IGF1 receptor crosstalk: Mechanism and clinical implications

IGF1 receptor crosstalk: Mechanism and clinical implications

JPT-107502; No of Pages 5 Pharmacology & Therapeutics xxx (2020) xxx Contents lists available at ScienceDirect Pharmacology & Therapeutics journal h...

855KB Sizes 0 Downloads 34 Views

JPT-107502; No of Pages 5 Pharmacology & Therapeutics xxx (2020) xxx

Contents lists available at ScienceDirect

Pharmacology & Therapeutics journal homepage: www.elsevier.com/locate/pharmthera

TSH/IGF1 receptor crosstalk: Mechanism and clinical implications Christine C. Krieger, Susanne Neumann, Marvin C. Gershengorn ⁎ Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, United States

a r t i c l e

i n f o

Available online xxxx

Keywords: TSH receptor stimulating antibodies Graves' ophthalmopathy fibroblasts Receptor crosstalk Thyroid-stimulating hormone receptor Insulin-like growth factor 1 receptor

a b s t r a c t Increasing evidence of interdependence between G protein-coupled receptors and receptor tyrosine kinase signaling pathways has prompted reevaluation of crosstalk between these receptors in disease and therapy. Investigations into thyroid-stimulating hormone (TSH) and insulin-like growth factor 1 (IGF1) receptor crosstalk, and its application to the clinic have in particular shown recent progress. In this review, we summarize current insights into the mechanism of TSH/IGF1 receptor crosstalk. We discuss evidence that crosstalk is one of the underlying causes of TSHR-based disease and the feasibility of using combinations of TSH receptor and IGF1 receptor antagonists to increase the therapeutic index for the treatment of Graves' hyperthyroidism and Graves' ophthalmopathy. © 2020 Published by Elsevier Inc.

Contents 1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . 2. TSH/IGF1 receptor crosstalk in cells in culture . . . . . . . . 3. The role of ERK and β-Arrestin 1 in TSH/IGF1 receptor crosstalk 4. Crosstalk in the pathogenesis and treatment of Graves' disease . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . .

1. Introduction Crosstalk between cell-surface G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs) is a ubiquitous phenomenon that leads to broadening of the signaling responses initiated by the specific activation of one receptor (Hilger, Masureel, & Kobilka, 2018). In the case of GPCRs and RTK, crosstalk may occur after activation of either receptor; this has been termed bidirectional

Abbreviations: GPCR, G protein-coupled receptor; RTK, receptor tyrosine kinases; TSH, thyrotropin, thyroid-stimulating hormone; TSHR, thyrotropin receptor; IGF1, insulin-like growth factor 1; IGF1R, insulin-like growth factor 1 receptor; GD, Graves' disease; GO, Graves' ophthalmopathy, thyroid eye disease; GOFs, Graves' orbital fibroblasts/ preadipocytes; HA, hyaluronan, hyaluronic acid; ERK, extracellular signal-regulated kinases; βARR, β-arrestin; PLA, Proximity Ligation Assay; GO-Igs, purified GO antibodies. ⁎ Corresponding author at: 50 South Dr., Building 50, Room 4134, Bethesda, MD 20892, United States. E-mail address: [email protected] (M.C. Gershengorn).

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

. . . . . .

0 0 0 0 0 0

transactivation (Delcourt, Bockaert, & Marin, 2007; Pyne & Pyne, 2011). Although these interactions may occur at several steps in the signal transduction pathways, a large number of these involve GPCR-RTK interactions at the plasma membrane. Crosstalk of this type may be dependent on the generation of a ligand for the second receptor by activation of the first receptor or may be independent of ligand generation (Di Liberto, Mudo, & Belluardo, 2019). Crosstalk independent of ligand generation may result from “(r)eceptor-receptor interactions: when the binding of a ligand to the orthosteric or allosteric sites of one receptor causes, via direct allosteric interactions a change in the ligand recognition, decoding and trafficking processes of another receptor” (Kenakin et al., 2010). However, an alternative to direct receptor-receptor interaction may be the formation of a multi-protein complex (“signalosome”) in which two receptors are brought in close proximity and may influence each other's signaling. This type of receptor-receptor interaction appears to be the mechanism of crosstalk between the thyrotropin (thyroid-stimulating hormone, TSH) receptor (TSHR), which is a GPCR, and the insulin-like growth factor 1 receptor (IGF1R), which is an RTK.

https://doi.org/10.1016/j.pharmthera.2020.107502 0163-7258/© 2020 Published by Elsevier Inc.

Please cite this article as: C.C. Krieger, S. Neumann and M.C. Gershengorn, TSH/IGF1 receptor crosstalk: Mechanism and clinical implications, Pharmacology & Therapeutics, https://doi.org/10.1016/j.pharmthera.2020.107502

C.C. Krieger et al. / Pharmacology & Therapeutics xxx (2020) xxx

In this review, we will describe findings that support the conclusion that TSH/IGF1 receptor crosstalk occurs and the molecular details involved in TSHR-IGF1R interaction. We will also summarize how TSH/ IGF1 receptor crosstalk explains the pathophysiology of Graves' hyperthyroidism, the most common cause of human hyperthyroidism, and Graves' ophthalmopathy (GO, thyroid eye disease), and suggests treatment approaches for Graves' disease. 2. TSH/IGF1 receptor crosstalk in cells in culture Functional interactions between TSHR and IGF1R in thyrocytes in vitro (Eggo, Bachrach, & Burrow, 1990; Kimura et al., 2001; Santisteban, Kohn, & Di Lauro, 1987; Tramontano, Cushing, Moses, & Ingbar, 1986) have been reported for many years; however, the majority of these studies were performed in rodent cell lines and whether they are accurate models of human cells is not clear. Functional interactions between TSHR and IGF1R have also been shown in orbital fibroblasts from patients with Graves' disease in vitro (Chen et al., 2014; Hoa et al., 2012; Kumar, Iyer, Bauer, Coenen, & Bahn, 2012; Tsui et al., 2008). Our own studies have been performed in primary cultures of Graves' orbital fibroblasts/preadipocytes (GOFs), primary cultures of normal, differentiated, human thyrocytes obtained from the contralateral lobe of patients at surgery for thyroid cancer, and a human bone osteosarcoma epithelial cell line (Krieger et al., 2016; Krieger et al., 2019; Krieger, Neumann, Place, MarcusSamuels, & Gershengorn, 2015; Morgan, Neumann, Marcus-Samuels, & Gershengorn, 2016). In many studies in GOFs, activation of TSHR- and IGF1R-stimulated secretion of hyaluronan (hyaluronic acid, HA) was measured as a biologic response because increased HA secretion in the orbit is a major component of the pathophysiology of GO. To determine whether TSHR-IGF1R interaction was dependent on the generation of IGF1 after stimulating TSHR, we measured the effect of an IGF1R blocking antibody, AF305. Fig. 1 illustrates that AF305, as expected, inhibits stimulation of HA secretion by IGF1 but had no effect on stimulation by TSH (Krieger, unpublished observation). Thus, stimulation of HA secretion initiated by TSHR activation is independent of autocrine/paracrine effects of IGF1 generation. A primary indication of TSHR-IGF1R interaction was our observation that simultaneous activation by TSH and IGF1 synergistically increased HA secretion in GOFs; there were increases in both potency and efficacy of TSH (Krieger et al., 2015) (Fig. 2). Further evidence in support of TSH/ IGF1 receptor crosstalk were our findings that an IGF1R small molecule antagonist, linsitinib, inhibited HA stimulation by TSH and that some IGF1R blocking antibodies inhibited stimulation by TSH (Chen et al., 2014). These findings were complemented by our results using a monoclonal TSHR-stimulating antibody M22, which was derived from a patient with Graves' hyperthyroidism (Sanders et al., 2003). We found that the dose-response to M22 was biphasic and that the IGF1R antagonist, linsitinib, inhibited the higher potency (lower M22 dose) phase but had no effect on the lower potency (higher M22 dose) phase (Fig. 3A). Fig. 3B illustrates a model of TSH/IGF1 receptor crosstalk stimulated by M22. We (Krieger et al., 2016; Place, Krieger, Neumann, & Gershengorn, 2017) and others (Chen et al., 2014; Kumar et al., 2012) provided additional evidence of crosstalk by observing that stimulation by M22 was inhibited by some, but not all, IGF1R blocking antibodies as described below. We found evidence for TSH/IGF1 receptor crosstalk in two other human cell systems in culture. In primary cultures of human thyrocytes, Morgan et al. (2016) found that linsitinib inhibited TSHinduced upregulation of the expression of sodium-iodide symporter, which is a critical mediator of thyroid hormone biosynthesis. In U2OS cells, an osteosarcoma epithelial cell line, overexpressing TSHRs, Krieger et al. (2019) showed that knockdown of IGF1R inhibited TSH induced secretion of osteopontin, which is a protein involved in bone homeostasis.

3. The role of ERK and β-Arrestin 1 in TSH/IGF1 receptor crosstalk Models of GPCR-RTK interaction have been proposed in which signalosomes comprised of GPCR, G protein and β-arrestin are in dynamical complexes, which may be influenced by ligand binding, are involved in GPCR/RTK crosstalk (Hilger et al., 2018; Pyne & Pyne, 2011). In GOFs, Krieger et al. (Krieger, Perry, Morgan, Kahaly, & Gershengorn, 2017) demonstrated that the increased potency for TSH seen for HA secretion was also found on ERK1/2 phosphorylation when cells were stimulated simultaneously by TSH and IGF1. β-arrestin 1 and β-arrestin 2 are key elements in the regulation of TSHR-mediated signaling (Kopp, 1997). While β-arrestin 2 plays a predominant role in TSHR-desensitization (Boutin, Eliseeva, Gershengorn, & Neumann, 2014; Frenzel, Voigt, & Paschke, 2006), β-arrestin 1 is primarily involved in activatory TSHR signaling. For example, TSHR stimulation of ERK1/2 phosphorylation was demonstrated to be mediated by β-arrestin 1 (Boutin et al., 2014). Likewise, activation of the IGF1R by IGF1 induces β-arrestin 1-mediated activation of the ERK pathway (Lin, Daaka, & Lefkowitz, 1998). β-arrestin knockdown was shown to inhibit TSH/IGF1 receptor crosstalk (Krieger et al., 2019). Krieger et al. (2019) showed that β-arrestin 1 was also a member of the preformed complex including TSHR and IGF1R and its presence was necessary for complex stability in GOFs, human thyrocytes and U2OS cells. This study used a Proximity Ligation Assay (PLA) to measure colocalization of TSHR and IGF1R (Fig. 4). PLA between TSHR and IGF1R revealed that these receptors co-localized (were within 40 nm of each other) under basal conditions in all three cell types; that is, ligand binding was not necessary for this signaling complex to form (Fig. 4A). This observation confirmed an earlier report of TSHR/IGF1R coimmunoprecipitation (Tsui et al., 2008) and indirectly supported the idea that crosstalk occurs proximal to the receptor, but did not demonstrate the necessity of receptor co-localization to crosstalk. Next, Krieger et al. (Krieger et al., 2019) used PLA to measure TSHR/IGF1R colocalization following knock-down of β-arrestins. In all studied cell types, knock-down of β-arrestin 1 reduced receptor co-localization (Fig. 4B). In thyrocytes and U2OS cells, these effects were partial. In GOFs co-localization of TSHR and IGF1R was completely prevented, coinciding with complete inhibition of functional crosstalk (Fig. 4C). These findings together indicate that TSHR and IGF1R are likely to be in a preformed signaling complex in order to crosstalk and demonstrate that β-arrestin 1 stabilizes this complex in the absence of ligand binding (Fig. 4D). We suggest that a subpopulation of receptors likely form TSHR/IGF1R signaling complexes in any tissue where both receptors are endogenously expressed. What other binding partners are members of this signaling complex and how it is regulated are open questions. 150

Hyaluronan (%TSH)

2

Control

AF305

125 100 75 50 25 0 Basal

TSH

IGF1

Fig. 1. TSH/IGF1 Receptor crosstalk in GOFs is independent of IGF1R ligand generation. One previously described mechanism for receptor transactivation between GPCRs and RTKs occurs when GPCR activation leads to the release of a RTK ligand (Delcourt et al., 2007). In this scenario, preventing RTK ligand from binding to its receptor would inhibit crosstalk. This model was tested in GOFs using an IGF1R blocking antibody, AF305. Here, TSHR was activated by its cognate ligand, TSH, without (gray bars) or with AF305 (black bars). Even though AF305 fully blocked IGF1 binding to and thereby activation of IGF1R, TSH induced hyaluronan secretion was unchanged in the presence of AF305. Thus, TSHR activation did not cause IGF1R activation by releasing IGF1.

Please cite this article as: C.C. Krieger, S. Neumann and M.C. Gershengorn, TSH/IGF1 receptor crosstalk: Mechanism and clinical implications, Pharmacology & Therapeutics, https://doi.org/10.1016/j.pharmthera.2020.107502

C.C. Krieger et al. / Pharmacology & Therapeutics xxx (2020) xxx

Hyaluronan (A.U.)

TSH + IGF1

TSH alone + IGF1 alone

C

on

tr

ol

TSH alone

-10

-9

-8

-7

-6

-5

TSH (log M)

Fig. 2. Synergistic activation of TSHR with simultaneous TSH and IGF1 treatment. Synergistic activation is often described as when the combination of two treatments is greater than the additive effects of each individual treatment. In GOFs, hyaluronan secretion was stimulated with increasing doses of TSH alone (black circles) or TSH with 100 ng/mL IGF1 (gray circles). The maximally effective dose of TSH + IGF1 was greater than TSH alone + IGF1 alone (white and black striped bar). Furthermore, in the presence of IGF1, TSH potency (dotted line) was approximately 100-fold higher than the potency of TSH in the absence of IGF1 (dashed line). This shift in potency was not seen in the reverse situation, the IGF1 dose-response with TSH, indicating that crosstalk originates at TSHR (Place et al., 2017).

4. Crosstalk in the pathogenesis and treatment of Graves' disease As described above, the M22 dose-response for stimulation of HA secretion is biphasic, suggesting that TSHR utilized at least two different signaling pathways to increase HA. Importantly, only one phase was inhibited by an IGF1R antagonist. We showed that M22 binds to TSHR but does not bind to IGF1R (Krieger, Neumann, Marcus-Samuels, & Gershengorn, 2017) and provided additional evidence that immunoglobulins from patients with GO (GO-Igs) do not directly activate IGF1R (Marcus-Samuels et al., 2018). Because M22 was specific for TSHR, we concluded that the linsitinib-sensitive phase originated from TSHRs engaged in crosstalk with IGF1R. Therefore, in the M22-GOF system, one could separately demonstrate effects on IGF1R-dependent and -independent signaling pathways. We used this to characterize different properties of IGF1R blocking antibodies (Krieger et al., 2016). Two different IGF1R antibodies, 1H7 and AF305, were compared in their ability to counter M22 stimulation in GOFs. Both these antibodies had the ability to block binding of IGF1 to IGF1R. Yet only 1H7 was able to inhibit the

3

linsitinib-sensitive phase of the M22 biphasic dose-response. 1H7, but not AF305, could therefore serve as an inhibitor of crosstalk originating from TSHR stimulation. To determine whether M22 was representative of the polyclonal antibodies that comprise the immunoglobulins in the blood of patients with GO (GO-Igs) and thereby determine whether TSH/IGF1 receptor crosstalk was involved in GO-Igs effects on GOFs, we compared the effects of 1H7 and AF305 to inhibit TSHR stimulation by GO-Igs (Krieger et al., 2016). In an attempt to exclude an effect of other active components in the patient's serum samples, Igs were purified from whole serum by thiophilic affinity chromatography, and subsequently the preparations were dialyzed. In the majority of samples, 1H7 partially inhibited GO-Igs stimulation while AF305 had little to no effect. We concluded that GO-Igs produced in these patients could activate crosstalk in TSHR/IGF1R signaling complexes, and activation of this pathway contributed to the pathogenesis of GO. Knowing that GO-Igs activate TSH/IGF1 receptor crosstalk may help us in designing new treatments and in understanding the mechanism (s) of action of new therapies currently being tested. There are advantages to developing combination treatments to target GPCR-RTK signaling platforms as suggested by Pyne & Pyne (Pyne & Pyne, 2011) and recently supported by Place et al. (2017) in GOFs. The question of this study was whether inhibiting TSH/IGF1 receptor crosstalk alone was sufficient for suppressing M22 induction of HA secretion. We found that an IGF1R antibody capable of inhibiting TSHR/IGF1R crosstalk cannot fully inhibit the induction of HA by TSHR-stimulating antibodies because stimulation proceeds via the IGF1R-independent pathway. In contrast, because TSH/IGF1 receptor crosstalk is initiated at TSHR, targeting that receptor with a small molecule antagonist (NCGC00242364) was fully efficacious. An intriguing finding by Place et al. (2017) was that treatment comprising low concentrations of TSHR and IGF1R antagonists was as efficacious as single treatment using a maximally effective concentration of a TSHR antagonist alone that is more effective than antagonism of IGF1R (Fig. 5). In other words, the putative clinically effective dose of each antagonist was reduced when used together (Fig. 6). This finding is significant in light of the fact that single therapy targeting IGF1R has advanced further in clinical trials (Kahaly, 2019; Patel, Yang, & Douglas, 2019) than single therapy targeting TSHR, currently in phase I clinical trials (Furmaniak et al., 2018; Singh, 2016). IGF1R targeted single therapy

Fig. 3. Model of M22-induced TSH/IGF1 receptor crosstalk in GOFs. (A) In GOFs, hyaluronan secretion in response to stimulating TSHR antibody M22 is biphasic (dashed line). In the presence of the IGF1R kinase inhibitor linsitinib, the high-potency phase is lost and the M22 dose-response becomes monophasic (solid line). Therefore, the high-potency phase is IGF1R-dependent and the low-potency phase is IGF1R-independent. (B) In our model, M22 can activate multiple signaling pathways through TSHR. When M22 activates TSH/IGF receptor crosstalk (gray arrows), TSHR uses IGF1R associated down-stream effectors. Alternatively, TSHR can use its own molecular machinery (black arrows). Both pathways converge further downstream to induce hyaluronan secretion (black and gray striped arrow).

Please cite this article as: C.C. Krieger, S. Neumann and M.C. Gershengorn, TSH/IGF1 receptor crosstalk: Mechanism and clinical implications, Pharmacology & Therapeutics, https://doi.org/10.1016/j.pharmthera.2020.107502

4

C.C. Krieger et al. / Pharmacology & Therapeutics xxx (2020) xxx

Fig. 4. TSHR/IGF1R signaling complex depends on β-arrestin 1. TSHR/IGF1R signaling complexes were detected using a Proximity Ligation Assay and visualized with fluorescent microscopy. Orange dots represent PLA signals, and a DNA/RNA stain (green) was used to determine cell number. (A) A representative confocal image of GOFs shows the number of signaling complexes under basal conditions. (B) In GOFs with reduced β-arrestin 1 protein expression, the number of dots is significantly reduced. (C) The bars depict the average Dots/cell from several images. *p b .05 (Krieger et al., 2019). (D) PLA shows that TSHR and IGF1R are within 40 nm of each other, and that distance would allow for a protein complex. A model of a TSHR/IGF1R signaling complex is consistent with the idea that β-arrestin's involvement is likely through an undefined protein complex rather than direct interactions with each receptor.

remain, including the fact that TSHR has not yet been crystallized making the design of small molecule ligands a trial and error process. Nevertheless, because TSHR engages in crosstalk with IGF1R, combination treatments could be designed to improve the therapeutic index by targeting both receptors.

100

Hyaluronan (% GO- Igs)

80 60 40

TSHR Antag + 1H7

1H7 (IC50)

TSHR Antag (IC50)

GO - Igs

0

M22

20 Baseline

with Teprotumumab™ has been effective in treating GO (Mohyi & Smith, 2018). The recent findings suggest that Teprotumumab™, like 1H7, acts to antagonize crosstalk in GOFs. However, since IGF1 signaling is integral to cellular processes in many cell types and IGF1R is expressed in many tissues and has a high degree of homology with the insulin receptor, IGF1R monotherapies have a high likelihood of off-target effects, recently reviewed by Simpson et al. (Simpson, Petnga, Macaulay, Weyer-Czernilofsky, & Bogenrieder, 2017), Osher et al. (Osher & Macaulay, 2019), and Manzella et al. (Manzella et al., 2019). We suggest these effects may be avoided at lower concentrations of Teprotumumab™ if used in combination with a TSHR antagonist (Fig. 6). This would expand the possible repertoire of therapeutic agents. A similar situation exists when considering recent efforts to develop drugs targeting β-arrestin. Because of β-arrestin's now widely accepted role in GPCR biased signaling (recently reviewed in (Bagnato & Rosano, 2019; Gurevich & Gurevich, 2019; Seyedabadi, Ghahremani, & Albert, 2019; Smith, Lefkowitz, & Rajagopal, 2018), this protein has become an attractive target for a variety of disorders involving GPCRs (Bond, Lucero Garcia-Rojas, Hegde, & Walker, 2019; van Gastel et al., 2018). However, β-arrestin is also ubiquitously expressed, therefore therapies targeted at β-arrestin have the potential to produce adverse side effects. In an analogous fashion, combination GPCR/β-arrestin therapies could increase specificity to tissues where only the two proteins of interest are expressed and lower possible adverse effects. In the long-term, treatments targeting TSHR will likely be better approaches for treatments of Graves' disease. However, much work needs to be done before these ideas can be translated to patient care. This model heavily relies on in vitro studies, which further rely on the availability of research tools such as IGF1R and TSHR inhibitory and stimulating antibodies. Efforts to expand the molecular toolkit are in progress (Pearce et al., 2019). While advances have been made, technical hurdles

+ GO-Igs Fig. 5. Combination treatments efficiently inhibit TSH/IGF1 receptor crosstalk. Like M22, antibodies purified from six different GO patient sera (GO-Igs) induce hyaluronan secretion through TSH/IGF1 receptor crosstalk in GOFs (dark gray bars). Inhibition of TSHR signaling or blocking TSHR/IGF1R crosstalk with a 50% inhibitory concentration (IC50) dose of a small molecule TSHR antagonist (TSHR antag) or the IGF1R antibody, 1H7, respectively, inhibits hyaluronan secretion, but inhibition is not complete (light gray bars). Yet, these same doses, when combined, are fully efficacious (black bar) (Place et al., 2017).

Please cite this article as: C.C. Krieger, S. Neumann and M.C. Gershengorn, TSH/IGF1 receptor crosstalk: Mechanism and clinical implications, Pharmacology & Therapeutics, https://doi.org/10.1016/j.pharmthera.2020.107502

C.C. Krieger et al. / Pharmacology & Therapeutics xxx (2020) xxx

Fig. 6. Model of inhibition of TSH/IGF1 receptor crosstalk to treat Graves' disease. This model is based primarily on studies with Graves' orbital fibroblasts in cell culture. TSHRstimulating antibodies, M22 or GO-Igs, activate either IGF1R-dependent (gray arrows) or IGF1R-independent (black arrows) signaling pathways, both of which converge to induce hyaluronan secretion (black/gray striped arrow). Destabilizing crosstalk with an IGF1R inhibitory antibody (IGF1R Ab) or kinase inhibitor alone may reduce hyaluronan secretion but cannot inhibit the IGF1R-independent pathway(s) activated by TSHR. Preventing activation of TSHR with either an antagonist or inhibitory antibody blocks both pathways.

Declaration of Competing Interest The authors declare that there are no conflicts of interest. Acknowledgements This work was funded by the Intramural Reserch Program of the National Institue of Diabetes and Digestive and Kidney Diseases, National Institutes of Health Z01 DK011006. References Bagnato, A., & Rosano, L. (2019). New routes in GPCR/beta-Arrestin-driven Signaling in Cancer progression and metastasis. Frontiers in Pharmacology 10, 114. Bond, R. A., Lucero Garcia-Rojas, E. Y., Hegde, A., & Walker, J. K. L (2019). Therapeutic potential of targeting ss-Arrestin. Frontiers in Pharmacology 10, 124. Boutin, A., Eliseeva, E., Gershengorn, M. C., & Neumann, S. (2014). Beta-Arrestin-1 mediates thyrotropin-enhanced osteoblast differentiation. The FASEB Journal 28, 3446–3455. Chen, H., Mester, T., Raychaudhuri, N., Kauh, C. Y., Gupta, S., Smith, T. J., & Douglas, R. S. (2014). Teprotumumab, an IGF-1R blocking monoclonal antibody inhibits TSH and IGF-1 action in fibrocytes. The Journal of Clinical Endocrinology and Metabolism 99, E1635–E1640. Delcourt, N., Bockaert, J., & Marin, P. (2007). GPCR-jacking: From a new route in RTK signalling to a new concept in GPCR activation. Trends in Pharmacological Sciences 28, 602–607. Di Liberto, V., Mudo, G., & Belluardo, N. (2019). Crosstalk between receptor tyrosine kinases (RTKs) and G protein-coupled receptors (GPCR) in the brain: Focus on heteroreceptor complexes and related functional neurotrophic effects. Neuropharmacology 152, 67–77. Eggo, M. C., Bachrach, L. K., & Burrow, G. N. (1990). Interaction of TSH, insulin and insulinlike growth factors in regulating thyroid growth and function. Growth Factors 2, 99–109. Frenzel, R., Voigt, C., & Paschke, R. (2006). The human thyrotropin receptor is predominantly internalized by beta-arrestin 2. Endocrinology 147, 3114–3122. Furmaniak, J., Ryder, M. M. R, Castro, M. R., Wentworth, M., Algeciras, A., Morris, J. C., ... Rees Smith, B. (2018). Blocking the TSH receptor with the human monoclonal autoantibody K1-70 improves graves’ ophthalmopathy and aids control of advanced follicular thyroid carcinoma – results of long-term treatment under the first in human single patient expanded use therapy. 41st annual meeting of the european thyroid association. Vol. 7. Newcastle, England. van Gastel, J., Hendrickx, J. O., Leysen, H., Santos-Otte, P., Luttrell, L. M., Martin, B., & Maudsley, S. (2018). Beta-Arrestin based receptor Signaling paradigms: Potential therapeutic targets for complex age-related disorders. Frontiers in Pharmacology 9, 1369. Gurevich, V. V., & Gurevich, E. V. (2019). GPCR Signaling regulation: The role of GRKs and Arrestins. Frontiers in Pharmacology 10, 125. Hilger, D., Masureel, M., & Kobilka, B. K. (2018). Structure and dynamics of GPCR signaling complexes. Nature Structural & Molecular Biology 25, 4–12. Hoa, N., Tsui, S., Afifiyan, N. F., Sinha Hikim, A., Li, B., Douglas, R. S., & Smith, T. J. (2012). Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves’ disease: Apparent role of ADAM17. PLoS One 7, e34173.

5

Kahaly, G. J. (2019). Immunotherapies for thyroid eye disease. Current Opinion in Endocrinology, Diabetes, and Obesity 26, 250–255. Kenakin, T., Agnati, L. F., Caron, M., Fredholm, B., Guidoli, D., Kobilka, B., ... Fuxe, K. (2010). International workshop at the Nobel forum, Karolinska Institutet on G proteincoupled receptors: Finding the words to describe monomers, oligomers, and their molecular mechanisms and defining their meaning. Can a consensus be reached? Journal of Receptor and Signal Transduction Research 30, 284–286. Kimura, T., Van Keymeulen, A., Golstein, J., Fusco, A., Dumont, J. E., & Roger, P. P. (2001). Regulation of thyroid cell proliferation by TSH and other factors: A critical evaluation of in vitro models. Endocrine Reviews 22, 631–656. Kopp, P. (1997). Key elements involved in the negative regulation of the TSH receptor: G protein-coupled receptor kinases, arrestin and inducible cAMP early repressor. European Journal of Endocrinology 136, 269–270. Krieger, C. C., Boutin, A., Jang, D., Morgan, S. J., Banga, J. P., Kahaly, G. J., ... Gershengorn, M. C. (2019). Arrestin-beta-1 physically scaffolds TSH and IGF1 receptors to enable crosstalk. Endocrinology 160, 1468–1479. Krieger, C. C., Neumann, S., Marcus-Samuels, B., & Gershengorn, M. C. (2017). TSHR/IGF1R cross-talk, not IGF-1R stimulating antibodies, mediates Graves’ Ophthalmopathy pathogenesis. Thyroid 27, 746–747. Krieger, C. C., Neumann, S., Place, R. F., Marcus-Samuels, B., & Gershengorn, M. C. (2015). Bidirectional TSH and IGF-1 receptor cross talk mediates stimulation of hyaluronan secretion by Graves’ disease immunoglobins. The Journal of Clinical Endocrinology and Metabolism 100, 1071–1077. Krieger, C. C., Perry, J. D., Morgan, S. J., Kahaly, G. J., & Gershengorn, M. C. (2017). TSH/IGF1 receptor cross-talk rapidly activates extracellular signal-regulated kinases in multiple cell types. Endocrinology 158, 3676–3683. Krieger, C. C., Place, R. F., Bevilacqua, C., Marcus-Samuels, B., Abel, B. S., Skarulis, M. C., ... Gershengorn, M. C. (2016). TSH/IGF-1 receptor cross talk in Graves’ Ophthalmopathy pathogenesis. The Journal of Clinical Endocrinology and Metabolism 101, 2340–2347. Kumar, S., Iyer, S., Bauer, H., Coenen, M., & Bahn, R. S. (2012). A stimulatory thyrotropin receptor antibody enhances hyaluronic acid synthesis in graves’ orbital fibroblasts: Inhibition by an IGF-I receptor blocking antibody. The Journal of Clinical Endocrinology and Metabolism 97, 1681–1687. Lin, F. T., Daaka, Y., & Lefkowitz, R. J. (1998). Beta-arrestins regulate mitogenic signaling and clathrin-mediated endocytosis of the insulin-like growth factor I receptor. The Journal of Biological Chemistry 273, 31640–31643. Manzella, L., Massimino, M., Stella, S., Tirro, E., Pennisi, M. S., Martorana, F., ... Vigneri, P. (2019). Activation of the IGF Axis in thyroid Cancer: Implications for tumorigenesis and treatment. International Journal of Molecular Sciences 20. Marcus-Samuels, B., Krieger, C. C., Boutin, A., Kahaly, G. J., Neumann, S., & Gershengorn, M. C. (2018). Evidence that Graves’ Ophthalmopathy immunoglobulins do not directly activate IGF-1 receptors. Thyroid 28, 650–655. Mohyi, M., & Smith, T. J. (2018). IGF1 receptor and thyroid-associated ophthalmopathy. Journal of Molecular Endocrinology 61, T29–T43. Morgan, S. J., Neumann, S., Marcus-Samuels, B., & Gershengorn, M. C. (2016). Thyrotropin and insulin-like growth factor 1 receptor crosstalk Upregulates sodium-iodide Symporter expression in primary cultures of human Thyrocytes. Thyroid 26, 1794–1803. Osher, E., & Macaulay, V. M. (2019). Therapeutic targeting of the IGF Axis. Cells 8. Patel, A., Yang, H., & Douglas, R. S. (2019). A new era in the treatment of thyroid eye disease. American Journal of Ophthalmology 208, 281–288. Pearce, S. H. S, Dayan, C., Wraith, D. C., Barrell, K., Olive, N., Jansson, L., ... Kahaly, G. J. (2019). Antigen-specific immunotherapy with Thyrotropin receptor peptides in Graves’ hyperthyroidism: A phase I study. Thyroid 29, 1003–1011. Place, R. F., Krieger, C. C., Neumann, S., & Gershengorn, M. C. (2017). Inhibiting thyrotropin/insulin-like growth factor 1 receptor crosstalk to treat Graves’ ophthalmopathy: Studies in orbital fibroblasts in vitro. British Journal of Pharmacology 174, 328–340. Pyne, N. J., & Pyne, S. (2011). Receptor tyrosine kinase-G-protein-coupled receptor signalling platforms: Out of the shadow? Trends in Pharmacological Sciences 32, 443–450. Sanders, J., Evans, M., Premawardhana, L. D., Depraetere, H., Jeffreys, J., Richards, T., ... Rees Smith, B. (2003). Human monoclonal thyroid stimulating autoantibody. Lancet 362, 126–128. Santisteban, P., Kohn, L. D., & Di Lauro, R. (1987). Thyroglobulin gene expression is regulated by insulin and insulin-like growth factor I, as well as thyrotropin, in FRTL-5 thyroid cells. The Journal of Biological Chemistry 262, 4048–4052. Seyedabadi, M., Ghahremani, M. H., & Albert, P. R. (2019). Biased signaling of G protein coupled receptors (GPCRs): Molecular determinants of GPCR/transducer selectivity and therapeutic potential. Pharmacology & Therapeutics 200, 148–178. Simpson, A., Petnga, W., Macaulay, V. M., Weyer-Czernilofsky, U., & Bogenrieder, T. (2017). Insulin-like growth factor (IGF) pathway targeting in cancer: Role of the IGF Axis and opportunities for future combination studies. Targeted Oncology 12, 571–597. Singh, D. (2016). K1–70 - a study in subjects with Graves’ disease. ClinicalTrials.gov. https://ClinicalTrials.gov/show/NCT02904330. Smith, J. S., Lefkowitz, R. J., & Rajagopal, S. (2018). Biased signalling: From simple switches to allosteric microprocessors. Nature Reviews. Drug Discovery 17, 243–260. Tramontano, D., Cushing, G. W., Moses, A. C., & Ingbar, S. H. (1986). Insulin-like growth factor-I stimulates the growth of rat thyroid cells in culture and synergizes the stimulation of DNA synthesis induced by TSH and Graves’-IgG. Endocrinology 119, 940–942. Tsui, S., Naik, V., Hoa, N., Hwang, C. J., Afifiyan, N. F., Sinha Hikim, A., ... Smith, T. J. (2008). Evidence for an association between thyroid-stimulating hormone and insulin-like growth factor 1 receptors: A tale of two antigens implicated in Graves’ disease. Journal of Immunology 181, 4397–4405.

Please cite this article as: C.C. Krieger, S. Neumann and M.C. Gershengorn, TSH/IGF1 receptor crosstalk: Mechanism and clinical implications, Pharmacology & Therapeutics, https://doi.org/10.1016/j.pharmthera.2020.107502