Abstract
A pharmacoperone (from “pharmacological chaperone”) is a small molecule that enters cells and serves as molecular scaffolding in order to cause otherwise-misfolded mutant proteins to fold and route correctly within the cell. Pharmacoperones have broad therapeutic applicability since a large number of diseases have their genesis in the misfolding of proteins and resultant misrouting within the cell. Misrouting may result in loss-of-function and, potentially, the accumulation of defective mutants in cellular compartments. Most known pharmacoperones were initially derived from receptor antagonist screens and, for this reason, present a complex pharmacology, although these are highly target specific. In this summary, we describe efforts to produce high throughput screens that identify these molecules from chemical libraries as well as a mouse model which provides proof-of-principle for in vivo protein rescue using existing pharmacoperones.
Keywords: pharmacoperone, protein trafficking, protein rescue, animal models, high throughput screens, therapeutic approaches
1. Introduction
For more than 20 years, there has been interest in gene therapy as a means to correct mutational disease. Issues related to the integration of therapeutic DNA into the genome, immune responses, technical problems with vectors (toxicity, immune, inflammatory responses, gene control and targeting issues), chances of inducing tumors, (insertional mutagenesis) and other problems, have made it challenging to reduce this approach to practice, however.
Correcting the folding of misfolded protein mutants and restoring them to function (with pharmacoperone drugs) is a potential alternative to inserting correctly folding proteins by gene therapy (Figure 1). This approach is generally referred to as “rescuing” the protein. It is likely that valuable drugs reside in chemical libraries, yet have been missed, since screening approaches that rely on identification of agonists and antagonists may have failed to identify existing pharmacoperones. There are several advantages to using pharmacoperone drugs; among these advantages are the ability to restore misfolded proteins to function without leaving residual non-functional proteins in other cellular compartments.
Fig. 1. The canonical pathway of protein translation from mRNA.

emphasizing that misfolded proteins are retained in the cell (ER and elsewhere) and can be rescued by target specific pharmacoperone drugs (red arrow) that correct misfolding and restore function. Modified from (24) and reprinted with permission of the American Society for Pharmacology and Experimental Therapeutics.
Protein rescue with agents that correct folding errors potentially applies to a diverse and vast array of human diseases that result from misfolding or instability of receptors, ion channels, enzymes and other proteins. These include cystic fibrosis (1-5), hypogonadotropic hypogonadism (HH) (6, 7), nephrogenic diabetes insipidus (8-10), retinitis pigmentosa (11), hypercholesterolemia (12), cataracts (13), neurodegenerative diseases (Alzheimer's, Huntington's and Parkinson's, (14-18)), cancer (19), α1 trypsin deficiency and lysosomal storage disease (20, 21), mucopolysaccharidosis type IIIC (22) and many others. One could envision drugs given in a prophylactic manner (in vitamins, for example) that prevent the misfolding that leads to neurodegenerative disorders (Alzheimer's (misfolded amyloid), (23)) Parkinson's (misfolded α-synuclein) and cataracts (misfolded lens crystalline). In this regard, diseases may be prevented before clinical signs present. In the case of certain proteins (e.g. the GnRHR, V2R and rhodopsin), this approach has succeeded with a striking number of different mutants (24) supporting the view that pharmacoperones will become powerful weapons in our therapeutic arsenal.
A large number of proteins of different classes and structures that have been rescued and restored to function in cell cultures is also suggestive of the broad applicability of this approach (25-39).
Our efforts have focused on G protein coupled receptors (GPCRs), which include the vasopressin type 2 receptor (V2R) and gonadotropin releasing hormone receptor (GnRHR). GPCRs comprise the largest family of validated drug targets — 35-50% of approved drugs derive their benefits by selectively targeting this family.
2. Mechanism of Pharmacoperone Action
GPCRs are subjected to a stringent quality control system (QCS) in the endoplasmic reticulum (ER); this system consists of both, protein chaperones that retain misfolded proteins and enzyme-like proteins that catalyze the folding process. The QCS (consisting of endogenous chaperone proteins and other factors) assesses structure but not function and insures that only correctly folded proteins enter the pathway leading to the plasma membrane (PM). Because of this, point mutations may result in the production of misfolded and disease-causing proteins that are unable to reach their functional destinations in the cell because they are retained by the QCS even though they may retain (or regain) function.
The functional rescue of misfolded mutant receptors by small non-peptide molecules, originally screened from libraries to serve as receptor antagonists, has now been demonstrated (24). A pharmacoperone is a “molecular scaffold” to promote correct folding of otherwise misfolded mutant proteins within the cell (6, 40). Misfolded proteins are frequently retained by the cellular quality control system (QCS) of the endoplasmic reticulum (ER), do not reach their normal site of function (9, 41) and may result in disease (42). Pharmacoperones can rescue misfolded receptor mutants and restore them to function, which is a potentially useful therapeutic approach when the target is a misfolded/misrouted protein (Figure 1).
We have summarized the literature for the gonadotropin releasing hormone (GnRH) and vasopressin type 2 (V2) receptor's pharmacoperones with a view toward moving these compounds in vivo (24). Science writers commenting on this approach (43, 44) have observed that rescue with pharmacoperones is a viable “alternative (to gene therapy)” since it serves as a means of “skirting gene therapy to correct genetic defects.” This view is supported by the consideration that correction of defective protein folding appears significantly less challenging than replacement of a defective gene (or gene product) by a perfect one. The QCS is not protein-specific; it recognizes general aspects of misfolding (e.g. exposure of hydrophobic plates in aqueous environments), frequently with relatively low affinity. Accordingly, GPCRs that retain ligand binding and effector coupling, but are recognized as misfolded by such general criteria, are often retained in the ER and degraded. Their rescue with pharmacoperones leads to proper folding, passage through the QCS, restoration to the proper site and return of function.
2.1 There are a number of principles that have guided our efforts
2.1.1 Most pharmacoperone drugs, used to date, were identified from hits in high throughput screens (HTS) for competitive receptor antagonists and then repurposed. These must be removed after rescue to preclude competition with agonists to avoid the complex pharmacology of trying to rescue function in the presence of its antagonist. This potential therapeutic problem can be addressed by identification of drug candidates in the proposed assays that are pharmacoperones but lack antagonistic activity. We believe that valuable drugs, which affect the trafficking of GPCRs, may have been overlooked because of this limitation (45, 46).
2.1.2 It has not been established that binding at or near the binding site of the natural ligand is a necessary pre-requisite for pharmacoperone activity and there is information to suggest otherwise (47-49). This would, in fact, be an unexpected requirement since one could imagine pharmacoperones that might stabilize the correctly routed form of the receptor and not show any antagonism (or agonism). Accordingly, identification of non-antagonistic (i.e. allosteric) pharmacoperones is a reasonable and therapeutically important goal.
2.1.3 Pharmacoperone drugs need not be present at the time of protein synthesis, but can rescue ER-retained proteins that have already accumulated (Figure 1) (50). This observation increases the therapeutic usefulness, since misfolded mutants need not be (first) degraded and then replaced by newly synthesized protein (i.e. the portion synthesized in the presence of pharmacoperone).
2.1.4 While pharmacoperones are specific for individual protein mutants, those that rescue one mutant of an individual protein typically rescue most mutants of the same protein, likely by stabilizing a core microdomain that makes the protein acceptable to the quality control system of the cell. This observation also improves the therapeutic reach of these drugs (24, 28, 51) since each mutant of an individual protein will not require a different drug.
3. Physiological Significance of the Targets Selected
3.1 GnRHR
The GnRHR resides in the gonadotrope cells of the pituitary and is responsible for producing responses to hypothalamic GnRH, such as the releasing of the gonadotropins, luteinizing hormone and follicle stimulating hormone. The hGnRHR has been a central focus of drug development and understanding the mechanism of GnRH action has already led to useful drugs (agonists and antagonists) for the treatment of disorders of reproduction and for cancer. When the function of this receptor is lost due to mutation, the disease hypogonadotropic hypogonadism results. Because this target is causally and mechanistically associated with pathophysiological responses, the proposed intervention with pharmacoperone drugs results in valid therapeutic approaches.
3.2 V2R
The V2 receptor is expressed in the distal convoluted tubule and the collecting ducts of the kidney. V2R responds to vasopressin by stimulating mechanisms that concentrate the urine and maintain water homeostasis in the organism. When the function of V2R is lost due to mutation, the disease nephrogenic diabetes insipidus results (8, 24). The current goal of treatment is to control the body's fluid levels and problems with electrolyte imbalances. Patients must drink large quantities of water to offset the loss. If the affected person does not drink enough fluids, high urine output may cause dehydration and high serum osmolarity. NDI is caused by an inborn error of metabolism that is present at birth; it is a chronic condition requiring lifelong treatment. Complications can include dilation of the ureters and bladder, hypernatremia, severe dehydration and shock. The importance of such non-antagonistic pharmacoperones was shown in a study of patients with X-linked NDI. Mutant vasopressin 2 receptors in NDI resulted in misrouted proteins that were trapped in the ER, degraded and did not reach the plasma membrane in the collecting ducts of the kidney where they would normally reabsorb water. In vitro studies indicated that a non-peptide V1a receptor antagonist rescued cell surface expression and function of mutant V2 receptors. When applied in vivo, a short-term treatment with a V1a receptor antagonist showed that patients given this molecule decreased both 24-h urine volume and water intake. Maximum increase in urine osmolality was observed on day 3 and sodium, potassium, creatinine excretions and plasma sodium were constant throughout the study (10). Unfortunately, the trade-off between antagonism and pharmacoperone activity resulted in complex pharmacology that did not allow full exploitation of pharmacoperone activity.
We have focused on two elements which we believe would be valuable for moving this field forward:
Reliable high throughput screens for identification of pharmacoperone drugs.
Animal models that enable optimization of rescue conditions with a view toward human use.
4. High throughput screening assays
4.1 Principle of the HTS
We first established an assay for pharmacoperones of the V2R. The primary assay uses a HeLa cell line constitutively expressing hV2R[L83Q] (the V2 mutant assay) expressed under the control of a tetracycline-regulated (TET-off) transactivator (52-54). In the absence of doxycycline (“Dox”) (stable analog of tetracycline), the mutant is expressed; it is then misrouted and retained in the ER. Following pretreatment with pharmacoperone, the mutant is rescued and trafficked to the plasma membrane. The rescued mutant is then responsive to native vasopressin and coupled to the production of cAMP for the mutant assay. In the HTS, the level of functional hV2R (mutant) present in each test well is quantitated using a luminescent-based cyclic adenosine monophosphate (cAMP) assay (cAMP-Glo, Promega). This allows the screen to identify compounds which increase the trafficking of mutant in our model system. To triage assay artifacts and compounds with intrinsic off-target activity, compounds are then counter screened with the same cell line as the primary assay in the presence of doxycycline, which shuts off the mutant expression. To eliminate artifacts resulting from the luminescence assay, an orthogonal radiolabeled cAMP RIA or IP assay is used. Compounds are profiled for nonspecific cytotoxicity and specificity for hV2R over other GPCR systems. Increase of total hV2R at the cell surface are confirmed using radioligand binding and fluorescently tagged receptor localization studies. All validated hits are screened for any direct hV2R agonist or antagonist activity and profiled for specificity using a panel of other cellular receptors.
In the hV2R[L83Q] assay, cells are challenged with an EC100 dose of vasopressin, then lysed and cAMP levels determined. The counter screen protocol is identical to the primary HTS assay protocol with the exception that the hV2R[L83Q] cells are incubated in the presence of doxycycline for 36 hours prior to assay. During this time the gene for the mutant is off and no measurable mutant remains in the cell. Accordingly the hV2R[L83Q] primary assay and counter screen protocols confirm the expected pharmacology of positive and negative controls (Figure 2).
Fig. 2. Responses of controls in the 384-well formatted hV2R[L83Q] Pharmacoperone Assay.

(a) Vasopressin titrations. In the absence of doxycycline (“Dox”), hV2R[L83Q] is synthesized and retained in the ER; a vasopressin challenge yields no cAMP response (triangles). Following pre-treatment with 100 nM of the pharmacoperone SR121463 (“SR”), hV2R[L83Q] is rescued and trafficked to the plasma membrane, and a robust cAMP response to vasopressin challenge is observed (circles). Also included are the results of challenging the cells with vasopressin after preincubation with SR and doxycycline (squares). (b) Pharmacoperone titrations. In the absence of Dox, increasing concentrations of pharmacoperone SR121463 increases the amount of hV2R[L83Q] on the surface of the cells, with the result of increasing cAMP responses after challenge with an EC100 of vasopressin (circles). An EC100 challenge of vasopressin to cells in the absence of doxycycline (squares) or pretreated with doxycycline (triangles) results in no appreciable cAMP response. Each dataset is normalized to either the 100% response of either VP (left) or SR (right) dose-response curve. Reprinted from (54) with permission of the publisher.
The SR121463 (originally developed as a receptor antagonist by Sanofi-Aventis Recherche & Développement, obtained with thanks to Dr. Claudine Serradeil) can be used as a pharmacoperone for the V2R mutant. It yields an EC50 of 1.97 ± 0.25 nM in the “-Dox” assay, and signal-to-background ratio (S/B) of ∼7 (n=4). Both protocols are high throughput compatible in both 384 and 1,536 well formats.
In the case of the hGnRHR mutants, the Merck compound IN3 (6, 28, 50) is an effective and target specific pharmacoperone; it was also initially developed as a GhRHR antagonist.
4.2 LOPAC (Library of Pharmacologically Active Compounds) pilot screen
To determine the performance of our optimized hV2R[L83Q] pharmacoperone assay in terms of robustness (Z′) under HTS conditions, we used the assay to screen the Sigma LOPAC (Library of Pharmacologically Active Compounds). Briefly, compounds were analyzed at a single concentration of 6 μM (0.4% DMSO) using SR121463 as a positive control. Each plate contained high and low signal control wells, which were used in Z′ factor calculations. An activity scatterplot of all compounds tested, as well as positive and negative controls, is shown in Figure 3.
Fig. 3. Scatter plot analysis of the high-throughput LOPAC pilot screen.

(a) All data from all assay plates (n=17, triplicate results), including controls, are displayed. Large separation in activity (Z′ =0.85±0.05, S/B=7.3±0.6) between wells dosed with SR121463 (“high control”) and DMSO (“low control”), as well as a broad distribution of hits indicate an excellent HTS assay. (b) Representative graph of compound activity correlating plate replicas. The best fit line has an r2=0.86, indicative of the high fidelity of the assay's hit identification. Reprinted from (54) with permission of the publisher.
As indicated from the positive and negative control scatterplots (Figure 3a), the assay demonstrated a high Z′ factor (0.85 ± 0.05) for the entire LOPAC screen, indicative of an excellent assay window. Day-to-day reproducibility of the assay is also excellent. As indicated in Figure 3b, a scatterplot of replicate measurements yields an r2= 0.86, indicating high reproducibility in hit identification. It is important to note that a high number of hits are expected from LOPAC screens due to the high proportion of pharmacologically active compounds present in the LOPAC itself. Using nominal hit-cutoffs (55) a hit rate of 0.78% was calculated. Based on results from previous cell-based LOPAC pilot screens, we anticipate an actual HTS hit rate <1%. The results presented above show a robust and reproducible HTS compatible assay, (S/B∼6 and Z′∼0.8).
Another important parameter to assess for HTS readiness is the susceptibility of an assay to artifact. For example, in the case of the hV2R[L83Q] pharmacoperone assay, compounds that non-specifically activate cyclase activity, agonists of endogenous GPCRs, cytotoxic compounds, and compounds that interfere with the luciferase-based detection reagents (via biochemical or optical means) may be falsely identified as hits. To assess the pharmacology of hits, as well as the contribution of artifact to the primary assay readout, we cherry-picked and titrated all hits from the LOPAC screen. Compounds were tested starting from 25 μM with 3-fold series dilution steps for 10 points (n=3) in both the primary hV2R[L83Q] pharmacoperone assay as well as its counter screen (i.e. pretreatment of the same cells with doxycycline 36 hours prior to assay). All hits confirmed activity in the titration assay, albeit with varying degrees of potency; a subset of the selected hits are shown in Table 1.
Table 1.
Primary assay and counter screen results for three hits from the LOPAC screen. For EC50 determination compounds were titrated starting from 25 μM with 3 fold series dilution steps for 10 points (n=3).
| Structure | Vendor information | LOPAC Screen Average % Response +/− Std Dev | EC50 (μM) −Dox | EC50 (μM) +Dox | Name | Selectivity | Dose Response Curves Triangles = −Dox Circles = +Dox |
|---|---|---|---|---|---|---|---|
|
Sigma Aldrich F6886 | 104+/−0 | 0.43 | 0.59 | Forskolin | Activates adenylate cyclase |
|
|
Sigma Aldrich L2540 | 74+/−2 | 8.30 | >25 | L-368,899 | Non-peptide oxytocin receptor antagonist |
|
|
Sigma Aldrich M6545 | 51+/−1 | >25 | >25 | Mitoxantrone | DNA synthesis inhibitor (cytotoxic) |
|
As expected, the counter screen was effective at identifying false positives such as forskolin, as well as cytotoxic compounds (such as Mitoxantrone). Some compounds showed activity only in the primary screen, such as L-368,899, an oxytocin receptor agonist. Given that the oxytocin receptor shares homology with the vasopressin receptor, this compound may be useful for further characterization as a V2R pharmacoperone. Combined, these results demonstrate the ability of the hV2R[L83Q] pharmacoperone assay to identify potent hits, and the usefulness of the counter screen to triage off-target hits.
5. SDDL (Scripps Drug Discovery Library) compounds
SDDL compounds are selected based on scaffold novelty, physical properties and spatial connectivity. A summary of select SDDL properties is shown in Figure 4.
Fig. 4. Distribution plots of SDDL properties.

Reprinted from (54) with permission of the publisher.
By design, the diversity of the SDDL mimics that of much larger collections found at major pharmaceutical companies, yet is responsive to lessons learned from successful drug discovery efforts and emerging trends in HTS library construction (56-60). The SDDL continues to be augmented with diverse small molecule scaffolds, as well as focused sub-libraries targeted to popular drug-discovery targets and compound collections provided by Scripps' distinguished chemistry faculty.
In its current state, the SDDL has several focused sub-libraries for screening popular drug-discovery target classes (e.g. kinases/transferases, GPCRs, ion channels, nuclear receptors, hydrolases, transporters), as well as diverse chemistries (e.g. click-chemistry, PAINS-free, Fsp3 enriched, and natural product collections) and physical properties (“rule-of-five,” “rule-of-three,” polar surface area, etc.) (60-65).
The novel screening approach presented here will provide a basis for other researchers interested in identifying small molecules that regulate the trafficking and folding of proteins. Regulation of these processes by pharmacoperones is not unique to GPCRs but pharmacoperones can be used to restore the functional expression of enzymes and ion channels as well (41, 42). Therapeutics based on this approach will become increasingly important in the coming years.
An analogous assay for pharmacoperones of the GnRHR is in development.
6. An In Vivo Proof-of-Principle for Pharmacoperone Action in Mice
We recently reported (66) the efficacy of pharmacoperone drugs in a newly developed mouse model expressing mutant GnRHR[E90K] in the pituitary and showing the hallmarks of HH. This disease is associated with reproductive failure in both males and females.
Because we are using receptor antagonist drugs that are repurposed as pharmacoperones, the rescue of all parameters is not 100% complete. Also, the drug must be given in a pulsatile fashion so that, after rescue of the receptor, it is washed out to enable occupancy by the endogenous ligand.
We use these drugs because non-antagonistic pharmacoperones are currently unavailable for HH (or other diseases, with modest exceptions, (47)). Our data show that available drugs will likely be useful to alleviate considerable suffering associated with diseases of misfolding. Pharmacoperone “IN3” (and other pharmacoperones that we use) have already been subjected to off-target and toxicology studies in humans and we are not assessing their toxicity in the mouse.
6.1 We chose GnRHR[E90K] as a model mutant since
6.1.1 The physiology of the GnRHR is well-characterized in humans, mice and many other animal models (67, 68); the mutant GnRHR[E90K] acts similarly in mice and humans (28, 69).
6.1.2 A great deal of information is also available regarding the cellular mechanism of action of the GnRHR (70). In addition, we have available, substantive information on the mechanism of misfolding (67, 68), mutant interactions with pharmacoperones (24, 71) and the molecular basis of the dominant-negative effect (50). This effect occurs when a misfolded mutant is retained by the ER and, because this receptor (normally) oligomerizes, it also retains WT receptor. This enhances the deleterious impact of the mutant and likely increases the severity in patients.
6.1.3 The mutant GnRHR[E90K] has, in cell cultures, already established general principles that are applicable to other diseases and misfolded molecules. Some examples are (a) The rescue of misfolded mutants by pharmacoperones, discovered in this system, appears to be generalizable to many diseases of misfolding (42). (b) The dominant-negative effect, which we first reported (72) to be based on ER-retention of WT protein by misrouted mutants, has emerged as an important feature of the regulation of other WT-mutant hetero-oligomers to the plasma membrane. For many GPCRs that oligomerize in order to traffic to the plasma membrane, a misfolded mutant also retains the WT molecule. Some examples are the delta-opiate receptor (73), histamine receptor (74), GHRH receptor (75), MC1 receptor, (76-78), TSH receptor (79), gonadotropin receptors (80-82) rhodopsin (83) and others (84, 85). (c) Constitutive oligomerization in the ER, first shown in this system (86), has also been demonstrated for receptors of melatonin (87), dopamine D2 (88), vasopressin (89), serotonin (90), δ-opioids (91), β2-adrenergics (92-94), GABAB (95-97), and follicle-stimulating hormone (98). (d) The observation that individual pharmacoperones rescue most mutants, noted above, (28) is generalizable, also applying to the V2R (99) and other receptors. (e) The observation that pharmacoperones rescue previously retained mutants (50) as well as newly synthesized ones appears generalizable to the V1 and V3 receptors (100). (f) We have shown that GFP and HA-tagging have a problematic impact on data interpretation (86), was subsequently observed by others as a concern (101, 102). It is reasonable and likely that we will continue to learn generalizable aspects applicable to rescue of other molecules from examination of this system.
- 6.1.4 Biochemical, Evolutionary and other Pragmatic Reasons for Mutation E90K selection: At the time we began design of this animal, sixteen mutations (N10K, T32I, E90K, Q106R, A129D, R139H, S168R, A171T, C200Y, S217R, R262Q, L266R, C279Y, Y284C, L314X and a splice junction mutation at the intron 1-exon 2 boundary) had been identified from patients with HH. Our selection was based on the following considerations:
- 6.1.4.1 In cell cultures mutant mouse GnRHR[E90K] is fully retained in the ER (72), identically as in the case of its human counterpart. This is important since the mouse orthologs of many misrouted human GnRHR mutants are correctly routed because trafficking of the mouse GnRHR (and its mutants) is less stringently controlled (69). S168R and S217R did not rescue with any class of pharmacoperone (46, 49, 103) so these were eliminated. We now know that this failure to rescue results from a substantial thermodynamic change in the transmembrane component of these mutants (68). We participated in the development of an animal expressing L117P by mutagenesis. This mutation has not been reported in human disease; subsequent to development of this animal, we learned (103) that it also cannot be rescued by pharmacoperones (due to the (covalent) right angle bend caused by the presence of proline) and it would have been excluded for this reason as well. Accordingly, this mutation could not be used in these studies.
- 6.1.4.2 We wanted mutants that rescued well with pharmacoperones. R139H, E90K, C279Y rescue best (i.e. high ratio of rescue (using pharmacoperone IN3, chemical structure below, section 6) to basal expression in cell culture models (28)).
-
6.1.4.3 We also wanted a mutation at a highly conserved site so that the mouse would have a high chance of being an effective model for the human.E90K met all our criteria and is found in human HH disease and we know a great deal about its behavior in COS cells (104).
6.1.5 E90K has a known biochemical mechanism of disruption of the receptor. The E90-K121 bridge is a highly conserved feature essential for trafficking of the GnRHR to the PM. This bridge establishes a physical interaction between transmembrane segment (TMS) 2 and TMS3. Because this salt bridge appears to be a fundamental requirement for correct routing, E90K results in a routing defect in both mouse and human GnRHR (6, 69, 105). In this mutant the E90-K121 salt bridge is broken (i.e. the mutant, K90-K121 opposes two positive charges and the bridge cannot form) (6, 106, 107). The strength of this model is increased by knowledge of the biochemical mechanism of action of the mutation (71, 107).
6.2 Development and genetic characterization of the model animal
We have published the technique for creating these animals (108). Briefly, GnrhrE90K “knock-in” mice (K90neo) were generated by homologous recombination in C57BL/6; 129SvEvTac hybrid RJ2.2 mouse embryonic stem (ES) cells (109). Previously, we removed neo and characterized the E90K phenotype (108). E90K/E90K males have slightly smaller testes compared to controls but are fertile. E90K/E90K females generate antral follicles but do not ovulate (108). Interestingly, when neo was left in the locus, the HH phenotype was more severe, making it a better mouse model for pharmacoperone trials.
6.3 Phenotypic characterization of the model animal
Circulating levels of gonadotropin and sex steroids were decreased in the homozygous mutant, compared with WT. Histological analysis (Fig. 5) of the ovaries of adult homozygous females showed absence of large antral follicles and corpora lutea. Mutant follicles were arrested at the early antral stage with only a few layers of granulosa cells and early antrum formation visible in about 30% of the follicles. Affected males showed small testes (Fig. 6).
Fig. 5. Phenotype of K90neo heterozygous and homozygous female mice.

Ovaries from mice of the indicated genotypes were collected at 90 days of age, imaged with a stereomicroscope and processed for histology (H&E staining). Wild-type (a) and heterozygotes (K90neo/+) (b) ovaries were indistinguishable. Both exhibited similar size, presence of follicles at all stages of development and presence of corpora lutea. The ovaries of K90neo/K90neo females (c) were smaller and exhibited no follicular development past the secondary follicle stage. The stereomicroscope image is shown on the left (scale bars= 0.5 mm) and the H&E stained sections are on the right (scale bars = 0.2 mm). Reprinted from (66) with permission of the publisher.
Fig. 6. Male phenotype.

Testes from the indicated genotypes were collected at 90 days of age, imaged with a stereomicroscope and stained. Wild-type (a) and K90neo/+ (b) testes were indistinguishable in size, appearance, abundance of Leydig cells and presence of spermatozoa in the seminiferous tubules. The testes of K90neo/K90neo males were smaller and exhibited varying degrees of hypogonadism. There were few to no Leydig cells or elongated spermatids (c). Treatment of K90neo/K90neo males with pharmacoperone IN3 (d) resulted in increased testis size and restored spermatogenesis. The stereomicroscopic image is shown in the left column (bar = 2 mm) and the H&E stained sections are shown on the middle and right (scale bars =0.1 mm (testis). Reprinted from (66) with permission of the publisher.
6.4 Surgical methods
A catheter was surgically inserted in the left carotid of the animal and the animal was attached to an infusion system in a specially designed cage, as shown (Fig. 7).
Fig. 7. Mouse in infusion tether.

A catheter was placed in the left carotid of a 60 day old male animal that was outfitted with a jacket and placed in a cage that allowed free-ranging while drug or saline containing heparin was infused. Pencil-drawn image made available by Instech and reprinted with permission.
6.5 Infusion and the pharmacology of drug administration
Currently available pharmacoperones for this system are repurposed (peptidomimetic) GnRHR antagonists. The molecules we used were developed by pharmaceutic companies as specific and orally bioavailable GnRHR antagonists and subsequently abandoned. In our search for structures that might bind and stabilize mutants of the GnRHR, we recognized that these molecules were candidates to be pharmacoperones since they bound the mutants with high affinity and selectivity yet did not activate it. Because these compounds are antagonists, they must be washed out of the system before (endogenous) agonist can occupy/activate the receptor.
Our proof of principle studies have relied on the Merck compound, “IN3”, ((2S)-2-[5-[2-(2-azabicyclo[2.2.2]oct-2-yl)-1,1-dimethyl-2-oxoethyl]-2-(3,5-dimethylphenyl)-1H-indol-3-yl]-N-(2-pyridin-4-ylethyl)propan-1-amine) at a concentration of 5 μg/ml, infused in heparin-saline at 25 μl/h. We have studied this compound extensively in cell cultures and pharmacological data is available with bioavailability of about 25% by i.m. or s.c. administration and a half-life of about 3 h (in humans, dogs and rats) (6, 28, 50, 71, 107, 110-112). We selected this drug since a half-life of this order is desirable as it allows sufficient time for the drug to access the vasculature of the pituitary gonadotrope without creating the need for a high concentration. This amount of time also allows for adequate washout in a 1-2 day pulse protocol.
6.6 Demonstration of In Vivo Rescue
We used an infusion system which relies on i.v. administration of the drug IN3, to demonstrate in vivo rescue of the mutant. Endogenous GnRH binds the GnRHR and results in LH release to the peripheral circulation. Because testis growth is closely related to circulating LH levels, testicular weight is a good and convenient indicator of its average level over time. The primary indication of human HH is the inability to elaborate pulsatile LH and testes remain small. The data (Fig. 6) shows the 90-day WT control (+/+) and K90neo/K90neo homozygous animals. A 60-day-old homozygous mutant was exposed to infusion of 5 μg/ml pharmacoperone IN3 for 30 days at a rate of 25 μl/h. The animal (Fig. 6) received pulses of 6 h every three days. Male K90neo/K90neo animals displayed decreased gonadal size compared to wild-type animals or heterozygotes at 60 days of age. Males were infused at a constant rate with saline-heparin (SH) or 5 μg/ml pharmacoperone IN3, were varied. The frequency (pulses/day) and pulse duration (hours) of pharmacoperone administration in the model animal to identify conditions which rescued the mutant. A selection of these conditions is shown (Fig 8). The optimum drug infusion conditions produced, in 30 days, an animal with a testis weight that was 72 fold the mean aged-matched untreated animals (0.045; N=5 g) at 90 days of age. We created a pool of “rescued” testes from these animals (0.09 ± 0.005/2 testes; N=14) to compare with untreated mutants, heterozygotes, or wild-type animals at 90 days of age. After 30 days of IN3 infusion, testis weight was markedly increased and spermatogenesis was restored to WT levels in all animals.
Fig. 8. Testicular weight.

was determined in 60-day-old animals after 30 days infusion with pharmacoperone IN3 (each 1, 2 or 3 days with pulse duration of 2, 4, 6 or 8 hours). Reprinted from (66) with permission of the publisher.
Because the mouse tether and jacket interfere with mating studies, we isolated sperm from the epididymis of a single IN3-rescued K90neo/K90neo male and performed sperm analysis and in vitro fertilization (IVF). Sperm from K90neo homozygotes possessed abnormal morphologies including loops in the sperm tails (Fig. 9), likely to be the cause of low sperm progression. In a single animal, IN3-rescue produced an animal with virtually normal sperm counts and good progression. Sperm from this single animal were used to produce blastocysts that were implanted into a surrogate female and resulted in a mouse pup with the expected (heterozygous) genotype.
Fig. 9. Morphology of Sperm.

Images are shown of normal sperm, as well as those from untreated K90neo/K90neo mutants. These lack heads, have thin heads or tail loops. Images by Drs. Eunju Kang and Shoukhrat Mitalipov, the Oregon National Primate Research Center.
Steroidogenic acute regulatory protein (StAR) mediates cholesterol transfer within the mitochondria, the rate-limiting step in the production of steroid hormones; its production is mediated by the luteinizing hormone levels in the testes. We examined StAR protein levels in 3 testes each of wild-type, K90neo homozygotes and IN3-treated K90neo homozygotes by Western blotting. Compared to the wild-type (arbitrarily 100% by densitometry), StAR protein was markedly decreased to 28% in K90neo homozygotes. The loss was substantially, but not totally, reversed by IN3 treatment (54%) (Fig. 10). Side chain cleavage cytochrome (P450scc or CYP11A1) is a mitochondrial enzyme that catalyzes conversion of cholesterol to pregnenolone. This is the first reaction in the process of steroidogenesis in all mammalian tissues that specialize in the production of various steroid hormones. Similar to that of StAR, there was a modest decrease in this enzyme in the K90neo homozygotes as assessed by Western blotting (34%) (Fig. 10). CYP11A1 protein expression was increased by IN3 treatment (50%), but not to wild-type levels (100%). The more modest loss of P450scc compared with StAR proteins may reflect the observation that this enzyme is always active, but its activity is limited by the supply of cholesterol in the inner mitochondrial membrane.
Fig. 10. Expression of StAR (4) and CYP11A1 (5) protein.

in wild-type, K90neo/K90neo and IN3-rescued K90neo/K90neo males. 50 μg of protein of testicular protein from wild-type, K90neo/K90neo and IN3-rescued K90neo/K90neo groups were analyzed by immunoblotting. Immunoblots show relative expression levels of StAR (A) and CYP11A1 (B) in testes from three different mice with β-actin expression as a loading control. Modified from (66) amd reprinted with permission of the publisher.
Testes were assessed by quantitative real-time PCR for mRNA levels of hydroxyl steroid dehydrogenase 17β (Fig. 11). This enzyme catalyzes conversion of the poorly bioactive 17-keto steroids to the highly bioactive 17β-hydroxysteroids (113). Levels were similar comparing WT and heterozygotes mRNA levels in E90Kneo homozygotes were markedly reduced compared with either group. Treatment with IN3 increased Hsd17b expression in homozygotes, but not the same levels as observed for WT. Despite the “sub-normal” level of this rate-limiting enzyme, circulating levels of testosterone were improved in IN3-treated homozygous mutants (Fig. 12).
Fig. 11. Hsd (hydroxysteroid dehydrogenase)17b3 qPCR.

Real-time PCR was performed using total RNA extracted from the testis. Data were analyzed by the ΔΔCT method and reported as copy number relative to that of Gapdh. Wild-type and K90neo heterozygotes, which are both phenotypically normal, exhibited similar levels of Hsd17b3 mRNA. K90neo/K90neo males, which exhibit hypogonadism, had reduced Hsd17b3 mRNA (P < 0.05). Hsd17b3 expression was increased in K90neo/K90neo males following pharmacoperone treatment (P < 0.05). Error bars show SEM. Reprinted from (66) with permission of the publisher.
Fig. 12. IN3 treatment restores testosterone levels in K90neo/K90neo males.

Serum testosterone was measured by RIA. K90neo/K90neo males exhibited less serum testosterone than heterozygotes. Testosterone levels were restored in K90neo/K90neo males after pharmacoperone treatment. Error bars show SEMs. IN3-treated animals were 60 days old, then treated for 30 additional days (90 days at euthanizing). Reprinted from (66) with permission of the publisher.
These studies show that mutant GnRHR[E90K] can be rescued in vivo and its translocation to the plasma membrane also restores other functions associated with the normal functioning of the GnRHR.
7. Conclusions
The studies that we summarize here describe the basis of development of high throughput screens for pharmacoperones of GPCRs, among the most common drug targets. In principle, these assays can be extended to other proteins. Of note, this approach is designed to identify drug leads that are specifically pharmacoperones and not simply antagonists that also show pharmacological activity. Such (non-antagonists) are valuable as they will show a more simple pharmacology that is afforded by pharmacoperones that are also antagonists. While such combination drugs can be shown to be useful in the mouse mutant model described, their use requires pulsatile administration so that the misrouted protein can first be rescued and then the drug removed to enable occupancy by endogenous agonist. We predict that, a drugs are identified that lack antagonistic activity, pharmacoperone rescue will provide a new approach for therapeutic development, useful for a range of diseases.
Acknowledgments
Supported by National Institutes of Health Grants OD012220, DK099090 and DK85040 (PMC).
Abbreviations
- GPCRs
G protein coupled receptors
- V2R
vasopressin type 2 receptor
- GnRHR
gonadotropin releasing hormone receptor
- QCS
quality control system
- ER
endoplasmic reticulum
- PM
plasma membrane
- GnRH
gonadotropin releasing hormone
- LH
luteinizing hormone
- FSH
follicle stimulating hormone
- HH
hypogonadotropic hypogonadism
- NDI
nephrogenic diabetes insipidus
- cAMP
adenosine monophosphate
- “Dox”
doxycycline
- S/B
signal-to-background ratio
- TMS
transmembrane segment
- IVF
in vitro fertilization
- StAR
Steroidogenic acute regulatory protein
Footnotes
Conflict of Interest: The authors have no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References Cited
- 1.Kerem E. Pharmacological induction of CFTR function in patients with cystic fibrosis: mutation-specific therapy. Pediatr Pulmonol. 2005;40:183–196. doi: 10.1002/ppul.20200. [DOI] [PubMed] [Google Scholar]
- 2.Dormer RL, Derand R, McNeilly CM, Mettey Y, Bulteau-Pignoux L, Metaye T, Vierfond JM, Gray MA, Galietta LJ, Morris MR, Pereira MM, Doull IJ, Becq F, McPherson MA. Correction of delF508-CFTR activity with benzo(c)quinolizinium compounds through facilitation of its processing in cystic fibrosis airway cells. J Cell Sci. 2001;114:4073–4081. doi: 10.1242/jcs.114.22.4073. [DOI] [PubMed] [Google Scholar]
- 3.Galietta LJ, Springsteel MF, Eda M, Niedzinski EJ, By K, Haddadin MJ, Kurth MJ, Nantz MH, Verkman AS. Novel CFTR chloride channel activators identified by screening of combinatorial libraries based on flavone and benzoquinolizinium lead compounds. The Journal of biological chemistry. 2001;276:19723–19728. doi: 10.1074/jbc.M101892200. [DOI] [PubMed] [Google Scholar]
- 4.Zhang XM, Wang XT, Yue H, Leung SW, Thibodeau PH, Thomas PJ, Guggino SE. Organic solutes rescue the functional defect in delta F508 cystic fibrosis transmembrane conductance regulator. The Journal of biological chemistry. 2003;278:51232–51242. doi: 10.1074/jbc.M309076200. [DOI] [PubMed] [Google Scholar]
- 5.Amaral MD. Therapy through chaperones: sense or antisense? Cystic fibrosis as a model disease. J Inherit Metab Dis. 2006;29:477–487. doi: 10.1007/s10545-006-0251-x. [DOI] [PubMed] [Google Scholar]
- 6.Janovick JA, Maya-Nunez G, Conn PM. Rescue of hypogonadotropic hypogonadism-causing and manufactured GnRH receptor mutants by a specific protein-folding template: misrouted proteins as a novel disease etiology and therapeutic target. J Clin Endocrinol Metab. 2002;87:3255–3262. doi: 10.1210/jcem.87.7.8582. [DOI] [PubMed] [Google Scholar]
- 7.Ulloa-Aguirre A, Janovick JA, Leanos-Miranda A, Conn PM. Misrouted cell surface GnRH receptors as a disease aetiology for congenital isolated hypogonadotrophic hypogonadism. Hum Reprod Update. 2004;10:177–192. doi: 10.1093/humupd/dmh015. [DOI] [PubMed] [Google Scholar]
- 8.Bichet DG. Nephrogenic diabetes insipidus. Adv Chronic Kidney Dis. 2006;13:96–104. doi: 10.1053/j.ackd.2006.01.006. [DOI] [PubMed] [Google Scholar]
- 9.Bernier V, Lagace M, Bichet DG, Bouvier M. Pharmacological chaperones: potential treatment for conformational diseases. Trends in endocrinology and metabolism TEM. 2004;15:222–228. doi: 10.1016/j.tem.2004.05.003. [DOI] [PubMed] [Google Scholar]
- 10.Bernier V, Morello JP, Zarruk A, Debrand N, Salahpour A, Lonergan M, Arthus MF, Laperriere A, Brouard R, Bouvier M, Bichet DG. Pharmacologic chaperones as a potential treatment for X-linked nephrogenic diabetes insipidus. J Am Soc Nephrol. 2006;17:232–243. doi: 10.1681/ASN.2005080854. [DOI] [PubMed] [Google Scholar]
- 11.Noorwez SM, Malhotra R, McDowell JH, Smith KA, Krebs MP, Kaushal S. Retinoids assist the cellular folding of the autosomal dominant retinitis pigmentosa opsin mutant P23H. The Journal of biological chemistry. 2004;279:16278–16284. doi: 10.1074/jbc.M312101200. [DOI] [PubMed] [Google Scholar]
- 12.Wang QY, Manicassamy B, Yu X, Dolmer K, Gettins PG, Rong L. Characterization of the LDL-A module mutants of Tva, the subgroup A Rous sarcoma virus receptor, and the implications in protein folding. Protein Sci. 2002;11:2596–2605. doi: 10.1110/ps.0219802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Benedek GB, Pande J, Thurston GM, Clark JI. Theoretical and experimental basis for the inhibition of cataract. Prog Retin Eye Res. 1999;18:391–402. doi: 10.1016/s1350-9462(98)00023-8. [DOI] [PubMed] [Google Scholar]
- 14.Heiser V, Scherzinger E, Boeddrich A, Nordhoff E, Lurz R, Schugardt N, Lehrach H, Wanker EE. Inhibition of huntingtin fibrillogenesis by specific antibodies and small molecules: implications for Huntington's disease therapy. Proc Natl Acad Sci U S A. 2000;97:6739–6744. doi: 10.1073/pnas.110138997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Forloni G, Terreni L, Bertani I, Fogliarino S, Invernizzi R, Assini A, Ribizzi G, Negro A, Calabrese E, Volonte MA, Mariani C, Franceschi M, Tabaton M, Bertoli A. Protein misfolding in Alzheimer's and Parkinson's disease: genetics and molecular mechanisms. Neurobiol Aging. 2002;23:957–976. doi: 10.1016/s0197-4580(02)00076-3. [DOI] [PubMed] [Google Scholar]
- 16.Permanne B, Adessi C, Saborio GP, Fraga S, Frossard MJ, Van Dorpe J, Dewachter I, Banks WA, Van Leuven F, Soto C. Reduction of amyloid load and cerebral damage in a transgenic mouse model of Alzheimer's disease by treatment with a beta-sheet breaker peptide. FASEB J. 2002;16:860–862. doi: 10.1096/fj.01-0841fje. [DOI] [PubMed] [Google Scholar]
- 17.Soto C, Kascsak RJ, Saborio GP, Aucouturier P, Wisniewski T, Prelli F, Kascsak R, Mendez E, Harris DA, Ironside J, Tagliavini F, Carp RI, Frangione B. Reversion of prion protein conformational changes by synthetic beta-sheet breaker peptides. Lancet. 2000;355:192–197. doi: 10.1016/s0140-6736(99)11419-3. [DOI] [PubMed] [Google Scholar]
- 18.Muchowskiand PJ, Wacker JL. Modulation of neurodegeneration by molecular chaperones. Nat Rev Neurosci. 2005;6:11–22. doi: 10.1038/nrn1587. [DOI] [PubMed] [Google Scholar]
- 19.Lubin DJ, Butler JS, Loh SN. Folding of tetrameric p53: oligomerization and tumorigenic mutations induce misfolding and loss of function. J Mol Biol. 2010;395:705–716. doi: 10.1016/j.jmb.2009.11.013. [DOI] [PubMed] [Google Scholar]
- 20.Fan JQ. A contradictory treatment for lysosomal storage disorders: inhibitors enhance mutant enzyme activity. Trends in pharmacological sciences. 2003;24:355–360. doi: 10.1016/S0165-6147(03)00158-5. [DOI] [PubMed] [Google Scholar]
- 21.Bottomley SP. The structural diversity in alpha1-antitrypsin misfolding. EMBO Rep. 2011;12:983–984. doi: 10.1038/embor.2011.187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Feldhammer M, Durand S, Pshezhetsky AV. Protein misfolding as an underlying molecular defect in mucopolysaccharidosis III type C. PLoS One. 2009;4:e7434. doi: 10.1371/journal.pone.0007434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sun Y, Breydo L, Makarava N, Yang Q, Bocharova OV, Baskakov IV. Site-specific conformational studies of prion protein (PrP) amyloid fibrils revealed two cooperative folding domains within amyloid structure. The Journal of biological chemistry. 2007;282:9090–9097. doi: 10.1074/jbc.M608623200. [DOI] [PubMed] [Google Scholar]
- 24.Conn PM, Ulloa-Aguirre A, Ito J, Janovick JA. G protein-coupled receptor trafficking in health and disease: lessons learned to prepare for therapeutic mutant rescue in vivo. Pharmacol Rev. 2007;59:225–250. doi: 10.1124/pr.59.3.2. [DOI] [PubMed] [Google Scholar]
- 25.Bernier V, Lagace M, Lonergan M, Arthus MF, Bichet DG, Bouvier M. Functional rescue of the constitutively internalized V2 vasopressin receptor mutant R137H by the pharmacological chaperone action of SR49059. Mol Endocrinol. 2004;18:2074–2084. doi: 10.1210/me.2004-0080. [DOI] [PubMed] [Google Scholar]
- 26.Conn PM. Rescue of Gonadotropin Hormone Receptor Mutants. US7695917. Oregon Health & Science University; United States: 2010. [Google Scholar]
- 27.Fanand ZC, Tao YX. Functional characterization and pharmacological rescue of melanocortin-4 receptor mutations identified from obese patients. J Cell Mol Med. 2009;13:3268–3282. doi: 10.1111/j.1582-4934.2009.00726.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Janovick JA, Goulet M, Bush E, Greer J, Wettlaufer DG, Conn PM. Structure-activity relations of successful pharmacologic chaperones for rescue of naturally occurring and manufactured mutants of the gonadotropin-releasing hormone receptor. J Pharmacol Exp Ther. 2003;305:608–614. doi: 10.1124/jpet.102.048454. [DOI] [PubMed] [Google Scholar]
- 29.Krebs MP, Holden DC, Joshi P, Clark CL, 3rd, Lee AH, Kaushal S. Molecular mechanisms of rhodopsin retinitis pigmentosa and the efficacy of pharmacological rescue. J Mol Biol. 2010;395:1063–1078. doi: 10.1016/j.jmb.2009.11.015. [DOI] [PubMed] [Google Scholar]
- 30.Loo TW, Bartlett MC, Clarke DM. Rescue of folding defects in ABC transporters using pharmacological chaperones. J Bioenerg Biomembr. 2005;37:501–507. doi: 10.1007/s10863-005-9499-3. [DOI] [PubMed] [Google Scholar]
- 31.Loo TW, Bartlett MC, Clarke DM. Rescue of DeltaF508 and other misprocessed CFTR mutants by a novel quinazoline compound. Molecular pharmaceutics. 2005;2:407–413. doi: 10.1021/mp0500521. [DOI] [PubMed] [Google Scholar]
- 32.Majtan T, Liu L, Carpenter JF, Kraus JP. Rescue of cystathionine beta-synthase (CBS) mutants with chemical chaperones: purification and characterization of eight CBS mutant enzymes. The Journal of biological chemistry. 2010;285:15866–15873. doi: 10.1074/jbc.M110.107722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Maupin CM, Castillo N, Taraphder S, Tu C, McKenna R, Silverman DN, Voth GA. Chemical rescue of enzymes: proton transfer in mutants of human carbonic anhydrase II. J Am Chem Soc. 2011;133:6223–6234. doi: 10.1021/ja1097594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Oueslati M, Hermosilla R, Schonenberger E, Oorschot V, Beyermann M, Wiesner B, Schmidt A, Klumperman J, Rosenthal W, Schulein R. Rescue of a nephrogenic diabetes insipidus-causing vasopressin V2 receptor mutant by cell-penetrating peptides. The Journal of biological chemistry. 2007;282:20676–20685. doi: 10.1074/jbc.M611530200. [DOI] [PubMed] [Google Scholar]
- 35.Qiao Y, Molina H, Pandey A, Zhang J, Cole PA. Chemical rescue of a mutant enzyme in living cells. Science. 2006;311:1293–1297. doi: 10.1126/science.1122224. [DOI] [PubMed] [Google Scholar]
- 36.Rivero-Muller A, Chou YY, Ji I, Lajic S, Hanyaloglu AC, Jonas K, Rahman N, Ji TH, Huhtaniemi I. Rescue of defective G protein-coupled receptor function in vivo by intermolecular cooperation. Proc Natl Acad Sci U S A. 2010;107:2319–2324. doi: 10.1073/pnas.0906695106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Singh LR, Chen X, Kozich V, Kruger WD. Chemical chaperone rescue of mutant human cystathionine beta-synthase. Mol Genet Metab. 2007;91:335–342. doi: 10.1016/j.ymgme.2007.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.White E, McKenna J, Cavanaugh A, Breitwieser GE. Pharmacochaperone-mediated rescue of calcium-sensing receptor loss-of-function mutants. Mol Endocrinol. 2009;23:1115–1123. doi: 10.1210/me.2009-0041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Breitwieser GE. Pharmacoperones and the calcium sensing receptor: Exogenous and endogenous regulators. Pharmacological research: the official journal of the Italian Pharmacological Society. 2013 doi: 10.1016/j.phrs.2013.11.006. [DOI] [PubMed] [Google Scholar]
- 40.Conn PM, Leanos-Miranda A, Janovick JA. Protein origami: therapeutic rescue of misfolded gene products. Mol Interv. 2002;2:308–316. doi: 10.1124/mi.2.5.308. [DOI] [PubMed] [Google Scholar]
- 41.Ulloa-Aguirre A, Janovick JA, Brothers SP, Conn PM. Pharmacologic rescue of conformationally-defective proteins: implications for the treatment of human disease. Traffic. 2004;5:821–837. doi: 10.1111/j.1600-0854.2004.00232.x. [DOI] [PubMed] [Google Scholar]
- 42.Castro-Fernandez C, Maya-Nunez G, Conn PM. Beyond the signal sequence: protein routing in health and disease. Endocr Rev. 2005;26:479–503. doi: 10.1210/er.2004-0010. [DOI] [PubMed] [Google Scholar]
- 43.Kresge N. Pharmacological Chaperones Show. Am Soc for Biochem Molec Bio Today. 2004;3:10–11. [Google Scholar]
- 44.Hunter PJ. Receptor redemption: skirting gene therapy to correct genetic defects. The Scientist. 2004;18:30. [Google Scholar]
- 45.Connand PM, Ulloa-Aguirre A. Trafficking of G-protein-coupled receptors to the plasma membrane: insights for pharmacoperone drugs. Trends in endocrinology and metabolism: TEM. 2010;21:190–197. doi: 10.1016/j.tem.2009.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Connand PM, Janovick JA. Drug development and the cellular quality control system. Trends in pharmacological sciences. 2009;30:228–233. doi: 10.1016/j.tips.2009.02.002. [DOI] [PubMed] [Google Scholar]
- 47.Janovick JA, Maya-Nunez G, Ulloa-Aguirre A, Huhtaniemi IT, Dias JA, Verbost P, Conn PM. Increased plasma membrane expression of human follicle-stimulating hormone receptor by a small molecule thienopyr(im)idine. Mol Cell Endocrinol. 2009;298:84–88. doi: 10.1016/j.mce.2008.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Janovick JA, Patney A, Mosley R, Goulet MT, Altman MD, Rush TS, 3rd, Cornea A, Conn PM. Molecular mechanism of action of pharmacoperone rescue of misrouted GPCR mutants: the GnRH receptor. Mol Endocrinol. 2009;23:157–168. doi: 10.1210/me.2008-0384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Connand PM, Janovick JA. Trafficking and quality control of the gonadotropin releasing hormone receptor in health and disease. Mol Cell Endocrinol. 2009;299:137–145. doi: 10.1016/j.mce.2008.10.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Janovick JA, Brothers SP, Cornea A, Bush E, Goulet MT, Ashton WT, Sauer DR, Haviv F, Greer J, Conn PM. Refolding of misfolded mutant GPCR: post-translational pharmacoperone action in vitro. Mol Cell Endocrinol. 2007;272:77–85. doi: 10.1016/j.mce.2007.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Morello JP, Salahpour A, Laperriere A, Bernier V, Arthus MF, Lonergan M, Petaja-Repo U, Angers S, Morin D, Bichet DG, Bouvier M. Pharmacological chaperones rescue cell-surface expression and function of misfolded V2 vasopressin receptor mutants. The Journal of clinical investigation. 2000;105:887–895. doi: 10.1172/JCI8688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Janovick JA, Park BS, Conn PM. Therapeutic rescue of misfolded mutants: validation of primary high throughput screens for identification of pharmacoperone drugs. PLoS One. 2011;6:e22784. doi: 10.1371/journal.pone.0022784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Smithson DC, Janovick JA, Conn PM. Therapeutic rescue of misfolded/mistrafficked mutants: automation-friendly high throughput assays for identification of pharmacoperone drugs of GPCRs. Methods in enzymology. 2012 doi: 10.1016/B978-0-12-391862-8.00001-6. In Press. [DOI] [PubMed] [Google Scholar]
- 54.Conn PM, Smith E, Hodder P, Janovick JA, Smithson DC. High-throughput screen for pharmacoperones of the vasopressin type 2 receptor. Journal of biomolecular screening. 2013;18:930–937. doi: 10.1177/1087057113483559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Calamini B, Silva MC, Madoux F, Hutt DM, Khanna S, Chalfant MA, Saldanha SA, Hodder P, Tait BD, Garza D, Balch WE, Morimoto RI. Small-molecule proteostasis regulators for protein conformational diseases. Nat Chem Biol. 2012;8:185–196. doi: 10.1038/nchembio.763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Leesonand PD, Springthorpe B. The influence of drug-like concepts on decision-making in medicinal chemistry. Nat Rev Drug Discov. 2007;6:881–890. doi: 10.1038/nrd2445. [DOI] [PubMed] [Google Scholar]
- 57.Axerio-Cilies P, Castaneda IP, Mirza A, Reynisson J. Investigation of the incidence of “undesirable” molecular moieties for high-throughput screening compound libraries in marketed drug compounds. Eur J Med Chem. 2009;44:1128–1134. doi: 10.1016/j.ejmech.2008.06.013. [DOI] [PubMed] [Google Scholar]
- 58.Harvey AL. Natural products in drug discovery. Drug Discov Today. 2008;13:894–901. doi: 10.1016/j.drudis.2008.07.004. [DOI] [PubMed] [Google Scholar]
- 59.Potashmanand MH, Duggan ME. Covalent modifiers: an orthogonal approach to drug design. J Med Chem. 2009;52:1231–1246. doi: 10.1021/jm8008597. [DOI] [PubMed] [Google Scholar]
- 60.Baelland JB, Holloway GA. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J Med Chem. 2010;53:2719–2740. doi: 10.1021/jm901137j. [DOI] [PubMed] [Google Scholar]
- 61.Ding S, Gray NS, Wu X, Ding Q, Schultz PG. A combinatorial scaffold approach toward kinase-directed heterocycle libraries. J Am Chem Soc. 2002;124:1594–1596. doi: 10.1021/ja0170302. [DOI] [PubMed] [Google Scholar]
- 62.Kolband HC, Sharpless KB. The growing impact of click chemistry on drug discovery. Drug Discov Today. 2003;8:1128–1137. doi: 10.1016/s1359-6446(03)02933-7. [DOI] [PubMed] [Google Scholar]
- 63.Lovering F, Bikker J, Humblet C. Escape from flatland: increasing saturation as an approach to improving clinical success. J Med Chem. 2009;52:6752–6756. doi: 10.1021/jm901241e. [DOI] [PubMed] [Google Scholar]
- 64.Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46:3–26. doi: 10.1016/s0169-409x(00)00129-0. [DOI] [PubMed] [Google Scholar]
- 65.Congreve M, Carr R, Murray C, Jhoti H. A ‘rule of three’ for fragment-based lead discovery? Drug Discov Today. 2003;8:876–877. doi: 10.1016/s1359-6446(03)02831-9. [DOI] [PubMed] [Google Scholar]
- 66.Janovick JA, Stewart MD, Jacob D, Martin LD, Deng JM, Stewart CA, Wang Y, Cornea A, Chavali L, Lopez S, Mitalipov S, Kang E, Lee HS, Manna PR, Stocco D, Behringer RR, Conn PM. Restoration of plasma membrane expression and activity of a misfolded GnRHR mutant in mice by a small molecule pharmacoperone. Proc Natl Acad Sci U S A. 2013 doi: 10.1073/pnas.1315194110. In press. 2013 Dec 9. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Conn PM, Janovick JA, Brothers SP, Knollman PE. ‘Effective inefficiency’: cellular control of protein trafficking as a mechanism of post-translational regulation. J Endocrinol. 2006;190:13–16. doi: 10.1677/joe.1.06771. [DOI] [PubMed] [Google Scholar]
- 68.Conn PM, Knollman PE, Brothers SP, Janovick JA. Protein folding as posttranslational regulation: evolution of a mechanism for controlled plasma membrane expression of a G protein-coupled receptor. Mol Endocrinol. 2006;20:3035–3041. doi: 10.1210/me.2006-0066. [DOI] [PubMed] [Google Scholar]
- 69.Knollman PE, Janovick JA, Brothers SP, Conn PM. Parallel regulation of membrane trafficking and dominant-negative effects by misrouted gonadotropin-releasing hormone receptor mutants. The Journal of biological chemistry. 2005;280:24506–24514. doi: 10.1074/jbc.M501978200. [DOI] [PubMed] [Google Scholar]
- 70.Jennes L, Ulloa-Aguirre A, Janovick JA, Adhan V, Conn PM. Gonadotropin releasing hormone and its receptor. In: Pfaff DW, Arnold AP, Etgen AM, Fahrbach SE, Rubin RT, editors. Hormones, Brain and Behavior. 2nd. Vol. 3. Academic Press; New York: 2009. pp. 1645–1668. [Google Scholar]
- 71.Janovick JA, Patny A, Mosley R, Goulet MT, Altman MD, Rush TS, 3rd, Cornea A, Conn PM. Molecular mechanism of action of pharmacoperone rescue of misrouted GPCR mutants: the GnRH receptor. Mol Endocrinol. 2009;23:157–168. doi: 10.1210/me.2008-0384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Brothers SP, Cornea A, Janovick JA, Conn PM. Human loss-of-function gonadotropin-releasing hormone receptor mutants retain wild-type receptors in the endoplasmic reticulum: molecular basis of the dominant-negative effect. Mol Endocrinol. 2004;18:1787–1797. doi: 10.1210/me.2004-0091. [DOI] [PubMed] [Google Scholar]
- 73.Leskela TT, Lackman JJ, Vierimaa MM, Kobayashi H, Bouvier M, Petaja-Repo UE. Cys-27 variant of human delta-opioid receptor modulates maturation and cell surface delivery of Phe-27 variant via heteromerization. The Journal of biological chemistry. 2012;287:5008–5020. doi: 10.1074/jbc.M111.305656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bakker RA, Lozada AF, van Marle A, Shenton FC, Drutel G, Karlstedt K, Hoffmann M, Lintunen M, Yamamoto Y, van Rijn RM, Chazot PL, Panula P, Leurs R. Discovery of naturally occurring splice variants of the rat histamine H3 receptor that act as dominant-negative isoforms. Molecular pharmacology. 2006;69:1194–1206. doi: 10.1124/mol.105.019299. [DOI] [PubMed] [Google Scholar]
- 75.McElvaineand AT, Mayo KE. A dominant-negative human growth hormone-releasing hormone (GHRH) receptor splice variant inhibits GHRH binding. Endocrinology. 2006;147:1884–1894. doi: 10.1210/en.2005-1488. [DOI] [PubMed] [Google Scholar]
- 76.Beaumont KA, Newton RA, Smit DJ, Leonard JH, Stow JL, Sturm RA. Altered cell surface expression of human MC1R variant receptor alleles associated with red hair and skin cancer risk. Human molecular genetics. 2005;14:2145–2154. doi: 10.1093/hmg/ddi219. [DOI] [PubMed] [Google Scholar]
- 77.Sanchez-Laorden BL, Herraiz C, Valencia JC, Hearing VJ, Jimenez-Cervantes C, Garcia-Borron JC. Aberrant trafficking of human melanocortin 1 receptor variants associated with red hair and skin cancer: Steady-state retention of mutant forms in the proximal golgi. J Cell Physiol. 2009;220:640–654. doi: 10.1002/jcp.21804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sanchez-Laorden BL, Sanchez-Mas J, Martinez-Alonso E, Martinez-Menarguez JA, Garcia-Borron JC, Jimenez-Cervantes C. Dimerization of the human melanocortin 1 receptor: functional consequences and dominant-negative effects. J Invest Dermatol. 2006;126:172–181. doi: 10.1038/sj.jid.5700036. [DOI] [PubMed] [Google Scholar]
- 79.Calebiro D, de Filippis T, Lucchi S, Covino C, Panigone S, Beck-Peccoz P, Dunlap D, Persani L. Intracellular entrapment of wild-type TSH receptor by oligomerization with mutants linked to dominant TSH resistance. Human molecular genetics. 2005;14:2991–3002. doi: 10.1093/hmg/ddi329. [DOI] [PubMed] [Google Scholar]
- 80.Apaja PM, Tuusa JT, Pietila EM, Rajaniemi HJ, Petaja-Repo UE. Luteinizing hormone receptor ectodomain splice variant misroutes the full-length receptor into a subcompartment of the endoplasmic reticulum. Molecular biology of the cell. 2006;17:2243–2255. doi: 10.1091/mbc.E05-09-0875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Tao YX, Johnson NB, Segaloff DL. Constitutive and agonist-dependent self-association of the cell surface human lutropin receptor. The Journal of biological chemistry. 2004;279:5904–5914. doi: 10.1074/jbc.M311162200. [DOI] [PubMed] [Google Scholar]
- 82.Zarinan T, Perez-Solis MA, Maya-Nunez G, Casas-Gonzalez P, Conn PM, Dias JA, Ulloa-Aguirre A. Dominant negative effects of human follicle-stimulating hormone receptor expression-deficient mutants on wild-type receptor cell surface expression. Rescue of oligomerization-dependent defective receptor expression by using cognate decoys. Mol Cell Endocrinol. 2010;321:112–122. doi: 10.1016/j.mce.2010.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rajanand RS, Kopito RR. Suppression of wild-type rhodopsin maturation by mutants linked to autosomal dominant retinitis pigmentosa. The Journal of biological chemistry. 2005;280:1284–1291. doi: 10.1074/jbc.M406448200. [DOI] [PubMed] [Google Scholar]
- 84.Bulenger S, Marullo S, Bouvier M. Emerging role of homo- and heterodimerization in G-protein-coupled receptor biosynthesis and maturation. Trends in pharmacological sciences. 2005;26:131–137. doi: 10.1016/j.tips.2005.01.004. [DOI] [PubMed] [Google Scholar]
- 85.Milligan G. G protein-coupled receptor dimerisation: molecular basis and relevance to function. Biochimica et biophysica acta. 2007;1768:825–835. doi: 10.1016/j.bbamem.2006.09.021. [DOI] [PubMed] [Google Scholar]
- 86.Brothers SP, Janovick JA, Conn PM. Unexpected effects of epitope and chimeric tags on gonadotropin-releasing hormone receptors: implications for understanding the molecular etiology of hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 2003;88:6107–6112. doi: 10.1210/jc.2003-031047. [DOI] [PubMed] [Google Scholar]
- 87.Ayoub MA, Couturier C, Lucas-Meunier E, Angers S, Fossier P, Bouvier M, Jockers R. Monitoring of ligand-independent dimerization and ligand-induced conformational changes of melatonin receptors in living cells by bioluminescence resonance energy transfer. The Journal of biological chemistry. 2002;277:21522–21528. doi: 10.1074/jbc.M200729200. [DOI] [PubMed] [Google Scholar]
- 88.Guo W, Shi L, Javitch JA. The fourth transmembrane segment forms the interface of the dopamine D2 receptor homodimer. The Journal of biological chemistry. 2003;278:4385–4388. doi: 10.1074/jbc.C200679200. [DOI] [PubMed] [Google Scholar]
- 89.Terrillon S, Barberis C, Bouvier M. Heterodimerization of V1a and V2 vasopressin receptors determines the interaction with beta-arrestin and their trafficking patterns. Proc Natl Acad Sci U S A. 2004;101:1548–1553. doi: 10.1073/pnas.0305322101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Herrick-Davis K, Grinde E, Mazurkiewicz JE. Biochemical and biophysical characterization of serotonin 5-HT2C receptor homodimers on the plasma membrane of living cells. Biochemistry. 2004;43:13963–13971. doi: 10.1021/bi048398p. [DOI] [PubMed] [Google Scholar]
- 91.McVey M, Ramsay D, Kellett E, Rees S, Wilson S, Pope AJ, Milligan G. Monitoring receptor oligomerization using time-resolved fluorescence resonance energy transfer and bioluminescence resonance energy transfer. The human delta -opioid receptor displays constitutive oligomerization at the cell surface, which is not regulated by receptor occupancy. The Journal of biological chemistry. 2001;276:14092–14099. doi: 10.1074/jbc.M008902200. [DOI] [PubMed] [Google Scholar]
- 92.Salahpour A, Angers S, Mercier JF, Lagace M, Marullo S, Bouvier M. Homodimerization of the beta2-adrenergic receptor as a prerequisite for cell surface targeting. The Journal of biological chemistry. 2004;279:33390–33397. doi: 10.1074/jbc.M403363200. [DOI] [PubMed] [Google Scholar]
- 93.Mercier JF, Salahpour A, Angers S, Breit A, Bouvier M. Quantitative assessment of beta 1- and beta 2-adrenergic receptor homo- and heterodimerization by bioluminescence resonance energy transfer. The Journal of biological chemistry. 2002;277:44925–44931. doi: 10.1074/jbc.M205767200. [DOI] [PubMed] [Google Scholar]
- 94.Angers S, Salahpour A, Joly E, Hilairet S, Chelsky D, Dennis M, Bouvier M. Detection of beta 2-adrenergic receptor dimerization in living cells using bioluminescence resonance energy transfer (BRET) Proc Natl Acad Sci U S A. 2000;97:3684–3689. doi: 10.1073/pnas.060590697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kaupmann K, Malitschek B, Schuler V, Heid J, Froestl W, Beck P, Mosbacher J, Bischoff S, Kulik A, Shigemoto R, Karschin A, Bettler B. GABA(B)-receptor subtypes assemble into functional heteromeric complexes. Nature. 1998;396:683–687. doi: 10.1038/25360. [DOI] [PubMed] [Google Scholar]
- 96.Margeta-Mitrovic M, Jan YN, Jan LY. A trafficking checkpoint controls GABA(B) receptor heterodimerization. Neuron. 2000;27:97–106. doi: 10.1016/s0896-6273(00)00012-x. [DOI] [PubMed] [Google Scholar]
- 97.Margeta-Mitrovic M. Assembly-dependent trafficking assays in the detection of receptor-receptor interactions. Methods. 2002;27:311–317. doi: 10.1016/s1046-2023(02)00088-9. [DOI] [PubMed] [Google Scholar]
- 98.Thomas RM, Nechamen CA, Mazurkiewicz JE, Muda M, Palmer S, Dias JA. Follice-stimulating hormone receptor forms oligomers and shows evidence of carboxyl-terminal proteolytic processing. Endocrinology. 2007;148:1987–1995. doi: 10.1210/en.2006-1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Robben JH, Sze M, Knoers NV, Deen PM. Functional rescue of vasopressin V2 receptor mutants in MDCK cells by pharmacochaperones: relevance to therapy of nephrogenic diabetes insipidus. Am J Physiol Renal Physiol. 2007;292:F253–260. doi: 10.1152/ajprenal.00247.2006. [DOI] [PubMed] [Google Scholar]
- 100.Robert J, Auzan C, Ventura MA, Clauser E. Mechanisms of cell-surface rerouting of an endoplasmic reticulum-retained mutant of the vasopressin V1b/V3 receptor by a pharmacological chaperone. The Journal of biological chemistry. 2005;280:42198–42206. doi: 10.1074/jbc.M510180200. [DOI] [PubMed] [Google Scholar]
- 101.Funakoshi T, Yanai A, Shinoda K, Kawano MM, Mizukami Y. G protein-coupled receptor 30 is an estrogen receptor in the plasma membrane. Biochemical and biophysical research communications. 2006;346:904–910. doi: 10.1016/j.bbrc.2006.05.191. [DOI] [PubMed] [Google Scholar]
- 102.Sanden C, Broselid S, Cornmark L, Andersson K, Daszkiewicz-Nilsson J, Martensson UE, Olde B, Leeb-Lundberg LM. G protein-coupled estrogen receptor 1/G protein-coupled receptor 30 localizes in the plasma membrane and traffics intracellularly on cytokeratin intermediate filaments. Molecular pharmacology. 2006;79:400–410. doi: 10.1124/mol.110.069500. [DOI] [PubMed] [Google Scholar]
- 103.Pask AJ, Kanasaki H, Kaiser UB, Conn PM, Janovick JA, Stockton DW, Hess DL, Justice MJ, Behringer RR. A novel mouse model of hypogonadotrophic hypogonadism: N-ethyl-N-nitrosourea-induced gonadotropin-releasing hormone receptor gene mutation. Mol Endocrinol. 2005;19:972–981. doi: 10.1210/me.2004-0192. [DOI] [PubMed] [Google Scholar]
- 104.Leanos-Miranda A, Ulloa-Aguirre A, Ji TH, Janovick JA, Conn PM. Dominant-negative action of disease-causing gonadotropin-releasing hormone receptor (GnRHR) mutants: a trait that potentially coevolved with decreased plasma membrane expression of GnRHR in humans. J Clin Endocrinol Metab. 2003;88:3360–3367. doi: 10.1210/jc.2003-030084. [DOI] [PubMed] [Google Scholar]
- 105.Soderlund D, Canto P, de la Chesnaye E, Ulloa-Aguirre A, Mendez JP. A novel homozygous mutation in the second transmembrane domain of the gonadotrophin releasing hormone receptor gene. Clinical endocrinology. 2001;54:493–498. doi: 10.1046/j.1365-2265.2001.01211.x. [DOI] [PubMed] [Google Scholar]
- 106.Jardon-Valadez E, Aguilar-Rojas A, Maya-Nunez G, Leanos-Miranda A, Pineiro A, Conn PM, Ulloa-Aguirre A. Conformational effects of Lys191 in the human GnRHR: mutagenesis and molecular dynamics simulations studies. J Endocrinol. 2009;201:297–307. doi: 10.1677/JOE-08-0527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Janovickand JA, Conn PM. Salt bridge integrates GPCR activation with protein trafficking. Proc Natl Acad Sci U S A. 2010;107:4454–4458. doi: 10.1073/pnas.0914261107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Stewart MD, Deng JM, Stewart CA, Mullen RD, Wang Y, Lopez S, Serna MK, Huang CC, Janovick JA, Pask AJ, Schwartz RJ, Conn PM, Behringer RR. Mice harboring Gnrhr E90K, a mutation that causes protein misfolding and hypogonadotropic hypogonadism in humans, exhibit testis size reduction and ovulation failure. Mol Endocrinol. 2012;26:1847–1856. doi: 10.1210/me.2012-1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lu L, Li Y, Kim SM, Bossuyt W, Liu P, Qiu Q, Wang Y, Halder G, Finegold MJ, Lee JS, Johnson RL. Hippo signaling is a potent in vivo growth and tumor suppressor pathway in the mammalian liver. Proc Natl Acad Sci U S A. 2010;107:1437–1442. doi: 10.1073/pnas.0911427107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Janovick JA, Knollman PE, Brothers SP, Ayala-Yanez R, Aziz AS, Conn PM. Regulation of G protein-coupled receptor trafficking by inefficient plasma membrane expression: molecular basis of an evolved strategy. The Journal of biological chemistry. 2006;281:8417–8425. doi: 10.1074/jbc.M510601200. [DOI] [PubMed] [Google Scholar]
- 111.Janovick JA, Pogozheva ID, Mosberg HI, Conn PM. Salt bridges overlapping the gonadotropin-releasing hormone receptor agonist binding site reveal a coincidence detector for g protein-coupled receptor activation. J Pharmacol Exp Ther. 2011;338:430–442. doi: 10.1124/jpet.111.180869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Janovick JA, Pogozheva ID, Mosberg HI, Cornea A, Conn PM. Rescue of misrouted GnRHR mutants reveals its constitutive activity. Mol Endocrinol. 2012;26:1179–1188. doi: 10.1210/me.2012-1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Saloniemi T, Jokela H, Strauss L, Pakarinen P, Poutanen M. The diversity of sex steroid action: novel functions of hydroxysteroid (17beta) dehydrogenases as revealed by genetically modified mouse models. J Endocrinol. 2012;212:27–40. doi: 10.1530/JOE-11-0315. [DOI] [PubMed] [Google Scholar]
