Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Gen Comp Endocrinol ; 177(1): 70-5, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22387984

ABSTRACT

Thyrotropin (TSH) is a pituitary glycoprotein hormone heterodimer that binds to its G-protein coupled receptor (TSH-R) at the thyroid to promote the synthesis and secretion of thyroid hormone. Very little is known about TSH-TSH-R interactions in teleost fish. Mammalian gonadotropins have been reported to have an intrinsic ability to activate teleost fish TSH-Rs, suggesting the TSH-R in teleost fish is more promiscuous than in other vertebrates. In this study we utilized the goldfish T(4)-release response and recombinant human TSH analogs as in vivo tools to evaluate the structural constraints on hormone-receptor interactions. We found that four positively charged lysines substituted for neutral or negatively charged amino acids within positions 11-20 of the glycoprotein hormone subunit α (GSUα) significantly increased biological activity of hTSH in fish, as it does in mammals. We further found that bovine follicle stimulating hormone but not luteinizing hormone, whose GSUα subunits also contain four lysine or arginine amino acid residues in the N-terminal portion of GSUα, was thyrotropic in goldfish, suggesting gonadotropin ß subunit contributes to the heterothyrotropic activity. Though recombinant human FSH did not produce a dose-dependent increase in T(4), thyrotropic activity could be acquired with the addition of positively charged amino acids at the N-terminal portion of its GSUα, confirming the importance of the charge on those amino acids for activation of the goldfish TSH-R. These studies demonstrate that mammalian glycoprotein hormone analogs can be utilized to evaluate the conservation of receptor binding and activation mechanisms between fish and mammals.


Subject(s)
Goldfish/metabolism , Receptors, Thyrotropin/metabolism , Animals , Evolution, Molecular , Goldfish/blood , Gonadotropins/blood , Gonadotropins/metabolism , Humans , Immunoassay , Thyrotropin/pharmacology , Thyroxine/blood
2.
Endocrinology ; 141(9): 3514-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10965926

ABSTRACT

Among glycoprotein hormone receptors the TSH receptor (TSHR) is the most susceptible to constitutive activation by mutations in various regions of the molecule, including mutations in the extracellular domain (ECD) and extracellular loops of the transmembrane domain (TMD). To understand the role of the ECD in TSHR activation we have tested several TSHR constructs with major deletions of the ECD. Previous studies reported very low expression of such truncated glycoprotein hormone receptors, which prevented reliable assessment of their ligand-binding and basal constitutive activities. We have eliminated this problem using TSHR tagged at its N-terminus with a hemagglutinin tag (HA) recognized by the HA-specific monoclonal antibody. Based on such quantitation the TSHR deletion mutant missing 386 N-terminal amino acid residues, constituting 98% of the entire ECD, showed 4-7 fold higher normalized basal activity compared to activity of the corresponding wild-type (WT) TSHR construct. This increase in basal activity was significantly inhibited by linking the common alpha-subunit of glycoprotein hormones at the N-terminus of the truncated TSH receptor. The role of a hypothetical activating fragment (409-418) in TSHR activation was further studied using peptides and mutagenesis of charged residues. This study provides important evidence supporting the "two-state" model of TSHR activation and the potential role of proteolytic cleavage for receptor activation. Accordingly, the mechanism of hormone-induced receptor activation is dependent, at least in part, on the elimination of inhibitory interactions within the receptor. Such intra-molecular inhibition of TSHR may include electrostatic interactions between the ECD and extracellular loops of TMD. Moreover, the truncated, constitutively active receptors described herein provide new insights valuable in the design of TSHR antagonists.


Subject(s)
Antithyroid Agents/pharmacology , Extracellular Space/metabolism , Receptors, Thyrotropin/antagonists & inhibitors , Receptors, Thyrotropin/metabolism , Animals , Antithyroid Agents/chemistry , COS Cells , Drug Design , Extracellular Space/drug effects , GTP-Binding Proteins/metabolism , Gene Deletion , Humans , Mutation/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Thyrotropin/genetics
3.
J Biol Chem ; 275(35): 27457-65, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10859314

ABSTRACT

We have previously engineered the first superactive analogs of human thyrotropin (hTSH) by using a novel design strategy. In this study, we have applied homology comparisons focusing on the alphaL3 loop of the common alpha-subunit of human glycoprotein hormones. Seven highly variable amino acid residues were identified, and charge-scanning mutagenesis revealed three previously unrecognized modification permissive domains and four gain-of-function lysine substitutions. Such gain-of-function mutations were hormone- and receptor-specific and dependent on location and basic charge. Cooperativity of individual substitutions was established in double and triple lysine mutants. In combinations of the most potent alphaL3 loop analog with two previously characterized loop analogs, a higher degree of cooperativity for the alphaL3 loop analog compared with both the alphaL1 loop analog and the hTSH-betaL3 loop analog was observed. We demonstrated that spatially distinct regions of the common alpha-subunit contribute differentially to the interaction of hTSH with its receptor and that combinations of two modified loops on the same and on opposite sides of the hTSH molecule display similar increases in in vitro biopotency. In addition, combination of all three superactive loops showed cooperativity in receptor binding and activation resulting in the most potent hTSH superactive analog described to date.


Subject(s)
Thyrotropin/analogs & derivatives , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Humans , Lysine/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Homology, Amino Acid , Thyrotropin/chemistry , Thyrotropin/genetics
4.
Methods ; 21(1): 67-81, 2000 May.
Article in English | MEDLINE | ID: mdl-10764608

ABSTRACT

This article provides the reader with an overview of methodological strategies to investigate structure-function relationships of human thyroid-stimulating hormone (hTSH). Various aspects of hTSH production, purification, and characterization described here in more detail are not only relevant to studies on other members of the glycoprotein hormone family, but also applicable to studies of other glycosylated proteins. Knowledge of structure-function relationships of specific hTSH domains is important for a better understanding of the molecular mechanisms of its action. New insights from such studies permit the design of glycoprotein hormone analogs with specific pharmacological properties and potential clinical applications.


Subject(s)
Thyrotropin/chemistry , Thyrotropin/physiology , Animals , Biological Assay/methods , Circular Dichroism , Databases, Factual , Humans , Male , Models, Biological , Mutation , Oligosaccharides/chemistry , Protein Engineering , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemical synthesis , Recombinant Proteins/isolation & purification , Structure-Activity Relationship , Thyrotropin/genetics , Time Factors
5.
Thyroid ; 9(5): 447-50, 1999 May.
Article in English | MEDLINE | ID: mdl-10365675

ABSTRACT

We have gained insight into the molecular mechanism of human thyrotropin (hTSH) action through cloning of the human TSHbeta subunit gene, development of recombinant TSH and novel analogues and chimeras produced by site-directed as well as cassette mutagenesis. A variety of loss of function mutations have shown several key domains in both the alpha- and beta-subunits that are important for high-affinity ligand interaction with the receptor. In contrast the specificity of receptor interaction was shown to be determined primarily by areas within the hTSH-beta "seat-belt" region. We have also designed various gain of function mutants (superagonists) using evolutionary considerations, homology modeling, and sequence comparisons within the cystine knot growth factor superfamily. Such superagonists resulted from increasing the positive charge by introduction of lysine or arginine residues or neutralization of negatively charged residues of the peripheral hairpin loops of each subunit in various combinations. Certain superagonists increased receptor binding, in vitro and in vivo bioactivity 100- to 1000-fold, more than that achieved previously for any other known protein ligand. In vivo metabolic clearance and biologic activity could be separately modulated by alteration of TSH carbohydrate structure including production of chimeras that added sites of O-glycosylation and/or covalently linked the alpha- and beta-subunits. These data suggest that electrostatic interactions resulting from net positive charge in TSH and net negative charge in its receptor play an important role in high-affinity TSH receptor binding and signal transduction. Insights gained from the design of such novel recombinant TSH analogues and chimeras should have many diagnostic and therapeutic applications. These include the design of improved in vitro assays for thyrotropic factors as well as the design of second generation recombinant TSH analogues for the detection and treatment of thyroid cancer.


Subject(s)
Thyrotropin/genetics , Thyrotropin/physiology , Glycosylation , Humans , Models, Molecular , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Protein Structure, Secondary , Receptors, Thyrotropin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thyrotropin/chemistry
6.
Nat Biotechnol ; 16(9): 871-5, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9743123

ABSTRACT

By combining evolutionary considerations, sequence comparisons and homology modeling we have designed recombinant human thyroid-stimulating hormone (hTSH) analogs with increased receptor binding and activity. The introduction of seven basic residues into the peripheral loops of hTSH resulted in up to a 50,000-fold increase in receptor binding affinity and 1300-fold increase in intrinsic activity. Such analogs are not only of potential clinical interest but can be tools to explore molecular aspects of conventional as well as nonclassical actions of glycoprotein hormones. These design strategies should be applicable to the development of novel analogs of other related hormones and growth factors with a variety of therapeutic and basic science applications, particularly for proteins that have undergone evolutionary decrease in bioactivity.


Subject(s)
Drug Design , Hormone Antagonists/chemical synthesis , Thyrotropin/antagonists & inhibitors , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Endothelial Growth Factors/metabolism , Hormone Antagonists/metabolism , Humans , Lymphokines/metabolism , Protein Conformation , Receptors, Thyrotropin/metabolism , Sequence Homology, Amino Acid , Thyrotropin/chemistry , Thyrotropin/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
J Biol Chem ; 272(34): 21312-6, 1997 Aug 22.
Article in English | MEDLINE | ID: mdl-9261143

ABSTRACT

The human thyroid-stimulating hormone (hTSH) subunits alpha and beta are transcribed from different genes and associate noncovalently to form the bioactive hTSH heterodimer. Dimerization is rate-limiting for hTSH secretion, and dissociation leads to hormone inactivation. Previous studies on human chorionic gonadotropin (hCG) and human follicle-stimulating hormone had shown that it was possible by subunit gene fusion to produce a bioactive, single chain hormone. However, neither the stability nor the clearance from the circulation of such fused glycoprotein hormones has been studied. We show here that genetic fusion of the hTSH alpha- and beta-subunits using the carboxyl-terminal peptide of the hCG beta-subunit as a linker created unimolecular hTSH whose receptor binding and bioactivity were comparable to native hTSH. Interestingly, the fused hTSH had higher thermostability and a longer plasma half-life than either native or dimeric hTSH containing the hCG beta-subunit-carboxyl-terminal peptide, suggesting that dimer dissociation may contribute to glycoprotein hormone inactivation in vivo. In addition, we show for the first time that synthesis of hTSH as a single polypeptide chain could overcome certain mutagenesis-induced defects in hTSH secretion, therefore enabling functional studies of such mutants. Thus, in addition to prolongation of plasma half-life, genetic fusion of hTSH subunits should be particularly relevant for the engineering of novel analogs where desirable features are offset by decreased dimer formation or stability. Such methods provide a general approach to expand the spectrum of novel recombinant glycoprotein hormones available for in vitro and in vivo study.


Subject(s)
Receptors, Thyrotropin/metabolism , Thyrotropin/chemistry , Animals , CHO Cells , Cell Membrane/metabolism , Cricetinae , Dimerization , Humans , Metabolic Clearance Rate , Mutagenesis, Site-Directed , Rats , Swine , Thyrotropin/metabolism , Thyrotropin/pharmacokinetics
9.
J Biol Chem ; 272(24): 15532-40, 1997 Jun 13.
Article in English | MEDLINE | ID: mdl-9182589

ABSTRACT

The region between the 10th and 12th cysteine (Cys88-Cys105 in human thyroid-stimulating hormone beta-subunit (hTSHbeta)) of the glycoprotein hormone beta-subunits corresponds to the disulfide-linked seat-belt region. It wraps around the common alpha-subunit and has been implicated in regulating specificity between human choriogonadotropin (hCG) and human follicle-stimulating hormone (hFSH), but determinants of hTSH specificity are unknown. To characterize the role of this region for hTSH, we constructed hTSH chimeras in which the entire seat-belt region Cys88-Cys105 or individual intercysteine segments Cys88-Cys95 and Cys95-Cys105 were replaced with the corresponding sequences of hCG and hFSH or alanine cassettes. Alanine cassette mutagenesis of hTSH showed that the Cys95-Cys105 segment of the seat-belt was more important for TSH receptor binding and signal transduction than the Cys88-Cys95 determinant loop region. Replacing the entire seat-belt of hTSHbeta with the hCG sequence conferred full hCG receptor binding and activation to the hTSH chimera, whereas TSH receptor binding and activation were abolished. Conversely, introduction of the hTSHbeta seat-belt sequence into hCGbeta generated an hCG chimera that bound to and activated the TSH receptor but not the CG/lutropin (LH) receptor. In contrast, an hTSH chimera bearing hFSH seat-belt residues did not possess any follitropic activity, and its thyrotropic activity was only slightly reduced. This may in part be due to the fact that the net charge of the seat-belt is similar in hTSH and hFSH but different from hCG. However, exchanging other regions of charge heterogeneity between hTSHbeta and hFSHbeta did not confer follitropic activity to hTSH. Thus, exchanging the seat-belt region between hTSH and hCG switches hormonal specificity in a mutually exclusive fashion. In contrast, the seat-belt appears not to discriminate between the TSH and the FSH receptors, indicating for the first time that domains outside the seat-belt region contribute to glycoprotein hormone specificity.


Subject(s)
Chorionic Gonadotropin/genetics , Follicle Stimulating Hormone/genetics , Luteinizing Hormone/metabolism , Recombinant Fusion Proteins/metabolism , Thyrotropin/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Follicle Stimulating Hormone/metabolism , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/genetics , Thyrotropin/genetics
10.
Endocrinology ; 138(1): 92-100, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8977390

ABSTRACT

To obtain large amounts of hTSH and to study the role of the N-linked oligosaccharides for its biological activity, hTSH was produced using recombinant baculovirus containing the human alpha-subunit and a hTSH beta-minigene, respectively, both under the control of the polyhedrin promoter. Expression in insect cells was 800-1000 ng/ml, 30-fold higher than in our optimized mammalian transient transfection system using Chinese hamster ovary (CHO) cells (20-50 ng/ml). The in vitro activity of insect-cell expressed hTSH (IC-hTSH) was increased 5-fold compared with CHO-hTSH, judged by the ability to induce cAMP production in CHO cells stably transfected with the hTSH receptor (JP09) and the rat thyroid cell line FRTL-5, as well as growth promotion in FRTL-5 cells. Lectin binding and enzymatic desialylation studies suggested that in contrast to CHO-hTSH, IC-hTSH lacked complex-type oligosaccharides terminating with sialic acid but contained predominantly high mannose-type oligosaccharides. The in vitro activity of CHO-hTSH also increased 5- to 6-fold upon treatment of the hTSH-producing cells with the oligosaccharide processing inhibitors swainsonine and castanospermine, which inhibit formation of complex, terminally sialylated oligosaccharides, and upon enzymatic desialylation. In contrast, insect cell-expression or treatment with processing inhibitors did not affect TSH receptor binding. Despite the higher in vitro activity, IC-hTSH had a much lower in vivo activity than CHO-hTSH, due to rapid clearance from the circulation. In summary, this study shows for the first time that relatively high levels of recombinant hTSH with high in vitro bioactivity can be produced in a baculovirus system. Cell-dependent glycosylation is a major factor that determines the final in vivo biopotency of recombinant glycoproteins, a finding that should be of general relevance for all insect cell-produced glycosylated proteins. Although not suitable for clinical use, highly bioactive recombinant hTSH derived from high expression in insect cells should be useful in defining structure-function relations of hormone analogs.


Subject(s)
Recombinant Proteins/biosynthesis , Thyrotropin/biosynthesis , Animals , Baculoviridae/genetics , CHO Cells , Cricetinae , Glycosylation , Humans , Lectins/metabolism , Male , Metabolic Clearance Rate , Mice , Rats , Receptors, Thyrotropin/metabolism , Spodoptera , Thyrotropin/pharmacology
11.
Trends Endocrinol Metab ; 7(8): 277-86, 1996 Oct.
Article in English | MEDLINE | ID: mdl-18406760

ABSTRACT

Recent progress in structure-function studies of glycoprotein hormones has provided new insights into the molecular mechanisms of action of these hormones and has further supported the concept that physiological modulation of assembly, bioactivity, and clearance of these hormones is dependent on specific structural components. This review emphasizes current advances in the structure-function relationships of human TSH, which have contributed to further elucidation of common and hormone specific features within the glycoprotein hormones family. Novel strategies are now being applied to investigate the role of individual structural elements. The principks discovered in such studies are essential to understand the physiological regulation of hormone bioactivity and allow for the rational design of novel analogs with potential therapeutic applications.

12.
Nat Biotechnol ; 14(10): 1257-63, 1996 Oct.
Article in English | MEDLINE | ID: mdl-9631089

ABSTRACT

We report the generation of superactive analogues of human glycoprotein hormones, with potential applications in thyroid and reproductive disorders. Current biological and structural data were used to rationalize mutagenesis. The 11-20 region in the alpha-subunit with a cluster of lysine residues forms a previously unrecognized domain critical for receptor binding and signal transduction, as well as an important motif in the evolution of glycoprotein hormone activities. The gradual elimination of basic residues in the alpha-subunit coincided with the evolutionary divergence of the hominids from the Old World monkeys. By selective reconstitution of certain critical residues present in homologous nonhuman hormones we have developed human thyroid stimulating hormone and chorionic gonadotropin analogues with substantial increases in receptor binding affinity and bioactivity, thus providing a paradigm for the design of novel therapeutic protein analogues.


Subject(s)
Glycoproteins/chemistry , Glycoproteins/genetics , Hormones/chemistry , Hormones/genetics , Amino Acid Sequence , Animals , Base Sequence , Biotechnology , Cell Line , Chorionic Gonadotropin/chemistry , Chorionic Gonadotropin/genetics , DNA Primers/genetics , Drug Design , Evolution, Molecular , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Engineering , Thyrotropin/analogs & derivatives , Thyrotropin/chemistry , Thyrotropin/genetics
13.
Mol Endocrinol ; 10(6): 769-79, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8776737

ABSTRACT

Amino acid residues 33-44 of the common alpha-subunit of the glycoprotein hormones have been implicated in heterodimerization as well as high affinity receptor binding of human (h) CG. In the present study, we compared the role of specific amino acids within this region for glycoprotein hormone heterodimer formation, using a transient transfection system to coexpress different mutant alpha-subunit constructs with the beta-subunit of either hTSH, hCG, or hFSH. Our results identified a crucial role for alpha Pro38 in the heterodimer expression of hTSH as well as hFSH, similiar to what had been described for hCG. In contrast, alpha Ala38, which had been critical for hCG, was not essential for hTSH heterodimer expression and less important for hFSH, whereas alpha Phe33 and alpha Arg35 appeared uniquely important for hFSH. Furthermore, we assessed the role of these residues for bioactivity and receptor binding of hTSH. Mutation of the surface-exposed residues alpha Arg42-Ser43-Lys44, which form part of a unique alpha-helical structure, to Ala42-Ala43-Ala44, decreased TSH receptor binding using porcine thyroid membranes as well as rat FRTL-5 cells. Residues alpha Phe33 and alpha Arg35, in contrast, were not important for high affinity binding of hTSH. In the signal transduction of hTSH, alpha Ala36 was necessary for efficient growth induction in FRTL-5 cells but not for cAMP production in either FRTL-5 cells or Chinese hamster ovary cells expressing the human TSH receptor (JP09). Similarly, residues alpha Arg42-Ser43-Lys44 were more important for hTSH-mediated induction of cell growth than cAMP production. Mutating alpha Arg35 to Ala reduced cAMP induction but not receptor binding of hTSH. In summary, using site-directed mutagenesis, we identified a domain, residues 33-44 of the common alpha-subunit, important in heterodimer expression, receptor binding, and activation of hTSH. The comparison of the relative roles of specific amino acids within this region in hTSH with hCG and hFSH highlights previously unrecognized differences in the structural requirements for heterodimer expression among the members of the glycoprotein hormone family. Moreover, our findings revealed a novel role for residues alpha 33-44 in triggering different postreceptor events, suggesting that cAMP production and growth promotion may, at least in part, be dissociable functions of hTSH.


Subject(s)
Mutagenesis, Site-Directed , Thyrotropin/chemistry , Thyrotropin/genetics , Amino Acid Sequence , Animals , CHO Cells/metabolism , Cell Division/genetics , Cells, Cultured , Chorionic Gonadotropin/chemistry , Chorionic Gonadotropin/metabolism , Cricetinae , Cyclic AMP/biosynthesis , Humans , Models, Molecular , Protein Conformation , Rats , Receptors, Thyrotropin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thyroid Gland/cytology , Thyroid Gland/metabolism , Thyrotropin/metabolism
14.
J Biol Chem ; 270(49): 29378-85, 1995 Dec 08.
Article in English | MEDLINE | ID: mdl-7493973

ABSTRACT

We used a novel approach to study the role of the Asn-linked oligosaccharides for human thyrotropin (hTSH) activity. Mutagenesis of Asn (N) within individual glycosylation recognition sequences to Gln (Q) was combined with expression of wild type and mutant hTSH in cell lines with different glycosylation patterns. The in vitro activity of hTSH lacking the Asn alpha 52 oligosaccharide (alpha Q52/TSH beta) expressed in CHO-K1 cells (sialylated oligosaccharides) was increased 6-fold compared with wild type, whereas the activities of alpha Q78/TSH beta and alpha/TSH beta Q23 were increased 2-3-fold. Deletion of the Asn alpha 52 oligosaccharide also increased the thyrotropic activity of human chorionic gonadotropin, in contrast to previous findings at its native receptor. The in vitro activity of wild type hTSH expressed in CHO-LEC2 cells (sialic acid-deficient oligosaccharides), CHO-LEC1 cells (Man5GlcNAc2 intermediates), and 293 cells (sulfated oligosaccharides) was 5-8-fold higher than of wild type from CHO-K1 cells. In contrast to CHO-K1 cells, there was no difference in the activity between wild type and selectively deglycosylated mutants expressed in these cell lines. Thus, in hTSH, the oligosaccharide at Asn alpha 52 and, specifically, its terminal sialic acid residues attenuate in vitro activity, in contrast to the previously reported stimulatory role of this chain for human chorionic gonadotropin and human follitropin activity. The increased thyrotropic activity of alpha Q52/CG beta suggests that receptor-related mechanisms may be responsible for these differences among the glycoprotein hormones. Despite their increased in vitro activity, alpha Q52/TSH beta, and alpha Q78/TSH beta from CHO-K1 cells had a faster serum disappearance rate and decreased effect on T4 production in mice. These findings highlight the importance of individual oligosaccharides in maintaining circulatory half-life and hence in vivo activity of hTSH.


Subject(s)
Oligosaccharides/chemistry , Thyrotropin/chemistry , Animals , CHO Cells , Cricetinae , Cyclic AMP/biosynthesis , Glycosylation , Half-Life , Humans , Male , Mice , Mutagenesis, Site-Directed , N-Acetylneuraminic Acid , Rats , Recombinant Proteins , Sialic Acids/physiology , Structure-Activity Relationship , Thyrotropin/physiology
15.
Proc Natl Acad Sci U S A ; 92(20): 9062-6, 1995 Sep 26.
Article in English | MEDLINE | ID: mdl-7568073

ABSTRACT

The recombinant human thyroid stimulating hormone (rhTSH) containing oligosaccharides terminated with NeuAc(alpha 2-3)Gal(beta 1-4)GlcNAc beta 1 showed higher in vivo activity and lower metabolic clearance rate (MCR) than pituitary human TSH (phTSH), which contains oligosaccharides terminating predominantly in SO(4)4GalNAc(beta 1-4)GlcNAc beta 1. To elucidate the relative contribution of the sulfated and sialylated carbohydrate chains of each subunit in the MCR and bioactivity of the hormone, the alpha and beta subunits of phTSH, rhTSH, and enzymatically desialylated rhTSH (asialo-rhTSH; asrhTSH) were isolated, their oligosaccharides were analyzed, and the respective subunits were dimerized in various combinations. The hybrids containing alpha subunit from phTSH or asrhTSH showed higher in vitro activity than those with alpha subunit from rhTSH, indicating that sialylation of alpha but not beta subunit attenuates the intrinsic activity of TSH. In contrast, hybrids with beta subunit from rhTSH displayed lower MCR compared to those with beta subunit from phTSH. The phTSH alpha-rhTSH beta hybrid had the highest in vivo bioactivity followed by rhTSH alpha-rhTSH beta, rhTSH alpha-phTSH beta, phTSH alpha-phTSH beta, and asrhTSH dimers. These differences indicated that hybrids with beta subunit from rhTSH displayed the highest in vivo activity and relatively low MCR, probably due to higher sialylation, more multiantennary structure, and/or the unique location of the beta-subunit oligosaccharide chain in the molecule. Thus, the N-linked oligosaccharides of the beta subunit of glycoprotein hormones have a more pronounced role than those from the alpha subunit in the metabolic clearance and thereby in the in vivo bioactivity. In contrast, the terminal residues of alpha-subunit oligosaccharides have a major impact on TSH intrinsic potency.


Subject(s)
Cyclic AMP/metabolism , Glycoprotein Hormones, alpha Subunit/metabolism , Glycoprotein Hormones, alpha Subunit/pharmacology , Oligosaccharides/chemistry , Thyrotropin/metabolism , Thyrotropin/pharmacology , Animals , CHO Cells , Carbohydrate Sequence , Carbohydrates/analysis , Cricetinae , Glycoprotein Hormones, alpha Subunit/chemistry , Humans , Macromolecular Substances , Male , Metabolic Clearance Rate , Molecular Sequence Data , Oligosaccharides/metabolism , Oligosaccharides/pharmacology , Protein Multimerization , Rats , Rats, Sprague-Dawley , Receptors, Thyrotropin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Structure-Activity Relationship , Thyrotropin/chemistry , Transfection , Triiodothyronine/blood , Triiodothyronine/pharmacokinetics
16.
Endocrinology ; 136(9): 3839-48, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7544273

ABSTRACT

Recombinant TSH is now successfully being used in clinical studies of thyroid cancer. Because of its therapeutic potential, we have constructed a longer acting analog of TSH by fusing the carboxy-terminal extension peptide (CTEP) of hCG beta onto TSH beta. When coexpressed either with alpha-subunit complementary DNA or alpha minigene in African green monkey (COS-7) and human embryonic kidney (293) cells, the chimera was fully bioactive in vitro and exhibited enhanced in vivo potency associated with a prolonged plasma half-life. The addition of 25 amino acids with 4 O-linked oligosaccharide chains did not affect the assembly and secretion of chimeric TSH. Wild-type (WT) and chimeric TSH secreted by COS-7 and 293 cells displayed wide differences in their plasma half-lives, presumably due to the presence of terminal sialic acid and SO4 on their oligosaccharide chains, respectively. Chimeric and WT TSH secreted by both cell lines demonstrated similar bioactivity in cAMP production, with some differences in [3H]thymidine incorporation. Chimeric TSH appears to be more effective in COS-7 cells than in 293 cells, as judged by growth assay. COS-7-produced chimeric TSH showed the maximum increase in half-life, indicating the importance of sialic acid in prolonging half-life and in vivo potency. Sulfation of both subunits, predominantly beta and to a lesser extent alpha, appears to be responsible at least in part for the increased metabolic clearance of WT and chimeric TSH secreted by 293 cells. Apart from its therapeutic potential, chimeric TSH produced in various cell lines can be used as a tool to delineate the roles of sulfate and sialic acid in the in vivo clearance and, thereby, the in vivo bioactivity.


Subject(s)
Carbohydrates/physiology , Chorionic Gonadotropin/analysis , Peptide Fragments/analysis , Recombinant Fusion Proteins/analysis , Thyrotropin/chemistry , Thyrotropin/pharmacokinetics , Animals , Base Sequence , CHO Cells , Chlorocebus aethiops , Chorionic Gonadotropin/chemistry , Chorionic Gonadotropin, beta Subunit, Human , Cricetinae , Cyclic AMP/metabolism , DNA Primers/analysis , DNA Primers/chemistry , DNA Primers/genetics , Half-Life , Humans , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Male , Methionine/analysis , Methionine/metabolism , Mice , Molecular Sequence Data , N-Acetylneuraminic Acid , Peptide Fragments/chemistry , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Sialic Acids/physiology , Sulfates/analysis , Sulfates/metabolism , Thymidine/analysis , Thymidine/metabolism , Thyrotropin/genetics , Tritium
17.
Endocrinology ; 136(8): 3325-30, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7628367

ABSTRACT

The recombinant human TSH (rhTSH) with highly sialylated oligosaccharide chains showed higher in vivo bioactivity and a lower MCR than the predominantly sulfated pituitary human TSH (phTSH). The aim of the present study was to investigate the role of terminal carbohydrate residues in organ distribution and metabolic clearance of TSH using an in vivo rat model. The different 125I-labeled TSH preparations with distinct carbohydrate composition were injected i.v. At various time points (5-180 min) after bolus TSH injection, blood, liver, kidney, spleen, lung, heart, and thyroid samples were collected. TSH uptake was determined by trichloroacetic acid precipitation of [125I]TSH in the organ homogenates. The rhTSH (solely sialylated) was distributed predominantly to the kidneys 5, 15, and 30 min after injection. In contrast, phTSH (sulfated/sialylated) and bovine TSH (bTSH; solely sulfated) were cleared predominantly by the liver (at 5 min), with a later renal phase of clearance (at 30 min). Asialo-rhTSH was cleared by the liver with only minor involvement of other organs. The early liver uptake (at 5 min) was proportionally highest for the asialo-rhTSH and bTSH preparations and lowest for rhTSH, which correlated inversely with the serum levels and the degree of sialylation. Blockade of the N-acetylgalactosamine (GalNAc) sulfate receptors by injection of bovine LH resulted in a significant decrease in liver uptake of phTSH. Similarly, liver uptake of asialo-rhTSH was significantly inhibited by injection of asialo-fetuin. Thus, phTSH and bTSH preparations containing sulfated oligosaccharide chains are cleared at least in part by the GalNAc sulfate-specific receptors in the liver. In contrast, rhTSH with highly sialylated oligosaccharides in both subunits accumulates predominantly in the kidneys, even at the early phase of clearance, indicating that sialylated glycoprotein hormones escape from specific receptor-mediated clearance mechanisms in the liver. These data indicate that terminal sialic acid and GalNAc sulfate residues, each to a different extent, determine glycoprotein hormone distribution and thereby plasma level, which as we have shown previously is a major factor in determining the in vivo potency of TSH.


Subject(s)
Asparagine/physiology , Oligosaccharides/metabolism , Pituitary Gland/metabolism , Thyrotropin/chemistry , Thyrotropin/metabolism , Acetylglucosamine/antagonists & inhibitors , Animals , Asialoglycoprotein Receptor , Asialoglycoproteins/metabolism , Asparagine/chemistry , Carbohydrate Metabolism , Humans , Kidney/metabolism , Liver/metabolism , Male , Oligosaccharides/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/antagonists & inhibitors , Recombinant Proteins , Thyrotropin/blood , Tissue Distribution
18.
Mol Endocrinol ; 9(8): 948-58, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7476992

ABSTRACT

The glycoprotein hormones TSH, CG, LH, and FSH are heterodimers consisting of a hormone-specific beta-subunit and a common alpha-subunit. The aim of the present study was to investigate the role of the carboxy terminus of the common alpha-subunit (amino acids Tyr89-His90-Lys91-Ser92), which has been shown to be important for human (h) CG and hFSH, for the activity of hTSH. Successive truncations of the alpha-carboxy terminus by site-directed mutagenesis revealed a stepwise reduction of bioactivity occurring at residues alpha Ser92 and alpha His90 to 64% and 13%, respectively. This contrasts with previous findings for hCG and hFSH, where loss of bioactivity occurred in a single step with the deletion of alpha Lys91 but alpha Ser92 was not important. The decreased bioactivities of the hTSH alpha-truncation mutants were reflected by concomitant reductions of cAMP production, thyroid hormone synthesis and cell growth and were accompanied by a loss of receptor binding. Substitution of residues alpha Lys91 or alpha His90 with either a hydrophobic or a bulkier residues resulted in a reduction of receptor binding and signal transduction, indicating that the alpha-carboxy terminus of hTSH may interact with the TSH receptor in a tight contact area. Conversely, substitution of alpha His90 with smaller residues enhanced bioactivity. In addition, the integrity of the alpha-carboxy terminus was essential for hTSH expression. Thus, we showed common and different roles of the alpha-carboxy-terminal residues for the glycoprotein hormones. The unique role of alpha Ser92 in hTSH activity explains the evolutionary constraint to preserve the alpha-carboxy-terminal Ser92 in all glycoprotein hormones.


Subject(s)
Thyrotropin/chemistry , Amino Acid Sequence , Animals , Cell Division , Cells, Cultured , Cyclic AMP/biosynthesis , Humans , Molecular Sequence Data , Rats , Receptors, Thyrotropin/physiology , Recombinant Proteins , Signal Transduction , Structure-Activity Relationship , Triiodothyronine/metabolism
19.
Horm Metab Res ; 27(4): 173-8, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7750899

ABSTRACT

Pulsatile secretion of thyrotropin (TSH) is a well characterized phenomenon in the human, yet only limited data are available from animal studies. We propose a combined model of chronic catheterization and suppression of endogenous TSH release in the rat allowing us to apply quantified pulses of different preparations of thyrotropin intravenously with minimal interference with endogenously produced hormone. Sprague-Dawley rats treated with drinking water containing 3,5,3'-triiodothyronine (T3) showed a significant 6-fold decrease of the rat TSH (rTSH) level after four days. Free thyroxine (FT4) levels were 9-fold below unsuppressed levels; FT4 response to exogenous TSH application above this suppressed baseline level was selected as an endpoint. Infusion studies compared pulsatile with continuous TSH administration. A low and a high dose of rTSH and recombinant human TSH (rec-hTSH) were administered. Levels of FT4 were compared after two and four days and responsiveness to a standard high bolus of rTSH 6 h after end of infusion was assessed. With regard to its potency for FT4 release, differences indicating a general trend as to superior efficiency of pulsatile TSH stimulation of the thyroid gland in comparison to continuous stimulation could be observed in all four experiments. More pronounced effects were seen after 2 and 4 days of high rTSH infusion and after 4 days of low rec-hTSH infusion. Comparison of FT4 responses to rTSH boli showed a 2- and 6-fold higher response in the pulsatile group compared to the continuous group after low and high rTSH infusion, respectively.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Thyrotropin/administration & dosage , Animals , Drinking , Humans , Kinetics , Male , Periodicity , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Thyroid Gland/drug effects , Thyrotropin/blood , Thyrotropin/pharmacology , Thyroxine/blood , Triiodothyronine/administration & dosage , Triiodothyronine/pharmacology
20.
Thyroid ; 5(1): 55-9, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7787435

ABSTRACT

A new and simple in vivo bioassay suitable for routine testing of pituitary and recombinant TSH preparations was developed. Male Albino Swiss CF-1 mice were given T3 in their drinking water to suppress endogenous TSH. T3, 3.0 micrograms/mL, given to mice for a period of 4 days decreased plasma total T4 (TT4) level to less than 10% of the nonsuppressed (control) level. Various doses of exogenous pituitary and recombinant TSH preparations were injected intraperitoneally and blood samples were obtained from the orbital sinus 6 h later. The TT4 level, measured by radioimmunoassay, served as the assay end-point. The assay required injection of approximately 3.0 micrograms of pituitary human TSH (phTSH), 1.0 microgram recombinant human TSH (rhTSH), 0.2 microgram bovine TSH (bTSH), and 0.1 microgram rat TSH (rTSH) to attain half-maximal response. The maximal level of TT4 after TSH stimulation was similar to that observed in normal, nonsuppressed mice. The procedure developed is relatively easy to perform, economical, and, unlike earlier TSH bioassays, does not require the use of radionuclides. This bioassay showed acceptable sensitivity and reliability in structure-function studies of pituitary TSH from different species as well as rhTSH.


Subject(s)
Biological Assay/methods , Thyrotropin/blood , Animals , Cattle , Male , Mice , Mice, Inbred Strains , Pituitary Gland/chemistry , Radioimmunoassay , Rats , Recombinant Proteins , Thyroxine/blood , Triiodothyronine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...