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










Publication year range
1.
Reprod Domest Anim ; 47 Suppl 4: 233-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22827376

ABSTRACT

Surgical sterilization is the mainstay of dog and cat population control, but its use is still often limited by the costs and effort involved, especially in developing countries. An ideal non-surgical sterilant that is safe, effective, permanent, administered as a single injection and capable of being manufactured inexpensively could have a significant impact on the world-wide dog and cat overpopulation problem. One approach towards developing such an agent is the targeting of pituitary gonadotrophic cells with cytotoxic agents using gonadotropin-releasing hormone (GnRH). GnRH is a peptide that binds to high-affinity receptors selectively expressed on gonadotrophs and some types of cancers. Both small molecules and proteins have been conjugated to GnRH analogues to generate targeted cytotoxic and imaging agents. Although most of these efforts have focused on development of human cancer therapeutics, available reproductive studies in rats and dogs suggest that current compounds do not have sufficient therapeutic windows for complete gonadotroph ablation, in part owing to poor stability of peptide targeting sequences. The only reported longer-term study of gonadotroph ablation in dogs reported suppression of serum testosterone for 8 months, but endocrine function then recovered, raising important questions about the mechanism of reproductive suppression and its recovery. Although studies to date suggest that this is a potentially attractive approach to non-surgical sterilization, ideal agents are yet to be developed, and important mechanistic questions remain to be answered.


Subject(s)
Contraceptive Agents/pharmacology , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Sterilization, Reproductive/veterinary , Animals , Contraceptive Agents/adverse effects , Female , Gonadotropin-Releasing Hormone/metabolism , Male , Receptors, LHRH/metabolism , Sterilization, Reproductive/methods
2.
Biochemistry ; 43(12): 3670-8, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15035637

ABSTRACT

CC chemokine receptor 7 (CCR-7) is expressed on mature dendritic cells and T-cells. Its ligands, CCL-19 (MIP-3beta) and CCL-21 (SLC), play an important role in the migration of these cells to secondary lymphoid organs where they are predominantly expressed. For most chemokines, the N-terminal domain preceding the first two conserved cysteines is involved in stabilizing the active conformation of its cognate receptors. We have chemically synthesized N-terminal analogues of CCL-19 with the aid of a native chemical ligation method to investigate structure function requirements of this ligand domain by performing ligand binding, GTP-gammaS binding, and chemotaxis assays. Successive truncations of the N-terminus of CCL-19 reduced the affinity of the receptor for the ligand in a size-dependent manner. Furthermore, Ala substitutions of Asn(3), Asp(4), and Asp(7) show that the side chains of these residues are important for high-affinity binding of CCL-19 to CCR-7. The effects observed were mirrored in both GTP-gammaS binding and chemotaxis assays, highlighting the functional importance of this ligand domain. We also describe two partial agonists of CCR-7 ([Nle(72)]CCL-19(6-77) and Ac-[Nle(72)]CCL-19(7-77)), and identify the first analogue of CCL-19 (Ac-[Nle(72)]CCL-19(8-77)) that acts as a functional antagonist in vitro (K(B) approximately 350 nM for GTP-gammaS binding assays). As mutations of both Glu(6) and Asp(7) to Ala did not dissociate effects on ligand binding from receptor activation, it is likely that the backbone of these two residues is crucial for agonist activity.


Subject(s)
Chemokines, CC/chemistry , Peptide Fragments/chemistry , Receptors, Chemokine/metabolism , Acetylation , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , CHO Cells , Cell Line, Tumor , Cell Migration Inhibition , Chemokine CCL19 , Chemokine CCL21 , Chemokines, CC/genetics , Cricetinae , Humans , Ligands , Methionine/genetics , Molecular Sequence Data , Norleucine/genetics , Peptide Fragments/genetics , Protein Binding/genetics , Receptors, CCR7 , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/chemistry , Receptors, G-Protein-Coupled/metabolism , Sequence Deletion
3.
J Med Chem ; 43(5): 784-96, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10715147

ABSTRACT

With the ultimate goal of identifying a consensus bioactive conformation of GnRH antagonists, the compatibility of a number of side chain to side chain bridges in bioactive analogues was systematically explored. In an earlier publication, cyclo[Asp(4)-Dpr(10)]GnRH antagonists with high potencies in vitro and in vivo had been identified.(1) Independently from Dutta et al. (2) and based on structural considerations, the cyclic [Glu(5)-Lys(8)] constraint was also found to be tolerated in GnRH antagonists. We describe here a large number of cyclic (4-10) and (5-8) and dicyclic (4-10/5-8) GnRH antagonists optimized for affinity to the rat GnRH receptor and in vivo antiovulatory potency. The most potent monocyclic analogues were cyclo(4-10)[Ac-DNal(1), DFpa(2),DTrp(3),Asp(4),DArg(6),Xaa(10)]GnRH with Xaa = D/LAgl (1, K(i) = 1.3 nM) or Dpr (2, K(i) = 0.36 nM), which completely blocked ovulation in cycling rats after sc administration of 2.5 microgram at noon of proestrus. Much less potent were the closely related analogues with Xaa = Dbu (3, K(i) = 10 nM) or cyclo(4-10)[Ac-DNal(1), DFpa(2),DTrp(3),Glu(4),DArg(6),D/LAgl(10)]GnRH (4, K(i) = 1.3 nM). Cyclo(5-8)[Ac-DNal(1),DCpa(2),DTrp(3),Glu(5),DArg++ +(6),Lys(8), DAla(10)]GnRH (13), although at least 20 times less potent in the AOA than 1 or 2 with similar GnRHR affinity (K(i) = 0.84 nM), was found to be one of the most potent in a series of closely related cyclo(5-8) analogues with different bridge lengths and bridgehead chirality. The very high affinity of cyclo(5,5'-8)[Ac-DNal(1), DCpa(2),DPal(3),Glu(5)(betaAla),DArg(6),(D or L)Agl,(8)DAla(10)]GnRH 14 (K(i) = 0.15 nM) correlates well with its high potency in vivo (full inhibition of ovulation at 25 microgram/rat). Dicyclo(4-10/5-8)[Ac-DNal(1),DCpa(2),DTrp(3),Asp (4),Glu(5),DArg(6), Lys(8),Dpr(10)]GnRH (24, K(i) = 0.32 nM) is one-fourth as potent as 1 or 2, in the AOA; this suggests that the introduction of the (4-10) bridge in 13, while having little effect on affinity, restores functional/conformational features favorable for stability and distribution. To further increase potency of dicyclic antagonists, the size and composition of the (5-8) bridge was varied. For example, the substitution of Xbb(5') by Gly (30, K(i) = 0.16 nM), Sar (31, K(i) = 0.20 nM), Phe (32, K(i) = 0.23 nM), DPhe (33, K(i) = 120 nM), Arg (36, K(i) = 0.20 nM), Nal (37, K(i) = 4.2 nM), His (38, K(i) = 0.10 nM), and Cpa (39, K(i) = 0.23 nM) in cyclo(4-10/5,5'-8)[Ac-DNal(1),DCpa(2),DPal(3),Asp(4),G lu(5)(Xbb(5')), DArg(6),Dbu,(8)Dpr(10)]GnRH yielded several very high affinity analogues that were 10, ca. 10, 4, >200, 1, ca. 4, >2, and 2 times less potent than 1 or 2, respectively. Other scaffolds constrained by disulfide (7, K(i) = 2.4 nM; and 8, K(i) = 450 nM), cyclo[Glu(5)-Aph(8)] (16, K(i) = 20 nM; and 17, K(i) = 0.28 nM), or cyclo[Asp(5)-/Glu(5)-/Asp(5)(Gly(5'))-Amp(8)] (19, K(i) = 1.3 nM; 22, K(i) = 3.3 nM; and 23, K(i) = 3.6 nM) bridges yielded analogues that were less potent in vivo and had a wide range of affinities. The effects on biological activity of substituting DCpa or DFpa at position 2, DPal or DTrp at position 3, and DArg, DNal, or DCit at position 6 are also discussed. Interestingly, monocyclo(5-8)[Glu(5), DNal(6),Lys(8)]GnRH (18, K(i) = 1. (ABSTRACT TRUNCATED)


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/chemical synthesis , Peptides, Cyclic/chemical synthesis , Animals , Cell Line , Chromatography, High Pressure Liquid , Female , Gonadotropin-Releasing Hormone/metabolism , Hormone Antagonists/chemistry , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Ovulation/drug effects , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Rats , Structure-Activity Relationship
4.
J Med Chem ; 43(5): 797-806, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10715148

ABSTRACT

Careful analysis of the NMR structures of cyclo(4-10)[Ac-Delta(3)Pro(1),DFpa(2),DTrp(3),Asp(4),DNal (6), Dpr(10)]GnRH, dicyclo(4-10/5-8)[Ac-DNal(1),DCpa(2),DTrp(3), Asp(4), Glu(5),DArg(6),Lys(8),Dpr(10)]GnRH, and dicyclo(4-10/5, 5'-8)[Ac-DNal(1),DCpa(2),DPal(3),Asp(4), Glu(5)(Gly),DArg(6),Dbu(8), Dpr(10)]GnRH showed that, in the N-terminal tripeptide, a type II beta-turn around residues 1 and 2 was probable along with a gamma-turn around DTrp(3)/DPal(3). This suggested the possibility of constraining the N-terminus by the introduction of a cyclo(1-3) scaffold. Optimization of ring size and composition led to the discovery of cyclo(1-3)[Ac-DAsp(1),DCpa(2),DLys(3),DNal(6), DAla(10)]GnRH (5, K(i) = 0.82 nM), cyclo(1,1'-3)[Ac-DAsp(1)(Gly), DCpa(2),DOrn(3),DNal(6),DAla(10)]GnRH (13, K(i) = 0.34 nM), cyclo(1, 1'-3)[Ac-DAsp(1)(Gly),DCpa(2),DLys(3),DNal(6),DA la(10)]GnRH (20, K(i) = 0.14 nM), and cyclo(1,1'-3)[Ac-DAsp(1)(betaAla), DCpa(2), DOrn(3),DNal(6),DAla(10)]GnRH (21, K(i) = 0.17 nM), which inhibited ovulation significantly at doses equal to or lower than 25 microgram/rat. These results were particularly unexpected in view of the critical role(s) originally ascribed to the side chains of residues 1 and 3.(1) Other closely related analogues, such as those where the [DAsp(1)(betaAla), DOrn(3)] cycle of 21 was changed to [DOrn(1)(betaAla), DAsp(3)] of cyclo(1,1'-3)[Ac-DOrn(1)(betaAla), DCpa(2),DAsp(3),DNal(6),DAla(10)]GnRH (22, K(i) = 2.2 nM) or where the size of the cycle was conserved and [DAsp(1)(betaAla), DOrn(3)] was replaced by [DGlu(1)(Gly), DOrn(3)] as in cyclo(1, 1'-3)[Ac-DGlu(1)(Gly),DCpa(2),DOrn(3),DNal(6),DA la(10)]GnRH (23, K(i) = 4.2 nM), were approximately 100 and 25 times less potent in vivo, respectively. Analogues with ring sizes of 18 ¿cyclo(1, 1'-3)[Ac-DGlu(1)(Gly),DCpa(2),DLys(3),DNal(6),DA la(10)]GnRH (24)¿ and 19 ¿cyclo(1,1'-3)[Ac-DGlu(1)(betaAla),DCpa(2),DLys( 3),DNal(6), DAla(10)]GnRH (25)¿ atoms were also less potent than 21 with slightly higher K(i) values (1.5 and 2.2 nM, respectively). These results suggested that the N-terminal tripeptide was likely to assume a folded conformation favoring the close proximity of the side chains of residues 1 and 3. The dicyclic analogue dicyclo(1-3/4-10)[Ac-DAsp(1),DCpa(2),DLys(3),Asp (4),DNal(6), Dpr(10)]GnRH (26) was fully active at 500 microgram, with a K(i) value of 1 nM. The in vivo potency of 26 was at least 10-fold less than that of monocyclic cyclo(1-3)[Ac-DAsp(1),DCpa(2),DLys(3),DNal(6), DAla(10)]GnRH (5); this suggested the existence of unfavorable interactions between the now optimized and constrained (1-3) and (4-10) cyclic moieties that must interact as originally hypothesized. Tricyclo(1-3/4-10/5-8)[Ac-DGlu(1),DCpa(2), DLys(3),Asp(4),Glu(5), DNal(6),Lys(8),Dpr(10)] GnRH (27) was inactive at 500 microgram/rat with a corresponding low affinity (K(i) = 4.6 nM) when compared to those of the most potent analogues (K(i) < 0.5 nM).


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/chemical synthesis , Peptides, Cyclic/chemical synthesis , Animals , Cell Line , Chromatography, High Pressure Liquid , Electrophoresis, Capillary , Female , Gonadotropin-Releasing Hormone/metabolism , Hormone Antagonists/chemistry , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Ovulation/drug effects , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Rats , Structure-Activity Relationship
5.
J Med Chem ; 43(5): 807-18, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10715149

ABSTRACT

In three earlier papers, the structures and biological potencies of numerous mono- and dicyclic antagonists of GnRH were reported. Among these, two families, each containing two to four members were identified that had very high antagonist potencies in an antiovulatory assay (within a factor of 2 of those of the most potent linear analogues) and high affinities (K(i) < 0.5 nM) for the rat GnRH receptor (rGnRHR). The most favored cycles bridged the side chains of residues (4-10),(1,2) (5-8),(2) (4-10/5-8),(2) (1-3),(3) and (1-3/4-10).(3) Our goal was to identify a consensus model of bioactive conformations of GnRH antagonists, yet these biocompatible constraints did not sufficiently restrain the spatial location of the N-terminal tripeptide with respect to the C-terminal heptapeptide, due largely to the rotational freedom about the bonds connecting these regions. Examination of models derived from NMR studies of cyclo(4-10) analogues suggested a large number of possible cyclic constraints such as cyclo (0-8), (1-8), or (2-8). All analogues tested with these substitutions were inactive as antiovulatory agents at 1 mg/rat (5-9) and had low affinity for rGnRHR. On the other hand, bridging positions 3 and 8 with a [DAsp(3)] to [Dbu(8)] (12, K(i) = 13 nM) or [Orn(8)] (13, K(i) = 14 nM) in the parent compound cyclo(3-8)[Ac-DNal(1),DCpa(2),DXaa(3), Arg(5),DNal(6),Xbb(8),DAla(10)]GnRH yielded analogues that blocked ovulation at 250 microgram/rat. Analogue 14 (K(i) = 2.3 nM), with a [DAsp(3), Lys(8)] bridge, was fully active at 50 microgram/rat. Loss of potency (>20-fold) was observed with the substitution of [DAsp(3)] in 14 by [DGlu(3)] in 15 (K(i) = 23 nM). Dicyclic analogues possessing the (4-10) cycle and selected (1-6), (2-6), and (2-8) cycles led to analogues that were inactive at doses of 500 microgram/rat or larger. Two analogues with (1-8/4-10) cycles (16, K(i) = 1.1 nM) or (3-8/4-10) cycles (22, K(i) = 17 nM) showed full antiovulatory potency at 250 microgram/rat. None of these substitutions yielded analogues potent enough (>80% inhibition of ovulation at 5 microgram/rat or less and K(i) < 0.5 nM) to be candidates for structural analysis by NMR. On the other hand, four dicyclic (1, 1'-5/4-10) analogues met this criterion: dicyclo(1, 1'-5/4-10)[Ac-Asp(1)(Gly),DCpa(2),DTrp(3),Asp(4),Dbu(5 ), DNal(6), Dpr(10)]GnRH (32, K(i) = 0.22 nM), dicyclo(1, 1'-5/4-10)[Ac-Asp(1)(Gly),DCpa(2),DNal(3),Asp(4),Dbu(5 ), DNal(6), Dpr(10)]GnRH (34, K(i) = 0.38 nM), dicyclo(1, 1'-5/4-10)[Ac-Asp(1)(betaAla),DCpa(2), DTrp(3),Asp(4),Dbu(5),DNal(6), Dpr(10)]GnRH (40, K(i) = 0.15 nM), and dicyclo(1, 1'-5/4-10)[Ac-Glu(1)(Gly), DCpa(2),DTrp(3),Asp(4),Dbu(5),DNal(6), Dpr(10)]GnRH (41, K(i) = 0.24 nM). Since they differed slightly in terms of the (1,1'-5) bridge length (21 and 22 atoms) and bridgehead configuration, we may hypothesize that they assume similar bioactive conformations that satisfy a very discriminating receptor, since many other closely related analogues were significantly less potent.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/chemical synthesis , Peptides, Cyclic/chemical synthesis , Animals , Cell Line , Chromatography, High Pressure Liquid , Electrophoresis, Capillary , Female , Gonadotropin-Releasing Hormone/metabolism , Hormone Antagonists/chemistry , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Ovulation/drug effects , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Rats , Structure-Activity Relationship
6.
J Med Chem ; 43(5): 819-28, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10715150

ABSTRACT

Little is known of the conformation of peptide hormones as they interact with their receptors for a number of reasons: peptide hormones are notoriously flexible in solution, their receptors are particularly complex, and there is strong evidence that receptor-ligand interaction leading to activation is a dynamic process. Insights into the active conformation of the decapeptide gonadotropin releasing hormone (GnRH) have been obtained previously from the solution structures of four constrained GnRH antagonists ¿cyclo(1-10)[Ac-Delta(3)-Pro(1),DCpa(2),DTrp(3,6),NMeLeu+ ++(7), betaAla(10)]GnRH (1), cyclo(4-10)[Ac-Delta(3)Pro(1),DFpa(2),DTrp(3), Asp(4),DNal(6),Dpr(10)]GnRH (2), dicyclo(4-10/5-8)[Ac-DNal(1), DCpa(2),DTrp(3),Asp(4),Glu(5),DArg(6),Lys(8),Dpr (10)]GnRH (3), and dicyclo(4-10/5-5'-8)[Ac-DNal(1),DCpa(2),DPal(3), Asp(4),Glu(5)(Gly), DArg(6),Dbu(8),Dpr(10)]GnRH (4)¿. However, the precise location of the N-terminal tripeptide in the highly potent (K(i) < 0.4 nM) 2-4 remained unclear due to the lack of constraints in this region. The NMR structure of the newly discovered and potent (K(i) = 0.24 nM) dicyclo(1-1'-5/4-10)[Ac-Glu(1)(Gly),DCpa(2),DTrp(3),As p(4),Dbu(5), DNal(6),Dpr(10)]GnRH (5) now allows the definition of the conformation of this region. A combined computational analysis (consensus forcing) of compounds 2-5, designed to explore the common conformations available to them that are simultaneously consistent with the NMR data corresponding to each compound, leads to a consensus structural model for the GnRH pharmacophore. This model shares some common features with the structure of the nonpeptidic GnRH mimetic T-98475. In the course of that comparative study, two additional contact points to those proposed by the authors are identified, suggesting that this model has predictive value.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/chemistry , Peptides, Cyclic/chemistry , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , In Vitro Techniques , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Ovulation/drug effects , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Pituitary Gland/cytology , Pituitary Gland/metabolism , Pyridones/chemistry , Pyridones/metabolism , Rats , Solutions , Thiophenes/chemistry , Thiophenes/metabolism
7.
Br Med Bull ; 56(3): 761-72, 2000.
Article in English | MEDLINE | ID: mdl-11255560

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is a decapeptide (pGlu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly.NH2) which is produced from a precursor polypeptide in hypothalamic neurons and secreted in a pulsatile fashion to stimulate the secretion of LH and FSH via its interaction with a cognate receptor on gonadotropes. Low doses of the native peptide delivered in a pulsatile manner to mimic that found in the hypothalamic portal vessels restore fertility in hypogonadal patients, and are also effective in treating cryptorchidism and delayed puberty. Administration of high doses of GnRH, or agonist analogues, causes desensitization of the gonadotrope with consequent decline in gonadal gametogenesis and steroid and peptide hormone synthesis. This phenomenon finds extensive therapeutic application in clinical medicine in a wide spectrum of disease (Table 1). In addition, GnRH analogues have promise as new generation male and female contraceptives in conjunction with steroid hormone replacement. GnRH antagonists inhibit the reproductive system through competition with endogenous GnRH for the receptor and, in view of their rapid effects, are being increasingly used for the above mentioned applications. The peptide agonists and antagonists currently available require parenteral administration, typically in the form of long-acting depots. A new generation of non-peptide GnRH antagonists are beginning to emerge which should allow oral administration and, therefore, may provide greater flexibility of dosing, lower costs and increased patient acceptance.


Subject(s)
Contraceptive Agents/pharmacology , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Administration, Oral , Animals , Contraceptive Agents/metabolism , Female , Forecasting , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/metabolism , Humans , Macaca fascicularis , Models, Animal , Protein Binding , Rats , Receptors, Gonadotropin/metabolism , Structure-Activity Relationship
8.
Proc Natl Acad Sci U S A ; 89(23): 11451-5, 1992 Dec 01.
Article in English | MEDLINE | ID: mdl-1454833

ABSTRACT

Activin A is a potent growth and differentiation factor related to transforming growth factor beta. In somatotrophs, activin suppresses the biosynthesis and secretion of growth hormone (GH) and cellular proliferation. We report here that, in MtTW15 somatotrophic tumor cells, activin decreased GH mRNA levels and inhibited expression of transfected GH promoter--chloramphenicol acetyltransferase fusion genes. Deletion mapping of nucleotide sequences mediating this inhibition led to the identification of a region that has previously been characterized as binding the pituitary-specific transcription factor Pit-1/GHF-1. Characterization of nuclear factor binding to this region demonstrated that binding of Pit-1 to the GH promoter is lost on activin treatment. These results indicate that activin-induced repression of GH biosynthesis is mediated by the loss of tissue-specific transcription factor binding to the GH promoter and suggest a possible general mechanism for other activin responses, whereby activin regulates the function of other POU- or homeodomain-containing transcription factors.


Subject(s)
DNA-Binding Proteins/metabolism , Growth Hormone/genetics , Inhibins/pharmacology , Promoter Regions, Genetic , Transcription Factors/metabolism , Activins , Animals , Female , Gene Expression Regulation/drug effects , Growth Hormone/biosynthesis , In Vitro Techniques , Nuclear Proteins/metabolism , Protein Binding , RNA, Messenger/genetics , Rats , Rats, Inbred WF , Recombinant Proteins/pharmacology , Transcription Factor Pit-1 , Tumor Cells, Cultured
9.
Nature ; 350(6319): 622-4, 1991 Apr 18.
Article in English | MEDLINE | ID: mdl-1826763

ABSTRACT

Most of the transcriptional effects of cyclic AMP are mediated by the cAMP response element binding protein (CREB). After activation of cAMP-dependent protein kinase A, the catalytic subunits of this enzyme apparently mediate the phosphorylation and activation of CREB. As cAMP serves as a mitogenic signal for anterior pituitary somatotrophic cells, we investigated whether CREB similarly regulates proliferation of these cells. We prepared transgenic mice expressing a transcriptionally inactive mutant of CREB (CREBM1), which cannot be phosphorylated, in cells of the anterior pituitary. If CREB activity is required for proliferation, the overexpressed mutant protein would effectively compete with wild-type CREB activity and thereby block the response to cAMP. As predicted, the CREBM1 transgenic mice exhibited a dwarf phenotype with atrophied pituitary glands markedly deficient in somatotroph but not other cell types. We conclude that transcriptional activation of CREB is necessary for the normal development of a highly restricted cell type, and that environmental cues, possibly provided by the hypothalamic growth hormone-releasing factor, are necessary for population of the pituitary by somatotrophic cells.


Subject(s)
DNA-Binding Proteins/genetics , Dwarfism/genetics , Growth Hormone/genetics , Pituitary Gland, Anterior/pathology , Amino Acid Sequence , Animals , Base Sequence , Cell Nucleus/metabolism , Cloning, Molecular , Crosses, Genetic , Cyclic AMP Response Element-Binding Protein , Female , Gene Expression , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Pedigree , Pituitary Gland, Anterior/metabolism , Promoter Regions, Genetic
10.
Proteins ; 9(1): 1-11, 1991.
Article in English | MEDLINE | ID: mdl-2017431

ABSTRACT

Alzheimer's disease is characterized by the deposition of amyloid beta-protein as plaques and tangles in the brains of its victims. The amyloid precursor can be expressed with or without the inclusion of a protease inhibitor domain, the potential role of which in amyloidogenesis has prompted the generation of a model of its three-dimensional structure based on the known structure of a related inhibitor. The model structure predicts that the mutated residues are almost entirely on the surface of the inhibitor domain, while conserved residues constitute the hydrophobic core. In addition, several pairs of structurally complementary, or concerted, mutations are seen. These structural features provide strong evidence for the validity of the modeled structure, and it is suggested that the presence of complementary mutations may be used as a criterion for evaluating protein structures built by homology, in addition to the (spatial) location of the mutations. The terminal residues delimiting the domain are among those furthest from the protease binding site and are in close proximity to one another, thus suggesting the ability of the domain to function as a structural cassette within the context of a larger protein. The electrostatic potentials of the inhibitor and of the related bovine pancreatic trypsin inhibitor reveal how two inhibitors with very different net charges can bind with approximately the same binding constant to trypsin and suggest a mutation of trypsin that might selectively enhance the binding of the amyloid inhibitor domain. The model provides a structural basis for understanding the functional roles of residues in the domain and for designing simpler molecules to test as pharmacologic agents for intervention in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/genetics , Trypsin Inhibitors/chemistry , Amino Acid Sequence , Amyloid beta-Peptides/biosynthesis , Binding Sites , Drug Design , Electronic Data Processing , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Reproducibility of Results , Stereoisomerism , Trypsin Inhibitors/pharmacology
11.
Proteins ; 8(4): 295-304, 1990.
Article in English | MEDLINE | ID: mdl-2091022

ABSTRACT

The introduction of conformational constraints into a flexible peptide hormone can be exploited to develop models for the conformation required for receptor binding and activity. In this review, we illustrate this approach to analog design using our work on antagonists of gonadotropin-releasing hormone (GnRH). Design of a conformationally constrained, competitive antagonist of GnRH, cyclo[delta 3,4 Pro-D4ClPhe-DTrp-Ser-Tyr-DTrp-NMeLeu-Arg-Pro-bet a Ala] led to the prediction of its bioactive conformation. Template forcing experiments show that this conformation is accessible to other active GnRH analogs. Two-dimensional NMR studies verified the predicted conformation in solution. The predicted binding conformation has recently been used to design two new analogs incorporating side chain-side chain linkages suggested by the conformational model: Ac-delta 3,4Pro-D4FPhe-DTrp-Dap-Tyr-DTrp-Leu-Arg-Asp-Gly- NH2 and Ac-delta 3,4Pro-D4FPhe-DTrp-Dap-Tyr-D2Nal-Leu-Arg-Pro-Asp -NH2. These analogs were synthesized and the one predicted to be most similar to the parent conformation had equivalent potency while the second, designed to refine the conformational hypothesis, was found to exhibit enhanced potency, thus confirming the original binding conformation hypothesis. These compounds and their derivatives now provide a new class of GnRH antagonists possessing both high biological potency and limited conformational flexibility, thus making them ideal for both biophysical and structure-activity studies.


Subject(s)
Gonadotropin-Releasing Hormone/chemistry , Peptides, Cyclic/chemistry , Amino Acid Sequence , Animals , Cells, Cultured , Computer Graphics , Computer Simulation , Drug Design , Gonadotropin-Releasing Hormone/pharmacology , Hormones/chemistry , In Vitro Techniques , Luteinizing Hormone/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Secretory Rate/drug effects , Structure-Activity Relationship
12.
Endocrinology ; 124(1): 24-9, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2535806

ABSTRACT

The specific binding of a GRF radioligand, [His1,125I-Tyr10,Nle27]hGRF-1-32NH2, to rat pituitary homogenates is reduced by the addition of GTP and its nonhydrolyzable analogs 5'-guanylylimidodiphosphate (GppNHp) and guanosine 5'-O-(3-thiotriphosphate) (GTP-gamma-S). GDP and cAMP had no effect while the nonhydrolyzable ATP analogs 5'-adenylylimidodiphosphate and adenosine 5'-O-(3-thiotriphosphate) did elicit a significant reduction in GRF binding. The effect of GppNHp was half-maximal at 0.2 microM, and the maximum inhibition achieved was 85%. The effect of 0.1 microM GppNHp on GRF competitive displacement experiments indicated a significant reduction in affinity for the ligand (Kd = 0.51 +/- 0.11 nM in the absence of GppNHp and 2.1 +/- 1.1 nM in its presence) without an effect on receptor number. The GRF radioligand dissociates slowly from its receptor (t1/2 = 250 +/- 50 min), but the addition of 0.1 microM GppNHp converts approximately half of the receptors present to a more rapidly dissociating form (t1/2 = 9 +/- 10 min). These results are consistent with existing models for receptor-G-protein interactions, and thus, we conclude that transduction of the GRF response across the cell membrane involves a guanine nucleotide-binding protein, presumably Gs.


Subject(s)
Growth Hormone-Releasing Hormone/metabolism , Nucleotides/pharmacology , Pituitary Gland/metabolism , Receptors, Neuropeptide , Receptors, Neurotransmitter/metabolism , Receptors, Pituitary Hormone-Regulating Hormone , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Animals , Cyclic AMP/pharmacology , Growth Hormone-Releasing Hormone/analogs & derivatives , Guanosine 5'-O-(3-Thiotriphosphate) , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/pharmacology , Guanylyl Imidodiphosphate/pharmacology , Kinetics , Male , Pituitary Gland/drug effects , Rats , Rats, Inbred Strains , Receptors, Neurotransmitter/drug effects , Thionucleotides/pharmacology
13.
Mol Endocrinol ; 2(2): 117-24, 1988 Feb.
Article in English | MEDLINE | ID: mdl-2840569

ABSTRACT

We have used a smooth muscle cell line isolated from rabbit carotid artery (RCA) as a model system with which to study the expression of atrial natriuretic peptide (ANP) receptors and, in addition, the receptor-mediated degradation of ANP. RCA cells bind rat alpha ANP-(1-28) reversibly at 37 C, apparently to a single class of high affinity (Kd approximately equal to 50 pM) binding sites (approximately equal to 20,000 sites per cell). Binding of rat alpha ANP-(1-28) elicits rapid accumulation of intracellular cGMP. However, the concentrations of rat alpha ANP-(1-28) and related peptides, abbreviated at the N- and C-terminals, required to stimulate cGMP synthesis are substantially greater than those required for binding. Analysis by HPLC of 125I-labeled rat alpha ANP-(1-28) bound to RCA cells at 37 C illustrates the rapid and continuous degradation of the radioiodinated rat alpha ANP-(1-28) to two radiolabeled products, one of which, 125I-labeled tyrosine is the major radiolabeled component that dissociates from the cells. Measurement of rat alpha ANP-(1-28) interaction with RCA cells by radioligand binding techniques therefore subsumes several processes. One of these processes is the rapid and continuous degradation of specifically bound ANP by these cells and perhaps also other target cells that respond to ANP.


Subject(s)
Atrial Natriuretic Factor/metabolism , Muscle, Smooth, Vascular/physiology , Receptors, Cell Surface/physiology , Animals , Carotid Arteries/cytology , Carotid Arteries/metabolism , Carotid Arteries/physiology , Chromatography, High Pressure Liquid , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rabbits , Receptors, Atrial Natriuretic Factor
14.
Am J Physiol ; 253(6 Pt 1): C809-16, 1987 Dec.
Article in English | MEDLINE | ID: mdl-2827497

ABSTRACT

We have used a smooth muscle cell line isolated from rabbit carotid artery (RCA) as a model system with which to study the ligand-mediated regulation of the atrial natriuretic peptide (ANP) receptor. RCA cells bind rat alpha-ANP-(1-28) semi-irreversibly at 4 degrees C, apparently to a single class of high-affinity binding sites (Kd approximately equal to 5 pM). In contrast, ANP initially bound to RCA cells at 37 degrees C is fully reversible after the cells have been cooled to 4 degrees C. Although the measured dissociation constant for binding rat alpha-ANP-(1-28) is 10-fold lower at 4 degrees C than at 37 degrees C, the maximum amount of specifically bound radioligand is equivalent at both temperatures. We infer this to be suggestive of ANP receptor recycling occurring at 37 degrees C. This hypothesis is substantiated by experiments demonstrating the rapid disappearance of ANP receptors (measured at 4 degrees C) after exposure of RCA cells to ANP at 37 degrees C. We believe this reflects a loss of cell-surface receptors that is reversible on removal of ANP from the incubation medium at 37 degrees C. The concentration dependence and rank order of potency of ANP in eliciting receptor redistribution resembles that for binding to the ANP receptor and not for eliciting cyclic GMP formation. In contrast to this striking loss of presumed cell-surface receptors, incubation of RCA cells with ANP elicits only limited desensitization of the cyclic GMP response.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Atrial Natriuretic Factor/physiology , Muscle, Smooth, Vascular/physiology , Receptors, Cell Surface/physiology , Animals , Cell Line , Cyclic GMP/biosynthesis , Endocytosis , Kinetics , Rabbits , Receptors, Atrial Natriuretic Factor , Structure-Activity Relationship , Temperature
15.
Biochemistry ; 26(9): 2642-56, 1987 May 05.
Article in English | MEDLINE | ID: mdl-3300777

ABSTRACT

Two-dimensional proton nuclear magnetic resonance spectroscopy at 500 MHz has been carried out on the cyclic decapeptide antagonist of gonadotropin-releasing hormone: cyclo-(delta 3-Pro1-D-pClPhe2-D-Trp3-Ser4-Tyr5-D-Trp6-NMeLeu7-Arg8- Pro9-beta-Ala 10). The antagonist exists in two slowly interconverting conformations. All data are consistent with the conclusion that one form has all-trans peptide bonds and the other has a cis beta-Ala10-delta3-Pro1 bond. With the use of sequential assignment methods, chemical shift assignments were obtained for all backbone and side-chain protons of both conformational isomers except for the serine and tyrosine hydroxyl groups and the C gamma, C delta, and guanidinium group protons of the arginine. Temperature dependence of spectral parameters and magnitudes of observed nuclear Overhauser effects support the interpretation that both conformers of the antagonist consist of two beta-turns (type II', D-Trp6-NMeLeu7; type II, delta 3-Pro1-D-pClPhe2) connected by extended antiparallel beta-like strands.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Peptides, Cyclic , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Protein Conformation , Thermodynamics
16.
Biochemistry ; 24(8): 1933-41, 1985 Apr 09.
Article in English | MEDLINE | ID: mdl-2861848

ABSTRACT

Peptide bonds between residues 7 and 8 residues 8 and 9, postulated internal cleavage sites of the peptide hormone somatostatin, were subjected to "pairwise" retro-inverso modification, where atoms of these peptide bonds were interchanged to give the analogues [gPhe7-m-(RS)-Trp8]somatostatin (I) and [gTrp8-m-(RS)-Lys9]somatostatin (II). Key fragments containing the modifications were synthesized by using [bis(trifluoroacetoxy)iodo]benzene for the generation of gem-diaminoalkyl-containing precursors from peptide amides. The versatility of solution synthetic methods was utilized to allow the incorporation of the modified segments. Protecting groups, removable selectively and under mild conditions, included tert-butyl-based groups for the side chains and the tert-butylmercapto group for the cysteine thiols. The excellent results obtained in the syntheses of analogues I and II, and previously of somatostatin on a larger scale [Moroder, L., Gemeiner, M., Goehring, W., Faeger, E., Thamm, P., & Wunsch, E. (1981) Biopolymers 20, 17-31], suggest the general feasibility of this route for the synthesis of centrally modified analogues. The purification of the products by Sephadex LH-20 chromatography afforded the separation of diastereomers of both analogues. The two isomers of I showed significant but different activities while those of analogue II were marginally active.


Subject(s)
Somatostatin/analogs & derivatives , Somatostatin/chemical synthesis , Amino Acid Sequence , Chromatography, Thin Layer , Indicators and Reagents , Optical Rotation , Structure-Activity Relationship
18.
Int J Pept Protein Res ; 21(1): 84-92, 1983 Jan.
Article in English | MEDLINE | ID: mdl-6826285

ABSTRACT

Partial retro-inverso modification of biologically active peptides is described as a topochemical alteration of the backbone to prevent enzymatic degradation. The preparation of gem-diaminoalkyl residues from peptide amides using the reagent [bis(trifluoroacetoxy)iodo]benzene (TIB) is discussed. Treatment of N-t-butyloxycarbonyl tyrosine and N-t-butyloxycarbonyl tryptophan with this reagent led to decomposition of the protected amino acids. Protecting the tyrosine and tryptophan residues by t-butyl ether and Nin-formyl groups, respectively, prevented decomposition and led to good yields of the desired products. Racemic 2-alkylmalonyl diastereomers were found to be separable by HPLC. The chiral stability of peptides containing optically active malonyl residues was investigated under simulated physiological conditions. Synthetic considerations for the incorporation of gem-diaminoalkyl and 2-alkylmalonyl residues into larger peptides to yield partially modified retro-inverso peptide analogs are presented.


Subject(s)
Peptides , Amino Acid Sequence , Peptides/chemical synthesis , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...