Elsevier

Pharmacology & Therapeutics

Volume 88, Issue 3, December 2000, Pages 437-463
Pharmacology & Therapeutics

Endomorphin-1 and endomorphin-2: pharmacology of the selective endogenous μ-opioid receptor agonists

https://doi.org/10.1016/S0163-7258(00)00100-5Get rights and content

Abstract

The recently discovered endogenous opioid peptides, endomorphins-1 and -2, appear to have properties consistent with neurotransmitter/neuromodulator actions in mammals. This review surveys the information gained so far from studies of different aspects of the endomorphins. Thus, the endomorphins have been found unequally in the brain; they are stored in neurons and axon terminals, with a heterogeneous distribution; they are released from synaptosomes by depolarization; they are enzymatically converted by endopeptidases; and they interact specifically and with high affinity with μ-opioid receptors. The most outstanding effect of the endomorphins is their antinociceptive action. This depends on both central and peripheral neurons. Additionally, the endomorphins cause vasodilatation by stimulating nitric oxide release from the endothelium. Their roles in different central and peripheral functions, however, have not been fully clarified yet. From a therapeutic perspective, therefore, they may be conceived at present as potent antinociceptive and vasodilator agents.

Introduction

It is well known that complex opioid systems modulate a number of physiological processes, including pain, reward, stress, immune responses, gastrointestinal, respiratory, and neuroendocrine functions, and cardiovascular control (Olson et al., 1998). These diverse effects are mediated by endogenous opioid peptides through the activation of specific membrane-bound opioid receptors. These receptors normally interact with opioid peptides, which are distributed in many regions of the mammalian brain. Since the demonstration of receptors for morphine, the μ-opioid receptors, over 25 years ago, neuroscientists have searched for substances produced by the nervous system that activate these receptors. The search led to the discovery of the enkephalins and the endorphins in the 1970s. These compounds display relatively low selectivity and efficacy at the μ-opioid receptors (Zadina et al., 1994), and the recently described endogenous peptides endomorphins-1 and -2 are the likely ligands for the morphine-preferring μ-opioid receptors in many brain regions. They exhibit the highest affinity and specificity for the μ-receptors of any compound found so far in the mammalian nervous system. Their μ characteristics have been supported by a variety of in vivo and in vitro studies. The endomorphin family includes two peptides that differ in one amino acid: endomorphin-1 (Tyr-Pro-Trp-Phe-NH2) and endomorphin-2 (Tyr-Pro-Phe-Phe-NH2) (Fig. 1), with molecular masses of 665.7 and 626.7 kDa, respectively. These endogenous ligands differ from conventional endogenous opioid receptor ligands in their N-terminal sequence, peptide length, and C-terminal amidation. The discovery of the endomorphins has opened up new possibilities in opioid research. In the 3 years that have elapsed since the original report of the isolation of endomorphins, close to 90 studies (until February 2000) have appeared on the binding profile, the release, the transduction mechanism, the distribution, and the in vitro and in vivo effects of these peptides. The purpose of this review is to summarize the wealth of information that has become available through the use of molecular biological, immunocytochemical, physiological, and pharmacological techniques, and to point to the perspectives for novel strategies, especially in pain therapy.

Section snippets

Identity of endomorphins

The route to the identification of the endomorphins was interesting in itself, because it involved an unusual interplay between pharmacology and combinatorial chemistry (Zadina et al., 1997). From a structural point of view, it has been established that naturally occurring opioid peptides consist of two components, a biologically important N-terminal tri- or tetrapeptide fragment (message sequence) and the remaining C-terminal fragment (address sequence) (Yamazaki et al., 1993). Endorphins,

Conformational analysis

To identify structural attributes unique to endomorphin-1, potential sites of recognition, and a possible correlation between the biological properties and the conformational preferences, a few conformational analysis studies have been performed by means of multidimensional NMR and molecular modeling techniques Fiori et al., 1999, Podlogar et al., 1999, Paterlini et al., 2000. The first spectroscopic results, from experiments in dimethylsulfoxide and in water, indicate that endomorphin-1 exists

Binding characteristics

Zadina et al. (1997) demonstrated that endomorphins-1 and -2 have high affinities (Ki=360 and 690 pM, respectively) and selectivities (4000- and 13,400-fold preference, respectively, over the δ-receptor, and 15,000- and 7600-fold preference, respectively, over the κ-receptor) for the μ-receptor of synaptic plasma membranes prepared from rat brain. They have equal affinities, but greater selectivities, for the μ-receptor than those of a μ-selective analog of enkephalin [[d-Ala(2), N

Transduction mechanisms

Recent cloning and expression studies have revealed that the μ-opioid receptors belong to the superfamily of 7-transmembrane domain receptors that are coupled to the inhibitory Gi/Go class of G-proteins (Chen et al., 1993). The binding of opioids to μ-opioid receptors produces coordinated changes at a cellular level, i.e., the closure of voltage-sensitive Ca2+ channels (Porzig, 1990), the activation of K+ channels (North, 1989), and a reduction in cyclic AMP (cAMP) formation (Childers, 1991).

Isolation and distribution of the endomorphins

In studies using radioimmunoassay and immunocytochemistry, endomorphin-1 and/or endomorphin-2 immunoreactivity has been isolated and localized within the bovine, human, rat, monkey, mice, and guinea pig nervous systems. The concentrations of the peptides (2.1 pmol/g) are below those of enkephalin, but similar to those of the less abundant opioids, β-endorphin and dynorphin, in bovine brain extracts (Zadina et al., 1997). The tetrapeptides, however, were found in much higher amounts (151 pmol/g)

Endomorphins and antinociception

Agonists that exert activity at the μ-opioid receptors are well known to be the most effective means of alleviating severe pain in a wide range of conditions. Their analgesic efficacy stems from their activity at the supraspinal, spinal, and peripheral levels. A number of studies have been performed to investigate the roles of endomorphins in pain transmission and in antinociception both in vitro and in vivo.

Endomorphins and the cardiovascular system

It is well known that opioids exhibit vasodilator actions through both central and peripheral sites (Bowman & Rand, 1980). The endogenous opioid system has been shown to play roles in regulation of the vascular smooth muscle tone, regional blood flow, and blood pressure in normal and hypertensive states, and morphine decreases the systemic arterial pressure in anesthetized cats and rats (Olson et al., 1998). This activity may be mediated by an action on the autonomic nervous system or by

Gastrointestinal functions

μ-Opioid receptors are also present in the peripheral nervous system, including the enteric nervous system, and opioids have long been recognized as having inhibitory effects on the gastrointestinal transit (Olson et al., 1998). There is little evidence as concerns the possible roles of the endomorphins in the gastrointestinal function.

The respiratory system

The opioid system significantly influences the respiratory system, although no data are available on the respiratory effects of centrally administered endomorphins. In the airways, opioids have been shown to modulate the release of mediators from the parasympathetic and sensory nerve fibers Bartho et al., 1987, Belvisi et al., 1990. The endomorphins induce a concentration-dependent inhibition of electrical-field stimulation-induced tachykinin-mediated contractions of the guinea pig bronchus (

Inflammation

Neurogenic inflammation is mediated via sensory peptides released from the peripheral terminals of the sensory nerves, and can be modulated by opioid peptides locally released at the site of injury. Khalil et al. (1999) used a vacuum-induced blister model in anesthetized rats to examine the effects of endomorphin-1 (0.01–10 μM) on the inflammatory responses. Local perfusion of endomorphin-1 significantly inhibited the inflammatory responses to both electrical stimulation of the sciatic nerve

The immune system

Endogenous opioid peptides possess a variety of immunomodulatory properties (Olson et al., 1998). It has been shown that the μ3-opioid receptor mediates inhibition by morphine of the activation of human peripheral blood monocytes and granulocytes (Stefano et al., 1993). Similar to other opioid peptides (Stefano et al., 1993), the endomorphins do not release NO from human monocytes and granulocytes, supporting the binding results (see Section 4; Rialas et al., 1998). The very surprising result

Miscellaneous

The central mechanisms regulating food intake are complex and still not understood completely. Opioid agonists have been shown to stimulate food intake, raising the possibility that opioid receptors are involved in the regulation of feeding (Olson et al., 1998). Intracerebroventricular injection of the endomorphins (0.03–30 nmol) enhanced the food intake for 4 hr in non-food-deprived mice in a dose-related manner (Asakawa et al., 1998). It is also well known that opioid agonists generally

Tolerance to endomorphins

Some in vivo data suggest the development of acute tolerance (or tachyphylaxis) against both endomorphins Stone et al., 1997, Higashida et al., 1998, Horvath et al., 1999. The development of acute spinal tolerance to pain sensitivity was observed 30 min following pretreatment with endomorphin-2 Stone et al., 1997, Horvath et al., 1999. Interestingly, endomorphin-1 required a longer pretreatment time (90 min) before tolerance was observed (Stone et al., 1997), which suggests that it is possible

Endomorphin analogs

One practical difficulty with the endomorphins is the short duration of their effects. One solution to this problem is to use stable analogs. Several analogs of the endomorphins have been synthesized to date. The first were d-Ala-endomorphin-2 [Tyr-d-Ala-Phe-Phe-NH2 (TAPP)] and d-Pro-endomorphin-2, where the l-Pro at Position 2 was substituted with d-Ala or d-Pro, respectively Champion & Kadowitz, 1998, Champion & Kadowitz, 1999. Next came β-MePhe4-endomorphins-1 and -2, where the endomorphins

Metabolism

The metabolic fate of the endomorphins either in vitro or in the organism remains relatively unknown. The endomorphin-induced [35S]GTPγS binding does not change in the presence of peptidase inhibitors Alt et al., 1998, Narita, 1998, implying that enzymatic peptide cleavage does not play a significant role under the conditions of these assays (Alt et al., 1998). In contrast, endomorphin-2 is an excellent substrate of different endopeptidases. It has been shown that DPP IV, a membrane-bound Ser

Conclusions

The substantial number of studies discussed in this review have clearly demonstrated that both endomorphins display potent bioactivities in vitro and in vivo. Their high affinities and selectivities for μ-opioid receptors and their potent antinociceptive and vasodilator activities have been firmly proven, although (as with all investigations) many questions remain to be addressed. At present, we possess some data on their catabolism, but we do not know the enzymes that take part in their

Acknowledgements

This work was supported by ETT 590/96. I am grateful to Drs. Ulrike Holzer-Petsche and György Benedek for their critical reading of the manuscript and helpful advice. I am indebted to Ildikó Dobos for technical contributions in the manuscript.

References (106)

  • L Hackler et al.

    Isolation of relatively large amounts of endomorphin-1 and endomorphin-2 from human brain cortex

    Peptides

    (1997)
  • Z Han et al.

    Endomorphin-1 mediates 2 Hz but not 100 Hz electroacupuncture analgesia in the rat

    Neurosci Lett

    (1999)
  • J.X Hao et al.

    Lack of cross tolerance between the antinociceptive effect of intrathecal orphanin FQ and morphine in the rat

    Neurosci Lett

    (1997)
  • S.L Hao et al.

    Isobolographic analysis of interaction between spinal endomorphin-1, a newly isolated endogenous opioid peptide, and lidocaine in the rat formalin test

    Neurosci Lett

    (1999)
  • C Harrison et al.

    Stimulatory effects of opioids

    Br J Anaesth

    (1998)
  • L.M Harrison et al.

    Differential effects of endomorphin-1, endomorphin-2, and Tyr-W-MIF-1 on activation of G-proteins in SH-SY5Y human neuroblastoma membranes

    Peptides

    (1998)
  • G Horvath et al.

    Antinociceptive effects of intrathecal endomorphin-1 and -2 in rats

    Life Sci

    (1999)
  • K Hosohata et al.

    Endomorphin-1 and endomorphin-2 are partial agonists at the human μ-opioid receptor

    Eur J Pharmacol

    (1998)
  • S.-X Jin et al.

    Endomorphin-1 reduces carrageenan-induced Fos expression in the rat spinal dorsal horn

    Neuropeptides

    (1999)
  • K Kakizawa et al.

    Parallel stimulations of in vitro and in situ [35S]GTPγS binding by endomorphin 1 and DAMGO in mouse brains

    Peptides

    (1998)
  • E.H Kwok et al.

    Endomorphins decrease heart rate and blood pressure possibly by activating vagal afferents in anesthetized rats

    Brain Res

    (1998)
  • H.H Loh et al.

    μ Opioid receptor knockout in mice: effects on ligand-induced analgesia and morphine lethality

    Mol Brain Res

    (1998)
  • S Martin-Schild et al.

    Endomorphin-2 is an endogenous opioid in primary sensory afferent fibers

    Peptides

    (1998)
  • K McConalogue et al.

    Activation and internalization of the μ-opioid receptor by the newly discovered endogenous agonists, endomorphin-1 and endomorphin-2

    Neuroscience

    (1999)
  • H Mizoguchi et al.

    The μ-opioid receptor gene-dose dependent reductions in G-protein activation in the pons/medulla and antinociception induced by endomorphins in μ-opioid receptor knockout mice

    Neuroscience

    (1999)
  • M Narita et al.

    Characterization of endomorphin-1 and -2 on [35S]GTPγS binding in the mouse spinal cord

    Eur J Pharmacol

    (1998)
  • I Nevo et al.

    Acute and chronic activation of the μ-opioid receptor with the endogenous ligand endomorphin differentially regulated adenylyl cyclase isoenzymes

    Neuropeptides

    (2000)
  • G.A Olson et al.

    Endogenous opiates: 1997

    Peptides

    (1998)
  • H.J Patel et al.

    Modulation of acetylcholine release from parasympathetic nerves innervating guinea-pig and human trachea by endomorphin-1 and -2

    Eur J Pharmacol

    (1999)
  • M.G Paterlini et al.

    Stereochemical requirements for receptor recognition of the μ-opioid peptide endomorphin-1

    Biophys J

    (2000)
  • A Péter et al.

    Liquid chromatographic study of the enzymatic degradation of endomorphins, with identification by electrospray ionization mass spectrometry

    J Chromatogr A

    (1999)
  • P.K Peterson et al.

    Endomorphin-1 potentiates HIV-1 expression in human brain cell cultures: implication of an atypical μ-opioid receptor

    Neuropharmacology

    (1999)
  • B Przewlocka et al.

    Spinal analgesic action of endomorphins in acute, inflammatory and neuropathic pain in rats

    Eur J Pharmacol

    (1999)
  • S Sakurada et al.

    Differential involvement of μ-opioid receptor subtypes in endomorphin-1 and -2-induced antinociception

    Eur J Pharmacol

    (1999)
  • R Shane et al.

    Modulation of endomorphin-2-induced analgesia by dipeptidyl peptidase IV

    Brain Res

    (1999)
  • M Spetea et al.

    In vitro binding and signaling profile of the novel μ opioid receptor agonist endomorphin 2 in rat brain membranes

    Biochem Biophys Res Commun

    (1998)
  • G.B Stefano et al.

    Presence of the μ3 opiate receptor in endothelial cells: coupling to nitric oxide production and vasodilation

    J Biol Chem

    (1995)
  • A Sugimoto-Watanabe et al.

    Antinociceptive effect and enzymatic degradation of endomorphin-1 in newborn rat spinal cord

    Jpn J Pharmacol

    (1999)
  • J.-L Wang et al.

    Distinct effect of intracerebroventricular and intrathecal injections of nociceptin/orphanin FQ in the rat formalin test

    Regul Pept

    (1999)
  • Y.-Q Wang et al.

    Effects of orphanin FQ on endomorphin-1 induced analgesia

    Brain Res

    (1999)
  • C.A Williams et al.

    Release of endomorphin-2 like substances from the rat spinal cord

    Neurosci Lett

    (1999)
  • S.Y Wu et al.

    Endomorphin-like immunoreactivity in the rat dorsal horn and inhibition of substantia gelatinosa neurons in vitro

    Neuroscience

    (1999)
  • A Alt et al.

    Stimulation of guanosine-5′-O-(3-[35S]thio)triphosphate binding by endogenous opioids acting at a cloned μ receptor

    J Pharmacol Exp Ther

    (1998)
  • A Asakawa et al.

    Endomorphins have orexigenic and anxiolytic activities in mice

    Neuroreport

    (1998)
  • G.A Barr et al.

    Maturation of endomorphin-2 in the dorsal horn of the medulla and spinal cord of the rat

    Neuroreport

    (1999)
  • G.A Barr et al.

    The ontogeny of endomorphin-1- and endomorphin-2-like immunoreactivity in rat brain and spinal cord

    Ann NY Acad Sci

    (2000)
  • L Bartho et al.

    Peripheral effects of opioid drugs on capsaicin-sensitive neurones of the guinea-pig bronchus and rabbit ear

    Naunyn Schmiedebergs Arch Pharmacol

    (1987)
  • M.G Belvisi et al.

    Modulation of cholinergic neurotransmission in guinea-pig airways by opioids

    Br J Pharmacol

    (1990)
  • A.R Bhatia et al.

    Spinal antinociceptive effects and G-protein activation induced by endomorphin-1 in the rat

    Anesth Analg

    (2000)
  • L.M Bohn et al.

    Evidence for κ- and μ-opioid receptor expression in C6 glioma cells

    J Neurochem

    (1998)
  • Cited by (114)

    • The opioid system in stress-induced memory disorders: From basic mechanisms to clinical implications in post-traumatic stress disorder and Alzheimer's disease

      2019, Progress in Neuro-Psychopharmacology and Biological Psychiatry
      Citation Excerpt :

      In general, β-endorphin binds to μ and δ opioid receptors, whereas dynorphin binds preferentially to κ receptors, and met- and leu-enkephalin bind to δ and μ receptors (Benarroch, 2012). Additional opioid peptides, such as nociceptin/orphanin FQ (N/OFQ) and endomorphin, which have respective affinities for ORL and μ receptors, have been also described (Chu et al., 1999; Horvath, 2000; Meunier, 1997). The opioid receptors are localized in regions of the central nervous system (CNS) that regulate the response to stress, including the hypothalamus, as well as in brain structures associated with emotional response, learning and memory (Contet et al., 2006).

    • Inhibition of proteases as a novel therapeutic strategy in the treatment of metabolic, inflammatory and functional diseases of the gastrointestinal tract

      2013, Drug Discovery Today
      Citation Excerpt :

      An intensive search for the endogenous opioid ligands led to the discovery of DOR-selective enkephalins (ENKs) [3], a natural ligand with activity at MOR and DOR β-endorphin [4] and KOR-selective dynorphins [5]. In 1997 endomorphins (EMs) [6], which possess the highest affinity and specificity at MORs of all endogenous compounds were localized in the mammalian nervous system [1]. Many structural and functional components are responsible for the proper function of the GI tract, including the enteric nervous system (ENS), GI smooth muscle cells, the intestinal mucosa and blood vessels.

    View all citing articles on Scopus
    View full text