Elsevier

European Journal of Pharmacology

Volume 763, Part B, 15 September 2015, Pages 184-190
European Journal of Pharmacology

Biased agonism at kappa opioid receptors: Implication in pain and mood disorders

https://doi.org/10.1016/j.ejphar.2015.07.018Get rights and content

Abstract

The kappa opioid receptor (k receptor) and its endogenous ligand dynorphin have received significant attention due to their involvement in pathophysiology of mood disorders, drug addiction, psychotic disorders and pain. Multiple lines of evidences suggest that the k receptor modulates overlapping neurocircuits connecting brainstem monoaminergic nuclei with forebrain limbic structures and thereby regulates neurobiological effects of stress and psychostimulants. The emerging concept of “biased agonism” (also known as functional selectivity) for G Protein Coupled Receptor (GPCR) ligands have provided new insights into overall response generated by a ligand, which could be exploited for drug discovery. According to this concept, every ligand possesses the unique ability (coded in its structure) that dictates distinct signalling pattern, and consequently beneficial or adverse response. Although still a long way to comprehend the clinical potential of biased GPCR ligands, such ligand could be vital pharmacological probes. This article highlights various lines of evidence, which indicates different ligands of k receptor as “biased”, and their potential implications in mood and pain disorders.

Introduction

The G Protein Coupled Receptors (GPCRs) are the largest receptor class in the human genome (Allen and Roth, 2011), and known to modulate almost every human physiological function. Due to being involved in such diverse physiological processes, GPCRs are the most frequently targeted receptor class for therapeutic interventions (Ma and Zemmel, 2002). However, the development of GPCR selective drugs is challenging due to following reasons: 1—high degree of homology among many receptors which regulates diverse physiologic functions; 2—one GPCR may couple to more than one type of G proteins; 3—allosteric modulation of receptor signalling via biophysical interactions with small molecules and other proteins present in the microenvironment. Thus, the predominant signalling pattern of a GPCR may differ from cell to cell in various tissues and organs.

Classically, drug development programs targeting GPCRs have focused on the concepts of agonism and antagonism of one receptor target, in which a ligand (agonist or antagonist) upon binding the receptor, stabilizes it in one conformation and dictates the same nature of the downstream effector signalling, and thus ligand efficacy in most of the systems. However, over the last decade, the emerging concept of “biased agonism”, also called as “functional selectivity” have revealed that the nature of GPCR signalling is not so rigid (Kenakin, 1995) and that ligand structure can direct (bias) signal output by stabilizing active receptor states in different proportions than the endogenous ligand. Thus, a biased or a functionally selective ligand is a novel chemical entity that holds the unique ability to qualitatively guide GPCR signalling, leading to distinct efficacy profile determined by ligand structure. Actually, the classical models of allosterism had already predicted the existence of multiple conformational states in the absence of ligand as a fundamental characteristic of allosteric proteins (Monod et al., 1965). The recently solved GPCRs structure support this previous theoretical notion that GPCRs exist in several micro-conformations and different ligands can stabilize different conformations favouring distinct signalling profiles (Deupi and Kobilka, 2010, Wacker et al., 2013). Furthermore, receptor interacting proteins, such as β-arrestins and G proteins, can allosterically modulate agonist binding affinity and therefore receptor conformations (Nygaard et al., 2013). Thus, bidirectional modulation of receptor conformation from both the ligand and interacting proteins regulate final outcome-physiological/pharmacological response. Finally, the promise of “biased agonism” lies in its ability to produce therapeutically beneficial signals while minimizing adverse effects. Due to the prevailing notion in the field that “biased agonists” might have superior therapeutic benefits, effort for many GPCR targets for drug discovery and developments have been revitalized.

Opioids have been used since ancient times for the treatment of pain and other human ailments (Brownstein, 1993), and are still the most effective and widely used analgesics. Most of the opioid analgesics are agonists of the mu (µ), delta (δ) and kappa (κ) opioid receptors (also known as µ receptor, δ receptor, and k receptor, respectively). Opioid receptors are activated by a family of endogenous peptides to inhibit neuronal activity as they are coupled with inhibitory G proteins (Gαi/o) in physiological conditions. Although opioid receptors are the most widely known therapeutic targets for the treatment of acute as well as chronic pain conditions, their clinical use is constrained by adverse side effects, such as development of tolerance and addiction (Williams et al., 2013). Therefore, improving the side effect profile and reducing the development of analgesic tolerance have remained major goals in the opioid receptor field. The k receptor belongs to the opioid system, a neuromodulatory system that is widely expressed throughout the central and peripheral nervous systems. Among opioid peptides, dynorphins (encoded by the Pdyn gene) primarily activate the k receptor and have very low affinity for µ or δ receptor. On the other hand, the other opioid peptides—endorphin and enkephalins, exhibit very low affinity with k receptor. Therefore, the dynorphin/k receptor signalling pathway forms a distinct process within the opioid system (Chavkin et al., 1982, Goldstein et al., 1979). In contrast to µ receptor and δ receptor agonists, k receptor agonists have long been recognized to be analgesics with no addiction and tolerance liability. However, almost all k receptor agonists cause dysphoria, anhedonia, and hallucinations (Carlezon et al., 1998, Pfeiffer et al., 1986, Roth et al., 2002). The present review is mainly focused on various lines of evidence that indicate different ligands of k receptor as “biased”, and their potential implications in mood and pain disorders.

Section snippets

Biased agonism at k receptor

The dynorphin/k receptor system is implicated in several psychiatric conditions such as depression, anxiety, drug addiction and schizophrenia (Bruchas et al., 2011, Schindler et al., 2012, Tejeda et al., 2013). High levels of k receptor expression were observed using the human genomic sequence analysis and RT-PCR technology in almost all regions of the human and rodent brain (ventral tegmental area, prefrontal cortex, claustrum, hippocampus, striatum, amygdala, locus coeruleus, dorsal raphe,

Quantification of “Bias”: challenges and limitations

Even though biased agonism offers the potential of better therapeutics, there are several limitations for its detection, quantification and translation into various physiological responses. The observations of ligand-specific activation of different effector system or second messenger system, were originally made in the 1980s (Gee and Yamamura, 1983). However, only in the last one decade this has become an established dimension of GPCR signaling (Reiter et al., 2012). Most of the studies, until

Implication of k receptor biased agonism in mood disorders

Despite decades of extensive research, the molecular and cellular mechanisms of mood disorders remain unclear. Research on mood and affective states had mainly focused on the roles of brain systems containing monoamines, such as dopamine (DA), norepinephrine (NE), and serotonin (5-hydroxytryptamine [5HT]) (Di Chiara and Imperato, 1988, Koch et al., 2002, Ritz et al., 1987). Although, historically opioids have received far less interest than monoamine systems, essential role of endogenous opioid

Implication of k receptor biased agonism in pain disorders

Pain is an unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage. Peripheral nerve injury elicits abnormal pain characterized by allodynia, where generally non-noxious stimuli (mild warming, cooling or touch) induces pain, and hyperalgesia, where noxious stimuli (skin heating, cooling or strong mechanical stimuli) are perceived as more painful (Wang et al., 2001). Allodynia and hyperalgesia experienced in neuropathic

Concluding remarks

In the present review, we have highlighted the accumulating evidences supporting the role of k receptor/dynorphin system in mood and pain processing. We have also described how k receptor is perfectly positioned to effectively modulate several psychiatric and pain disorders. Previously, major aspirations in the GPCR field had been to identify distinct signalling pathways that may operate to control specific behavioural responses. Future studies using ¯ receptor ligands should combine

Acknowledgments

The authors thank Mrs Deepmala and Mrs. Swati N. Yadav for critically reading this manuscript and the Department of Science and Technology (Govt of India) for research grant (Ramanujan Fellowship to PNY) and University Grant commission of India for Fellowship (JRF and SRF to SD).

References (95)

  • C. Hoffmann et al.

    Agonist-selective, receptor-specific interaction of human P2Y receptors with beta-arrestin-1 and -2

    J. Biol. Chem.

    (2008)
  • K.C. Kajander et al.

    Dynorphin increases in the dorsal spinal cord in rats with a painful peripheral neuropathy

    Peptides

    (1990)
  • T. Kenakin

    Agonist-receptor efficacy. II. Agonist trafficking of receptor signals

    Trends Pharmacol. Sci.

    (1995)
  • T. Kenakin et al.

    Replicated, replicable and relevant-target engagement and pharmacological experimentation in the 21st century

    Biochem. Pharmacol.

    (2014)
  • T. Kenakin et al.

    Defining and characterizing drug/compound function

    Biochem. Pharmacol.

    (2014)
  • S. Koch et al.

    R-fluoxetine increases extracellular DA, NE, as well as 5-HT in rat prefrontal cortex and hypothalamus: an in vivo microdialysis and receptor binding study

    Neuropsychopharmacology

    (2002)
  • T.M. Laughlin et al.

    Spinally administered dynorphin A produces long-lasting allodynia: involvement of NMDA but not opioid receptors

    Pain

    (1997)
  • Y.W. Lee et al.

    Systemic and supraspinal, but not spinal, opiates suppress allodynia in a rat neuropathic pain model

    Neurosci. Lett.

    (1995)
  • P. Leone et al.

    Morphine-dopamine interaction: ventral tegmental morphine increases nucleus accumbens dopamine release

    Pharmacol. Biochem. Behav.

    (1991)
  • T.P. Malan et al.

    Extraterritorial neuropathic pain correlates with multisegmental elevation of spinal dynorphin in nerve-injured rats

    Pain

    (2000)
  • J.P. McLaughlin et al.

    Prolonged kappa opioid receptor phosphorylation mediated by G-protein receptor kinase underlies sustained analgesic tolerance

    J. Biol. Chem.

    (2004)
  • J.P. McLaughlin et al.

    Phosphorylation of a carboxyl-terminal serine within the kappa-opioid receptor produces desensitization and internalization

    J. Biol. Chem.

    (2003)
  • J. Monod et al.

    On the nature of allosteric transitions: a plausible model

    J. Mol. Biol.

    (1965)
  • M. Narita et al.

    Regulations of opioid dependence by opioid receptor types

    Pharmacol. Ther.

    (2001)
  • R. Nygaard et al.

    The dynamic process of beta(2)-adrenergic receptor activation

    Cell

    (2013)
  • R. Przewlocki et al.

    Prodynorphin gene expression in spinal cord is enhanced after traumatic injury in the rat

    Brain Res.

    (1988)
  • M.L. Rives et al.

    6′-Guanidinonaltrindole (6′-GNTI) is a G protein-biased kappa-opioid receptor agonist that inhibits arrestin recruitment

    J. Biol. Chem.

    (2012)
  • C.F. Robles et al.

    Effects of kappa opioid receptors on conditioned place aversion and social interaction in males and females

    Behav. Brain Res.

    (2014)
  • S.S. Schattauer et al.

    Ligand directed signaling differences between rodent and human kappa-opioid receptors

    J. Biol. Chem.

    (2012)
  • C.L. Schmid et al.

    Functional selectivity of 6′-guanidinonaltrindole (6′-GNTI) at kappa-opioid receptors in striatal neurons

    J. Biol. Chem.

    (2013)
  • A.N. Schoffelmeer et al.

    Mu-, delta- and kappa-opioid receptor-mediated inhibition of neurotransmitter release and adenylate cyclase activity in rat brain slices: studies with fentanyl isothiocyanate

    Eur. J. Pharmacol.

    (1988)
  • T.S. Shippenberg et al.

    Dynorphin and the pathophysiology of drug addiction

    Pharmacol. Ther.

    (2007)
  • S.H. Snyder et al.

    Historical review: opioid receptors

    Trends Pharmacol. Sci.

    (2003)
  • M. Tallent et al.

    The cloned kappa opioid receptor couples to an N-type calcium current in undifferentiated PC-12 cells

    Neuroscience

    (1994)
  • J.L. Whistler et al.

    Functional dissociation of mu opioid receptor signaling and endocytosis: implications for the biology of opiate tolerance and addiction

    Neuron

    (1999)
  • J.A. Allen et al.

    Strategies to discover unexpected targets for drugs active at G protein-coupled receptors

    Annu. Rev. Pharmacol. Toxicol.

    (2011)
  • J.A. Allen et al.

    Discovery of beta-arrestin-biased dopamine D2 ligands for probing signal transduction pathways essential for antipsychotic efficacy

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

    (2011)
  • U. Arvidsson et al.

    The kappa-opioid receptor is primarily postsynaptic: combined immunohistochemical localization of the receptor and endogenous opioids

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

    (1995)
  • P.M. Beardsley et al.

    Differential effects of the novel kappa opioid receptor antagonist, JDTic, on reinstatement of cocaine-seeking induced by footshock stressors vs cocaine primes and its antidepressant-like effects in rats

    Psychopharmacology

    (2005)
  • K.A. Berg et al.

    Effector pathway-dependent relative efficacy at serotonin type 2A and 2C receptors: evidence for agonist-directed trafficking of receptor stimulus

    Mol. Pharmacol.

    (1998)
  • J.W. Black et al.

    Operational models of pharmacological agonism

    Proc. R. Soc. Lond. B Biol. Sci.

    (1983)
  • M.J. Brownstein

    A brief history of opiates, opioid peptides, and opioid receptors

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

    (1993)
  • M.R. Bruchas et al.

    Stress-induced p38 mitogen-activated protein kinase activation mediates kappa-opioid-dependent dysphoria

    J. Neurosci.

    (2007)
  • W.A. Carlezon et al.

    Regulation of cocaine reward by CREB

    Science

    (1998)
  • G.V. Carr et al.

    Antidepressant-like effects of kappa-opioid receptor antagonists in Wistar Kyoto rats

    Neuropsychopharmacology

    (2010)
  • C. Chavkin

    The therapeutic potential of kappa-opioids for treatment of pain and addiction

    Neuropsychopharmacology

    (2011)
  • C. Chavkin et al.

    Dynorphin is a specific endogenous ligand of the kappa opioid receptor

    Science

    (1982)
  • Cited by (44)

    • The G-protein biased kappa opioid agonists, triazole 1.1 and nalfurafine, produce non-uniform behavioral effects in male rhesus monkeys

      2022, Pharmacology Biochemistry and Behavior
      Citation Excerpt :

      Some have interpreted the observed relation between signaling bias and improved side-effect profiles to be an indication that the G-protein signaling pathway is responsible for therapeutic-like effects (e.g., antinociception) while β-arrestin-2 recruitment mediates adverse effects such as dysphoria and sedation (Bedini et al., 2020; Brust et al., 2016; Mores et al., 2019). However, this dichotomy has not held up across all studies (Dogra and Yadav, 2015; Mores et al., 2019; White et al., 2015). Moreover, cell-signaling results that are used to quantify “bias factors” for test ligands vary between approaches and reports, which precludes precise scaling of drugs along the continuum of bias (Brust et al., 2016; Dunn et al., 2019; Dunn et al., 2018; Kaski et al., 2019).

    • An updated assessment of the translational promise of G-protein-biased kappa opioid receptor agonists to treat pain and other indications without debilitating adverse effects

      2022, Pharmacological Research
      Citation Excerpt :

      Two pathways can be compared to generate a single scalar measure of bias, or “bias factor”, or several pathways compared simultaneously in spiderweb/radial plots [26] or forest plots [8]. Biased signaling at the κOR has been reviewed recently [27–29] and for this review we mostly limit our discussion to studies investigating bias for G protein signaling over β-arrestin recruitment. As stated above, bias factors are always calculated with respect to a reference agonist, which ideally taken to be the endogenous agonist of the receptor, or an unbiased synthetic compound that activates and recruits both G protein and β-arrestin pathways to the same degree as the endogenous ligand.

    View all citing articles on Scopus
    View full text