, histologic analysis revealed that GM 6001 decreased both the inflammatory cellular infiltrates and epidermal hyperplasia induced by PdiBu. Whereas similar results were found for GM 1489, an analog of GM 6001, acetohydroxamic acid, containing the critical metal ligand group but without the amino acid side chains necessary for binding to the MMPs, did not alter the response to PdiBu inflammation/hyperplasia. These results show that the MMP inhibitors, GM 6001 and GM 1489, are effective in reducing both the inflammatory and hyperproliferative responses that occur following topical phorbol ester application, suggesting a potential role for MMPs in cutaneous inflammatory dermatoses. Moreover, the delivery of this class of inhibitors across intact stratum corneum implies that MMP inhibition could provide an approach to the topical treatment of inflammatory dermatoses.


Subject(s)
Dermatitis, Contact/pathology , Extracellular Matrix/enzymology , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Skin/drug effects , Skin/pathology , Animals , Dermatitis, Contact/etiology , Dipeptides/pharmacology , Hyperplasia , Mice , Mice, Hairless , Phorbol 12,13-Dibutyrate/pharmacology , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
6.
J Immunol ; 158(1): 414-9, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-8977217

ABSTRACT

TNF-alpha is a pleiotropic cytokine that exists both as a 26-kDa cell-associated and a 17-kDa soluble form. Recently, a class of matrix metalloproteinase inhibitors has been identified that can prevent the processing by TNF convertase of 26-kDa TNF-alpha to its 17-kDa form and can reduce mortality from normally lethal doses of D-galactosamine plus LPS (D-GalN/LPS). Here we report that a matrix metalloproteinase inhibitor, GM-6001, improves survival but does not protect against liver injury from D-GalN/LPS-induced shock in the mouse. In Con A-induced hepatitis, GM-6001 actually exacerbates hepatocellular necrosis and apoptosis despite greater than 90% reduction in plasma TNF-alpha concentrations. Treatment with GM-6001 also has minimal effect on the concentration of membrane-associated TNF-alpha in the livers of animals with Con A induced hepatitis. In contrast, a TNF binding protein (TNF-bp), which neutralizes both membrane-associated and soluble TNF-alpha, prevents D-GalN/LPS- and Con A-induced hepatitis. Our studies suggest that cell-associated TNF-alpha plays a role in the hepatocellular necrosis and apoptosis that accompany D-GalN/LPS- or Con A-induced hepatitis, and that matrix metalloproteinase inhibitors are ineffective in preventing this hepatic injury.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Dipeptides/toxicity , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/toxicity , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/toxicity , Animals , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/mortality , Concanavalin A/toxicity , Drug Synergism , Galactosamine/chemistry , Galactosamine/toxicity , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/injuries , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor/blood , Tumor Necrosis Factor-alpha/analysis
7.
Shock ; 6(5): 377-82, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8946655

ABSTRACT

Although matrix metalloproteinase inhibitors prevent the increase in soluble tumor necrosis factor-alpha during endotoxemia, it remains unknown whether a novel matrix metalloproteinase inhibitor, GM6001, improves cardiovascular and hepatocellular function after trauma and hemorrhage. To determine this, rats underwent laparotomy (i.e., trauma-induced), and were bled to and maintained at a mean arterial pressure of 40 mmHg until 40% of maximal shed volume was returned in the form of Ringer's lactate. The animals were then resuscitated with 3 times the volume of maximal bleedout with Ringer's lactate over 45 min, followed by 2 times Ringer's lactate over 60 min. GM6001, at a dose of 100 mg/kg or an equal volume of normal saline, was administered subcutaneously 15 min before the completion of resuscitation. At 2 and 4 h after resuscitation, cardiac output was measured by indocyanine green (ICG) dilution. Hepatocellular function (i.e., maximum velocity and the efficiency of ICG clearance) was determined by in vivo ICG clearance. Microvascular blood flow in various organs was assessed by laser Doppler flowmetry. The results indicate that cardiac output, hepatocellular function, and tissue microvascular blood flow decreased significantly at 2 and 4 h after resuscitation. GM6001 treatment, however, significantly improved the depressed cardiovascular and hepatocellular function. Since GM6001 improves cardiovascular and hepatocellular function, this agent may be a useful adjunct to fluid resuscitation after trauma and hemorrhagic shock.


Subject(s)
Cardiovascular System/drug effects , Dipeptides/pharmacology , Hemorrhage/drug therapy , Liver/drug effects , Protease Inhibitors/pharmacology , Animals , Cardiac Output/drug effects , Disease Models, Animal , Hemodynamics/drug effects , Liver/blood supply , Male , Metalloendopeptidases/antagonists & inhibitors , Microcirculation/drug effects , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Resuscitation , Wounds and Injuries/drug therapy
8.
J Clin Invest ; 94(6): 2177-82, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7989572

ABSTRACT

Gelatinases, belonging to the matrix metalloproteases, contribute to tissue destruction in inflammatory demyelinating disorders of the central nervous system such as multiple sclerosis. We used experimental autoimmune encephalomyelitis (EAE) as an animal model to evaluate the effect of a hydroxamate matrix metalloprotease inhibitor (GM 6001) on inflammatory demyelination. A single dose of the inhibitor, given intraperitoneally, provided sufficient levels in the cerebrospinal fluid of animals with EAE to induce at least a partial inhibition of the gelatinase activity in the cerebrospinal fluid. When administered daily either from the time of disease induction or from the onset of clinical signs, GM 6001 suppressed the development or reversed clinical EAE in a dose-dependent way, respectively. Animals returned to the same clinical course as the nontreated group after cessation of treatment. Animals treated from the onset of clinical signs had normal permeability of the blood-brain barrier, compared with the enhanced permeability in nontreated animals. These results indicate that matrix metalloprotease inhibition can reverse ongoing EAE. This effect appears to be mediated mainly through restoration of the damaged blood-brain barrier in the inflammatory phase of the disease, since, the degree of demyelination and inflammation did not differ between the treatment groups.


Subject(s)
Autoimmune Diseases/therapy , Demyelinating Diseases/therapy , Dipeptides/therapeutic use , Encephalomyelitis/therapy , Metalloendopeptidases/antagonists & inhibitors , Animals , Blood-Brain Barrier , Dipeptides/cerebrospinal fluid , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gelatinases/cerebrospinal fluid , Injections, Intraperitoneal , Mice , Mice, Inbred Strains , Permeability
9.
Circ Res ; 75(3): 539-45, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8062427

ABSTRACT

We have characterized matrix metalloproteinase expression in the rat carotid artery after two forms of arterial injury, balloon catheter denudation and nylon filament denudation. Gelatinolytic enzymes with molecular masses of 70 and 62 kD were produced constitutively in the rat carotid. Production of an 88-kD gelatinase was induced after balloon catheter injury, and proteinase production continued during the period of migration of smooth muscle cells from the media to the intima, from 6 hours to 6 days after balloon catheter injury. In addition, a marked increase in 62-kD gelatinolytic activity was observed between 4 and 14 days after arterial injury. Gelatinase activities (88 and 62 kD) were also increased after nylon filament denudation but were markedly less after this injury than after balloon catheter injury. These results suggested a correlation between gelatinase activity and smooth muscle cell migration after arterial injury. Administration of a metalloproteinase inhibitor after balloon catheter injury resulted in a 97% reduction in the number of smooth muscle cells migrating into the intima. Therefore, we hypothesize that gelatinase expression directly facilitates smooth muscle cell migration within the media and into the intima. These results suggest that gelatinases are involved in the vascular smooth muscle cell activation and neointimal formation that characterize arterial tissue remodeling after injury.


Subject(s)
Carotid Artery Injuries , Collagenases/biosynthesis , Endothelium, Vascular/physiology , Gene Expression , Muscle, Smooth, Vascular/injuries , Animals , Carotid Artery, Common/enzymology , Carotid Artery, Common/physiology , Catheterization , Cell Movement , Collagenases/analysis , Dipeptides/pharmacology , In Situ Hybridization , Male , Matrix Metalloproteinase 9 , Matrix Metalloproteinase Inhibitors , Molecular Weight , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Time Factors
11.
Cancer Res ; 54(17): 4715-8, 1994 Sep 01.
Article in English | MEDLINE | ID: mdl-7520359

ABSTRACT

The inhibitor N-[2R-2-(hydroxamidocarbonymethyl)-4-methylpentanoyl)]-L- tryptophan methylamide specifically blocks several matrix metalloproteases, enzymes which are thought to be involved in angiogenesis. An extract of Walker 256 carcinoma in Hydron pellets implanted in the corneas of Sprague-Dawley rats was used to stimulate angiogenesis from the vessels of the limbus. Angiogenesis was graded visually as the distance penetrated into the cornea and the number of vessels generated. The vessel area was also measured by image analysis using Image 1 software. Continuous i.v. administration of N-[2-(hydroxamidocarbonymethyl)-4-methylpentanoyl)]- L-tryptophan methylamide at 32 mg/kg/day (n = 17) via syringe pump reduced vessel number [25.06 +/- 5.9 (SEM) compared to 65.33 +/- 9.0] and vessel area (26.14 +/- 3.2 mm2 compared with 40.96 +/- 4.6 mm2), but not distance penetrated, compared to vehicle-treated control eyes after 6 days. These results confirm the suspected role for matrix metalloproteases in angiogenesis and suggest that inhibitors of these enzymes may be angiostatic agents.


Subject(s)
Carcinoma 256, Walker/blood supply , Dipeptides/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Neovascularization, Pathologic/prevention & control , Animals , Cornea , Metalloendopeptidases/physiology , Neoplasm Transplantation , Rats , Rats, Sprague-Dawley
12.
J Neurooncol ; 21(2): 141-50, 1994.
Article in English | MEDLINE | ID: mdl-7861190

ABSTRACT

The function of proteases in brain tumor invasion is currently not well established. For tumors of epithelial and fibromatous origin collagenase production can enhance the invasive capacity of cells to penetrate basement membranes. We showed previously that a c-Ha-ras transformed glial cell line (CxT24neo3) invaded hamster brain tissue in vivo. These cells were also capable of invading reconstituted basement membrane and embryonic chick hearts in vitro. Since the histopathology of CxT24neo3 tumors mimics that of glioblastoma multiforme in humans, CxT24neo3 was used as the model in vitro for this type of brain tumor. Presently, we detected by zymogram analysis a gelatinase that was secreted by CxT24neo3 and that had an apparent molecular mass of 62 kD. To verify whether gelatinase affected invasion in vitro of these glial cells we determined the efficacy of a substrate specific collagenase inhibitor on invasion in vitro. GM6001 is a synthetic polypeptide that specifically occupies the substrate binding sites of metalloprotease. Since this drug did not show cytotoxicity, its specificity for metalloprotease is a valuable tool to evaluate the physiological function of these enzymes on invasion. We found that treatment of CxT24neo3 with GM6001 reduced the fraction of invading CxT24neo3 cells through reconstituted basement membrane. These data suggest that metalloproteases can stimulate brain tumor invasion.


Subject(s)
Amides/pharmacology , Dipeptides/pharmacology , Gelatinases/antagonists & inhibitors , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neuroglia/pathology , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/physiology , Tyrosine/analogs & derivatives , Animals , Basement Membrane/pathology , Cell Line, Transformed , Cell Movement/drug effects , Chick Embryo , Collagen , Cricetinae , Culture Media, Conditioned/chemistry , Culture Media, Serum-Free , Drug Combinations , Gelatinases/metabolism , Heart/embryology , Laminin , Mesocricetus , Myocardium/pathology , Neoplasm Proteins/metabolism , Neuroglia/enzymology , Proteoglycans , Recombinant Fusion Proteins , Transfection , Tyrosine/pharmacology
13.
Invest Ophthalmol Vis Sci ; 33(12): 3325-31, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1385350

ABSTRACT

Healing of corneal alkali injuries remains a severe clinical challenge. The authors evaluated the effect of a new synthetic inhibitor of matrix metalloproteinases (GM6001 or N-[2(R)-2-(hydroxamido carbonylmethyl)-4-methylpentanoyl]-L-tryptophane methylamide) on preventing ulceration of rabbit corneas after alkali injury. Topical treatment of corneas with severe alkali injuries with 400 micrograms/ml or 40 micrograms/ml GM6001 alone prevented ulceration for 28 days, although 8 of 10 corneas treated with vehicle perforated. Corneas treated with 4 micrograms/ml GM6001 had midstromal depth ulcers. Corneas treated with 400 micrograms/ml of GM6001 contained very few inflammatory cells and had significantly reduced vessel ingrowth compared with vehicle-treated corneas. Epithelial regeneration after moderate alkali injuries also was investigated. Persistent epithelial defects developed 4 days after moderate alkali injury in rabbit corneas treated with vehicle and progressively increased to an average of 20% of the original 6 mm diameter wound by 27 days after moderate alkali injury. By contrast, epithelial regeneration was complete and persisted for 21 days for corneas treated with a formulation containing GM6001 (400 micrograms/ml), epidermal growth factor (10 micrograms/ml), fibronectin (500 micrograms/ml), and aprotinin (400 micrograms/ml). Sporadic punctate staining developed in 20% of the corneas treated with the combination of agents between days 21-28 after moderate alkali injury. These results demonstrate that topical application of GM6001 prevented corneal ulceration after severe alkali injury and that a combination containing GM6001, epidermal growth factor, fibronectin, and aprotinin promoted stable regeneration of corneal epithelium after moderate alkali injury.


Subject(s)
Alkalies , Burns, Chemical/drug therapy , Corneal Injuries , Extracellular Matrix/enzymology , Eye Burns/chemically induced , Metalloendopeptidases/antagonists & inhibitors , Animals , Aprotinin/pharmacology , Burns, Chemical/pathology , Cornea/pathology , Cornea/physiopathology , Corneal Ulcer/prevention & control , Dipeptides/chemistry , Dipeptides/therapeutic use , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Eye Burns/drug therapy , Eye Burns/pathology , Fibronectins/pharmacology , Rabbits , Regeneration
14.
Biochemistry ; 31(31): 7152-4, 1992 Aug 11.
Article in English | MEDLINE | ID: mdl-1322694

ABSTRACT

The hydroxamic acid HONHCOCH2CH(i-Bu)CO-L-Trp-NHMe, isomer 6A (GM 6001), inhibits human skin fibroblast collagenase with Ki of 0.4 nM using the synthetic thiol ester substrate Ac-Pro-Leu-Gly-SCH(i-Bu)CO-Leu-Gly-OEt at pH 6.5. The other isomer, 6B, which has the opposite configuration at the CH2CH(i-Bu)CO alpha-carbon atom, has a Ki of 200 nM for this enzyme. GM 6001 is one of the most potent inhibitors of human skin fibroblast collagenase yet reported. GM 6001 has a Ki of 20 nM against thermolysin and Pseudomonas aeruginosa elastase. Isomer 6B has a Ki of 7 nM against thermolysin and 2 nM against the elastase. 6A and 6B are the most potent hydroxamate inhibitors reported for these bacterial enzymes. The pattern of inhibition for all three enzymes suggests that isomer 6A is the (R,S) compound, stereochemically analogous to the L,L-dipeptide, and isomer 6B is the (S,S) compound, analogous to the DL-dipeptide. The tolerance of the D configuration by thermolysin and the elastase allows these inhibitors to discriminate between the human and bacterial enzymes simply by inversion of configuration at the CH2CH(i-Bu)CO alpha-carbon atom. Substitution of the potential metal liganding groups carboxylate and hydrazide for the hydroxamate group yields much weaker inhibitors for all three enzymes.


Subject(s)
Hydroxamic Acids/pharmacology , Microbial Collagenase/antagonists & inhibitors , Pancreatic Elastase/antagonists & inhibitors , Pseudomonas aeruginosa/enzymology , Skin/enzymology , Thermolysin/antagonists & inhibitors , Fibroblasts/enzymology , Humans , Kinetics , Skin/cytology
15.
Matrix Suppl ; 1: 259-62, 1992.
Article in English | MEDLINE | ID: mdl-1480035

ABSTRACT

Substitution of the phosphonamidate linkage (PO2-NH) for the peptide bond (CO-NH) in substrate-like sequences produces inhibitors of human skin fibroblast collagenase with Ki's far below Km for the native collagen substrate. Using a thiol ester substrate at pH 6.5, phthaloyl-GlyP-Ile-Trp-(S)NHCH-(Me)Ph, the phosphonamidate analog of phthaloyl-Gly-Ile-Trp-(S)NHCH(Me)Ph, has a Ki of 20 nM. Peptide phosphonamidates with amino acid sequences extended further to the right or the left of the Gly-Ile-Trp sequence had higher Ki's. Substitution of the phosphinate linkage (PO2-CH2) for the peptide bond also gives potent inhibitors such as napthoyl-GlyP-C-Leu-Trp-NHBzl, the phosphinate analog of naphtholyl-Gly-Leu-Trp-NHBzl, which has a Ki of 10 nM. Some of the phosphonamidates and phosphinates are also excellent inhibitors of the bacterial zinc metalloproteases thermolysin and Pseudomonas aeruginosa elastase.


Subject(s)
Enzyme Inhibitors/pharmacology , Fibroblasts/enzymology , Matrix Metalloproteinase Inhibitors , Peptides/pharmacology , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Humans , Kinetics , Metalloendopeptidases/antagonists & inhibitors , Molecular Sequence Data , Molecular Structure , Peptides/chemical synthesis , Skin/cytology , Structure-Activity Relationship , Thermolysin/antagonists & inhibitors
16.
Invest Ophthalmol Vis Sci ; 32(11): 2997-3001, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1655675

ABSTRACT

Normal and abnormal processes of cellular invasion often are initiated by degradation of basement membranes. The process of corneal ulceration might operate via similar mechanisms; degradation of the corneal stroma is not seen until after the basement membrane underlying the corneal epithelium in the preulcerative lesion is lost. Recent data implicate a member of the matrix metalloproteinase (MMP) family of enzymes, 92 kD gelatinase/type IV collagenase (MMP-9) in both cellular invasion processes and degradation of epithelial basement membrane before corneal ulceration. This suggests that use of nontoxic substances that block activity of MMP-9 might be useful in preventing or inhibiting pathologic invasion processes in vivo. An agent that fits these criteria is N-[D,L-2-isobutyl-3(N'-hydroxycarbonylamido)-propanoyl]-O- methyl-L-tyrosine methylamide, which previously has been characterized as an inhibitor of tumor cell collagenases. In this study, the authors show that the inhibitor can efficiently block activity of MMP-9 purified from cultures of rabbit corneal epithelial cells. Results suggest that the recently reported efficacy of a closely related inhibitor in blocking progression of alkali burns to ulceration might be attributable to its action against MMP-9.


Subject(s)
Amides/pharmacology , Cornea/enzymology , Microbial Collagenase/antagonists & inhibitors , Tyrosine/analogs & derivatives , Animals , Cells, Cultured , Chromatography, Affinity , Dipeptides , Electrophoresis, Polyacrylamide Gel , Epithelium/enzymology , Fibroblasts/enzymology , Gelatin/metabolism , Humans , Matrix Metalloproteinase 9 , Microbial Collagenase/isolation & purification , Rabbits , Tyrosine/pharmacology
17.
Biochem Biophys Res Commun ; 169(3): 1111-6, 1990 Jun 29.
Article in English | MEDLINE | ID: mdl-2114105

ABSTRACT

The phosphinic acid isosteres of di-, tetra- and hexapeptides containing a hydrophobic amino acid side chains at the P1-P'1 positions are powerful inhibitors of Human Immunodeficiency Virus protease. Ki's ranged from 0.4 nM to 26 microM at pH 6.5 and were lower at pH 4.5. The compounds showed no activity against trypsin, weak activity against renin at pH 6.5, moderate activity against pepsin at pH 2.0 (Ki values in the microM range) and substantial activity against cathepsin D at pH 3.5 (Ki values from 9 to 300 nM).


Subject(s)
Endopeptidases/metabolism , Gene Products, pol/metabolism , HIV-1/enzymology , Oligopeptides/pharmacology , Phosphinic Acids/pharmacology , Protease Inhibitors , Cathepsin D/antagonists & inhibitors , Esters , HIV Protease , Hydrogen-Ion Concentration , Pepsin A/antagonists & inhibitors , Structure-Activity Relationship
18.
J Med Chem ; 33(1): 263-73, 1990 Jan.
Article in English | MEDLINE | ID: mdl-2153207

ABSTRACT

An extensive series of N-(monoethylphosphoryl)peptides was synthesized and their inhibition of purified human skin fibroblast collagenase examined. At the cleavage site S1 all reported compounds have the (EtO)(OK)P(O) group and the peptide side chain extended toward the C-terminal end (up to P5') of the substrate sequence. These phosphoramidates with a tetrahedrally hybridized phosphorus atom are thought to be transition state analogue inhibitors. They exhibited fair inhibitory potency against this vertebrate collagenase having Ki values in the micromolar range. The most potent of these, (EtO)(OK)P(O)-Ile-TrpNHCH3 (68), inhibits with a Ki value of 1.5 microM and is nearly 100 times stronger than (EtO)(OK)P(O)-Ile-Ala-GlyOK (51) (Ki of 140 microM), which has the sequence matching that of the alpha 1 (I) chain of collagen in P1', P2', P3' after the cleavage site. Several compounds were prepared in an attempt to identify the nature of the S2', S3', and S4' binding sites. Alanine at the P2' position was replaced by leucine, phenylalanine, tryptophan, or tyrosine derivatives, resulting in Ki values in a significantly lower range, 1.0-40 microM, compared to 51. No upper size limitation or specificity has been found at this position, yet similar replacements at the P3' position, which is occupied naturally by a glycine residue, gave weaker inhibitors: (EtO)(OK)P(O)-Ile-Tyr(OBzl)-PheOK (57) had a Ki of 120 microM. Hexapeptide derivatives had weaker activities in the 270 microM-2 mM range. All inhibitors were evaluated by using the synthetic thio peptolide spectrophotometric assay.


Subject(s)
Dipeptides/pharmacology , Fibroblasts/enzymology , Microbial Collagenase/antagonists & inhibitors , Oligopeptides/pharmacology , Organophosphorus Compounds/pharmacology , Amino Acid Sequence , Chemical Phenomena , Chemistry , Dipeptides/chemical synthesis , Humans , Hydrogen-Ion Concentration , Molecular Sequence Data , Oligopeptides/chemical synthesis , Organophosphorus Compounds/chemical synthesis , Structure-Activity Relationship
19.
Biochemistry ; 28(12): 4948-51, 1989 Jun 13.
Article in English | MEDLINE | ID: mdl-2765520

ABSTRACT

The importance of a specific hydrogen bond between thermolysin and a phosphonamidate inhibitor, Z-NHCH2-PO(O-)-Leu-Leu (1) [Bartlett, P. A., & Marlowe, C. K. (1987) Science (Washington D.C.) 235, 569-571], has been reevaluated. We have determined the inhibition constants (binding free energies) for thermolysin of phosphonamidate n-hexyl-P(O)(O-)-Leu-Trp-NHMe (4), phosphonate n-hexyl-P-(O)(O-)OCH(iBu)CO-Trp-NHMe (5), and phosphinates n-hexyl-P(O)(O-)CH2CH(iBu)CO-Trp-NHMe (6) and Z-NHCH2PO(O-)CH2CH(iBu)CO-Leu (3). Replacement of the P-NH group by P-CH2 (1----3 and 4----6) weakens the overall binding free energy by about 1.5 kcal/mol. A negligible difference in solvation energy has been measured for these pairs, and the basicity of the P-O- ligand for zinc in each pair remains nearly unchanged as determined by pH titration of their 31P NMR resonances. Therefore, this value of 1.5 kcal/mol can be assigned to the specific hydrogen bond known to exist between the P-NH of 1 and thermolysin [Tronrud, D. E., Holden, H. M., & Matthews, B. W. (1987) Science (Washington, D.C.) 235, 871-574] and inferred to exist between 4 and the enzyme. Substitution of P-O for P-NH (1----2 [Bartlett, P. A., & Marlowe, C. K. (1987) Science (Washington, D.C.) 235, 569-571] and 4----5) weakens the overall binding free energy by 4.1 kcal/mol for each pair as the basicity of the P-O- ligand decreases by about 1.6 pH units. The measured solvation energy difference between 4 and 5 (and by inference between 1 and 2) is negligible.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Organophosphorus Compounds , Thermolysin/antagonists & inhibitors , Binding Sites , Energy Transfer , Hydrogen Bonding , Hydrogen-Ion Concentration , Kinetics , Ligands , Magnetic Resonance Spectroscopy , Organophosphorus Compounds/chemical synthesis , Solubility , Thermodynamics
20.
Biochem Biophys Res Commun ; 159(2): 426-31, 1989 Mar 15.
Article in English | MEDLINE | ID: mdl-2539108

ABSTRACT

The ketone cinnamoyl-(1-13C-Phe)-CGly-Pro-Pro [(4-13C-5-cinnamido-4-oxo-6-phenylhexanoyl)-Pro-Pro 2] competitively inhibits a mixture of collagenases from Clostridium histolyticum with a Ki of 40 +/- 6 nM. 13C-nmr spectroscopy of the ketone in the presence of this collagenase shows a bound 13C resonance at 102.6 ppm and the resonance of the free ketone at 212 ppm. Ketone alone shows no trace (less than 0.5%) of a resonance in the region around 100 ppm. The bound resonance is displaceable by another competitive inhibitor. This ketone is thus a transition state analog which is rehybridized from trigonal planar to tetrahedral upon binding to collagenase.


Subject(s)
Clostridium/enzymology , Dipeptides/pharmacology , Ketones/pharmacology , Microbial Collagenase/antagonists & inhibitors , Binding Sites , Binding, Competitive , Kinetics , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...