Review
Engineering receptors activated solely by synthetic ligands (RASSLs)

https://doi.org/10.1016/S0165-6147(00)01743-0Get rights and content

Abstract

The functional and molecular diversity of G-protein-coupled receptors presents a significant challenge to understanding the connection between a single receptor signaling pathway and a specific physiological or pathological response. To gain control over the timing and specificity of a G-protein signal, receptors activated solely by synthetic ligands (RASSLs) have been developed. These engineered receptors no longer respond to endogenous peptides, but can still be activated by a specific small-molecule drug. Further control over the location of the signal can be achieved by using RASSLs in conjunction with tissue-specific expression systems in vivo. Existing RASSLs have clarified the role of Gi signaling in cardiac physiology and are currently being used to study cardiomyopathy, muscle remodeling, sensory transduction and complex neurobehavioral responses.

Section snippets

RASSL construction

For our first RASSL, we modified the KOR. This receptor responds to endogenous peptides and signals via the well-characterized Gi pathway. Because of the importance of this receptor family for pain modulation, the pharmaceutical industry has developed many high-affinity opioid receptor agonists. Small-molecule ligands of the KOR are structurally distinct from the endogenous peptide ligands, including dynorphin 11, 12. Unlike mu or delta opioid receptor agonists, KOR agonists are nonaddictive 13.

Control of RASSL expression in vivo

The expression of RASSLs in specific tissues of whole animals is a potentially valuable tool for studying both normal and pathological signaling cascades. To take full advantage of the signaling control offered by a RASSL, it is also important to control the timing and location of RASSL expression in vivo. To achieve this, we have used the tetracycline-controlled inducible expression system (tet system) developed by Gossen and Bujard 20. Briefly, transgene expression is driven by a minimal

Controlling RASSL signaling in vivo

When the receptor is expressed, it should be functionally silent until the administration of spiradoline stimulates the RASSL and activates signaling pathways rapidly and specifically. Gi signaling in the heart has been shown to reduce heart rate 26. Therefore, when Ro1 is expressed in the heart (using the α-myosin heavy chain promoter), receptor activation can be studied by simply measuring changes in heart rate. Wild-type animals have few KORs in the heart and therefore show no change in

Detecting RASSLs in vivo

For many experiments that use RASSLs to study the role of Gi signaling in vivo, it is essential to pinpoint the receptor localization, sometimes down to the subcellular level. To this end, we have developed several tagged RASSLs*.

Small epitope tags have been added to the N-terminus of the RASSL protein for subsequent detection with antibody staining. Common tags such as hemagglutinin (HA)

Applications of RASSL technology

Acute activation of RASSLs expressed in discrete tissues or cell types will allow researchers to probe the role of receptor-mediated Gi signaling in normal cell function. Correspondingly, extended activation of RASSLs can be used to explore disease models related to hyperactive G-protein signaling.

Tissue-specific expression and activation of RASSLs

To date, RASSLs have been expressed in multiple tissues of transgenic mice (Table 1), including heart, liver, salivary glands, smooth muscle, adipose tissue and specific brain regions. Ro1-mediated Gi activation has been shown to slow heart rate 5. RASSLs are being used to study the effects of G proteins on smooth muscle contractility, and mouse lines that express RASSLs in specific brain regions are being used to study how activation of Gi signaling in specific brain nuclei can affect

Future directions: new RASSLs

Existing RASSLs will allow study of the effects of Gi-mediated signaling in specific tissues under specific circumstances. In the future, RASSLs could be made to control all the major G-protein signaling pathways, individually or in combination (Gs, Gi, Gq, G12, G13). These new RASSLs can be developed using many of the same principles used to develop the existing Gi-coupled RASSLs. The ideal RASSL would have orally available, nanomolar affinity agonists and antagonists that are safe for

References (34)

  • G.J. Kilpatrick

    7TM receptors: the splicing on the cake

    Trends Pharmacol. Sci.

    (1999)
  • C.D. Strader

    Structure and function of G protein-coupled receptors

    Annu. Rev. Biochem.

    (1994)
  • A.M. Spiegel

    Receptor–effector coupling by G proteins: implications for normal and abnormal signal transduction

    Endocrinol. Rev.

    (1992)
  • D. Hoyer

    VII. International union of pharmacology classification of receptors for 5-hydroxytryptamine (serotonin)

    Pharmacol. Rev.

    (1994)
  • P. Coward

    Controlling signaling with a specifically designed Gi-coupled receptor

    Proc. Natl. Acad. Sci. U. S. A.

    (1998)
  • C.H. Redfern

    Conditional expression and signaling of a specifically designed Gi-coupled receptor in transgenic mice

    Nat. Biotechnol.

    (1999)
  • C.H. Redfern

    Conditional expression of a Gi-coupled receptor causes ventricular conduction delay and a lethal cardiomyopathy

    Proc. Natl. Acad. Sci. U. S. A.

    (2000)
  • Cited by (48)

    • CNO Administration Increases Dopamine and Glutamate in the Medial Prefrontal Cortex of Wistar Rats: Further Concerns for the Validity of the CNO-activated DREADD Procedure

      2022, Neuroscience
      Citation Excerpt :

      In contrast, the ability to modulate G-coupled protein receptors (GCPRs) has lagged because of the lack of specificity of compounds for these targets. In 2000, a novel procedure created receptors activated solely by synthetic ligands (RASSLs) was developed to target select GCPRs (Bishop et al., 2000; Scearce-Levie et al., 2001). The RASSLs procedure involved the mutation of native receptors with the desired goal to create a receptor with functions identical to the native receptor.

    • Novel designer receptors to probe GPCR signaling and physiology

      2013, Trends in Pharmacological Sciences
      Citation Excerpt :

      Thus, to better understand the physiological and, potentially, pathophysiological roles of specific GPCR signaling pathways, an experimental system that allows the activation (in vivo) of a particular GPCR in a cell type- or tissue-specific fashion would be highly desirable. In this regard, the development of designer GPCRs referred to as RASSLs (receptors activated solely by synthetic ligand) has provided important novel tools to study the in vivo relevance of distinct GPCR signaling pathways [3,4]. In most cases, these first-generation RASSLs represent engineered GPCRs that can be efficiently activated by a preexisting synthetic drug but are much less sensitive to activation by its endogenous ligand.

    • Pharmacological profile of engineered 5-HT<inf>4</inf> receptors and identification of 5-HT<inf>4</inf> receptor-biased ligands

      2013, Brain Research
      Citation Excerpt :

      To overcome this problem, different GPCRs have been engineered over the last decade. First, receptors activated solely by synthetic ligands (RASSLs) were generated by site-directed mutagenesis and Gq-, Gs- and Gi-coupled RASSLs are now available (Conklin et al., 2008; Coward et al., 1998; Scearce-Levie et al., 2001). These GPCRs are insensitive to their endogenous ligand and can be activated only by specific synthetic ligands.

    • Rapid and reversible chemical inactivation of synaptic transmission in genetically targeted neurons

      2005, Neuron
      Citation Excerpt :

      Silencing by expression of K+ or Cl− channels has been useful in invertebrate and cultured neurons (Burrone et al., 2002; Nitabach et al., 2002; Paradis et al., 2001; White et al., 2001), but has yielded mixed results in the mammalian brain in vivo (Nadeau et al., 2000; Sutherland et al., 1999). To introduce temporal control, K+ or Cl− channels have been coupled to ligands without endogenous receptors (Cully et al., 1994; Lechner et al., 2002; Li et al., 2002; Scearce-Levie et al., 2001; Slimko and Lester, 2003; Slimko et al., 2002), but these promising approaches have as yet only been demonstrated in vitro. An elegant alternative involves blocking sodium channels by expression of membrane-tethered toxins (Ibanez-Tallon et al., 2004).

    • DREADDs in Epilepsy Research: Network-Based Review

      2022, Frontiers in Molecular Neuroscience
    View all citing articles on Scopus
    View full text