当前位置: 首页 > 期刊 > 《内分泌学杂志》 > 2003年第1期 > 正文
编号:10582334
Specificity of Cognate Ligand-Receptor Interactions: Fusion Proteins of Human Chorionic Gonadotropin and the Heptahelical Receptors for Human Luteiniz
http://www.100md.com 《内分泌学杂志》2003年第1期
     Abstractn-zp^, 百拇医药

    The family of glycoprotein hormones and their homologous heptahelical receptors represent an excellent system for comparative structure-function studies. We have engineered single chain molecules of human chorionic gonadotropin (hCG) fused to its cognate receptor, LH receptor (LHR), and to the noncognate receptors, TSH receptor (TSHR) and FSH receptor (FSHR; N-ß--receptor-C), to create the yoked (Y) complexes YCG/LHR, YCG/TSHR, and YCG/FSHR. The expression and bioactivity of these fusion proteins were examined in transiently transfected HEK 293 cells. Western blot analysis and antibody binding assays demonstrated that each of the proteins was expressed. In the case of YCG/LHR, minimal binding of exogenous hormone was observed due to the continued occupation of receptor by the fused ligand. The presence of hCG in the YCG/TSHR and YCG/FSHR, however, did not prevent binding of exogenous cognate ligand, presumably due to the lower affinity of hCG. The basal cAMP levels in cells expressing the YCG/LHR complex was approximately 20-fold higher than that in cells expressing LHR. Increases in basal cAMP production were also observed with YCG/TSHR and YCG/FSHR, e.g. 13- and 4-fold increases, respectively. Whereas the affinity and specificity of hCG for LHR are extraordinarily high, the hormone is capable of binding to and activating both TSHR and FSHR under these conditions that mimic high ligand concentrations. These findings were confirmed by adding high concentrations of hCG to cells expressing TSHR and FSHR. Although the functional interaction of hCG and TSHR has been recognized in gestational hyperthyroidism, there are no reports linking hCG to FSHR activation. This study, however, suggests that such a functional interaction is capable of occurring under conditions of high circulating levels of hCG, e.g. the first trimester of pregnancy and in patients with hCG-secreting tumors.

    Introductioni2x], http://www.100md.com

    THERE ARE FOUR hormones that comprise the family of human (h) glycoprotein hormones, human chorionic gonadotropin (hCG), LH, TSH, and FSH (1, 2). These hormones are heterodimers, consisting of a common -subunit and a receptor-specific ß-subunit, and confer activity by binding to their respective G protein-coupled receptors (GPCRs) (2). The glycoprotein hormone receptors are a special class of GPCRs, characterized by their relatively large ectodomain, which is responsible for high affinity hormone binding (3). Because of the high sequence similarity in their ß-subunits, both hCG and LH bind to the same receptor, LH receptor (LHR), whereas TSH and FSH bind to unique receptors, TSHR and FSHR, respectively (3). There is a high degree of sequence homology among the glycoprotein hormone ß-subunits (Fig. 1A) and among the glycoprotein hormone receptor ectodomains (Fig. 1B).i2x], http://www.100md.com

    fig.ommitteedi2x], http://www.100md.com

    Figure 1. Amino acid sequences of the human glycoprotein hormone ß-subunits (A) and glycoprotein hormone receptor ectodomains (B). The dashes indicate identical sequence, and the dots indicate gaps in the sequence as analyzed by CLUSTAL alignment (4 ). A, The amino acid sequences of the common -subunit and aligned ß-subunits are shown. , The seat belt region of the ß-subunits. B, The alignment of the receptor ectodomains is separated to show the N-terminal region (30 amino acid residues), the 9 (imperfect) LRR predicted by Bhowmick et al. (5 ) for rat LHR, and the Cys-rich region before transmembrane helix 1. , Identical residues in the three receptors; , conservative replacements, e.g. hydrophobic, identically charged residues, and T/S. The frequently observed (N/C) at position 9 (6 ) occurs in only three of the predicted LRRs of the LHR extracellular domain, LRR5–7 (5 ). The amino acid residues encoded by exons 1–10 and part of exon 11 are 1–31, 32–54, 55–80, 81–104, 105–129, 130–155, 156–178, 179–203, 204–265, 266–292, and 293–339, respectively.

    In 1994, the crystal structure of aglyco-hCG was determined independently by two groups (7, 8), which was later followed by that of partially glycosylated hFSH (9). This information has provided a molecular framework for structure-function studies designed to elucidate the regions of these gonadotropins that impart receptor binding and activation.td, http://www.100md.com

    We and others have used fusion protein techniques to further delve into structure-function studies within the families of glycoprotein hormones and their GPCRs (10, 11). Single chain, or yoked (Y), hormones have been designed in the manner N-hCGß--C (10, 11, 12) and N--hCGß-C (13, 14, 15). Our group (10) and that of Boime (11) first designed YhCG1 with hCGß at the N terminus; we then created YhCG3 in which hCGß is located at the C-terminus (13). Both forms of single chain hCG were effective in activating LHR; however, YhCG1 was able to bind LHR with a higher affinity than that found with YhCG3. This fusion protein technique was later expanded to include yoked hormone-receptor complexes in which YhCG1 and YhCG3 were fused to the N terminus of rat LHR: N-hCGß--CTP-LHR-C and N--CTP-hCGß-LHR-C, respectively (13, 16). This approach of using a fused ligand-receptor complex permits the study of receptor binding and activation by individual subunits, subunit mutants that fail to efficiently form holoproteins, and ligand mutants that bind with low affinity to their cognate receptor.

    As the glycoprotein hormones are comprised of a common -subunit and homologous ß-subunits, and their receptors are homologous as well, we have prepared and characterized fusion proteins of single chain hCG with the three human receptors using the configuration N-ß-TP-R to yield YCG/LHR, YCG/TSHR, and YCG/FSHR (Fig. 2) to investigate the anticipated weak binding of hCG to and the activation of its noncognate receptors, TSHR and FSHR. Although hCG stimulation of TSHR results in pathophysiological conditions such as gestational hyperthyroidism (17), there are no known clinical cases involving hCG activation of FSHR. However, the noncognate hormone-receptor interaction involving hCG and TSHR documents that cross-reactivity occurs within these families.vo(^, http://www.100md.com

    fig.ommitteedvo(^, http://www.100md.com

    Figure 2. Schematics of the YCG/R fusion protein constructs. YhCG was fused to the N terminus of each of the glycoprotein hormone receptors using the hCGß CTP, designated with vertical stripes, as a flexible linker region to give YCG/LHR, YCG/TSHR, and YCG/FSHR.

    The goal of the current study was to comparatively evaluate the ability of hCG to activate each of the human glycoprotein hormone receptors: hLHR, hTSHR, and hFSHR. This information is important in elucidating basic structure-function relationships of these families of hormones and receptors; furthermore, the information gained should aid in the design of agonists and antagonists for the glycoprotein hormone receptors.7, http://www.100md.com

    Materials and Methods7, http://www.100md.com

    Construction of yoked constructs7, http://www.100md.com

    PCR was used to add a portion of the CTP to the 5' end of each of the human glycoprotein hormone receptors as well as an XhoI restriction site to the 3' end of each receptor using the following primer designs: 5' primer, 5'-(AflIII/CTP/factor Xa/human receptor sequence)-3'; and 3' primer, 5'-(human receptor sequence/XhoI)-3'. Each of the human glycoprotein hormone receptors was digested with EcoRI and XhoI and then ligated to both YhCG1 that had been digested with BamHI and EcoRI, and pcDNA3 that had been digested with BamHI and XhoI to create vectors containing YCG/LHR, YCG/TSHR, and YCG/FSHR.

    Cell culture and transient transfections#24, http://www.100md.com

    HEK 293 cells were grown in monolayer culture in growth medium consisting of DMEM fortified with 10% (vol/vol) newborn calf serum, 10 mM HEPES buffer (pH 7.4), 50 U/ml penicillin, 50 µg/ml streptomycin, 50 µg/ml fungizone, and 0.125 µg/ml amphotericin B. Cells were grown and sustained at 37 C in humidified air containing 5% CO2. The cDNAs were transiently transfected using Lipofectamine 2000 as recommended by the manufacturer (Life Technologies, Inc., Grand Island, NY) into 75-cm2 tissue culture flasks containing HEK 293 cells (5 µg cDNA for each fusion protein).#24, http://www.100md.com

    Western blot analysis#24, http://www.100md.com

    Solubilized membrane fractions were electrophoresed on 10% SDS-PAGE and transferred to polyvinylidene difluoride membranes. The membrane was probed with a 1:1000 dilution of an anti-CTP polyclonal antibody (provided by Dr. Vernon Stevens, Ohio State University, Columbus, OH) and visualized by chemiluminescence via a secondary horseradish peroxidase-labeled antirabbit antibody.

    Hormone binding.dy, 百拇医药

    Cells were resuspended in 36 ml growth medium approximately 16–18 h after transfection, and 1 ml medium was added to each well of a 12-well tissue culture plate coated with 0.1% gelatin in PBS. The cells were assayed for 125I-labeled hormone binding approximately 24 h later. To perform competitive binding assays, increasing concentrations of unlabeled urinary hCG, bovine (bTSH; both hCG and bTSH were provided by Dr. Albert Parlow and the NIDDK), or hFSH (Sigma, St. Louis, MO) in Waymouth’s medium with 0.1% BSA (wt/vol) and 50 pM [125I]hCG, 50 pM [125I]bTSH, or 100 pM [125I]hFSH, respectively, were added to each well containing cells expressing the YCG/R protein or cognate receptor and incubated at 37 C for 6 h. Limited competitive binding studies were also performed with recombinant hCG (Organon, Oss, The Netherlands) and each of the 125I-labeled hormones and their cognate receptors. Nonspecific binding was determined by the addition of 1 µg/ml unlabeled hCG and bTSH and 3 µg/ml unlabeled hFSH. The incubation medium was aspirated, and the cells washed with PBS and then lysed by the addition of 0.5 ml 1 N NaOH. All binding assays were performed in duplicate. Mock-transfected cells treated in the same manner were used as negative controls.

    Cell surface antibody binding assay0@, 百拇医药

    Approximately 16–18 h after transfection, the cells were processed as described above, assayed for binding of a receptor-specific primary antibody, raised against a synthetic peptide, and then detected by secondary [125I]antirabbit antibody. For hLHR, a 1:4000 dilution of an antirat LHR antibody raised against residues 15–38 (provided by Dr. Patrick Roche, Mayo Medical School, Rochester, MN) was used. Dr. Mariusz Szkudlinski (Trophogen, Inc., Rockville, MD) donated an anti-hTSHR antibody raised against residues 352–366, and Dr. Mario Ascoli (University of Iowa, Iowa City, IA) provided an antirat FSHR antibody raised against residues 19–29. Dilutions of 1:1000 and 1:500 were made of the TSHR and FSHR antibodies, respectively. The cells were incubated in the appropriate dilution of the primary antibody, as indicated, in Waymouth’s medium with 0.3% (wt/vol) BSA for the LHR antibody and 0.1% (wt/vol) BSA for the TSHR and FSHR antibodies for 4 h at room temperature while shaking. After washing twice with Waymouth’s medium containing 0.3% (wt/vol) BSA for the LHR antibody and 0.1% (wt/vol) BSA for the TSHR and FSHR antibodies, the cells were incubated with [125I]antirabbit secondary antibody (400,000 cpm/well) in Waymouth’s medium containing 0.3% or 0.1% BSA for 1 h at room temperature with shaking. The medium was aspirated, and the cells were washed with PBS after the second incubation period. Lysates were prepared as described above, and the samples were counted in a -counter. Each binding experiment was performed in duplicate.

    cAMP assay;zrz9p, 百拇医药

    The cells were replated and washed as described above some 16–18 h after transfection, then incubated in Waymouth’s medium with 0.1% (wt/vol) BSA and 0.8 mM isobutylmethylxanthine for 15 min at 37 C. The cells were incubated with increasing concentrations of cognate hormone, urinary hCG, bTSH, or hFSH, for 30 min at 37 C in Waymouth’s medium containing 0.1% (wt/vol) BSA and 0.8 mM isobutylmethylxanthine immediately following the first incubation. In other studies increasing concentrations of recombinant hCG were added to cells expressing each of the three glycoprotein hormone receptors. The incubation medium was removed, and the cells were lysed in 100% ethanol at -20 C overnight. The extract was collected, dried under vacuum, and cAMP concentrations were determined by RIA as recommended (NEN Life Science Products, Boston, MA). Each data point was performed in duplicate, and mock-transfected cells were used as negative controls.;zrz9p, 百拇医药

    Data analysis

    Competitive binding and cAMP results were analyzed by nonlinear regression using the PRISM software program (GraphPad Software, Inc., San Diego, CA). The results are given as the mean ± SEM, based on three to five independent transfections, and significance was determined by t test. The figures showing competitive binding and hormone-mediated cAMP production are representative experiments. The results in Table 1 refer to the average of each individual parameter from multiple transfections.\%9rj0, 百拇医药

    fig.ommitteed\%9rj0, 百拇医药

    Table 1. Summary of binding and signaling parameters of YCG/Rs and human receptors\%9rj0, 百拇医药

    Results\%9rj0, 百拇医药

    Cell surface expression of the yoked hormone-receptor complexes\%9rj0, 百拇医药

    After transient transfection into HEK 293 cells, membrane preparations of cells expressing YCG/R complexes were prepared. Western blot analysis using a 1:1000 dilution of an anti-CTP antibody showed that all YCG/R proteins were expressed in the molecular mass range of 120–150 kDa (Fig. 3A).

    fig.ommitteed#, 百拇医药

    Figure 3. Expression of the YCG/R proteins. A, Western blot analysis using a 1:1000 dilution of CTP antibody and a 1:5000 of secondary antirabbit HRP antibody shows that all three YCG/R proteins are expressed. B, Cell surface expression is documented by receptor-specific primary antibody detected with [125I]antirabbit secondary antibody. There is no difference in cell surface expression when comparing the expression of each YCG/R protein with the corresponding wild-type human glycoprotein hormone receptor. The data are corrected for binding of antibody to mock transfected cells (n = 3). C, Specific binding of [125I]hCG, [125I]bTSH, and [125I]hFSH to their cognate receptors and yoked constructs. Cells expressing YCG/LHR showed minimal apparent binding of [125I]hCG due to the occupation of binding sites by the fused hormone. There was no difference in [125I]bTSH binding to cells expressing either YCG/TSHR or hTSHR, whereas cells expressing YCG/FSHR showed reduced binding of [125I]hFSH compared with those expressing hFSHR. In each case, specific binding was measured with mock-transfected cells, and the values obtained averaged 0–10 cpm; thus, no corrections were necessary.

    To compare cell surface expression levels of the YCG/R complexes with those of their corresponding receptors, the transfected cells were incubated in a solution containing a primary antibody against the extracellular portion of the receptors, whose binding was detected with an 125I-labeled antirabbit secondary antibody. As shown in Fig. 3B, there were no significant differences in the expression of YCG/R complexes and their corresponding wild-type receptors, signifying that any differences in the functional assays were not due to differences in receptor densities.!:, 百拇医药

    Specific binding results for each of the YCG/R complexes and their corresponding wild-type receptors are depicted in Fig. 3C. There was no apparent binding of [125I]hCG to cells expressing YCG/LHR, probably due to the presence of the fused ligand. Cells expressing TSHR and YCG/TSHR were able to bind [125I]bTSH at approximately the same level, whereas those expressing YCG/FSHR displayed reduced binding of [125I]hFSH compared with cells expressing FSHR.

    Yoked hCG-receptor complexes: characterization of single chain ligand receptor complexesih/s|w!, http://www.100md.com

    hLHR and YCG/LHR.ih/s|w!, http://www.100md.com

    Cells expressing YCG/LHR showed essentially no binding of 50 pM [125I]hCG due to the occupancy of binding sites by the attached hormone (Fig. 3C), as expected (13, 16). These cells, however, exhibited a high constitutive level of cAMP production that was not elevated upon addition of exogenous hormone (Fig. 4). Comparisons of the binding and signaling parameters of cells expressing hLHR and YCG/LHR cells are shown in Table 1.ih/s|w!, http://www.100md.com

    fig.ommitteedih/s|w!, http://www.100md.com

    Figure 4. Characterization of hLHR and YCG/LHR proteins. cAMP production in cells expressing hLHR and YCG/LHR in response to increasing concentrations of hCG. ED50 values for cells expressing hLHR are given in Table 1. Values could not be determined for cells expressing YCG/LHR because of the inability of exogenous hCG to bind YCG/LHR.ih/s|w!, http://www.100md.com

    hTSHR and YCG/TSHR.

    Competitive binding studies with [125I]bTSH and bTSH were conducted with cells expressing hTSHR and YCG/TSHR (Fig. 5A), and no differences were found in the IC50 values (Table 1). An approximately 13-fold increase in basal cAMP in cells expressing YCG/TSHR compared with that seen in cells expressing hTSHR was found. Interestingly, these cells were further stimulated upon addition of exogenous bTSH, and the same maximal cAMP production was observed as that obtained in cells expressing hTSHR (Fig. 5B). Moreover, there was no difference between the ED50 values for cells expressing hTSHR and YCG/TSHR (Table 1).&30r:k, http://www.100md.com

    fig.ommitteed&30r:k, http://www.100md.com

    Figure 5. Characterization of hTSHR and YCG/TSHR proteins. A, Competitive binding of cells expressing hTSHR and YCG/TSHR. [125I]bTSH (50 pM) with increasing concentrations of unlabeled bTSH was used to determine the binding affinity of exogenous bTSH to cells expressing hTSHR or YCG/TSHR. B, cAMP production of cells expressing hTSHR and YCG/TSHR upon stimulation with increasing amounts of bTSH; mock-transfected cells were used as a negative control. IC50 and ED50 values are given in Table 1.

    hFSHR and YCG/FSHR.${m, http://www.100md.com

    For competitive binding of cells expressing hFSHR and YCG/FSHR, 100 pM [125I]hFSH was used with increasing concentrations of unlabeled hFSH (Fig. 6A). Again, there was no difference in the IC50 values for hFSHR and YCG/FSHR (Table 1). Cells expressing YCG/FSHR exhibited an approximately 4-fold increase in basal cAMP production compared with that of hFSHR; moreover, as with cells expressing YCG/TSHR, those cells were further stimulated after the addition of exogenous hFSH, reaching a maximum cAMP level comparable to that of cells expressing hFSHR (Fig. 6B). No differences in the ED50 values were observed (Table 1).${m, http://www.100md.com

    fig.ommitteed${m, http://www.100md.com

    Figure 6. Characterization of hFSHR and YCG/FSHR proteins. A, Competitive binding of cells expressing hFSHR and YCG/FSHR. [125I]hFSH (200 pM) with increasing concentrations of unlabeled hFSH was used to determine the binding affinity of hFSH for each of the expressed proteins. B, Cells expressing hFSHR and YCG/FSHR were assayed for the production of cAMP after the addition of increasing concentrations of hFSH, with mock-transfected cells serving as a negative control. IC50 and ED50 values are listed in Table 1.

    hCG and hLHR, hTSHR, and hFSHR.$eg?hn, 百拇医药

    As YhCG was able to increase cAMP production in the noncognate receptors, hTSHR and hFSHR, experiments were performed to ascertain whether high concentrations of free hCG would also activate these receptors. Figure 7A shows a representative dose-response curve of cAMP production in hLHR-expressing cells at various concentrations of recombinant hCG. Interestingly, high doses of hCG can also lead to minimal activation of hTSHR and hFSHR (Fig. 7B). Although the responses were too limited to obtain ED50 and maximal cAMP values, these results document that the observed increase in basal cAMP in YCG/TSHR and YCG/FSHR is attributable to hCG and not to a nonspecific effect from fusing hCG to the N terminus of each noncognate receptor. Limited competitive binding studies were also conducted with recombinant hCG and [125I]bTSH plus hTSHR and [125I]hFSH plus hFSHR (data not shown). At about 300 µg/ml hCG, i.e. the highest dose used in Fig. 7B, competition with 50 pM [125I]bTSH and 100 pM [125I]hFSH and the cognate receptors reduced specific binding of the cognate hormone-receptor pairs by 30–40%.

    fig.ommitteed675a#-., 百拇医药

    Figure 7. Stimulation of hLHR, hTSHR, and hFSHR by hCG. A, cAMP production by cells expressing hLHR in response to different concentrations of hCG. B, cAMP production by cells expressing hTSHR and hFSHR in response to different concentrations of hCG (note the difference in scales of both axes relative to A). In all experiments purified recombinant hCG was used.675a#-., 百拇医药

    Discussion675a#-., 百拇医药

    The results reported herein demonstrate that there is indeed recognition of hCG by the three glycoprotein hormone receptors. In the yoked hormone-receptor system, hCG binds to and activates not only its cognate receptor, LHR, but also the noncognate receptors, TSHR and FSHR. These findings were confirmed using high concentrations of free, i.e. nonyoked, purified recombinant hCG. Both YCG/TSHR and YCG/FSHR can be further stimulated by the addition of exogenous cognate ligand, indicating that the interactions of hCG, even when fused with a noncognate receptor, were insufficient to prevent cognate hormone binding, as occurs in the homologous system, i.e. [125I]hCG binding to YCG/LHR. However, as all of the glycoprotein hormones share a common -subunit, and there is a high degree of sequence homology in the glycoprotein hormone ß-subunits and the glycoprotein hormone receptor ectodomains, some degree of cross-reactivity is evidently allowed within this hormone receptor family. An understanding of the differences and similarities between the abilities of these hormones to bind to and activate cognate and noncognate receptors will provide important information not only about the sequence determinants responsible for specificity, but also about common binding and receptor activation elements, e.g. and conserved ß contact sites to identical receptor domains. Such results will help guide the development of small molecule agonists and antagonists for these GPCRs.

    Figure 1B is arranged to depict the three glycoprotein hormone receptor ectodomains in the context of the model proposed by Bhowmick et al. (5) for that of rat LHR. In their homology model based on the leucine-rich repeat (LRR) structure of pancreatic ribonuclease (18), residues 27–235 of rat LHR were proposed to form nine LRRs as shown in Fig. 1B for the human receptors. The N- and C-terminal regions of the ectodomain do not exhibit an LRR structure, but it was suggested that a portion of the hinge region may adopt a chemokine-like fold (5). Identical amino acid residues in the ectodomains of the three glycoprotein hormone receptors are shown by open boxes, and conserved residues, e.g. hydrophobic, ionizable/identical charge (K/R and D/E), and hydroxylated (S/T), are indicated by shaded boxes.159]t-, 百拇医药

    We know from several studies that high affinity hCG binding to LHR occurs primarily in the region encoded by exons 1–8 (3, 19, 20). There is, of course, no assurance that the binding of TSH and FSH to their cognate receptor ectodomains will be comparable to that of hCG and the LHR ectodomain. Indeed, in a systematic comparison of the roles of the LRRs in the ectodomains of LHR and FSHR, Song et al. (21), focusing on LRR1–8, concluded that the Leu/Ile-x-Leu/Ile motifs of the ß-strands are important, but not equally. Using Ala-scanning mutagenesis of the Leu/Ile residues in the above motif, where there is only one difference between LHR and FSHR in the aligned sequences, they found that hCG binding was eliminated or greatly reduced with Ala replacements of either of the ß-strand Leu/Ile residues in all eight LRRs of FSHR, whereas LRR5 and -6 of LHR contributed less than the others. Thus, even identical amino acid residues in the three receptor ectodomains may have distinct functions in binding cognate ligands. The same group also showed that LRR4 of LHR contained some amino acid residues critical for hCG binding and others important in signaling (22). Whether such distinctions will hold for FSHR and TSHR remains to be determined.

    A careful examination of Fig. 1B shows that the regions encoded by exons 1 (most of the N-terminal Cys-rich region), 10, and a portion of 11 (most of the C-terminal Cys-rich region) are the most divergent in structure. The three ectodomains contain a highly conserved region encoded by exon 9, L(T/S)YPSHCCAFXN, and an identical region encoded by exon 11, FNPCEDIMGY. Both of these domains have been shown to influence hCG-mediated LHR activation, but not ligand binding (23, 24, 25, 26). Within the region of the LHR ectodomain believed to be responsible for high affinity ligand binding, the N-terminal Cys-rich region and that of LRR1 and -7 are the most variable and LRR2–6 and -8 exhibit about 55 ± 7% amino acid sequence identity/homology in the three receptors. It is tempting to assign some of the binding specificity to the hormone-specific regions of the ß-subunits making contacts with the N-terminal Cys-rich region, LRR1 and -7, and portions of LRR2–6 and -8, whereas the -subunit and the identical regions of the ß-subunits are more likely to have contact sites in the identical/conserved regions of LRR1–8. Those are only suggestions, but they provide a useful framework for data interpretation and to guide further experiments.

    A number of amino acid residues on the - and ß-subunits have been suggested as possible contact sites for cognate receptor binding (2). The seatbelt regions of the ß-subunits, identified in the crystal structures of hCG (7, 8) and hFSH (9), have been studied by several groups using chimeras of the ß-subunits (27, 28, 29, 30). The results suggest that the N-terminal portion of the seatbelt, i.e. the determinant loop (2), appears more important in hCG binding to LHR, whereas the C-terminal region of the seatbelt has more of an influence on FSH and TSH binding to their cognate receptors. It will be of great interest to determine the contact sites of the seatbelts, if any (29), with the specific regions of the receptors discussed above.cl!8v, http://www.100md.com

    It is interesting that when yoked to hTSHR and hFSHR, hCG is capable of activating the receptors with minimal, if any, interference with binding of the cognate ligand. This observation may indicate that yoked hCG is capable of partially activating the endodomain or may bind to and activate a region of the ectodomain independent of the site for cognate ligand binding. It is also possible that the low affinity of hCG for hTSHR and hFSHR is amplified in the yoked system, where hCG cannot diffuse far from the receptor ectodomain due to the constraint of the covalent linkage. As the reduced affinity of hCG to the noncognate receptors, relative to the cognate hormones, is probably manifested to some degree by the time of occupancy of the hormone on the receptor-binding site, one would expect a higher rate constant of dissociation of hCG from hTSHR and hFSHR than those of the cognate ligands. In the 30-min cAMP assay, any level of productive binding of yoked hCG to the noncognate receptors could lead to an increase in basal cAMP production. On the other hand, in a 6-h binding assay, the greater affinity of cognate ligand to the yoked hCG-receptor complex, e.g. expected Kd differences on the order of 104–105, would favor occupancy by the natural ligand rather than YhCG. If YhCG inhibits some of the binding of cognate ligand, it would probably reduce the apparent binding capacity and not the Kd, unless of course YhCG alters the conformation of the receptor-binding region on the ectodomain. The inherent difficulties of quantifying and comparing results from cognate ligand binding and receptor antibody binding prohibit sufficient accuracy to ascertain whether the apparent binding capacity values are altered.

    It is known that in cases of gestational hyperthyroidism hCG is able to bind to and activate TSHR (17), consistent with our YCG/TSHR findings. Comparative studies of the abilities of hCG and hLH to activate hTSHR expressed in CHO cells showed that both were functional at high concentrations, with hLH being more effective than hCG (31). The difference was attributable to the hCGß-CTP. In addition, Rodien et al. (32) showed that a mutation in TSHR, in which Lys183 is replaced with Arg, increases the receptor’s affinity for hCG significantly above that of wild-type hTSHR. Again, this finding supports our studies and exemplifies the importance of the slight differences in primary structure within these families to direct hormone binding and allow for receptor activation. To the best of our knowledge, there are no reported clinical cases in which hypersecretion of hCG activates FSHR, but our results suggest that this can occur.p'81n3u, http://www.100md.com

    Yoked hormones and yoked hormone-receptor systems have been used to study interactions within the families of glycoprotein hormones and the glycoprotein hormone receptors. We and others have created YhCG complexes in which the fusion protein has been found to be active in either orientation, N-hCGß--C (10, 11, 12) and N--hCGß-C (13, 14, 15). In addition to single chain TSH (33) and FSH (34) hormones, both of which are as active as the heterodimers, Boime’s group has created multifunctional fusion proteins (35, 36). In one such protein, the common -subunit was fused to FSHß and hCGß, creating a hybrid that could react with FSHR as well as LHR (35). These studies were then expanded to include a fusion protein that included all three glycoprotein hormone ß-subunits and a single -subunit to create a trifunctional protein. This protein was found to bind with high affinity to each of the glycoprotein hormone receptors and result in cAMP production (36).

    Although we cannot discount the possibility that the presence of hCG in the YCG/R complexes forces a constraint on receptor conformation that promotes an increase in basal cAMP levels, we do not believe that this happens, based on the documentation of hCG-TSHR interactions (17) as well as the absence of receptor activation when the -subunit, as a monomer or homodimer, was fused to hTSHR (37). In addition, recent studies from our group showed that the human -subunit, hCGß, CTP, or PRL, an unrelated protein, when fused to LHR, produced no increase in basal cAMP production (38). Given these data, we believe that the increases in basal cAMP levels that we have observed are caused by specific interactions of hCG with each of the glycoprotein hormone receptors. Of interest is the observation that YCG/LHR yields a high basal cAMP that is not increased by the addition of exogenous hormone. This mimics the functional response of many activating mutations of LHR and may reflect the possibility that the conformation of the partially activated receptor is somewhat different from that of the cognate (nonyoked) ligand-receptor complex (39). There is, unfortunately, no information available on the effects of yoking ligand to receptors (cognate and noncognate) on desensitization and internalization. In summary, the findings from this study have indicated an ability of hCG to bind to and activate its noncognate receptors, hTSHR and hFSHR.

    Acknowledgments4[wyc, http://www.100md.com

    We acknowledge all members of the laboratory for their helpful and insightful suggestions and support. We particularly thank Drs. Krassimira Angelova and Lisa Kelly for their careful review of this manuscript.4[wyc, http://www.100md.com

    Received August 8, 2002.4[wyc, http://www.100md.com

    Accepted for publication October 1, 2002.4[wyc, http://www.100md.com

    References4[wyc, http://www.100md.com

    Pierce JG, Parsons TF 1981 Glycoprotein hormones: structure and function. Annu Rev Biochem 50:465–4954[wyc, http://www.100md.com

    Hearn MTW, Gomme PT 2000 Molecular architecture and biorecognition process of the cysteine knot protein superfamily: part I. The glycoprotein hormones. J Mol Recognition 13:223–2784[wyc, http://www.100md.com

    Ascoli M, Fanelli F, Segaloff DL 2002 The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocr Rev 23:141–1744[wyc, http://www.100md.com

    Thompson JD, Higgins DG, Gibson TJ 1994 CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res 22:4673–4680

    Bhowmick N, Huang, J, Puett, D, Isaacs, NW, and Lapthorn, AJ 1996 Determination of residues important in hormone binding to the extracellular domain of the luteinizing hormone/chorionic gonadotropin receptor by site-directed mutagenesis and modeling. Mol Endocrinol 10:1147–1159$g9a8e, 百拇医药

    Kobe B, Kajava AV 2001 The leucine-rich repeat as a protein recognition motif. Curr Opin Struct Biol 11:725–732$g9a8e, 百拇医药

    Lapthorn AJ, Harris DC, Littlejohn A, Lustbader JW, Canfield RE, Machin KJ, Morgan FJ, Isaacs NW 1994 Crystal structure of human chorionic gonadotropin. Nature 369:455–461$g9a8e, 百拇医药

    Wu H, Lustbader JW, Liu Y, Canfield RE, Hendrickson WA 1994 Structure of human chorionic gonadotropin at 2.6 Å resolution from MAD analysis of the selenomethionyl protein. Structure 2:545–558$g9a8e, 百拇医药

    Fox KM, Dias JA, Van Roey P 2001 Three-dimensional structure of human follicle-stimulating hormone. Mol Endocrinol 15:378–389$g9a8e, 百拇医药

    Narayan P, Wu C, Puett D 1995 Functional expression of yoked human chorionic gonadotropin in baculovirus-infected insect cells. Mol Endocrinol 9:1720–1726

    Sugahara T, Pixley MR, Minami S, Perlas E, Ben-Menahem D, Hsueh AJ, Boime I 1995 Biosynthesis of a biologically active single peptide chain containing the human common and chorionic gonadotropin ß subunits in tandem. Proc Natl Acad Sci USA 92:2041–2045w3, http://www.100md.com

    Heikoop JC, Van Beuningen-De VMM, Van Den Boogaart P, Grootenhusi PDJ 1997 Evaluation of subunit truncation and the nature of the spacer for single chain human gonadotropins. Eur J Biochem 245:656–662w3, http://www.100md.com

    Narayan P, Gray J, Puett D 2000 A biologically active single chain human chorionic gonadotropin analog with altered receptor binding properties. Endocrinology 141:67–71w3, http://www.100md.com

    Sen Gupta C, Dighe RR 2000 Biological activity of single chain choriogonadotropin, hCGß is decreased upon deletion of five carboxyl terminal amino acids of the subunit without affecting its receptor binding. J Mol Endocrinol 24:157–164w3, http://www.100md.com

    Ben-Menahem D, Jablonka-Shariff A, Hyde RK, Pixley MR, Srivastava S, Berger P, Boime I 2001 The position of the and ß subunits in a single chain variant of human chorionic gondotropin affects the heterodimeric interaction of the subunits and receptor-binding epitopes. J Biol Chem 276:29871–29879

    Wu C, Narayan P, Puett D 1996 Protein engineering of a novel constitutively active hormone-receptor complex. J Biol Chem 271:31638–31642d2, 百拇医药

    Burrow G 1993 Thyroid function and hyperfunction during gestation. Endocr Rev 14:194–201d2, 百拇医药

    Kobe B, Deisenhofer J 1993 Crystal structure of porcine ribonuclease inhibitor, a protein with leucine-rich repeats. Nature 366:751–756d2, 百拇医药

    Thomas D, Rozell TG, Segaloff DL 1996 Mutational analyses of the extracellular domain of the full-length lutropin/choriogonadotropin receptor suggest leucine-rich repeats 1–6 are involved in hormone binding. Mol Endocrinol 10:760–768d2, 百拇医药

    Hong S, Phang T, Ji I, Ji TH 1998 The amino-terminal region of the luteinizing hormone/choriogonadotropin receptor contacts both subunits of human choriogonadotropin. I. Mutational analysis. J Biol Chem 273:13835–13840d2, 百拇医药

    Song YS, Ji I, Beauchamp J, Isaacs NW, Ji TH 2001 Hormone interactions to Leu-rich repeats in gonadotropin receptors. I. Analysis of Leu-rich repeats of human luteinizing hormone/chorionic gonadotropin receptor and follicle-stimulating hormone receptor. J Biol Chem 276:3426–3435

    Song YS, Ji I, Beauchamp J, Isaacs NW, Ji TH 2001 Hormone interactions to Leu-rich repeats in the gondotropin receptors. II. Analysis of Leu-rich repeat 4 of human luteinizing hormone/chorionic gonadotropin receptor. J Biol Chem 276:3436–3442#h3sh, http://www.100md.com

    Huang J, Puett D 1995 Identification of two amino acid residues on the extracellular domain of the lutropin/choriogonadotropin receptor important in signaling. J Biol Chem 270:30023–30028#h3sh, http://www.100md.com

    Alvarez CA, Narayan P, Huang J, Puett D 1999 Characterization of a region of the lutropin receptor extracellular domain near transmembrane helix 1 that is important in ligand-mediated signaling. Endocrinology 140:1775–1782#h3sh, http://www.100md.com

    Nakabayashi K, Kudo M, Kobilka B, Hsueh AJW 2000 Activation of the luteinizing hormone receptor following substitution of Ser-277 with selective hydrophobic residues in the ectodomain hinge region. J Biol Chem 275:30264–30271#h3sh, http://www.100md.com

    Zeng H, Phang T, Song YS, Ji I, Ji TH 2001 The role of the hinge region of the luteinizing hormone receptor in hormone interaction and signal generation. J Biol Chem 276:3451–3458

    Campbell RK, Dean-Emig DM, Moyle WR 1991 Conversion of human choriogonadotropin into a follitropin by protein engineering. Proc Natl Acad Sci USA 88:760–764ee[], 百拇医药

    Dias JA, Zhang Y, Liu X 1994 Receptor binding and functional properties of chimeric human follitropin perpared by an exchange between a small hydrophilic intercystine loop of human follitropin and human lutropin. J Biol Chem 269:25289–25294ee[], 百拇医药

    Han Y, Bernard MP, Moyle WR 1996 hCGß residues 94–96 alter LH activity without appearing to make key contacts [erratum Mol Cell Endocrinol 129:237–239, 1997]. Mol Cell Endocrinol 124:151–161ee[], 百拇医药

    Grossmann M, Szkudlinski MW, Wong R, Dias JA, Ji TH, Weintraub BD 1997 Substitution of the seat-belt region of the thyroid-stimulating hormone (TSH) ß-subunit with the corresponding regions of choriogonadotropin or follitropin confers luteotropic but not follitropic activity to chimeric TSH. J Biol Chem 272:15532–15540ee[], 百拇医药

    Yoshimura M, Hershman JM, Pang X-P, Berg L, Pekary AE 1993 Activation of the thyrotropin (TSH) receptor by human chorionic gonadotropin and luteinizing hormone in Chinese hamster ovary cells expressing functional human TSH receptors. J Clin Endocrinol Metab 77:1009–1013

    Rodien P, Bremont C, Sanson ML, Parma J, Van Sande J, Costagliola S, Luton JP, Vassart G, Duprez L 1998 Familial gestational hyperthyroidism caused by a mutant thyrotropin receptor hypersensitive to human chorionic gonadotropin. N Engl J Med 339:1823–1826r\q?fsf, http://www.100md.com

    Fares FA, Yamabe S, Ben-Menahem D, Pixley M, Hsueh AJ, Boime I 1998 Conversion of thyrotropin heterodimer to a biologically active single-chain. Endocrinology 139:2459–2464r\q?fsf, http://www.100md.com

    Sugahara T, Sato A, Kudo M, Ben-Menahem D, Pixley MR, Hsueh AJ, Boime I 1996 Expression of biologically active fusion genes encoding the common subunit and the follicle-stimulating hormone ß subunit. Role of a linker sequence. J Biol Chem 271:10445–10448r\q?fsf, http://www.100md.com

    Kanda M, Jablonka-Shariff A, Sato A, Pixley M, Bos E, Hiro’oka T, Ben-Menahem D, Boime I 1999 Genetic fusion of an -subunit gene to the follicle-stimulating hormone and chorionic gonadotropin-ß subunit genes: production of a bifunctional protein. Mol Endocrinol 13:1873–1881r\q?fsf, http://www.100md.com

    Garcia-Campayo V, Kumar TK, Boime I 2002 Thyrotropin, follitropin, and chorionic gonadotropin expressed as a single multifunctional unit reveal remarkable permissiveness in receptor-ligand interactions Endocrinology 143:3773–3778r\q?fsf, http://www.100md.com

    Angelova K, Jain R, Puett D, Szkudlinski MW, Narayan P, Fusion of the common glycoprotein hormone -subunit to the TSH receptor results in partial antagonism of TSH activation. Program of the 83rd Annual Meeting of The Endocrine Society, Denver, CO, 2001, p 265r\q?fsf, http://www.100md.com

    Narayan P, Gray J, Puett D 2002 Yoked complexes of human choriogonadotropin and the lutropin receptor: evidence that monomeric individual subunits are inactive. Mol Endocrinol 16:2733–2745r\q?fsf, http://www.100md.com

    Angelova K, Fanelli F, Puett D 2002 A model for constitutive lutropin receptor activation based on molecular simulation and engineered mutations in transmembrane helices 6 and 7. J Biol Chem 277:32202–32213(Rebecca L. Schubert Prema Narayan and David Puett)