Abstract

Due to their low expression levels, complex multi-pass transmembrane structure, and the current lack of highly specific antibodies, the assessment of endogenous G protein-coupled receptors (GPCRs) remains challenging. While most of the research regarding their functions was performed in heterologous systems overexpressing the receptor, recent advances in genetic engineering methods have allowed the generation of several unique mouse models. These animals proved to be useful to investigate numerous aspects underlying the physiological functions of GPCRs, including their endogenous expression, distribution, interactome, and trafficking processes. Given their significant pharmacological importance and central roles in the nervous system, opioid peptide receptors (OPr) are often referred to as prototypical receptors for the study of GPCR regulatory mechanisms. Although only a few GPCR knock-in mouse lines have thus far been generated, OPr are strikingly well represented with over 20 different knock-in models, more than half of which were developed within the last 5 years. In this review, we describe the arsenal of OPr (mu-, delta-, and kappa-opioid), as well as the opioid-related nociceptin/orphanin FQ (NOP) receptor knock-in mouse models that have been generated over the past years. We further highlight the invaluable contribution of such models to our understanding of the in vivo mechanisms underlying the regulation of OPr, which could be conceivably transposed to any other GPCR, as well as the limitations, future perspectives, and possibilities enabled by such tools.

Highlights

  • Characterized by a common topology exhibiting an extracellular N-terminal domain, seven hydrophobic membrane α-helices and a cytosolic C-terminus, G protein-coupled receptors (GPCR) form the largest family of transmembrane proteins (Hauser et al, 2017)

  • Ozawa et al (2015) have mapped the expression of the nociceptin receptor (NOPr)-enhanced green fluorescent protein (eGFP) in the spinal cord, dorsal root ganglia (DRG) neurons and numerous brain regions involved in both pain and reward (e.g., nucleus accumbens (NAc), ventral tegmental area (VTA), medial habenula (MHb), amygdala, hippocampus and interpeduncular nucleus (IPN)), which supports a role for this receptor in such circuitries

  • In a complete Freund’s adjuvant (CFA)-induced chronic pain model, our results indicated that the antihyperalgesic effects of deltorphin II, a delta-opioid receptor (DOPr) specific agonist, were partially reinstated 6 weeks post viral infection, thereby supporting a role for DOPr localized on primary afferents in the control of pain induced by a thermal stimulus (Degrandmaison et al, 2020)

Read more

Summary

Introduction

Characterized by a common topology exhibiting an extracellular N-terminal domain, seven hydrophobic membrane α-helices and a cytosolic C-terminus, G protein-coupled receptors (GPCR) form the largest family of transmembrane proteins (Hauser et al, 2017). A novel MOPr-Venus-YFP (yellow FP) KI mouse line was generated by homologous recombination, resulting in the expression of the MOPr-Venus-YFP replacing the native murine receptor, in order to monitor the agonist-induced differential trafficking of the receptor (Figure 1G; Ehrlich et al, 2019).

Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call