Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Nat Med ; 6(11): 1248-52, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11062536

ABSTRACT

We covalently linked doxorubicin with a peptide that is hydrolyzable by prostate-specific antigen. In the presence of prostate tumor cells secreting prostate-specific antigen, the peptide moiety of this conjugate, L-377,202, was hydrolyzed, resulting in the release of leucine-doxorubicin and doxorubicin, which are both very cytotoxic to cancer cells. However, L-377,202 was much less cytotoxic than conventional doxorubicin to cells in culture that do not secrete prostate-specific antigen. L-377,202 was approximately 15 times more effective than was conventional doxorubicin at inhibiting the growth of human prostate cancer tumors in nude mice when both drugs were used at their maximally tolerated doses. Nude mice inoculated with human prostate tumor cells secreting prostate-specific antigen showed considerable reductions in tumor burden with minimal total body weight loss when treated with L-377, 202. This improvement in therapeutic index correlated with the selective localization of leucine-doxorubicin and free doxorubicin in tissues secreting prostate-specific antigen after exposure to L-377,202.


Subject(s)
Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Oligopeptides/therapeutic use , Prodrugs/therapeutic use , Prostate-Specific Antigen/physiology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Animals , Doxorubicin/pharmacokinetics , Humans , Male , Mice , Mice, Nude , Oligopeptides/pharmacokinetics , Prodrugs/pharmacokinetics , Prostate-Specific Antigen/analysis , Prostate-Specific Antigen/blood , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
J Cell Biol ; 111(4): 1713-23, 1990 Oct.
Article in English | MEDLINE | ID: mdl-2211834

ABSTRACT

The venom protein, s-echistatin, originally derived from the saw-scaled viper Echis carinatus, was found to be a potent inhibitor of bone resorption by isolated osteoclasts. This Arg24-Gly25-Asp26-(RGD)-containing protein inhibited the excavation of bone slices by rat osteoclasts (IC50 = 0.1 nM). It also inhibited the release of [3H]proline from labeled bone particles by chicken osteoclasts (IC50 = 100 nM). By comparison, the tetrapeptide Arg-Gly-Asp-Ser (RGDS) inhibited resorption by rat or chicken osteoclasts with an IC50 of 0.1 mM while ala24-echistatin was inactive. Video microscopy showed that rat osteoclast attachment to substrate was more sensitive to s-echistatin than was the attachment of mononuclear cells or chicken osteoclasts. The difference in sensitivity of rat and chicken osteoclasts to s-echistatin may be due to differences between receptors on rat and chicken osteoclasts for s-echistatin. Antibody localization of echistatin on these cells showed much greater echistatin binding to rat osteoclasts than to chicken osteoclasts. Laser scanning confocal microscopy after immunohistochemical staining showed that s-echistatin binds to osteoclasts, that s-echistatin receptors are most abundant at the osteoclast/glass interface, and that s-echistatin colocalizes with vinculin. Confocal interference reflection microscopy of osteoclasts incubated with s-echistatin, demonstrated colocalization of s-echistatin with the outer edges of clusters of grey contacts at the tips of some lamellipodia. Identification of the echistatin receptor as an integrin was confirmed by colocalization of echistatin fluorescence with staining for an alpha-like subunit. Attachment of bone particles labeled with [3H]proline to chicken osteoclasts confirmed that the mechanism of action of echistatin was to inhibit osteoclast binding to bone presumably by disrupting adhesion structures. These data demonstrate that osteoclasts bind to bone via an RGD-sequence as an obligatory step in bone resorption, that this RGD-binding integrin is at adhesion structures, and that it colocalizes with vinculin and has an alpha-like subunit.


Subject(s)
Bone Resorption , Osteoclasts/drug effects , Viper Venoms/pharmacology , Amino Acid Sequence , Animals , Cell Adhesion/physiology , Chickens , In Vitro Techniques , Integrins/metabolism , Intercellular Signaling Peptides and Proteins , Molecular Sequence Data , Oligopeptides/pharmacology , Osteoclasts/metabolism , Peptides/pharmacology , Rats
3.
Mol Cell Biol ; 12(5): 1905-14, 1992 May.
Article in English | MEDLINE | ID: mdl-1314947

ABSTRACT

The human papillomavirus E7 gene can transform murine fibroblasts and cooperate with other viral oncogenes in transforming primary cell cultures. One biochemical property associated with the E7 protein is binding to the retinoblastoma tumor suppressor gene product (pRB). Biochemical properties associated with pRB include binding to viral transforming proteins (E1A, large T, and E7), binding to cellular proteins (E2F and Myc), and binding to DNA. The mechanism by which E7 stimulates cell growth is uncertain. However, E7 binding to pRB inhibits binding of cellular proteins to pRB and appears to block the growth-suppressive activity of pRB. We have found that E7 also inhibits binding of pRB to DNA. A 60-kDa version of pRB (pRB60) produced in reticulocyte translation reactions or in bacteria bound quantitatively to DNA-cellulose. Recombinant E7 protein used at a 1:1 or 10:1 molar ratio with pRB60 blocked 50 or greater than 95% of pRB60 DNA-binding activity, respectively. A mutant E7 protein (E7-Ala-24) with reduced pRB60-binding activity exhibited a parallel reduction in its blocking of pRB60 binding to DNA. An E7(20-29) peptide that blocks binding of E7 protein to pRB60 restored the DNA-binding activity of pRB60 in the presence of E7. Peptide E7(2-32) did not block pRB60 binding to DNA, while peptide E7(20-57) and an E7 fragment containing residues 1 to 60 partially blocked DNA binding. E7 species containing residues 3 to 75 were fully effective at blocking pRB60 binding to DNA. These studies indicate that E7 protein specifically blocks pRB60 binding to DNA and suggest that the E7 region responsible for this property lies between residues 32 and 75. The functional significance of these observations is unclear. However, we have found that a point mutation in pRB60 that impairs DNA-binding activity also blocks the ability of pRB60 to inhibit cell growth. This correlation suggests that the DNA-binding activity of retinoblastoma proteins contributes to their biological properties.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Retinoblastoma Protein/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Cell Division , Chromatography, Affinity , Chromosome Deletion , Cloning, Molecular , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Genes, Retinoblastoma , Genes, Viral , Humans , Kinetics , Mice , Molecular Sequence Data , Oligonucleotide Probes , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Peptides/chemical synthesis , Protein Binding , Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Retinoblastoma Protein/genetics
4.
Mol Cell Biol ; 13(2): 953-60, 1993 Feb.
Article in English | MEDLINE | ID: mdl-7678696

ABSTRACT

Human papillomaviruses (HPVs) are the etiological agents for genital warts and contribute to the development of cervical cancer in humans. The HPV E7 gene product is expressed in these diseases, and the E7 genes from HPV types 16 and 18 contribute to transformation in mammalian cells. Mutation and deletion analysis of this gene suggests that the transforming activity of the protein product resides in the same domain as that which is directly involved in complex formation with the retinoblastoma gene product (pRB). This domain is one of two conserved regions (designated CRI and CRII) shared by E7 and other viral oncoproteins which bind pRB, including adenovirus E1A protein. Binding of HPV type 16 E7 protein to pRB has previously been shown to affect pRB's ability to bind DNA and to form complexes with other cellular proteins. In the current study, we map the functional interaction between E7 protein and pRB by monitoring the association between a 60-kDa version of the pRB, pRB60, and the cellular transcription factor E2F. We observe that CRII of E7 (amino acids 20 to 29), which completely blocks binding of full-length E7 protein, is necessary but not sufficient to inhibit E2F/pRB60 complex formation. While CRI of E1A (amino acids 37 to 55) appears to be sufficient to compete with E2F for binding to pRB60, the equivalent region of E7 is neither necessary nor sufficient. Only E7 fragments that contained both CRII and at least a portion of the zinc-binding domain (amino acids 60 to 98) inhibited E2F/pRB60 complex formation. These results suggest that pRB60 associates with E7 and E2F through overlapping but distinct domains.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Oncogene Proteins, Viral/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Base Sequence , Binding, Competitive , DNA/metabolism , E2F Transcription Factors , Epitopes , HeLa Cells , Humans , Molecular Sequence Data , Oncogene Proteins, Viral/chemistry , Papillomavirus E7 Proteins , Peptide Fragments/metabolism , Retinoblastoma Protein/chemistry , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/chemistry
5.
Mol Cell Biol ; 8(8): 2999-3007, 1988 Aug.
Article in English | MEDLINE | ID: mdl-2850475

ABSTRACT

Transforming growth factor alpha (TGF-alpha) is a 50-amino-acid peptide that stimulates cell proliferation via binding to cell surface receptors. To identify the structural features of TGF-alpha that govern receptor-ligand interactions, we prepared synthetic peptide fragments and recombinant mutant proteins of TGF-alpha. These TGF-alpha derivatives were tested in receptor binding and mitogenesis assays. Synthetic peptides representing the N terminus, the C terminus, or the individual disulfide constrained rings of TGF-alpha did not exhibit receptor-binding or mitogenic activity. Replacement of the cysteines with alanines at positions 8 and 21, 16 and 32, and 34 and 43 or at positions 8 and 21 and 34 and 43 yielded inactive mutant proteins. However, mutant proteins containing substitutions or deletions in the N-terminal region retained significant biologic activity. Conservative amino acid changes at residue 29 or 38 or both and a nonconservative amino acid change at residue 12 had little effect on binding or mitogenesis. However, nonconservative amino acid changes at residues 15, 38, and 47 produced dramatic decreases in receptor binding (23- to 71-fold) and mitogenic activity (38- to 125-fold). These studies indicate that at least three distinct regions of TGF-alpha contribute to biologic activity.


Subject(s)
Cell Division/drug effects , Recombinant Proteins/pharmacology , Transforming Growth Factors/pharmacology , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , DNA Restriction Enzymes , Escherichia coli/genetics , Molecular Sequence Data , Nucleic Acid Hybridization , Plasmids , Protein Conformation , Structure-Activity Relationship , Transforming Growth Factors/chemical synthesis , Transforming Growth Factors/genetics
6.
Biochim Biophys Acta ; 1092(2): 226-32, 1991 Apr 17.
Article in English | MEDLINE | ID: mdl-1850303

ABSTRACT

In crosslinking experiments, [125I]endothelin-1 was treated with N-hydroxysuccinimidyl-4-azidobenzoate, purified by HPLC, allowed to bind to bovine aortic membranes and then photoactivated. Autoradiography of sodium dodecyl sulfate polyacrylamide gel electrophoretograms of the products of this reaction showed that a component of apparent Mr = 42,000 was specifically labelled by endothelin-1 under reducing conditions. Under nonreducing conditions, a small amount of 125I-labelled endothelin-1 specifically labelled a component of apparent Mr = 45,900 in the absence of crosslinking agent. Non-radiolabelled endothelin analogues with a wide range of binding affinities inhibited specific labelling of the Mr = 42,000 and 45,900 components in parallel over the concentration ranges which inhibited binding of radiolabelled endothelin. Specific labelling of these components was also observed in parallel in membranes from bovine heart and kidney. The components labelled in the presence and absence of crosslinker appear to be the same, and the small difference in apparent Mr in the labelled components is likely due to a difference in conformational constraints arising from the two labelling processes, with a true, corrected Mr of 43,400. Since the specific labelling of this component is related to physiologically relevant binding in several bovine tissues, we conclude that it is a component of the bovine endothelin receptor.


Subject(s)
Receptors, Cell Surface/chemistry , Affinity Labels , Animals , Aorta/metabolism , Azides , Binding, Competitive , Cattle , Cell Membrane/metabolism , Cross-Linking Reagents , Electrophoresis, Polyacrylamide Gel , Endothelins/metabolism , Ethylmaleimide/pharmacology , Molecular Weight , Photochemistry , Receptors, Cell Surface/metabolism , Receptors, Endothelin
7.
Endocrinology ; 103(1): 81-5, 1978 Jul.
Article in English | MEDLINE | ID: mdl-744088

ABSTRACT

A somatostatin analog, [D-Ala5, D-Trp8]-somatostatin, has been found to selectively inhibit insulin and GH release in rats. The release of these hormones is inhibited by an analog dose of 5 microgram/kg in short term experiments (15 min from analog administration to blood sampling), while glucagon levels are not lowered by analog doses as high as 500 microgram/kg. The lowered insulin to glucagon ratio results in hyperglycemia. [D-Ala5, D-Trp8]Somatostatin is also long acting; a 1 mg/kg dose results in hypoinsulinemia for 2 h and hyperglycemia for 3 h.


Subject(s)
Insulin/blood , Somatostatin/analogs & derivatives , Alanine , Animals , Blood Glucose/metabolism , Glucagon/blood , Kinetics , Male , Rats , Somatostatin/pharmacology , Tryptophan
8.
Endocrinology ; 132(3): 1411-3, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8440195

ABSTRACT

Osteoclastic bone resorption requires the formation of a tightly sealed compartment between the osteoclast and the mineralized bone matrix. This compartment functions as an extracellular "lysosome" which contains proteolytic enzymes and acids. Vitronectin receptors (VnR, integrin alpha v beta 3) displayed on the osteoclast cell surface may play a role in the attachment of osteoclasts to the resorption surface. VnR are known to bind to arginyl-glycyl-aspartyl (RGD)-containing matrix proteins and it has recently been reported that soluble peptides containing RGD sequences can block osteoclast attachment to bone and inhibit bone resorption in vitro. In this study echistatin, a naturally-occurring protein containing an RGD-sequence motif, was shown to completely inhibit osteoclast-mediated bone resorption in vivo. Echistatin or smaller derivative peptides may prove useful in the treatment of disorders characterized by excess bone resorption, such as osteoporosis and metastatic bone disease.


Subject(s)
Bone Resorption/prevention & control , Oligopeptides/pharmacology , Osteoclasts/drug effects , Parathyroid Hormone/pharmacology , Peptides , Viper Venoms/pharmacology , Animals , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Calcium/blood , Intercellular Signaling Peptides and Proteins , Male , Osteoclasts/metabolism , Osteoclasts/pathology , Parathyroidectomy , Rats , Rats, Sprague-Dawley , Thyroidectomy
9.
J Med Chem ; 36(26): 4276-92, 1993 Dec 24.
Article in English | MEDLINE | ID: mdl-8277510

ABSTRACT

A series of 3-(arylureido)-5-phenyl-1,4-benzodiazepines, nonpeptidal antagonists of the peptide hormone cholecystokinin (CCK), are described. Derived by reasoned modification of the CCK-A selective 3-carboxamido-1,4-benzodiazepine, MK-329, this paper chronicles the development of potent, orally effective compounds in which selectivity for the CCK-B receptor subtype was achieved. The principal lead structure that emerged from these studied is L-365,260, a compound which has been submitted for clinical evaluation. Details of the ability to modulate the receptor interactions of these benzodiazepines by appropriate structure modifications are discussed which imply the possibility of further refining the CCK-B receptor affinity and selectivity of this class of compounds.


Subject(s)
Benzodiazepines/chemical synthesis , Benzodiazepinones/chemical synthesis , Phenylurea Compounds , Receptors, Cholecystokinin/antagonists & inhibitors , Animals , Benzodiazepines/pharmacokinetics , Benzodiazepines/pharmacology , Benzodiazepinones/chemistry , Benzodiazepinones/pharmacokinetics , Benzodiazepinones/pharmacology , Biological Availability , Cerebral Cortex/metabolism , Devazepide , Guinea Pigs , Molecular Structure , Pancreas/metabolism , Rats , Sincalide/metabolism , Structure-Activity Relationship
10.
J Med Chem ; 35(21): 3905-18, 1992 Oct 16.
Article in English | MEDLINE | ID: mdl-1331448

ABSTRACT

A new structural class of cyclic hexapeptide oxytocin antagonists derived from Streptomyces silvensis and typified by L-365,209 (cyclo-[L-prolyl1-D-phenylalanyl2-L- isoleucyl3-D-dehydropiperazyl4-L-dehydroperazyl5-D-(N- methyl)phenylalanyl6]) was recently reported. In this paper we further delineate the structure-activity profile for this new class by systematic study of L-365,209 analogs obtained by total synthesis. The optimal combination of cyclic amino acid ring sizes at positions 1, 4, and 5 and the role of the N-alkyl substituent at position 6 was elucidated. The lipophilic amino acids at positions 2 and 3 and the unusual amino acid D-dehydropiperazic acid at position 4 were found to be the most critical residues for obtaining good oxytocin receptor affinity. Analogs containing a basic side chain at the less critical 5- and 6-positions maintained good receptor affinity and also had useful levels of water solubility for intravenous formulation. By combining potency- and solubility-enhancing substitutions, several analogs were identified that have the desired combination of properties in vitro (22, cyclo-[L-prolyl-D-tryptophanyl-L-isoleucyl-D-pipecolyl-L-pipeco lyl-D- histidyl]; 25, cyclo-[L-prolyl-D-2-naphthylalanyl-L-isoleucyl-D-pipecolyl-L -pipecolyl-D- histidyl]; 26, cyclo-[L-prolyl-D-tryptophanyl-L-isoleucyl-D-dehydropiperazyl-L-++ pipecolyl-D-histidyl]; 33, cyclo-[L-prolyl-D-tryptophanyl-L-isoleucyl-D-pipecolyl-L- piperazinylcarboxy-D-(N-methyl)phenylalanyl]; 34, cyclo-[L-prolyl-D-phenylalanyl-L-isoleucyl-D-dehydropiperazyl-L-or nithyl- D-(N-methyl)phenylalanyl]). In general, this class exhibited good selectivity for binding to the oxytocin receptor versus the arginine vasopressin V1a and V2 receptor subtypes, although increased V2 receptor affinity was observed in one case (32, cyclo[L-prolyl-D-2-naphthylalanyl-L-isoleucyl-D-pipecolyl-L- lysyl-D-(N- methyl)phenylalanyl]). Unexpectedly, compound 33 was found to stimulate contractions of the isolated rat uterus via activation of the uterine bradykinin receptor. Compounds 22, 25, 26, 33, and 34 were found to be potent antagonists of oxytocin-stimulated contraction of the rat uterus in vitro and in vivo. Compounds 22 and 25 were additionally characterized as potent antagonists of oxytocin-stimulated uterine contractions in the near-term pregnant rhesus monkey. These studies thus demonstrate the selectivity and efficacy of certain members of this novel class of antagonists and suggest their use as pharmacological tools in further defining the role of oxytocin in both term and preterm labor.


Subject(s)
Oxytocin/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Amino Acid Sequence , Animals , Female , Haplorhini , In Vitro Techniques , Male , Molecular Sequence Data , Oxytocin/metabolism , Peptides, Cyclic/chemistry , Radioligand Assay , Rats , Receptors, Angiotensin/metabolism , Receptors, Oxytocin , Streptomyces/metabolism , Structure-Activity Relationship , Substrate Specificity
11.
J Med Chem ; 44(24): 4216-24, 2001 Nov 22.
Article in English | MEDLINE | ID: mdl-11708923

ABSTRACT

Doxorubicin (Dox) can provide some stabilization in prostate cancer; however, its use is limited because of systemic toxicities, primarily cardiotoxicity and immunosuppression. The administration of a prodrug of doxorubicin, designed to permit selective activation by the tumor, would reduce general systemic exposure to the active drug and would thereby increase the therapeutic index. Prostate specific antigen (PSA) is a serine protease with chymotrypsin-like activity that is a member of the kallikrein gene family. PSA's putative physiological role is the liquefaction of semen by virtue of its ability to cleave the seminal fluid proteins semenogelins I and II. Serum PSA levels have been found to correlate well with the number of malignant prostate cells. The use of a prodrug which is cleaved by the enzyme PSA in the prostate should in principle produce high localized concentrations of the cytotoxic agent at the tumor site while limiting systemic exposure to the active drug. Cleavage maps following PSA treatment of human semenogelin were constructed. Systematic modification of the amino acid residues flanking the primary cleavage site led to the synthesis of a series of short peptides which were efficiently hydrolyzed by PSA. Subsequent coupling of selected peptides to doxorubicin provided a series of doxorubicin-peptide conjugates which were evaluated in vitro and in vivo as targeted prodrugs for PSA-secreting tumor cells. From these studies we selected Glutaryl-Hyp-Ala-Ser-Chg-Gln-Ser-Leu-Dox, 27, as the peptide-doxorubicin conjugate with the best profile of physical and biological properties. Compound 27 has a greater than 20-fold selectivity against human prostate PSA-secreting LNCaP cells relative to the non-PSA-secreting DuPRO cell line. In nude mouse xenograft studies, 27 reduced PSA levels by 95% and tumor weight by 87% at a dose below its MTD. Both doxorubicin and Leu-Dox (13) were ineffective in reducing circulating PSA and tumor burden at their maximum tolerated doses. On the basis of these results, we selected 27 for further study to assess its ability to inhibit human prostate cancer cell growth and tumorigenesis.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Oligopeptides/chemistry , Peptide Fragments/chemistry , Prodrugs/chemistry , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/toxicity , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Doxorubicin/chemical synthesis , Doxorubicin/pharmacology , Doxorubicin/toxicity , Drug Screening Assays, Antitumor , Humans , Male , Mass Spectrometry , Mice , Mice, Nude , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Oligopeptides/toxicity , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Prodrugs/toxicity , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Structure-Activity Relationship , Transplantation, Heterologous , Tumor Cells, Cultured
12.
Thromb Haemost ; 64(4): 576-81, 1990 Dec 28.
Article in English | MEDLINE | ID: mdl-2084943

ABSTRACT

A model of acute, platelet-dependent canine coronary artery thrombosis was utilized to assess the antithrombotic effect of a synthetic, RGD-containing 49-residue protein termed echistatin. This protein is derived from the venom of the viper, Echis carinatus. In vitro, echistatin inhibited ADP (10 microM)-induced platelet aggregation with IC50 values in human and canine platelet-rich plasma of 101 +/- 4 and 127 +/- 32 nM, respectively. In vivo, in the dog, infusion of echistatin for 30 min at 20 micrograms kg-1 min-1 or 2.6 nM kg-1 min-1 resulted in total abolition of acute platelet-dependent coronary thrombus formation in all dogs tested (n = 5). Infusion of a lower dose (10 micrograms kg-1 min-1) was not effective in prevention of thrombus formation. Blood samples were taken before and after infusion of echistatin in order to determine ex vivo platelet aggregatory responses. Echistatin (20 micrograms kg-1 min-1, i.v.) attenuated ex vivo platelet aggregation elicited by ADP, U-46619 and collagen and increased bleeding time by 2.9 +/- 0.5-fold over control. Thus, in the dog, echistatin is an effective antithrombotic agent inhibiting both platelet aggregation in vivo in the coronary artery as well as ex vivo with a concomitant increase in bleeding time. Furthermore, the effects of echistatin on platelet aggregation and bleeding time are reversible with restoration to control levels occurring 30-60 min after termination of the infusion.


Subject(s)
Coronary Thrombosis/prevention & control , Peptides , Platelet Aggregation Inhibitors/therapeutic use , Viper Venoms/therapeutic use , Animals , Bleeding Time , Disease Models, Animal , Dogs , Female , Hemodynamics/drug effects , Intercellular Signaling Peptides and Proteins , Male
13.
Ann N Y Acad Sci ; 853: 63-78, 1998 Sep 16.
Article in English | MEDLINE | ID: mdl-10603937

ABSTRACT

The cardiac sarcoplasmic reticulum (SR) protein phospholamban (PLB) is an endogenous inhibitor of the SR Ca(2+)-ATPase. Phosphorylation of PLB relieves this inhibition and up-regulates calcium transport. PLB has proved remarkably difficult to study by conventional solution-state nuclear magnetic resonance (NMR) methods, due primarily to the extreme hydrophobic nature of the protein and its propensity to form pentamers. That the C-terminal domain of PLB is helical and membrane spanning is now well established; the structure of the cytoplasmic domain is relatively ill defined. In order to discern the effect of phosphorylation on the structure of the cytoplasmic domain, we have characterized a variety of model peptides in several structure-inducing and/or lipid-mimicking environments using circular dichroism and solution-state NMR. The resolution of peptide structures obtained in aqueous trifluoroethanol was markedly improved by the incorporation of 15N labels into the peptide backbone, allowing a variety of isotope edited, filtered, and resolved techniques to be applied. Molecular dynamics simulations on the full-length protein were combined with an analysis of published data to suggest a revised model for the structure of PLB.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/metabolism , Amino Acid Sequence , Computer Simulation , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Phosphorylation , Protein Structure, Quaternary , Protein Structure, Secondary , Software
14.
Eur J Pharmacol ; 163(1): 199-203, 1989 Apr 12.
Article in English | MEDLINE | ID: mdl-2663529

ABSTRACT

Administration of endothelin (0.03-3.0 micrograms/kg i.v.) caused transient depressor responses followed by sustained pressor responses in anesthetized spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto rats (WKY). The initial depressor response occurred at lower doses (0.1 versus 0.3 micrograms/kg i.v.) in SHR versus WKY. The secondary pressor response was attenuated in SHR compared to WKY in both the threshold dose (3.0 versus 0.1 microgram/kg i.v.) and maximum effect at high doses (52 versus 91% at 3.0 micrograms/kg i.v.). In conscious SHR and WKY, endothelin elicited comparable initial depressor responses with increases in heart rate; the secondary pressor responses were attenuated compared to those in anesthetized rats. Therefore endothelin elicits a prominent depressor response, which may be associated with afterload reduction, in SHR.


Subject(s)
Blood Pressure/drug effects , Peptides/pharmacology , Animals , Endothelins , Heart Rate/drug effects , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY
16.
Horm Metab Res ; 13(12): 675-8, 1981 Dec.
Article in English | MEDLINE | ID: mdl-6119282

ABSTRACT

The method of administration of [D-Ala5,D-Trp8] somatostatin is of central importance in determining the degree and duration of suppression of insulin and glucagon release. The analog decreased insulin levels in rats when injected by s.c. or i.v. routes, with a nadir 15 minutes following injection. After i.v. injection, insulin levels rapidly returned to basal values while s.c. injection produced significant suppression for 60 minutes. Neither type of injection altered glucagon levels. Intravenous infusion resulted in inhibition of both insulin and glucagon release, with rebound hyperglucagonemia, but not hyperinsulinemia in the post-infusion period. Plasma glucose levels reflected these hormonal changes. Thus, dramatic alterations in the specificity of this somatostatin analog may be achieved by employing different methods of administration.


Subject(s)
Glucagon/blood , Insulin/blood , Somatostatin/analogs & derivatives , Animals , Blood Glucose/metabolism , Infusions, Parenteral , Injections, Intravenous , Injections, Subcutaneous , Kinetics , Male , Rats , Somatostatin/administration & dosage
17.
Biochemistry ; 40(19): 5633-41, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11341828

ABSTRACT

The gp41 subunit of the human immunodeficiency virus type 1 envelope glycoprotein mediates fusion of the cellular and viral membranes. The gp41 ectodomain is a trimer of alpha-helical hairpins, where N-terminal helices form a parallel three-stranded coiled-coil core and C-terminal helices pack around the core. A deep hydrophobic pocket on the N-terminal core represents an attractive target for antiviral therapeutics. We have employed a soluble derivative of the gp41 core ectodomain and small cyclic disulfide D-peptide inhibitors to define the stoichiometry, affinity, and thermodynamics of ligand binding to this pocket using isothermal titration calorimetry. These inhibitors bind with micromolar affinity to the pocket with the expected stoichiometry of three peptides per gp41 core trimer. There are no cooperative interactions among the three binding sites. Linear eight- or nine-residue D-peptides derived from the pocket-binding domain of the cyclic molecules also bind specifically. A negative heat capacity change is observed and is consistent with burial of hydrophobic surface upon binding. Contrary to expectations for a reaction dominated by the classical hydrophobic effect, peptide binding is enthalpically driven and is opposed by an unfavorable negative entropy change. The calorimetry data support models whereby dominant negative inhibitors bind to a transiently exposed surface on the prefusion intermediate state of gp41 and disrupt subsequent resolution to the fusion-active six-stranded hairpin conformation.


Subject(s)
Anti-HIV Agents/metabolism , HIV Envelope Protein gp41/metabolism , HIV-1/metabolism , Peptides, Cyclic/metabolism , Amino Acid Sequence , Anti-HIV Agents/chemical synthesis , Binding, Competitive , Calorimetry , Hot Temperature , Humans , Ligands , Molecular Sequence Data , Peptides, Cyclic/chemical synthesis , Protein Binding , Protein Conformation , Solutions , Thermodynamics , Ultracentrifugation
18.
J Biol Chem ; 271(50): 31957-63, 1996 Dec 13.
Article in English | MEDLINE | ID: mdl-8943242

ABSTRACT

Site-specific substitutions of as few as four amino acids (M46I/L63P/V82T/I84V) of the human immunodeficiency virus type 1 (HIV-1) protease engenders cross-resistance to a panel of protease inhibitors that are either in clinical trials or have recently been approved for HIV therapy (Condra, J. H., Schleif, W. A., Blahy, O. M. , Gadryelski, L. J., Graham, D. J., Quintero, J. C., Rhodes, A., Robbins, H. L., Roth, E., Shivaprakash, M., Titus, D., Yang, T., Teppler, H., Squires, K. E., Deutsch, P. J., and Emini, E. A. (1995) Nature 374, 569-571). These four substitutions are among the prominent mutations found in primary HIV isolates obtained from patients undergoing therapy with several protease inhibitors. Two of these mutations (V82T/I84V) are located in, while the other two (M46I/L63P) are away from, the binding cleft of the enzyme. The functional role of these mutations has now been delineated in terms of their influence on the binding affinity and catalytic efficiency of the protease. We have found that the double substitutions of M46I and L63P do not affect binding but instead endow the enzyme with a catalytic efficiency significantly exceeding (110-360%) that of the wild-type enzyme. In contrast, the double substitutions of V82T and I84V are detrimental to the ability of the protease to bind and, thereby, to catalyze. When combined, the four amino acid replacements institute in the protease resistance against inhibitors and a significantly higher catalytic activity than one containing only mutations in its active site. The results suggest that in raising drug resistance, these four site-specific mutations of the protease are compensatory in function; those in the active site diminish equilibrium binding (by increasing Ki), and those away from the active site enhance catalysis (by increasing kcat/KM). This conclusion is further supported by energy estimates in that the Gibbs free energies of binding and catalysis for the quadruple mutant are quantitatively dictated by those of the double mutants.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV Protease/genetics , Carbamates , Clinical Trials as Topic , Furans , Fusion Proteins, gag-pol/metabolism , HIV Protease/chemistry , Humans , Hydrolysis , Indinavir/pharmacology , Kinetics , Mutagenesis , Ritonavir/pharmacology , Sulfonamides/pharmacology
19.
Biochem Biophys Res Commun ; 73(4): 911-6, 1976 Dec 20.
Article in English | MEDLINE | ID: mdl-15625860

ABSTRACT

A nonreducible cyclic analog of somatostatin (SRIF) was prepared by a combination of solid phase and solution peptide synthesis. The compound, gamma-Abu-Lys-Asn-Phe-Phe-Trp-Lys-Thr-Phe-Thr-Ser-Asp-OH, was tested for its effect on the release of growth hormone, glucagon and insulin in rats. It significantly suppressed pentobarbital-stimulated growth hormone release but showed no effect on arginine-stimulated glucagon or insulin release. The linear form, NH2-gamma-Abu-Lys-Asn-Phe-Phe-Trp-Lys-Thr-Phe-Thr-Ser-Asp-OH, was also prepared and tested in vivo. It was shown to have only slight activity.


Subject(s)
Growth Hormone/metabolism , Somatostatin/chemistry , Somatostatin/pharmacology , Amino Acid Sequence , Animals , Glucagon/blood , Glucagon/metabolism , Growth Hormone/blood , Insulin/blood , Insulin/metabolism , Insulin Secretion , Male , Molecular Sequence Data , Rats , Somatostatin/analogs & derivatives , Somatostatin/chemical synthesis
20.
Pharmacology ; 42(6): 340-8, 1991.
Article in English | MEDLINE | ID: mdl-1946598

ABSTRACT

We evaluated the effect of the RGD-containing peptide, echistatin, on thrombolysis time and acute reocclusion in a canine model of coronary thrombosis/thrombolysis. Occlusive thrombus formation was induced by electrical injury, via a stimulating electrode, to the endothelial surface of the circumflex coronary artery in the open-chest, anesthetized dog in the presence of a critical stenosis. Fifteen minutes after occlusive thrombus formation, dogs received either an intravenous infusion of vehicle (saline at 0.1 ml/min) or echistatin (15 micrograms/kg/min i.v.). Heparin was given as an initial bolus (100 U/kg i.v.) 15 min after thrombus formation and repeated at hourly intervals (50 U/kg). This dose of heparin increased activated partial thromboplastin time to 1.5- to 2.5- fold over control. Thrombolysis was induced with recombinant tissue-type plasminogen activator (tPA) at a total dose of 1 mg/kg, intravenously administered over 90 min with 10% given as an initial bolus. The vehicle-treated animals reperfused at 48 +/- 9 min with a reperfusion incidence of 60% (3/5). The echistatin-treated animals reperfused at 46 +/- 5 min with a reperfusion incidence of 100% (5/5). After stopping the tPA infusion, acute reocclusion occurred in 100% (3/3) of the vehicle-treated dogs and in only 20% (1/5) of the echistatin-treated dogs. Echistatin caused a greater than 5-fold increase in buccal mucosa bleeding time and almost completely inhibited ex vivo platelet aggregation to ADP, collagen, and U-46619. Residual thrombus wet weight, determined at the end of the experiment, was significantly lower for the echistatin group (2.1 +/- 0.2 mg) compared to the vehicle group (5.8 +/- 0.7 mg).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Coronary Thrombosis/prevention & control , Peptides , Platelet Aggregation Inhibitors/therapeutic use , Thrombolytic Therapy , Tissue Plasminogen Activator/therapeutic use , Viper Venoms/therapeutic use , Animals , Coronary Thrombosis/drug therapy , Disease Models, Animal , Dogs , Female , Fibrinolysis/drug effects , Hemodynamics , Intercellular Signaling Peptides and Proteins , Male , Perfusion , Platelet Aggregation/drug effects , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL