Endogenous morphine and its metabolites in mammals: History, synthesis, localization and perspectives

Endogenous morphine and its metabolites in mammals: History, synthesis, localization and perspectives

Neuroscience 233 (2013) 95–117 REVIEW ENDOGENOUS MORPHINE AND ITS METABOLITES IN MAMMALS: HISTORY, SYNTHESIS, LOCALIZATION AND PERSPECTIVES A. LAUX-B...

1MB Sizes 0 Downloads 17 Views

Neuroscience 233 (2013) 95–117

REVIEW ENDOGENOUS MORPHINE AND ITS METABOLITES IN MAMMALS: HISTORY, SYNTHESIS, LOCALIZATION AND PERSPECTIVES A. LAUX-BIEHLMANN, J. MOUHEICHE, J. VE´RIE`PE AND Y. GOUMON *

The biosynthetic pathway of endogenous opiates in mammals: a comparison with plants 98 L-tyrosine to dopamine 98 The formation of norlaudanosoline and norcoclaurine 98 Three O-methylation steps to form (R)-reticuline 100 (R)-reticuline to thebaine 100 Thebaine to morphine: two parallel pathways 100 Conclusion: conservation throughout evolution 100 Investigation of new endogenous opiate compounds of interest 100 Precursors 100 Morphine catabolites 101 In humans 101 In other mammals 103 Localization of endogenous opiates in mammals 103 The central nervous system 103 Endogenous morphine in astrocytes 103 Endogenous morphine in GABAergic cells 103 In the periphery 105 Different cellular partners may be involved in endogenous opiate biosynthesis 106 Is EM biosynthesis restricted to catecholaminergic cells? 106 The precursor-uptake hypothesis 106 Localization of enzymes involved in endogenous morphine biosynthesis 107 What are the roles of endogenous opiates? 107 Endogenous opiate levels 107 At the periphery: stress and immune responses 107 Endogenous opiates in the adrenal medulla 107 Endogenous opiates in the immune system 107 Proposed roles in the CNS 108 Do endogenous opiates act as neurotransmitters or neuromediators? 108 Endogenous opiates and control of nociception 108 Endogenous opiates and memory 108 Endogenous opiates and addiction 108 Endogenous opiates, neurogenesis and structural plasticity 109 Endogenous opiates and neuroinflammation 109 Endogenous opiates and CNS pathologies 109 Schizophrenia 109 A ‘‘hypermorphinergic’’ pathology? 109 Endogenous morphine, schizophrenia and nociception 110 Parkinson’s disease 110 Conclusion 110 Acknowledgements 110 References 110

Nociception and Pain Department, Institut des Neurosciences Cellulaires et Inte´gratives, CNRS UPR3212 and Universite´ de Strasbourg, F-67084 Strasbourg, France

Abstract—Morphine derived from Papaver somniferum is commonly used as an analgesic compound for pain relief. It is now accepted that endogenous morphine, structurally identical to vegetal morphine–alkaloid, is synthesized by mammalian cells from dopamine. Morphine binds mu opioid receptor and induces antinociceptive effects. However, the exact role of these compounds is a matter of debate although different links with infection, sepsis, inflammation, as well as major neurological pathologies (Parkinson’s disease, schizophrenia) have been proposed. The present review describes endogenous morphine and morphine derivative discovery, synthesis, localization and potential implications in physiological and pathological processes. Ó 2012 IBRO. Published by Elsevier Ltd. All rights reserved.

Key words: morphine, morphine-glucuronide, dopamine, opioid receptor, analgesia. Contents General introduction and nomenclature 96 Endogenous opiates: a history of their discovery 96 Discovery of endogenous morphine-like compounds 96 Identification of endogenous opiates 96 Evidence of de novo morphine biosynthesis in mammals 98

*Corresponding author. E-mail address: [email protected] (Y. Goumon). Abbreviations: 4-HPAA, 4-hydroxyphenylacetaldehyde; 4-HPP, 4hydroxyphenylpyruvate; AADC, aromatic L-amino acid decarboxylase; COMT, catechol-O-methyltransferase; CSF, cerebrospinal fluid; CYP2D6, cytochrome P450 2D6; DOPAL, 3,4-dihydroxyphenylacetaldehyde; DOR, delta opioid receptor; EM, endogenous morphine; EMM, endogenous morphine metabolites; HPLC, high-performance liquid chromatography; KOR, kappa opioid receptor; M3G, morphine-3glucuronide; M3S, morphine-3-sulfate; M6G, morphine-6-glucuronide; M6S, morphine-6-sulfate; MAO, monoamine oxidase; MD2, myeloid differentiation protein 2; MLC, morphine-like compound; MOR, mu opioid receptor; MS, mass spectrometry; NCS, norcoclaurine synthase; PNMT, phenylethanolamine N-methyltransferase; RIA, radioimmunoassay; TH, tyrosine hydroxylase; THP, tetrahydropapaveroline; TLR4, toll-like receptor 4; TYDC, tyrosine decarboxylase; TyrAT, tyrosine aminotransferase; UGT, UDP-glucuronosyl-transferase enzymes. 0306-4522/12 $36.00 Ó 2012 IBRO. Published by Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.neuroscience.2012.12.013 95

96

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

GENERAL INTRODUCTION AND NOMENCLATURE In the early 1950s, the presence of specific morphinebinding receptors was hypothesized on the basis of morphine’s analgesic effects. In the 1970s, three different receptors for opioid and opiate compounds were discovered: the Mu (l), Delta (d) and Kappa (j) opioid receptors (MOR, DOR and KOR, respectively) (Pert et al., 1973; Pert and Snyder, 1973). More recently, a fourth opioid receptor named nociceptin/ orphanin FQ (NOR) was cloned. These opioid receptors have seven transmembrane domains coupled to G proteins (for review: Kieffer and Evans, 2002, 2009; Trescot et al., 2008; Dietis et al., 2011; Al-Hasani and Bruchas, 2012; Feng et al., 2012). The presence of opioid receptors has led to the characterization of several endogenous ligands called opioids due to their peptidic nature: enkephalins (Hughes et al., 1975a,b; Simantov and Snyder, 1976), b-endorphin (Bradbury et al., 1976; Graf et al., 1976; Lazarus et al., 1976; Li and Chung, 1976; Li et al., 1976), dynorphin (Cox et al., 1975; Goldstein et al., 1979; Lowney et al., 1979), nociceptin/orphanin FQ (Meunier et al., 1995; Reinscheid et al., 1995) and endomorphins (Hackler et al., 1997; Zadina et al., 1997). In addition to endogenous opioid peptides, endogenous morphine-like molecules, which are known as endogenous opiates due to their alkaloid nature have been discovered (Gintzler et al., 1976a,b; Blume et al., 1977; Shorr et al., 1978). Nevertheless, these molecules remain unfamiliar to most scientists, with only a small number of laboratories focusing on this particular research area. The data on endogenous opiates are scattered among the thousands of articles on ‘‘exogenous morphine’’ and endogenous opioid peptides. Furthermore, confusion arises when scientists use the term ‘‘opiates’’ instead of ‘‘opioids’’ for endogenous opioid peptides. Because no clear consensus exists, the following conventions will be used in this review: ‘‘opioids’’ will refer to peptides having an affinity for opioid receptors, whereas ‘‘opiates’’ will refer to natural or synthetic morphine-derived alkaloids (morphine, codeine, morphine-glucuronides. . .). The present review summarizes the findings from the available literature on endogenous opiates (endogenous morphine and endogenous morphine metabolites). Their history, synthesis in mammals, tissue/cellular localization and potential functions in physiological and pathological states will be addressed in the following paragraphs. We will also discuss exciting fundamental and therapeutic perspectives that have recently emerged from cutting-edge research.

Endogenous opiates: a history of their discovery Discovery of endogenous morphine-like compounds. In 1903, the French scientist Dr. Mavrojannis made the observation that morphine injections in rats led to symptoms related to catalepsy and subsequently

hypothesized that such symptoms in mammals are the consequences of endogenous ‘‘morphine-like’’ compounds (Mavrojannis, 1903). In 1970, Davis and Walsh (1970) were the first to propose the presence of ‘‘true’’ morphine in mammals that potentially arose from a synthetic pathway similar to the biosynthetic pathway described in plants. Around the time of the discovery of the first endogenous opioid peptides, the group of Pr. S. Spector demonstrated the existence of an endogenous non-peptide ‘‘morphine-like compound’’ (MLC) (Gintzler et al., 1976a,b, 1978). This MLC was detected in rabbit and cat brains using a radioimmunoassay (RIA) directed against morphine. Once extracted, this compound displayed pharmacological properties identical to those of morphine. Furthermore, this MLC, which bound opioid receptors, was resistant to peptidase/protease treatments (Gintzler et al., 1976a,b, 1978). In parallel, other studies described the presence of a MLC in guinea-pig blood and small intestines (Schulz et al., 1977) as well as in human blood (Pert et al., 1976). A year later, Blume et al. (1977) described an antimorphine antibody that was able to bind specifically to MLC; the fact that it did not interfere with endogenous opioid peptides ruled out any possible artifacts. This group postulated that MLC has a molecular structure similar to that of morphine and hypothesized the existence of two families of compounds (peptide and non-peptide) that bind opioid receptors. The presence of MLC was described in the mouse brain and immunolocalized in neuronal perikarya and processes of the cerebellum and the raphe nuclei (Gintzler et al., 1978). Its presence was also described in the cerebrospinal fluid (CSF), urine and brain extracts of patients naı¨ ve for exogenous morphine or morphine derivatives, suggesting its endogenous origin (Shorr et al., 1978; Wuster et al., 1978). In 1981, Killian et al. (1981) confirmed the existence of a non-peptide compound immunoprecipitated by anti-morphine antibodies in the calf brain, with an analgesic activity that could be blocked by naloxone, an opioid receptor antagonist. Identification of endogenous opiates. In 1985, Goldstein and collaborators described the presence of four different MLCs in the bovine brain and adrenal gland. Using a NMR (nuclear magnetic resonance) approach, they demonstrated that one of the four MLCs was structurally identical to morphine extracted from poppies (Goldstein et al., 1985). Morphine endogenously present in mammals was named ‘‘endogenous morphine’’ (EM) as opposed to morphine from plants (exogenous morphine). The same group subsequently described in the bovine hypothalamus, the presence of endogenous codeine, a morphine precursor (Weitz et al., 1986). In the years that followed, several studies focused on the presence of EM and endogenous codeine using high-performance liquid chromatography (HPLC), NMR and mass spectrometry (MS) approaches in tissues from different species (for review: Meijerink et al., 1999; Stefano et al., 2000).

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

97

Fig. 1. Morphine biosynthetic pathway in mammals and plants: from L-tyrosine to reticuline. Common intermediates (in italic) present in both kingdoms are in black font. Specific compounds present in plants appear in green whereas mammals’ ones are in red font. Identified enzymes (i.e., cloned) appear in bold font in green for plants and in red for mammals. Unknown or putative enzymes are in black font. References: 1, Facchini and De Luca (1994); 2, Wassenberg et al. (2010); 3, Lee and Facchini (2011); 4, Bilkova et al. (2005); 5, Florang et al. (2007); 6, Deitrich and Erwin (1980); 7 , Samanani and Facchini (2002); 8, Morishige et al. (2000); 9, Loeffler et al. (1995); 10, Ziegler et al. (2005); 11, Silber et al. (1991); 12, Facchini and Park (2003); 13, Morishige et al. (2000); 14, Grobe et al. (20110); 15, Ziegler et al. (2009); 16, Hirata et al. (2004). TYDC, tyrosine/dopa decarboxylase; 4-HPP, 4-hydroxyphenylpyruvate; 4-HPAA, 4-hydroxyphenylacetaldehyde; L-DOPA, 3,4-dihydroxy-L-phenylalanine; DOPAL, 3,4-dihydroxyphenylacetaldehyde; NCS, norcoclaurine synthase; AO, amine oxydase; CNMT, (S)-coclaurine-N-methyl-transferase; 6-OMT, 6-O-methyltransferase; DRS, 1,2-dehydroreticuline synthase; DRR, 1,2-dehydroreticuline reductase; 40 -OMT, 40 -O-methyltransferase; NMCH, (S)-N-methylcoclaurine-30 hydroxylase; TH, tyrosine hydroxylase; AADC, aromatic L-amino acid decarboxylase; MAO, monoamine oxydase; NMT, N-methyltransferase; TyrAT, tyrosine aminotransferase; THP, tetrahydropapaveroline; COMT, catechol-O-methyltransferase. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

98

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Evidence of de novo morphine biosynthesis in mammals. In 1970, Davis and Walsh (1970) proposed that morphine biosynthesis occurred in mammals through a biosynthetic pathway similar to that described in plants. In 1986, three precursors of plant morphine synthesis (salutaridine, thebaine and codeine) were shown to increase EM levels in the rat brain, revealing the presence of enzymes involved in the de novo biosynthesis of morphine in mammals (Donnerer et al., 1986). The conversion of reticuline to salutaridine, which is regarded as the key step in the formation of the morphine skeleton, was later demonstrated in vivo in the rat liver (Weitz et al., 1987). Additionally, the presence of thebaine, a morphine precursor, was described in the bovine brain (Kodaira et al., 1989). Conclusive evidence of de novo morphine synthesis in mammals was provided by the laboratory of Pr. M. H. Zenk. By cultivating the human neuroblastoma cell line SHSY5Y in the presence of 18O2, this group was able to isolate different radiolabeled intermediates of morphine biosynthesis along with radiolabeled morphine, which together account for the entire synthetic pathway of morphine in this human cell line (Poeaknapo et al., 2004; Boettcher et al., 2005). More recently, an in vivo study showed that mice with a disrupted tyrosine hydroxylase gene were unable to synthesize EM in the brain (Neri et al., 2008). As tyrosine hydroxylase is the key enzyme for dopamine formation, these results indicate that morphine synthesis requires dopamine. Finally, the discovery of EM and several precursors in mouse urine confirmed the synthesis of morphine in animals in vivo (Grobe et al., 2010).

L-tyrosine

is hydroxylated to L-DOPA by tyrosine hydroxylase (TH), and L-DOPA is subsequently decarboxylated by the aromatic L-amino acid decarboxylase (AADC) to form dopamine. Cytochrome P450 2D6 (CYP2D6) has been shown to catalyze the hydroxylation of tyramine to form dopamine and thus represents an alternative biosynthetic pathway for dopamine formation (Hiroi et al., 1998a,b; Bromek et al., 2011). In mammals, DOPAL formation is catalyzed by monoamine oxidase (MAO) from dopamine (Wachtel and Abercrombie, 1994). In opium poppy, the same intermediates are involved in the formation of dopamine. However, only tyrosine decarboxylase (TYDC), which catalyzes the same reactions as AADC, has been characterized in plants. Plants can form DOPAL, but it is its monohydroxylated homolog, 4-hydroxyphenylacetaldehyde (4-HPAA), that is involved in morphine biosynthesis. 4-HPAA is synthesized from 4-hydroxyphenylpyruvate (4-HPP) formed by the transamination of L-tyrosine by the tyrosine aminotransferase (TyrAT) (Lee and Facchini, 2011). The 4-HPAA intermediate exists in mammals and may represent a new intermediate in EM biosynthesis. Indeed, in humans, neutrophils and macrophages produce 4HPAA from L-tyrosine via a myeloperoxidase (Hazen et al., 1999; Whitman et al., 1999; Exner et al., 2001). Moreover, the production of 4-HPAA by these cells occurs under specific physiological or pathological conditions that have been shown to increase EM blood levels (Hazen et al., 2000; Heller et al., 2000). The formation of norlaudanosoline and norcoclaurine

THE BIOSYNTHETIC PATHWAY OF ENDOGENOUS OPIATES IN MAMMALS: A COMPARISON WITH PLANTS The morphine synthetic pathway in mammals appears to be highly similar to the pathway described in opium poppy on the basis of common intermediates. The synthetic pathway of morphine in opium poppy has been well characterized in the literature (for review: Kirby, 1967). However, the discovery of enzymes involved in morphine synthesis in plants has recently occurred, and only half of these enzymes have been cloned and characterized during the last decade. It is therefore not surprising that only a few enzymes involved in the mammalian morphine synthetic pathway have been described and that their discovery was based on sequence similarities. This chapter presents, in five steps, the morphine biosynthetic pathway (Figs. 1 and 2), highlighting the similarities and differences between the pathways in mammals (Poeaknapo et al., 2004; Boettcher et al., 2005) and opium poppy (for review: Facchini, 2001; Ziegler et al., 2009). L-tyrosine

to dopamine

In mammals, morphine biosynthesis begins with the formation of two compounds, dopamine and 3,4dihydroxyphenylacetaldehyde (DOPAL) from L-tyrosine.

In mammals, the production of morphine has been found to derive from norlaudanosoline, which is also known as tetrahydropapaveroline (THP). Norlaudanosoline, considered as dopamine catabolite, is formed by a spontaneous Picted–Spengler condensation reaction between dopamine and DOPAL (for review: Marchitti et al., 2007). It is important to note that both enantiomers, (R)-norlaudanosoline and (S)norlaudanosoline, are present in mammals (Cashaw et al., 1983; Cashaw, 1993b,a; Sango et al., 2000). Surprisingly, high levels of THP have been associated with pathological states such as Parkinson’s disease and alcoholism. In plants, it was originally hypothesized that (S)norlaudanosoline was the precursor of morphinan alkaloids. However, only (S)-norcoclaurine was found to be involved in morphine biosynthesis. (S)-norcoclaurine is yielded by the condensation of dopamine with 4HPAA, which is catalyzed by (S)-norcoclaurine synthase (NCS) (Ilari et al., 2009). In mammals, the involvement of norcoclaurine cannot be excluded from the EM biosynthetic pathway. Hence, 4HPAA is produced in human neutrophils and macrophages from L-tyrosine via a myeloperoxidase (Hazen et al., 1999; Whitman et al., 1999; Exner et al., 2001). Accordingly, endogenously formed norcoclaurine has been detected in the brains of rats treated with alcohol (Haber et al., 1997).

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

99

Fig. 2. Morphine biosynthetic pathway in mammals and plants: from reticuline to morphine. Common intermediates (in italic) present in both kingdoms are in black font. Specific compounds present in plant appear in green whereas mammals’ ones are in red font. Identified enzymes (i.e., cloned) appear in bold font in green for plants and in red for mammals. References: 1, Gesell et al. (2009); 2, Grobe et al. (2009); 3, Gerardy and Zenk (1993); 4, Grothe et al. (2001); 5, Hagel and Facchini (2010); 6, Lenz and Zenk (1995); 7, Zhu (20080. SalSyn, salutaridine synthase; SalR, salutaridine 7-oxidoreductase; SalAT, salutaridinol-7-O-acetate; COR, codeine reductase; T6ODM, thebaine-6-O-demethylase; CODM, codeine-Odemethylase. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

100

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Three O-methylation steps to form (R)-reticuline In mammals, two pathways may lead to the formation of (R)-reticuline. (i) In 2005, Boettcher et al. (2005) demonstrated the presence of (S)-norlaudanosoline and (S)-reticuline and discussed their possible roles in EM biosynthesis. The enzymes involved in these three methylation steps are still unknown, although it has been proposed that COMT (catechol-O-methyltransferase) is one of these enzymes (Mantione et al., 2008). (S)-reticuline is finally converted to the (R)enantiomer by an unknown enzyme. (ii) The second possible pathway involves the formation of (R)-norlaudanosoline and its conversion into (R)-reticuline. Indeed, N-methyltransferase (NMT; E.C. 2.1.1.49) was recently found to perform the last (R)-norlaudanosoline methylation step in a manner specific to the enantiomer configuration (Grobe et al., 2011). This second pathway is more interesting from an evolutionary point of view, and the formation of an (R)-enantiomer of norlaudanosoline requires fewer intermediates to form (R)reticuline. In plants, (S)-norcoclaurine is converted into (R)reticuline after three methylations and one hydroxylation. The four enzymes implicated and the order of these steps have been described and characterized in the literature (Fig. 1). (R)-reticuline to thebaine The conversion of (R)-reticuline to salutaridine has been shown to occur in vivo in the rat liver (Weitz et al., 1987). This reaction is the critical step in the formation of the morphine skeleton and therefore represents the limiting step. This step is likely performed by the CYP2D6 enzyme in humans and CYP3A4 in rats (Grobe et al., 2009). Salutaridine is then converted to thebaine through several identified intermediates. However, the enzymes involved in these reactions remain unknown in mammals. In plants, identical intermediates have been observed, and the different enzymes implicated have already been described and characterized in the literature (Fig. 2). Thebaine to morphine: two parallel pathways As in plants, two parallel pathways exist in mammals for the formation of morphine from thebaine. Furthermore, all of the intermediates that lead to morphine are present in both kingdoms. The enzymes involved in this pathway were only recently characterized in plants (Lenz and Zenk, 1995; Hagel and Facchini, 2010). In mammals, only CYP2D6, which demethylates thebaine to oripavine and codeine to morphine, is known. Conclusion: conservation throughout evolution A comparison between the morphine synthetic pathways described in mammals and in plants indicates a strong

conservation throughout evolution (Meijerink et al., 1999; Kream et al., 2007). In addition to its existence in plants and mammals, morphine is also found in other vertebrates, such as eels (Epple et al., 1993) and toads (Oka et al., 1985). Surprisingly, EM is also present in invertebrates, including molds (Stefano et al., 1993) (Zhu et al., 2001a), mollusks (Goumon et al., 2001; Sonetti et al., 2005), arthropods (Casares et al., 2005), annelids (Pryor and Elizee, 2000), nematodes (Goumon et al., 2000b) and flatworms (Pryor and Elizee, 2000; Zhu et al., 2002). This conservation of morphine biosynthesis throughout evolution suggests not only a conservation of the enzymatic machinery but also a crucial physiological role of EM.

INVESTIGATION OF NEW ENDOGENOUS OPIATE COMPOUNDS OF INTEREST The presence of the EM biosynthetic pathway in mammals poses numerous questions. Indeed, mammalian cells and tissues may be exposed to EM precursors and/or derivatives. Most of the studies cited in this section describe the pharmacological properties of morphine precursors and catabolites without considering the endogenously produced molecules. Nevertheless, these studies help introduce the possible effects and pharmacological properties of these compounds.

Precursors The formation of THP (also named norlaudanosoline), a dopamine catabolite, has been well characterized and corresponds to the first steps of the EM biosynthetic pathway. The neurotoxic effect of norlaudanosoline has been found to involve the inhibition of mitochondrial respiration and increased reactive oxygen species (ROS) production (for review: Surh and Kim, 2010). Norlaudanosoline decreases dopamine biosynthesis by inhibiting TH (Kim et al., 2005; Yao et al., 2010) and inhibits dopamine uptake by acting on its plasma membrane transporter (Okada et al., 1998). Through a blockade of L-type Ca++ channels, reticuline induces central depressant effects (Morais et al., 1998), anti-platelet aggregation (Chen et al., 2000) and hypotensive effects (Dias et al., 2004; Medeiros et al., 2009). Furthermore, two studies have characterized salutaridine as a partial agonist of GABA(A) receptors (Kardos et al., 1984; Eisenreich et al., 2003). Thebaine induces antinociceptive effects (Aceto et al., 1999), whereas oripavine, codeinone and morphinone have analgesic effects but are potentially toxic (Yeh, 1981; Fang et al., 1984; Aceto et al., 1989). More recently, intermediates identified between thebaine and morphine were found to bind opioid receptors and induce G protein activation (Nikolaev et al., 2007; Zhang et al., 2012).

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Morphine catabolites

In humans. Morphine catabolism mainly occurs in the liver (Fig. 3), but it has also been reported in the brain and kidneys. In the liver, up to 60% of morphine is converted into morphine-3-glucuronide (M3G), 10% into morphine6-glucuronide (M6G) and less than 1% into morphine3,6-diglucuronide (Yeh et al., 1977a; Osborne et al., 1990; Hasselstrom and Sawe, 1993; Lotsch et al., 1996). In mammals, the glucuronidation of morphine is dependent on the family of UDP-glucuronosyltransferase enzymes (UGT). In humans, 31 members of the UGT family have been characterized (Mackenzie et al., 1997, 2005), and of these, the UGT1A1, 1A3, 1A6, 1A8, 1A9, 1A10, 2A1 and 2B7 isoforms have been found to be involved in morphine glucuronidation (Jedlitschky et al., 1999; Stone et al., 2003). These UGT isoforms form M3G, whereas only the UGT2B7

101

isoform appears to produce both M3G and M6G. In addition to their localization in the liver, the UGT1A6 and 2B7 isoforms are also expressed in neurons and astrocytes, suggesting that morphine can be glucuronidated in CNS cells (Suleman et al., 1998; King et al., 1999; Nagano et al., 2000; Yamada et al., 2003; Buckley and Klaassen, 2007). Concerning their activities, M6G is 1–600 times more analgesic than morphine, whereas M3G has long been considered devoid of any activity (reviewed in: (Lotsch and Geisslinger, 2001; Coller et al., 2009)). However, it was shown that M3G counteracts the analgesic effects of morphine (Smith et al., 1990; Gong et al., 1992) and induces allodynia after intrathecal administration (Lewis et al., 2010). This effect occurs through its binding to toll-like receptor 4 (TLR4) and, more importantly, to its accessory protein myeloid differentiation protein 2 (MD2) (Komatsu et al., 2009; Hutchinson et al., 2010a; Lewis et al., 2010, 2012). Besides, morphine binds both MOR

Fig. 3. Morphine catabolites in humans. Synthetic representation of compounds detected in the blood or urine of humans after morphine administration.

102

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Fig. 4. Morphine catabolites found in other mammals. These compounds are present in the blood or urine of rodents after morphine administration. None of these molecules has been found so far in humans.

and TLR4 (Komatsu et al., 2009; Lewis et al., 2012; Stevens et al., 2012). Alternatively, morphine can be converted into hydromorphone (morphinone) (Yeh et al., 1977b; Oyler et al., 2000; Cone et al., 2006, 2008; McDonough et al., 2008; Wasan et al., 2008; Hughes et al., 2012) and exhibits high analgesic activities through its binding to MOR (Kumar et al., 2008; Volpe et al., 2011). The

sulfoconjugated morphine-derived compounds morphine-3-sulfate (M3S) and morphine-6-sulfate (M6S) have also been described. However, only M3S is detected in humans after morphine administration (Yeh et al., 1977a,b,c). Several articles have studied phenolsulphotransferase enzymes that may be involved in morphine sulfo-derivative formation (Foldes and Meek, 1973; Rein et al., 1982; Donnerer et al., 1987;

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Nagano et al., 2000). M6S, but not M3S, has been described as a more potent analgesic than morphine after intracerebroventricular injections (Brown et al., 1985; Zuckerman et al., 1999), an effect that may be related to its ability to bind and activate MOR (Frolich et al., 2011). A recent article reported the presence of 3.47% of morphine-3-glucoside and 0.4% of morphine-6-glucoside in the urine of morphine-treated cancer patients (Chen et al., 2003). However, there is no available data on the formation or potential activities of these compounds. Morphine has also been shown to be demethylated into normorphine. Still, only 1% of this compound is found in urine (Yeh, 1975; Yeh et al., 1977a,b). Normorphine can also be detected in two glucuronidated forms in urine: normorphine-3-glucuronide and normorphine-6-glucuronide (Chen et al., 2003). Normorphine binds and activates MOR and therefore exhibits analgesic activity (Sullivan et al., 1989; Frolich et al., 2011). In other mammals. In addition to morphine derivatives found in humans, other morphine catabolism products have been reported in non-human mammalian species (Daly et al., 1965; Misra et al., 1974a,b; Yeh et al., 1979; Frolich et al., 2011) (summarized in Fig. 4). It has been proposed that an O-methylation of morphine subsequently yields codeine (Borner and Abbott, 1973), but such a conversion is still a matter of debate (Yeh, 1974). In the bovine brain, endogenous 6acetylmorphine has been reported, suggesting a possible acetylation of EM (Weitz et al., 1988). The enzymes implicated and the ratios of the compounds formed remain unknown.

LOCALIZATION OF ENDOGENOUS OPIATES IN MAMMALS The central nervous system Immunohistochemistry, HPLC, MS and radioimmuno assay (RIA) approaches have improved the characterization of EM in the mammalian CNS. Several studies have specifically focused on the quantification of endogenous opiate levels in different areas of the CNS (Table 1, adapted from (Perea-Sasiaı´ n, 2008)). At the cellular level, Bianchi and collaborators provided evidence for the immunolocalization of endogenous opiates to the hippocampus, striatum, cortex and cerebellum cells (Bianchi et al., 1994). More recently, a precise mapping of the immunoreactivity of endogenous opiates was performed in the mouse CNS (Fig. 5), revealing their localization in both astrocytes and GABA neurons (Fig. 6). In the brain, endogenous opiate immunoreactivity has been observed in various structures, including the hippocampus, olfactory bulb, band of Broca, basal ganglia and cerebellum. In the spinal cord, endogenous opiates are mainly found in GABA neurons in the gray matter of the ventral horn and in astrocytes. While these compounds are present in presynaptic terminals in the cerebellum, they are

103

found in postsynaptic terminals in the rest of the CNS (Laux et al., 2011, 2012). Endogenous morphine in astrocytes. Astrocytes do not express TH (Jaeger, 1985) and are therefore unable to synthesize dopamine or, by extension, endogenous opiates de novo. Two hypotheses have been proposed to explain the presence of endogenous opiates in astrocytes (Laux et al., 2011, 2012). (i) The first proposes that astrocytes can uptake morphine precursors and thus accomplish morphine biosynthesis. However, further studies of uptake mechanisms and enzyme expression are required to confirm this hypothesis. (ii) The second hypothesis proposes that astrocytes are key cells involved in neuromediator recycling from the extracellular space and may therefore be involved in morphine catabolism. Accordingly, transporters may mediate an uptake of endogenous opiates from the extracellular space. Opiate (endogenous or exogenous) transport mechanisms in the CNS remain poorly understood, and only a few transporters are known to carry morphine. The uptake of morphine by astrocytes is further supported by their expression of the P-glycoprotein (PgP) transporter (Gaillard et al., 2000; Calatozzolo et al., 2005; Chen and Sommer, 2009), which transports morphine across the blood–brain barrier (Dagenais et al., 2001; Seelbach et al., 2007). It is important to note that astrocytes express UGT1A6 and that the formation of endogenous M3G may therefore occur in these specific cells (Suleman et al., 1998; Heurtaux et al., 2006). Moreover, astrocytes express multidrug-resistant proteins (MRP)-1, -3, -4 and -5, as well as the glucose transporter (GLUT), which are potentially involved in morphine/M6G/M3G/ codeine uptake/efflux (Hirrlinger et al., 2002; Bourasset et al., 2003; Somogyi et al., 2007; van de Wetering et al., 2007). Hence, astrocytes may excrete glucuronidated derivatives upon the glucuronidation of morphine by UGTs. The present hypothesis of morphine catabolism (endogenous and exogenous) by astrocytes presents several avenues for future research, particularly in the field of tolerance/addiction to opiates. It is also important to note that endogenous opiates are present in astrocytic foot processes around blood vessels, suggesting a possible capture and/or release of morphine and its derivatives in the blood (Laux et al., 2011, 2012). Endogenous morphine in GABAergic cells. In addition to astrocytes, endogenous opiates are present in GABAergic cells throughout the CNS (Laux et al., 2011, 2012). However, not all GABAergic neurons are concerned, indicating that only a subpopulation of these cells is able to produce EM or its derivatives. In the CNS, GABAergic cells represent the most widespread cell population. As an example, more than 16 subtypes of GABAergic neurons have been characterized in the hippocampus (Somogyi and Klausberger, 2005). No information is currently available, however, regarding the neurochemical characteristics of GABAergic cells containing endogenous opiates. Nevertheless,

104

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Table 1. Amounts of EM, codeine, M3G and M6G present in CNS. ELISA, enzyme-linked immunosorbent assay; GC–MS, gas chromatography–mass spectrometry; HPLC, high-performance liquid chromatography; IP, immunoprecipitation; LC-MS/MS, liquid chromatography tandem mass-spectrometry; QTOF-MS/MS, quadrupole time-of-flight tandem mass spectrometry; RIA, radioimmunoassay. Adapted from Perea-Sasiaı´ n (2008) Structure

Species

Amounts

References

Detection techniques

Donnerer et al. (1986) Guarna et al. (1998) Molina et al. (1995a) Goumon et al. (2000) Guarna et al. (2002) Laux et al. (2011) Laux et al. (2011)

HPLC, RIA HPLC, GC/MS HPLC, RIA HPLC, GC/MS HPLC, GC/MS IP, LC/MS-MS HPLC, LC/MS-MS

Charlet et al. (2010) Horak et al. (1993)

ELISA HPLC, RIA

Monkey

26 ± 6 fmol/g 79 ± 16 fmol/g 0.23 ± 0.013 ng/g 329 ± 93 fmol/g 31 ± 9 fmol/g 7 ± 3.2 ng/g 0.31 ± 0.012 ng/g 6.3 fmol 17.1 fmol 1.01 ± 0.31 pmol/g 0.61 ± 0.22 pmol/g M6G: 0.25 ± 0.15 pmol/g/M3G: 0.21 ± 0.22 pmol/g 0.25 ± 0.23 ng/g 0.39 pmol/g (9 h) 1.16 pmol/g (17 h) 0.11 ng/g 0.04 ng/g

Neri et al. (2004)

GC/MS

Temporal lobe

Human

3.4 ng/g

Fricchione et al. (2008)

HPLC, RIA

Cortex

Rat

0.62 ± 0.32 pmol/g 0.43 ± 0.09 pmol/g 1 pmol/g 1.47 pmol/g 0.62 fmol/mg

HPLC, HPLC, HPLC, ELISA HPLC, RIA HPLC, RIA HPLC,

Morphine Brain (global)

Rat

Mouse

Codeine

Bovine Cat Dog

0.05 ± 0.01 pmol/g 1.5 ± 0.7 ng/g 0.15 pmol/g

Donnerer et al. (1987) Lee and Spector (1991) Meijerink et al. (1999) Muller et al. (2008) Laux et al. (2011) Gintzler et al. (1976a) Oka et al. (1985) Gintzler et al. (1976a) Meijerink et al. (1999)

Hippocampus

Mouse Rabbit Cat

7.46 pmol/g 8 ± 3 ng/g 5 ± 2 ng/g

Muller et al. (2008) Gintzler et al. (1976a) Gintzler et al. (1976a)

ELISA RIA RIA

Basal ganglia

Rabbit Calf Cat

33 ± 10 ng/g 14 ng/g 24 ± 7 ng/g

Gintzler et al. (1976a) Killian et al. (1981) Gintzler et al. (1976a)

RIA IP RIA

Olfactory bulb

Mouse

2.27 pmol/g 2 fmol/mg

Muller et al. (2008) Laux et al. (2011)

ELISA HPLC, LC/MS-MS

Hypothalamus

Rat Rabbit Bovine Cat Dog

7.22 pmol/g 14 ± 4 ng/g 0.25 a` 4.9 pmol/g 12 ± 6 ng/g 0.22 pmol/g

Meijerink et al. (1999) Gintzler et al. (1976a) Goldstein et al. (1985) Gintzler et al. (1976a) Meijerink et al. (1999)

HPLC, RIA RIA HPLC, RIA RIA HPLC, RIA

Cerebellum

Rat

0.63 ± 0.28 pmol/g 0.37 ± 0.07 pmol/g 1.474 pmol/g 1.13 ± 0.59 ng/g 1.48 pmol/g 12 ± 3 ng/g 0.09 pmol/g 14 ± 4 ng/g 0.134 pmol/g

0.80 ± 0.26 pmol/g 0.48 ± 0.08 pmol/g

Donnerer et al. (1987) Lee and Spector (1991) Meijerink et al. (1999) Charlet et al. (2010) Muller et al. (2008) Gintzler et al. (1976a) Oka et al. (1985) Gintzler et al. (1976a) Meijerink et al. (1999)

HPLC, HPLC, HPLC, ELISA ELISA RIA HPLC, RIA HPLC,

0.93 ± 0.36 pmol/g 0.52 ± 0.07 pmol/g 4 ± 1 ng/g 2.7 ± 1 ng/g

1.84 ± 0.57 pmol/g 0.38 ± 0.09 pmol/g

Donnerer et al. (1987) Lee and Spector (1991) Gintzler et al. (1976a) Gintzler et al. (1976a)

HPLC, RIA, MS HPLC, RIA RIA RIA

0.67 ± 0.18 pmol/g 0.59 ± 0.10 pmol/g 0.346 pmol/g 0.54 ± 0.44 ng/g 3.39 pmol/g

1.20 ± 0.27 pmol/g 0.47 ± 0.08 pmol/g

Donnerer et al. (1987) Lee and Spector (1991) Meijerink et al. (1999) Charlet et al. (2010) Muller et al. (2008)

HPLC, RIA, MS HPLC, RIA HPLC, RIA ELISA ELISA

Mouse

Mouse Rabbit Bovine Cat Dog Midbrain

Rat Rabbit Cat

Brainstem

Rat

Mouse

0.81 ± 0.11 pmol/g 0.63 ± 0.08 pmol/g

RIA, MS RIA QTOF MS/MS LC/MS-MS RIA RIA

RIA, MS RIA QTOF MS/MS

RIA RIA

105

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117 Table 1 (continued) Structure

Species

Amounts Morphine

Spinal cord

Rabbit Dog

4 ± 1 ng/g 0.388 pmol/g

Rat Rat Mouse Dog

0.13 pmol/g 1.64 ± 0.49 pmol/g 0.05 ± 0.01 ng/g 0.16 pmol/g

References

Detection techniques

Gintzler et al. (1976a) Meijerink et al. (1999)

RIA HPLC, RIA

Meijerink et al. (1999) Donnerer et al. (1987) Charlet et al. (2010) Meijerink et al. (1999)

HPLC, RIA HPLC, RIA, MS ELISA HPLC, RIA

Codeine

3.22 ± 1.24 pmol/g

Fig. 5. Localization of endogenous morphine and/or metabolites in adult mouse brain and spinal cord. ACB, accumbens nucleus; AH, Ammon’s horn; AON, anterior olfactory nucleus; BST, bed nucleus of the stria terminalis; CBX, cerebellum; CP, caudate putamen; CTX, cortex; DCO, dorsal cochlear nucleus; DG, dentate gyrus; DR, dorsal raphe nucleus; FN, fastigial nucleus; FS, fundus striati; gl, glomerular layer; GP, Globus pallidus; gr, granular cell layer; HY, hypothalamus; IC, inferior colliculus; INC, interstitial nucleus of Cajal; LV, lateral ventricle; MB, midbrain; mi, mitral cell layer; ml, molecular layer; MR, median raphe nucleus; MY, medulla; ND, nucleus of Darkschewitsch; NDB, nucleus of the diagonal band; NLL, nucleus of the lateral lemniscus; NOT, nucleus of the optic tract; OB, olfactory bulb; OT, olfactory tubercle; P, pons; PAG, periaqueductal gray; PB, parabrachial nucleus; PIR, piriform cortex; pl, purkinje layer; PON, periolivary nucleus; PVT, paraventricular thalamic nucleus; RN, red nucleus; RT, reticular thalamic nucleus; SC, superior colliculus; sgz, subgranular zone; SI, substantia innominata; SNc, substantia nigra pars compacta; SNr, substantia nigra pars reticulata; STN, subthalamic nucleus; SUB, subiculum; TH, thalamus; TSN, triangular septal nucleus; VN, vestibular nucleus; VTN, ventral tegmental nucleus; ZI, zona Incerta. In the spinal cord (lower left), laminae are labeled using roman number.

endogenous opiates specifically occur in a well-defined morphological subpopulation of GABAergic cells corresponding to the basket cells of the cerebellum and the sub-granular layer of the dentate gyrus (Muller et al., 2008; Laux et al., 2011, 2012). This neuronal population may therefore have the transporters and enzymes necessary for the capture of morphine precursors (THP. . .) and completion of the EM synthesis reactions. Interestingly, pluricellular interaction is required for morphine biosynthesis in opium poppy, and three specific cell types are implicated in this process (Bird et al., 2003). Therefore, the identification and study of this subpopulation could provide important information about the EM synthesis pathway.

In the periphery In mammals, EM and endogenous morphine metabolites (EMM) have been detected in the adrenal gland (Oka et al., 1985; Donnerer et al., 1987; Molina et al., 1995a; Goumon and Stefano, 2000), which is one of the major organs implicated in stress responses (Goumon et al., 2009). Moreover, EM has been found in tumoral rat pheochomocytoma PC-12 (Goumon et al., 2000c; Poeaknapo et al., 2004), while the presence of M6G has been reported in the secretory granules of bovine primary chromaffin cells (Goumon et al., 2006). EM has also been detected in lung cells (dog and human (Munjal et al., 1995)); rat and human hepatocytes

106

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Fig. 6. Immunolocalization of endogenous opiates in adult mouse cortex. An immunolabel is observed in GABAergic neurons (black arrow) and astrocytes (white arrow). The antibody recognizes endogenous codeine, morphine and its glucuronidated derivatives (Laux et al., 2011, 2012). Scale bar = 20 lm.

(Donnerer et al., 1986, 1987; Molina et al., 1995b; Poeaknapo et al., 2004); and the rat spleen, thymus and kidneys (Molina et al., 1995b). Morphine and its derivatives have been reported in the rat pancreas (Lee and Spector, 1991), and the formation of morphine intermediates such as THP and reticuline has been observed in the human pancreatic cell line DAN-G (Poeaknapo et al., 2004). Surprisingly, rabbit (Oka et al., 1985), rat (Donnerer et al., 1987), toad (Oka et al., 1985), and human skin cells (Weinstein et al., 2002), as well as the human keratinocyte cell line HaCaT (Poeaknapo et al., 2004) contain EM. EM has also been detected in the mouse (Horak et al., 1993), rat (Donnerer et al., 1987) and human hearts (Zhu et al., 2001b) as well as in granulocytes (Zhu et al., 2005; Boettcher et al., 2006; Glattard et al., 2010) and mononuclear cells (Boettcher et al., 2006). The reported amounts of EM in these different tissues and cells vary from one study to another. Such variation may result from several physiological conditions known to increase EM levels in plasma or tissues. In mice models in particular, the EM levels detected in the brain, heart and spleen have been found to be up to six times higher in the afternoon (16–17 h) than in the morning (9–10 h), indicating a circadian influence on EM synthesis (Horak et al., 1993). Many of the other conditions discussed in the following paragraphs also increase EM levels in the plasma or tissues. These conditions are described in some detail to help determine the roles of EM. Different cellular partners may be involved in endogenous opiate biosynthesis

Is EM biosynthesis restricted to catecholaminergic cells? Since the characterization in 2005 of the EM biosynthetic pathway in the catecholaminergic human neuroblastoma SH-SY5Y (Poeaknapo et al., 2004; Boettcher et al., 2005), it has been postulated that EM

synthesis occurs in dopaminergic/catecholaminergic cells specifically at the periphery surrounded by chromaffin (Goumon and Stefano, 2000; Goumon et al., 2000c; Goumon et al., 2006), polymorphonuclear (PMN) (Zhu et al., 2005) and mononuclear (MN) cells (Boettcher et al., 2006). Moreover, dopamine is necessary for EM formation (Neri et al., 2008). As described in this chapter, endogenous opiates are present in non-catecholaminergic cells; more specifically, they are found in the CNS in astrocytes and GABAergic cells (Muller et al., 2008; Laux et al., 2011, 2012). Hence, the synthesis of EM within the CNS appears to be more complex than expected and cannot simply be extrapolated from data limited to the neuroblastoma cell line SH-SY5Y (Muller et al., 2008; Boettcher et al., 2005). The precursor-uptake hypothesis. In contrast to the periphery, where EM synthesis appears to occur in single dopaminergic and/or catecholaminergic cells, the presence of endogenous alkaloids in non-dopaminergic cells of the CNS suggests a possible uptake of EM precursors. Indeed, various precursors involved in different stages of the EM synthetic pathway could be taken up from the extracellular space, cerebrospinal fluid or blood by cells expressing the enzymes necessary to complete the synthesis of EM. One possibility is that in the CNS, astrocytes and GABAergic neurons uptake morphine precursors to finalize the synthesis of EM. Indeed, the main dopamine transporter DAT is found at the synaptic terminals of dopaminergic neurons but is also expressed by astrocytes (Takeda et al., 2002) and non-dopaminergic neurons (Ciliax et al., 1995; Freed et al., 1995). This dopamine uptake may also rely on OCT-3 (organic cation transporter 3), which is expressed by astrocytes (Gasser et al., 2009) and able to transport dopamine to non-dopaminergic cells (Chemuturi and Donovan, 2007). The PMAT (plasma membrane monoamine transporter), which can also transport dopamine, is

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

present in many CNS neurons (Dahlin et al., 2007). Finally, EMT (extraneuronal monoamine transporter) and NET (norepinephrine transporter) are also involved in dopamine uptake by astrocytes (Inazu et al., 2003a,b) and may contribute to precursor uptake. Until now, only one study has reported an interaction between THP and DAT in which THP blocks dopamine transport (Okada et al., 1998). It is also unclear whether THP may itself be transported by DAT. Unfortunately, no information is available concerning the possible transport of other precursors of EM, and the possibility of morphine capture by GABAergic neurons cannot be excluded. Localization of enzymes involved in endogenous morphine biosynthesis. In mammals, the presence of enzymes involved in dopamine synthesis (TH and AADC) is necessary for EM formation. Nevertheless, these enzymes, which are mainly present in dopaminergic neurons, are not expressed by ‘‘morphinergic’’ neurons or astrocytes (Jaeger, 1985; Laux et al., 2011, 2012). The capture of precursors described in the previous paragraph therefore seems to represent an essential step for EM synthesis in CNS cells. The formation of DOPAL from dopamine by an MAO is a crucial step in the formation of THP and the consequent generation of EM. Two types of MAOs (MAO-A and MAOB) are expressed by neurons and astrocytes in many CNS structures (Ekblom et al., 1993; Ikemoto et al., 1997a,b; Jahng et al., 1997), including nondopaminergic cells, and may thus allow cells to produce THP. Following THP formation, three methylation steps are necessary for the formation of reticuline. In the past few years, it has been proposed, albeit without demonstration, that catechol-O-methyltransferase (COMT) and phenylethanolamine N-methyltransferase (PNMT) may be involved in at least one of these steps (Mantione et al., 2008). It was also recently reported that N-methyltransferase (NMT) transforms (R)norreticuline to (R)-reticuline in mammals, while PNMT has no activity (Grobe et al., 2011). NMT was initially reported to catalyze the N-methylation of tryptamine (Thompson et al., 1999) present within the CNS (Boarder and Rodnight, 1976). Nevertheless, the cellular localization of NMT remains unknown. Among the members of the cytochrome P450 enzyme family, which is involved in many enzymatic reactions, CYP2D6 is widely expressed in CNS neurons (Norris et al., 1996; Siegle et al., 2001) but not expressed in human astrocytes (Siegle et al., 2001). Because CYP2D6 catalyzes four steps of the EM biosynthetic pathway, the presence of this enzyme in cells appears to be crucial for the de novo synthesis of EM. The absence of this enzyme in astrocytes therefore suggests that this synthesis does not occur in astrocytes. However, the existence of another p450 enzyme involved in morphine biosynthesis cannot be ruled out. At the present time, none of these enzymes can be used for a mouse knock-out model depleted for endogenous opiates because they are implicated in a

107

number of crucial physiological processes. Additional investigations are required for the discovery of specific EM biosynthesis enzymes and the generation of a relevant animal model with disrupted EM synthesis.

WHAT ARE THE ROLES OF ENDOGENOUS OPIATES? Endogenous opiate levels The effects of exogenous morphine on the CNS at concentrations commonly used for many therapeutic purposes (lM range) have been extensively detailed in the literature. These concentrations are far higher than those detected for EM (pM-nM). It is therefore difficult to extrapolate the potential effects of EM from the effects observed for exogenous morphine (Perea-Sasiaı´ n, 2008). Under healthy conditions in humans, peripheral EM concentrations are barely detectable (pM). In contrast, EM blood levels increase to the nM range under pathological conditions, particularly under states of infection or inflammation (see next paragraph) (Glattard et al., 2010; Laux-Biehlmann et al., 2012). Within the CNS, EM has been found to increase from fmoles to pmoles per gram of tissue, making it difficult to determine the effective local concentrations. The effects of EM in the CNS have not been well documented and are often extrapolated from results obtained with low amounts of administered exogenous morphine. At the periphery: stress and immune responses Endogenous opiates in the adrenal medulla. As mentioned above, EM and endogenous M6G are present in chromaffin cell granules of the adrenal medulla, a major stress response organ, by releasing catecholamines (e.g., adrenaline) into the blood. Studies have shown that primary bovine chromaffin cell cultures contain M6G in their secretory granules, which is secreted upon nicotine stimulation (Goumon et al., 2005, 2006). The involvement of these cells in response to stress suggests that M6G is co-secreted with catecholamines and could therefore be involved in stress responses. As no studies have been performed in human chromaffin cells, the presence of EM or endogenous M6G in these cells is unknown. Endogenous opiates in the immune system. EM synthesis has been demonstrated in immune cells including human monocytes and polymorphonuclear cells, suggesting an involvement of EM in immune responses (Boettcher et al., 2005; Zhu et al., 2005; Glattard et al., 2010; Laux-Biehlmann et al., 2012). Accordingly, EM levels were found to be significantly higher in the blood of patients and pigs having had coronary bypass or laparotomy surgeries compared to healthy donors and control animals. For example, a coronary bypass surgery performed in humans increased blood concentrations of EM from 0.28 to 3.9 nM (Brix-Christensen et al., 1997). Similarly, coronary bypass surgery in piglets induced EM blood

108

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

levels of 10 nM, whereas no EM was detected in the control group (Brix-Christensen et al., 2000). Interestingly, two studies carried out in humans have shown that open cholecystectomy significantly increased EM levels in the blood (0.2 nM) compared to a laparoscopic procedure (0.018 nM) (Yoshida et al., 2000; Madbouly et al., 2010). Together, these data suggest that this EM increase induces particular effects as the blood concentrations during these states are compatible with MOR affinity (nM). Alternatively, lipopolysaccharide injections in rats or injections of muramyl dipeptide in mice to mimic bacterial infection have been shown to increase EM levels in tissues (Horak et al., 1993; Goumon et al., 2000a). Other stresses such as fasting cause a similar increase in EM concentrations in the blood and the spinal cord (Molina et al., 1995a; Meijerink et al., 1999; Goumon et al., 2000a). The immunosuppressive effects of morphine via NO production by immune cells inhibit the synthesis of proinflammatory cytokines, chemotaxis and phagocytosis (Stefano et al., 2000). In addition, morphine-induced NO production by the endothelium reduces the expression of adhesion molecules, allowing for the recruitment of immune cells toward inflammation sites and activating the hypothalamic–pituitary–adrenal axis, which promotes glucocorticoid release (Mellon and Bayer, 1998a). Morphine also inhibits immune stem cell differentiation into lymphocytes, T-cell proliferation (Mellon and Bayer, 1998b) and the cytolytic activity of natural killer cells (Mellon and Bayer, 1999). It is important to note that EM levels during sepsis (8 nM) significantly inhibit the secretion of IL-8 in vitro from human polymorphonuclear cells primed with LPS (Glattard et al., 2010). The release of EM in the blood under stress conditions may represent a physiological mechanism that reduces inflammatory responses (i.e., anti-inflammatory effects). Proposed roles in the CNS

Do endogenous opiates act as neurotransmitters or neuromediators? The role of EM as a neurotransmitter has been considered for many years (for review: Guarna et al., 2005). In fact, EM satisfies several neurotransmitter criteria: neuronal synthesis, presynaptic vesicular localization, release upon potassium depolarization (Guarna et al., 1998) or nicotine stimulation (Muller et al., 2008) and reuptake by neurons (Bianchi et al., 1993). However, recent studies have shown that EM and EMM are primarily localized in the postsynaptic compartment, except in the cerebellum, where they are present in the presynaptic terminals of basket cells (BC) innervating the cell bodies of Purkinje neurons (Muller et al., 2008; Laux et al., 2011). This particular location, which is likely exclusive to the cerebellum, casts doubt on the hypothesis that morphine functions as a neurotransmitter. However, it is possible that only a minority of neurons, such as basket cells, are capable of EM storage in their presynaptic terminals (Bianchi et al., 1994; Muller et al., 2008; Laux et al., 2011).

In neurons, where EM is located in the postsynaptic compartment, another role for this alkaloid warrants consideration. While neurotransmission ‘‘traditionally’’ occurs by the release of neurotransmitters from the presynaptic terminal, some postsynaptic terminals release neurotransmitters or neuromediators in a ‘‘retrograde’’ manner to activate presynaptic receptors (examples include nitric oxide and endocannabinoids; for review: Ludwig and Pittman, 2003). The presence of EM and its derivatives in dendrites and the postsynaptic endings of many neurons throughout the CNS is not trivial and may be indicative of potential retrograde release. Moreover, the presence of opioid receptors, especially MORs, in the presynaptic regions of some neurons strengthen this idea (Drake and Milner, 1999; Svingos et al., 2001a,b; Zhang and Pan, 2010). Endogenous opiates and control of nociception. Based on the analgesic effects of exogenous morphine, one of the first roles that comes to mind for its endogenous counterpart is the control of nociception processes. Accordingly, multiple studies have suggested that EM may be involved in these processes. First, immunohistochemical approaches have indicated the presence of EM in the neurons and processes of CNS areas involved in the control of nociception, including the rostroventral medulla, periaqueductal gray matter, raphe magnus nuclei and superficial laminae of the spinal cord (Stefano et al., 2000; Laux et al., 2011, 2012). Moreover, when anti-morphine antibodies were injected into the CSF of mice, Guarna et al. (2002) observed reduced levels of cerebral EM. This treatment reduced the latency of paw licking, indicating that thermal hyperalgesia occurred when the EM level was reduced. On the other hand, KO mice for MOR have an increased sensitivity to thermal nociceptive stimuli but not mechanical stimuli (Kieffer and Gaveriaux-Ruff, 2002). Endogenous opiates and memory. EM has been described in neurons and astrocytes in the hippocampus, suggesting a possible role of EM in memory processes. More precisely, EM is present in GABAergic neurons of Amon’s horn and in GABAergic basket cells of the subgranular zone (SGZ) of the dentate gyrus (Laux et al., 2011). A study using injections of anti-morphine antibodies into the CSF of mice suggested a link between EM and memory (Guarna et al., 2004). In this experiment, EM CNS levels were significantly elevated after 12 h of fasting compared to control animals. When these animals were tested for memory, they showed impaired memory acquisition and consolidation due to increased EM levels. Moreover, this impairment was abolished when the EM was immunodepleted. These data suggest that EM might, under stress conditions such as fasting, inhibit memory processes. Endogenous opiates and addiction. The role of dopamine, a precursor of EM, as a key neurotransmitter for addictive processes prompted the consideration of a possible link between EM and addiction. Work

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

performed on two invertebrate models, Mytilus edulis and Homarus americanus, showed that exposure to addictive substances (ethanol, nicotine or cocaine) increases EM release by a factor of two in the nervous systems of these organisms (Zhu et al., 2006a,b). In mammals, a release of EM by human immune cells after an administration of cocaine, alcohol or nicotine has been reported (Zhu et al., 2006a). Interestingly, alcoholism induces an overexpression of CYP2D6, an enzyme involved in four steps of the EM synthesis pathway (Miksys et al., 2002), in different cerebral structures. Finally, it has been shown that alcoholism dramatically increases circulating levels of THP, which is a precursor of EM synthesis in mammals (Haber et al., 1997; Sango et al., 2000; McCoy et al., 2003). On the basis of these findings, a link between EM and addiction has been proposed (Stefano et al., 2007). Endogenous opiates, neurogenesis and structural plasticity. In the adult mammalian brain, the subventricular zone (SVZ) bordering the lateral ventricle and the SGZ of the hippocampus dentate gyrus are both adult neurogenesis niches. Exogenously administered morphine regulates the survival and production of neural progenitors and also affects neuronal plasticity. Indeed, morphine inhibits neurogenesis in the SGZ of mice and adult rats through its action on MOR (Harburg et al., 2007; Arguello et al., 2008; Fischer et al., 2008; Sargeant et al., 2008). Morphine causes structural changes in different areas of the CNS (e.g., the nucleus accumbens), affecting dendrites and the dendritic spine structure (Harston et al., 1980; Ikeda et al., 2000, 2010; Badiani and Robinson, 2004; Robinson and Kolb, 2004). Morphine has also been found to promote axonal and synaptic regeneration in the spinal cord of injured mice (Zeng et al., 2007). Together, these effects of exogenously administered morphine on neurogenesis and structural plasticity highlight a potential role for EM. Additionally, EM and EMM are present in the cells of the SGZ and the SVZ. In both cases, EM and its derivatives are notably present in radial astrocytes (Laux et al., 2012). Using nestin, an immunohistochemical marker for radial glialike progenitors (von Bohlen Und Halbach, 2007), unpublished data from our lab have confirmed EM localization in these cells. The presence of EM in radial glia-like progenitors of the SGZ and SVZ suggest an involvement of this endogenous alkaloid in neurogenesis. Furthermore, these cells express MOR (Persson et al., 2003a,b), and the action of endogenous opioids on MOR has been found to inhibit neurogenesis induced by an ischemia in the SGZ (Kolodziej et al., 2008). Interestingly, the dentate gyrus of MOR-KO mice contains a greater quantity of immature postmitotic neurons (Harburg et al., 2007). Exogenous morphine has an effect on structural plasticity in vitro. A high morphine concentration (1 mM) inhibits the neurite outgrowth of hippocampal neurons or of granular cerebellum neurons in primary culture

109

(Brailoiu et al., 2004). Conversely, a low concentration (<1 pM) stimulates the neurite outgrowth of PC-12 cells and spinal cord or cortex neurons independently of MOR (Tenconi et al., 1996; Brailoiu et al., 2004). In the absence of direct evidence, EM or EMM control of neurogenesis and brain plasticity cannot be excluded, particularly during pathological states leading to increased biosynthesis. Endogenous opiates and neuroinflammation. Recent data suggest that morphine and M3G bind TLR4 and, more specifically, its accessory protein MD2 (Hutchinson et al., 2010b,c). TLR4 is a pathogenassociated molecular pattern receptor involved in neuroinflammatory responses to various CNS stimuli through the activation of microglial cells and astrocytes. TLR4 is mainly present in immune cells and microglia. However, its presence has been reported on other CNS cells including astrocytes and neurons (for review: Buchanan et al., 2010). The action of morphine and M3G on TLR4 presents new research perspectives in the area of endogenous opiates. It would be particularly interesting, for example, to investigate the effect of endogenous opiates (morphine, M6G and M3G ratios) on neuroinflammation. EM may act on both MOR and TLR4 present on astrocytes and microglial cells, whereas the action of endogenous M3G would be restricted to the TLR4 receptor. Surprisingly, activated astrocytes over-express UGT1A6 under infectious states (Heurtaux et al., 2006) and accordingly, the EM/ M3G ratio would be altered during a neuroinflammatory state and directly activate MOR/TLR4.

ENDOGENOUS OPIATES AND CNS PATHOLOGIES Schizophrenia A ‘‘hypermorphinergic’’ pathology? Schizophrenia is a psychosis manifested by signs of mental dissociation, emotional discordance and delusional activity. It is a major cause of disability worldwide (Murray and Lopez, 1996). The dopamine deregulation hypothesis for schizophrenia is often proposed as the underlying neurobiological mechanism of this disease. This hypothesis postulates that an excess of dopamine in the mesolimbic pathway is responsible for some schizophrenia symptoms (for review: Cousins et al., 2009; Heinz and Schlagenhauf, 2010; Miyake et al., 2011). Because dopamine is an EM precursor, an increase in EM levels in CNS tissues would be expected. Using a mouse model of synaptic and hyperdopaminergic aspects of schizophrenia (Andrieux et al., 2002; Brun et al., 2005), we recently observed increased levels of brain EM in STOP mice compared to wild-type mice (Charlet et al., 2010). Interestingly, COMT, a potential factor in the methylation of morphine precursors, is more active in certain brain regions of schizophrenic patients. This increase in activity is primarily due to the presence of the Val105/158 Met polymorphism (reviewed in: (Sagud et al., 2010)).

110

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

However, no data concerning the EM levels in schizophrenic patients are available. It is interesting to note that in some cases, opioid antagonists (e.g., naloxone, naltrexone) were previously used to treat schizophrenia associated with addiction, implicating MOR (McNicholas and Martin, 1984; Welch and Thompson, 1994; Wonodi et al., 2004). Thus, high levels of EM may be related to some of the symptoms observed in this pathology. Endogenous morphine, schizophrenia and nociception. Among the symptoms of schizophrenia, clinical observations reveal a lack of pain expression during normally painful situations in schizophrenic patients (burns, fractures, etc.). However, this phenomenon is poorly understood, and two hypotheses have been proposed: (i) a modification of endogenous pain controls and (ii) differing conscious interpretations of nociceptive signals (Blumensohn et al., 2002; Autie et al., 2009; Bonnot et al., 2009). The higher levels of EM potentially present in schizophrenic patients may explain functional changes in endogenous pain control. The A118G polymorphism of the OPRM1 gene occurs more frequently in patients with schizophrenia, suggesting the involvement of MOR and the opioid/opiate system in schizophrenic pain symptoms (Sery et al., 2010). Interestingly, patients with this polymorphism are described to be less sensitive to morphine (for review: Mague and Blendy, 2010). More investigations are needed to determine the precise role of EM in schizophrenia and may lead to the development of new treatments for schizophrenic symptoms. Parkinson’s disease Parkinson’s disease is primarily characterized by movement disorders, such as slowness (akinesia), rigid movements (hypertonia) and tremors, which are consequences of the degeneration of dopaminergic neurons in the substantia nigra pars compacta. Ultimately, these changes result in a depletion of dopamine in the CNS. We recently reported an increase in the levels of EM and its metabolites in the CNS of Parkinson’s disease patients and in different animal models (Charron et al., 2011). This increase may be the result of increased dopaminergic catabolism and higher amounts of the morphine precursor THP. Higher levels of endogenous opiates in the brain may have varying implications: (i) they may be responsible for some motor symptoms of Parkinson’s disease or (ii) they may be one of the many compensation mechanisms established by the CNS (Bezard and Gross, 1998; Bezard et al., 2003). Because EM and its metabolites are present in CNS areas involved in motor control, movement and posture, these alkaloids may be implicated in motor function. Thus, a change in the levels or ratios of endogenous opiates may result in an impairment of these functions (Laux et al., 2011, 2012). In addition to motor symptoms, Parkinson’s disease is also associated with pain symptoms at a prevalence of approximately 40% (for review: Chaudhuri and Odin, 2010; Ford, 2010; Meissner et al., 2011). Warm thermal

hyperalgesia is observed in some patients (Schestatsky et al., 2007), while thermal hypoalgesia to cold is observed in others (Brefel-Courbon et al., 2005). In both cases, an improvement in these symptoms is observed after L-DOPA treatment. We have similarly observed a return to normal EM levels following L-DOPA treatments in patients and in Parkinson’s disease models (Charron et al., 2011). Because EM and its derivatives may be involved in nociception control, increases in EM or specific EMM species may be contributing factors or the cause of pain symptoms observed in Parkinson’s disease patients. The correlation between L-DOPA treatments and pain relief as well as reduced EM further supports the hypothesis of EM involvement in these symptoms. At the present time, it is not possible to determine whether increased levels of endogenous opiates are a cause or consequence of parkinsonian symptoms. Further investigations are required to reveal whether these endogenous compounds represent therapeutic targets.

CONCLUSION Although no clear function has yet been attributed to EM and EMM in the CNS, their presence in specific neurons and astrocytes cannot be considered as fortuitous. Besides its potential implication in pain-regulatory processes, the presence of these endogenous compounds in brain regions that are not usually involved in pain modulation opens exciting perspectives to extend the role and function of this endogenous opiate far beyond analgesic functions. Acknowledgements—We are most grateful to Mrs. Elise Savier and Mr. Leny teyssier for their advice on the manuscript.

REFERENCES Aceto MD, Bowman ER, Harris LS, May EL (1989) Dependence studies on new compounds in the rhesus monkey, rat and mouse. NIDA Res Monogr 95:578–631. Aceto MD, Harris LS, Abood ME, Rice KC (1999) Stereoselective muand delta-opioid receptor-related antinociception and binding with (+)thebaine. Eur J Pharmacol 365:143–147. Al-Hasani R, Bruchas MR (2012) Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology 115:1363–1381. Andrieux A, Salin PA, Vernet M, Kujala P, Baratier J, Gory-Faure S, Bosc C, Pointu H, Proietto D, Schweitzer A, Denarier E, Klumperman J, Job D (2002) The suppression of brain coldstable microtubules in mice induces synaptic defects associated with neuroleptic-sensitive behavioral disorders. Genes Dev 16:2350–2364. Arguello AA, Harburg GC, Schonborn JR, Mandyam CD, Yamaguchi M, Eisch AJ (2008) Time course of morphine’s effects on adult hippocampal subgranular zone reveals preferential inhibition of cells in S phase of the cell cycle and a subpopulation of immature neurons. Neuroscience 157:70–79. Autie A, Montreuil M, Moulier V, Braha S, Wojakiewicz A, Januel D (2009) Pain and schizophrenia: myth and reality. Encephale 35:297–303.

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117 Badiani A, Robinson TE (2004) Drug-induced neurobehavioral plasticity: the role of environmental context. Behav Pharmacol 15:327–339. Bezard E, Gross CE (1998) Compensatory mechanisms in experimental and human parkinsonism: towards a dynamic approach. Prog Neurobiol 55:93–116. Bezard E, Gross CE, Brotchie JM (2003) Presymptomatic compensation in Parkinson’s disease is not dopamine-mediated. Trends Neurosci 26:215–221. Bianchi E, Alessandrini C, Guarna M, Tagliamonte A (1993) Endogenous codeine and morphine are stored in specific brain neurons. Brain Res 627:210–215. Bianchi E, Guarna M, Tagliamonte A (1994) Immunocytochemical localization of endogenous codeine and morphine. Adv Neuroimmunol 4:83–92. Bilkova A, Bezakova L, Bilka F, Psenak M (2005) An amine oxidase in seedlings of Papaver somniferum L.. Biol Plant 49:389–394. Bird DA, Franceschi VR, Facchini PJ (2003) A tale of three cell types: alkaloid biosynthesis is localized to sieve elements in opium poppy. Plant Cell 15:2626–2635. Blume AJ, Shorr J, Finberg JP, Spector S (1977) Binding of the endogenous nonpeptide morphine-like compound to opiate receptors. Proc Natl Acad Sci U S A 74:4927–4931. Blumensohn R, Ringler D, Eli I (2002) Pain perception in patients with schizophrenia. J Nerv Ment Dis 190:481–483. Boarder MR, Rodnight R (1976) Tryptamine-N-methyltransferase activity in brain tissue: a re-examination. Brain Res 114:359–364. Boettcher C, Fellermeier M, Boettcher C, Drager B, Zenk MH (2005) How human neuroblastoma cells make morphine. Proc Natl Acad Sci U S A 102:8495–8500. Boettcher C, Fischer W, Zenk MH (2006) Comment on ‘‘human white blood cells synthesize morphine: CYP2D6 modulation’’. J Immunol 176:5703–5704. Bonnot O, Anderson GM, Cohen D, Willer JC, Tordjman S (2009) Are patients with schizophrenia insensitive to pain? A reconsideration of the question. Clin J Pain 25:244–252. Borner U, Abbott S (1973) New observations in the metabolism of morphine. The formation of codeine from morphine in man. Experientia 29:180–181. Bourasset F, Cisternino S, Temsamani J, Scherrmann JM (2003) Evidence for an active transport of morphine-6-beta-D-glucuronide but not P-glycoprotein-mediated at the blood–brain barrier. J Neurochem 86:1564–1567. Bradbury AF, Smyth DG, Snell CR (1976) Biosynthetic origin and receptor conformation of methionine enkephalin. Nature 260:165–166. Brailoiu E, Hoard J, Brailoiu GC, Chi M, Godbolde R, Dun NJ (2004) Ultra low concentrations of morphine increase neurite outgrowth in cultured rat spinal cord and cerebral cortical neurons. Neurosci Lett 365:10–13. Brefel-Courbon C, Payoux P, Thalamas C, Ory F, Quelven I, Chollet F, Montastruc JL, Rascol O (2005) Effect of levodopa on pain threshold in Parkinson’s disease: a clinical and positron emission tomography study. Mov Disord 20:1557–1563. Brix-Christensen V, Tonnesen E, Sanchez RG, Bilfinger TV, Stefano GB (1997) Endogenous morphine levels increase following cardiac surgery as part of the antiinflammatory response? Int J Cardiol 62:191–197. Brix-Christensen V, Goumon Y, Tonnesen E, Chew M, Bilfinger T, Stefano GB (2000) Endogenous morphine is produced in response to cardiopulmonary bypass in neonatal pigs. Acta Anaesthesiol Scand 44:1204–1208. Bromek E, Haduch A, Golembiowska K, Daniel WA (2011) Cytochrome P450 mediates dopamine formation in the brain in vivo. J Neurochem 118:806–815. Brown CE, Roerig SC, Burger VT, Cody Jr RB, Fujimoto JM (1985) Analgesic potencies of morphine 3- and 6-sulfates after intracerebroventricular administration in mice: relationship to structural characteristics defined by mass spectrometry and nuclear magnetic resonance. J Pharm Sci 74:821–824.

111

Brun P, Begou M, Andrieux A, Mouly-Badina L, Clerget M, Schweitzer A, Scarna H, Renaud B, Job D, Suaud-Chagny MF (2005) Dopaminergic transmission in STOP null mice. J Neurochem 94:63–73. Buchanan MM, Hutchinson M, Watkins LR, Yin H (2010) Toll-like receptor 4 in CNS pathologies. J Neurochem 114:13–27. Buckley DB, Klaassen CD (2007) Tissue- and gender-specific mRNA expression of UDP-glucuronosyltransferases (UGTs) in mice. Drug Metab Dispos 35:121–127. Calatozzolo C, Gelati M, Ciusani E, Sciacca FL, Pollo B, Cajola L, Marras C, Silvani A, Vitellaro-Zuccarello L, Croci D, Boiardi A, Salmaggi A (2005) Expression of drug resistance proteins Pgp, MRP1, MRP3, MRP5 and GST-pi in human glioma. J Neurooncol 74:113–121. Casares FM, McElroy A, Mantione K, Baggermann G, Zhu W, Stefano GB (2005) The American lobster, Homarus americanus, contains morphine that is coupled to nitric oxide release in its nervous and immune tissues: evidence for neurotransmitter and hormonal signaling. Neuro Endocrinol Lett 26:89–97. Cashaw JL (1993a) Determination of tetrahydropapaveroline in the urine of parkinsonian patients receiving L-dopa-carbidopa (Sinemet) therapy by high-performance liquid chromatography. J Chromatogr 613:267–273. Cashaw JL (1993b) Tetrahydropapaveroline in brain-regions of rats after acute ethanol administration. Alcohol 10:133–138. Cashaw JL, Ruchirawat S, Nimit Y, Davis VE (1983) Regioselective O-methylation of tetrahydropapaveroline and tetrahydroxyberbine in vivo in rat brain. Biochem Pharmacol 32:3163–3169. Charlet A, Muller AH, Laux A, Kemmel V, Schweitzer A, Deloulme JC, Stuber D, Delalande F, Bianchi E, Van Dorsselaer A, Aunis D, Andrieux A, Poisbeau P, Goumon Y (2010) Abnormal nociception and opiate sensitivity of STOP null mice exhibiting elevated levels of the endogenous alkaloid morphine. Mol Pain 6:96. Charron G, Doudnikoff E, Laux A, Berthet A, Porras G, Canron MH, Barroso-Chinea P, Li Q, Qin C, Nosten-Bertrand M, Giros B, Delalande D, Van Dorsselaer A, Vital A, Goumon Y, Bezard E (2011) Endogenous morphine-like compound immunoreactivity increases in Parkinsonism. Brain 134:2321–2338. Chaudhuri KR, Odin P (2010) The challenge of non-motor symptoms in Parkinson’s disease. Prog Brain Res 184:325–341. Chemuturi NV, Donovan MD (2007) Role of organic cation transporters in dopamine uptake across olfactory and nasal respiratory tissues. Mol Pharm 4:936–942. Chen Y, Sommer C (2009) The role of mitogen-activated protein kinase (MAPK) in morphine tolerance and dependence. Mol Neurobiol 40:101–107. Chen JJ, Chang YL, Teng CM, Chen IS (2000) Anti-platelet aggregation alkaloids and lignans from Hernandia nymphaeifolia. Planta Med 66:251–256. Chen XY, Zhao LM, Zhong DF (2003) A novel metabolic pathway of morphine: formation of morphine glucosides in cancer patients. Br J Clin Pharmacol 55:570–578. Ciliax BJ, Heilman C, Demchyshyn LL, Pristupa ZB, Ince E, Hersch SM, Niznik HB, Levey AI (1995) The dopamine transporter: immunochemical characterization and localization in brain. J Neurosci 15:1714–1723. Coller JK, Christrup LL, Somogyi AA (2009) Role of active metabolites in the use of opioids. Eur J Clin Pharmacol 65:121–139. Cone EJ, Heit HA, Caplan YH, Gourlay D (2006) Evidence of morphine metabolism to hydromorphone in pain patients chronically treated with morphine. J Anal Toxicol 30:1–5. Cone EJ, Caplan YH, Moser F, Robert T, Black D (2008) Evidence that morphine is metabolized to hydromorphone but not to oxymorphone. J Anal Toxicol 32:319–323. Cousins DA, Butts K, Young AH (2009) The role of dopamine in bipolar disorder. Bipolar disord 11:787–806. Cox BM, Opheim KE, Teschemacher H, Goldstein A (1975) A peptide-like substance from pituitary that acts like morphine. 2. Purification and properties. Life Sci 16:1777–1782.

112

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Dagenais C, Zong J, Ducharme J, Pollack GM (2001) Effect of mdr1a P-glycoprotein gene disruption, gender, and substrate concentration on brain uptake of selected compounds. Pharm Res 18:957–963. Dahlin A, Xia L, Kong W, Hevner R, Wang J (2007) Expression and immunolocalization of the plasma membrane monoamine transporter in the brain. Neuroscience 146:1193–1211. Daly J, Inscoe JK, Axelrod J (1965) The formation of O-methylated catechols by microsomal hydroxylation of phenols and subsequent enzymatic catechol O-methylation. Substrate specificity. J Med Chem 8:153–157. Davis VE, Walsh MJ (1970) Alcohol, amines, and alkaloids: a possible biochemical basis for alcohol addiction. Science 167:1005–1007. Deitrich R, Erwin V (1980) Biogenic amine–aldehyde condensation products: tetrahydroisoquinolines and tryptolines (betacarbolines). Annu Rev Pharmacol Toxicol 20:55–80. Dias KL, Da Silva Dias C, Barbosa-Filho JM, Almeida RN, De Azevedo Correia N, Medeiros IA (2004) Cardiovascular effects induced by reticuline in normotensive rats. Planta Med 70:328–333. Dietis N, Rowbotham DJ, Lambert DG (2011) Opioid receptor subtypes: fact or artifact? Br J Anaesth 107:8–18. Donnerer J, Oka K, Brossi A, Rice KC, Spector S (1986) Presence and formation of codeine and morphine in the rat. Proc Natl Acad Sci U S A 83:4566–4567. Donnerer J, Cardinale G, Coffey J, Lisek CA, Jardine I, Spector S (1987) Chemical characterization and regulation of endogenous morphine and codeine in the rat. J Pharmacol Exp Ther 242:583–587. Drake CT, Milner TA (1999) Mu opioid receptors are in somatodendritic and axonal compartments of GABAergic neurons in rat hippocampal formation. Brain Res 849:203–215. Eisenreich WJ, Hofner G, Bracher F (2003) Alkaloids from Croton flavens L. and their affinities to GABA-receptors. Nat Prod Res 17:437–440. Ekblom J, Jossan SS, Bergstrom M, Oreland L, Walum E, Aquilonius SM (1993) Monoamine oxidase-B in astrocytes. Glia 8:122–132. Epple A, Navarro I, Horak P, Spector S (1993) Endogenous morphine and codeine: release by the chromaffin cells of the eel. Life Sci 52:PL117–PL121. Exner M, Alt E, Hermann M, Hofbauer R, Kapiotis S, Quehenberger P, Speiser W, Minar E, Gmeiner B (2001) Phydroxyphenylacetaldehyde, the major product of tyrosine oxidation by the activated myeloperoxidase system can act as an antioxidant in LDL. FEBS Lett 490:28–31. Facchini PJ (2001) Alkaloid biosynthesis in plants: biochemistry, cell biology, molecular regulation, and metabolic engineering applications. Annu Rev Plant Physiol Plant Mol Biol 52:29–66. Facchini PJ, De Luca V (1994) Differential and tissue-specific expression of a gene family for tyrosine/dopa decarboxylase in opium poppy. J Biol Chem 269:26684–26690. Facchini PJ, Park SU (2003) Developmental and inducible accumulation of gene transcripts involved in alkaloid biosynthesis in opium poppy. Phytochemistry 64:177–186. Fang S, Takemori AE, Portoghese PS (1984) Activities of morphinone and N-(cyclopropylmethyl)normorphinone at opioid receptors. J Med Chem 27:1361–1363. Feng Y, He X, Yang Y, Chao D, Lazarus LH, Xia Y (2012) Current research on opioid receptor function. Curr Drug Targets 13:230–246. Fischer SJ, Arguello AA, Charlton JJ, Fuller DC, Zachariou V, Eisch AJ (2008) Morphine blood levels, dependence, and regulation of hippocampal subgranular zone proliferation rely on administration paradigm. Neuroscience 151:1217–1224. Florang VR, Rees JN, Brogden NK, Anderson DG, Hurley TD, Doorn JA (2007) Inhibition of the oxidative metabolism of 3,4dihydroxyphenylacetaldehyde, a reactive intermediate of dopamine metabolism, by 4-hydroxy-2-nonenal. Neurotoxicology 28:76–82.

Foldes A, Meek JL (1973) Rat brain phenolsulfotransferase: partial purification and some properties. Biochim Biophys Acta 327:365–374. Ford B (2010) Pain in Parkinson’s disease. Mov Disord 25(Suppl. 1):S98–S103. Freed C, Revay R, Vaughan RA, Kriek E, Grant S, Uhl GR, Kuhar MJ (1995) Dopamine transporter immunoreactivity in rat brain. J Comp Neurol 359:340–349. Fricchione G, Zhu W, Cadet P, Mantione KJ, Bromfield E, Madsen J, DeGirolami U, Dworetzky B, Vaccaro B, Black P, Stefano GB (2008) Identification of endogenous morphine and a mu3-like opiate alkaloid receptor in human brain tissue taken from a patient with intractable complex partial epilepsy. Med Sci Monit 14:CS45–CS49. Frolich N, Dees C, Paetz C, Ren X, Lohse MJ, Nikolaev VO, Zenk MH (2011) Distinct pharmacological properties of morphine metabolites at G(i)-protein and beta-arrestin signaling pathways activated by the human mu-opioid receptor. Biochem Pharmacol 81:1248–1254. Gaillard PJ, van der Sandt IC, Voorwinden LH, Vu D, Nielsen JL, de Boer AG, Breimer DD (2000) Astrocytes increase the functional expression of P-glycoprotein in an in vitro model of the blood– brain barrier. Pharm Res 17:1198–1205. Gasser PJ, Orchinik M, Raju I, Lowry CA (2009) Distribution of organic cation transporter 3, a corticosterone-sensitive monoamine transporter, in the rat brain. J Comp Neurol 512:529–555. Gerardy R, Zenk MH (1993) Purification and characterization of salutaridine: NADPH 7-oxidoreductase from Papaver somniferum. Phytochemistry 34:125–132. Gesell A, Rolf M, Ziegler J, Diaz Chavez ML, Huang FC, Kutchan TM (2009) CYP719B1 is salutaridine synthase, the C–C phenolcoupling enzyme of morphine biosynthesis in opium poppy. J Biol Chem 284:24432–24442. Gintzler AR, Levy A, Spector S (1976a) Antibodies as a means of isolating and characterizing biologically active substances: presence of a non-peptide, morphine-like compound in the central nervous system. Proc Natl Acad Sci U S A 73:2132–2136. Gintzler AR, Mohacsi E, Spector S (1976b) Radioimmunoassay for the simultaneous determination of morphine and codeine. Eur J Pharmacol 38:149–156. Gintzler AR, Gershon MD, Spector S (1978) A nonpeptide morphinelike compound: immunocytochemical localization in the mouse brain. Science 199:447–448. Glattard E, Welters ID, Lavaux T, Muller AH, Laux A, Zhang D, Schmidt AR, Delalande F, Laventie BJ, Dirrig-Grosch S, Colin DA, Van Dorsselaer A, Aunis D, Metz-Boutigue MH, Schneider F, Goumon Y (2010) Endogenous morphine levels are increased in sepsis: a partial implication of neutrophils. PLoS One 5:e8791. Goldstein A, Tachibana S, Lowney LI, Hunkapiller M, Hood L (1979) Dynorphin-(1-13), an extraordinarily potent opioid peptide. Proc Natl Acad Sci U S A 76:6666–6670. Goldstein A, Barrett RW, James IF, Lowney LI, Weitz CJ, Knipmeyer LL, Rapoport H (1985) Morphine and other opiates from beef brain and adrenal. Proc Natl Acad Sci U S A 82:5203–5207. Gong QL, Hedner J, Bjorkman R, Hedner T (1992) Morphine-3glucuronide may functionally antagonize morphine-6-glucuronide induced antinociception and ventilatory depression in the rat. Pain 48:249–255. Goumon Y, Stefano GB (2000) Identification of morphine in the rat adrenal gland. Brain Res Mol Brain Res 77:267–269. Goumon Y, Bouret S, Casares F, Zhu W, Beauvillain JC, Stefano GB (2000a) Lipopolysaccharide increases endogenous morphine levels in rat brain. Neurosci Lett 293:135–138. Goumon Y, Casares F, Pryor S, Ferguson L, Brownawell B, Cadet P, Rialas CM, Welters ID, Sonetti D, Stefano GB (2000b) Ascaris suum, an intestinal parasite, produces morphine. J Immunol 165:339–343. Goumon Y, Zhu W, Weeks BS, Casares F, Cadet P, Bougaeva M, Brownawell B, Stefano GB (2000c) Identification of morphine in

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117 the adrenal medullary chromaffin PC-12 cell line. Brain Res Mol Brain Res 81:177–180. Goumon Y, Casares F, Zhu W, Stefano GB (2001) The presence of morphine in ganglionic tissues of Modiolus deminissus: a highly sensitive method of quantitation for morphine and its derivatives. Brain Res Mol Brain Res 86:184–188. Goumon Y, Strub JM, Stefano GB, Van Dorsselaer A, Aunis D, MetzBoutigue MH (2005) Characterization of a morphine-like molecule in secretory granules of chromaffin cells. Med Sci Monit 11:MS31–MS34. Goumon Y, Muller A, Glattard E, Marban C, Gasnier C, Strub JM, Chasserot-Golaz S, Rohr O, Stefano GB, Welters ID, Van Dorsselaer A, Schoentgen F, Aunis D, Metz-Boutigue MH (2006) Identification of morphine-6-glucuronide in chromaffin cell secretory granules. J Biol Chem 281:8082–8089. Goumon Y, Laux A, Muller A, Aunis D (2009) Central and peripheral endogenous morphine. An R Acad Nac Farm 75:389–418. Graf L, Ronai A, Bajusz S, Csheh G, Szekely JI (1976) Opioid agonist activity of beta-lipotropin fragments: a possible biological source of morphine-like substances in the pituitary. FEBS Lett 64:181–184. Grobe N, Zhang B, Fisinger U, Kutchan TM, Zenk MH, Guengerich FP (2009) Mammalian cytochrome P450 enzymes catalyze the phenol-coupling step in endogenous morphine biosynthesis. J Biol Chem 284:24425–24431. Grobe N, Lamshoft M, Orth RG, Drager B, Kutchan TM, Zenk MH, Spiteller M (2010) Urinary excretion of morphine and biosynthetic precursors in mice. Proc Natl Acad Sci U S A 107:8147–8152. Grobe N, Ren X, Kutchan TM, Zenk MH (2011) An (R)-specific Nmethyltransferase involved in human morphine biosynthesis. Arch Biochem Biophys 506:42–47. Grothe T, Lenz R, Kutchan TM (2001) Molecular characterization of the salutaridinol 7-O-acetyltransferase involved in morphine biosynthesis in opium poppy Papaver somniferum. J Biol Chem 276:30717–30723. Guarna M, Neri C, Petrioli F, Bianchi E (1998) Potassium-induced release of endogenous morphine from rat brain slices. J Neurochem 70:147–152. Guarna M, Bianchi E, Bartolini A, Ghelardini C, Galeotti N, Bracci L, Neri C, Sonetti D, Stefano G (2002) Endogenous morphine modulates acute thermonociception in mice. J Neurochem 80:271–277. Guarna M, Ghelardini C, Galeotti N, Bartolini A, Noli L, Neri C, Stefano GB, Bianchi E (2004) Effects of endogenous morphine deprivation on memory retention of passive avoidance learning in mice. Int J Neuropsychopharmacol 7:311–319. Guarna M, Ghelardini C, Galeotti N, Stefano GB, Bianchi E (2005) Neurotransmitter role of endogenous morphine in CNS. Med Sci Monit 11:RA190–RA193. Haber H, Roske I, Rottmann M, Georgi M, Melzig MF (1997) Alcohol induces formation of morphine precursors in the striatum of rats. Life Sci 60:79–89. Hackler L, Zadina JE, Ge LJ, Kastin AJ (1997) Isolation of relatively large amounts of endomorphin-1 and endomorphin-2 from human brain cortex. Peptides 18:1635–1639. Hagel JM, Facchini PJ (2010) Dioxygenases catalyze the Odemethylation steps of morphine biosynthesis in opium poppy. Nat Chem Biol 6:273–275. Harburg GC, Hall FS, Harrist AV, Sora I, Uhl GR, Eisch AJ (2007) Knockout of the mu opioid receptor enhances the survival of adultgenerated hippocampal granule cell neurons. Neuroscience 144:77–87. Harston CT, Morrow A, Kostrzewa RM (1980) Enhancement of sprouting and putative regeneration of central noradrenergic fibers by morphine. Brain Res Bull 5:421–424. Hasselstrom J, Sawe J (1993) Morphine pharmacokinetics and metabolism in humans. Enterohepatic cycling and relative contribution of metabolites to active opioid concentrations. Clin Pharmacokinet 24:344–354. Hazen SL, Heller J, Hsu FF, d’Avignon A, Heinecke JW (1999) Synthesis, isolation, and characterization of the adduct formed in

113

the reaction of p-hydroxyphenylacetaldehyde with the amino headgroup of phosphatidylethanolamine and phosphatidylserine. Chem Res Toxicol 12:19–27. Hazen SL, Gaut JP, Crowley JR, Hsu FF, Heinecke JW (2000) Elevated levels of protein-bound p-hydroxyphenylacetaldehyde, an amino-acid-derived aldehyde generated by myeloperoxidase, are present in human fatty streaks, intermediate lesions and advanced atherosclerotic lesions. Biochem J 352(Pt. 3):693–699. Heinz A, Schlagenhauf F (2010) Dopaminergic dysfunction in schizophrenia: salience attribution revisited. Schizophr Bull 36:472–485. Heller JI, Crowley JR, Hazen SL, Salvay DM, Wagner P, Pennathur S, Heinecke JW (2000) P-hydroxyphenylacetaldehyde, an aldehyde generated by myeloperoxidase, modifies phospholipid amino groups of low density lipoprotein in human atherosclerotic intima. J Biol Chem 275:9957–9962. Heurtaux T, Benani A, Moulin D, Muller N, Netter P, Minn A (2006) Induction of UGT1A6 isoform by inflammatory conditions in rat astrocytes. Neuropharmacology 50:317–328. Hirata K, Poeaknapo C, Schmidt J, Zenk MH (2004) 1,2Dehydroreticuline synthase, the branch point enzyme opening the morphinan biosynthetic pathway. Phytochemistry 65:1039–1046. Hiroi T, Imaoka S, Chow T, Funae Y (1998a) Tissue distributions of CYP2D1, 2D2, 2D3 and 2D4 mRNA in rats detected by RT-PCR. Biochim Biophys Acta 1380:305–312. Hiroi T, Imaoka S, Funae Y (1998b) Dopamine formation from tyramine by CYP2D6. Biochem Biophys Res Commun 249:838–843. Hirrlinger J, Konig J, Dringen R (2002) Expression of mRNAs of multidrug resistance proteins (Mrps) in cultured rat astrocytes, oligodendrocytes, microglial cells and neurones. J Neurochem 82:716–719. Horak P, Haberman F, Spector S (1993) Endogenous morphine and codeine in mice – effect of muramyl dipeptide. Life Sci 52:PL255–PL260. Hughes J, Smith T, Morgan B, Fothergill L (1975a) Purification and properties of enkephalin – the possible endogenous ligand for the morphine receptor. Life Sci 16:1753–1758. Hughes J, Smith TW, Kosterlitz HW, Fothergill LA, Morgan BA, Morris HR (1975b) Identification of two related pentapeptides from the brain with potent opiate agonist activity. Nature 258:577–580. Hughes MM, Atayee RS, Best BM, Pesce AJ (2012) Observations on the metabolism of morphine to hydromorphone in pain patients. J Anal Toxicol 36:250–256. Hutchinson MR, Lewis SS, Coats BD, Rezvani N, Zhang Y, Wieseler JL, Somogyi AA, Yin H, Maier SF, Rice KC, Watkins LR (2010a) Possible involvement of toll-like receptor 4/myeloid differentiation factor-2 activity of opioid inactive isomers causes spinal proinflammation and related behavioral consequences. Neuroscience 167:880–893. Hutchinson MR, Loram LC, Zhang Y, Shridhar M, Rezvani N, Berkelhammer D, Phipps S, Foster PS, Landgraf K, Falke JJ, Rice KC, Maier SF, Yin H, Watkins LR (2010b) Evidence that tricyclic small molecules may possess toll-like receptor and myeloid differentiation protein 2 activity. Neuroscience 168:551–563. Hutchinson MR, Zhang Y, Shridhar M, Evans JH, Buchanan MM, Zhao TX, Slivka PF, Coats BD, Rezvani N, Wieseler J, Hughes TS, Landgraf KE, Chan S, Fong S, Phipps S, Falke JJ, Leinwand LA, Maier SF, Yin H, Rice KC, Watkins LR (2010c) Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behav Immun 24:83–95. Ikeda K, Kobayashi T, Kumanishi T, Niki H, Yano R (2000) Involvement of G-protein-activated inwardly rectifying K (GIRK) channels in opioid-induced analgesia. Neurosci Res 38:113–116. Ikeda H, Miyatake M, Koshikawa N, Ochiai K, Yamada K, Kiss A, Donlin MJ, Panneton WM, Churchill JD, Green M, Siddiqui AM, Leinweber AL, Crews NR, Ezerskiy LA, Rendell VR, Belcheva MM, Coscia CJ (2010) Morphine modulation of thrombospondin

114

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

levels in astrocytes and its implications for neurite outgrowth and synapse formation. J Biol Chem 285:38415–38427. Ikemoto K, Kitahama K, Jouvet A, Arai R, Nishimura A, Nishi K, Nagatsu I (1997a) Demonstration of L-dopa decarboxylating neurons specific to human striatum. Neurosci Lett 232: 111–114. Ikemoto K, Kitahama K, Seif I, Maeda T, De Maeyer E, Valatx JL (1997b) Monoamine oxidase B (MAOB)-containing structures in MAOA-deficient transgenic mice. Brain Res 771:121–132. Ilari A, Franceschini S, Bonamore A, Arenghi F, Botta B, Macone A, Pasquo A, Bellucci L, Boffi A (2009) Structural basis of enzymatic (S)-norcoclaurine biosynthesis. J Biol Chem 284:897–904. Inazu M, Takeda H, Matsumiya T (2003a) Expression and functional characterization of the extraneuronal monoamine transporter in normal human astrocytes. J Neurochem 84:43–52. Inazu M, Takeda H, Matsumiya T (2003b) Functional expression of the norepinephrine transporter in cultured rat astrocytes. J Neurochem 84:136–144. Jaeger CB (1985) Cytoarchitectonics of substantia nigra grafts: a light and electron microscopic study of immunocytochemically identified dopaminergic neurons and fibrous astrocytes. J Comp Neurol 231:121–135. Jahng JW, Houpt TA, Wessel TC, Chen K, Shih JC, Joh TH (1997) Localization of monoamine oxidase A and B mRNA in the rat brain by in situ hybridization. Synapse 25:30–36. Jedlitschky G, Cassidy AJ, Sales M, Pratt N, Burchell B (1999) Cloning and characterization of a novel human olfactory UDPglucuronosyltransferase. Biochem J 340(Pt. 3):837–843. Kardos J, Blasko G, Simonyi M, Szantay C (1984) Pharmacodynamic investigation of (+/)-salutaridine. Arzneimittelforschung 34:1758–1759. Kieffer BL, Evans CJ (2002) Opioid tolerance-in search of the holy grail. Cell 108:587–590. Kieffer BL, Evans CJ (2009) Opioid receptors: from binding sites to visible molecules in vivo. Neuropharmacology 56:205–212. Kieffer BL, Gaveriaux-Ruff C (2002) Exploring the opioid system by gene knockout. Prog Neurobiol 66:285–306. Killian AK, Schuster CR, House JT, Sholl S, Connors M, Wainer BH (1981) A non-peptide morphine-like compound from brain. Life Sci 28:811–817. Kim YM, Kim MN, Lee JJ, Lee MK (2005) Inhibition of dopamine biosynthesis by tetrahydropapaveroline. Neurosci Lett 386:1–4. King CD, Rios GR, Assouline JA, Tephly TR (1999) Expression of UDP-glucuronosyltransferases (UGTs) 2B7 and 1A6 in the human brain and identification of 5-hydroxytryptamine as a substrate. Arch Biochem Biophys 365:156–162. Kirby GW (1967) Biosynthesis of the morphine alkaloids. Science 155:170–173. Kodaira H, Lisek CA, Jardine I, Arimura A, Spector S (1989) Identification of the convulsant opiate thebaine in mammalian brain. Proc Natl Acad Sci U S A 86:716–719. Kolodziej A, Stumm R, Becker A, Hollt V (2008) Endogenous opioids inhibit ischemia-induced generation of immature hippocampal neurons via the mu-opioid receptor. Eur J Neurosci 27:1311–1319. Komatsu T, Sakurada S, Katsuyama S, Sanai K, Sakurada T (2009) Mechanism of allodynia evoked by intrathecal morphine-3glucuronide in mice. Adv Neuropharmacol 85:207–219. Kream RM, Sheehan M, Cadet P, Mantione KJ, Zhu W, Casares F, Stefano GB (2007) Persistence of evolutionary memory: primordial six-transmembrane helical domain mu opiate receptors selectively linked to endogenous morphine signaling. Med Sci Monit 13:SC5–SC6. Kumar P, Sunkaraneni S, Sirohi S, Dighe SV, Walker EA, Yoburn BC (2008) Hydromorphone efficacy and treatment protocol impact on tolerance and mu-opioid receptor regulation. Eur J Pharmacol 597:39–45. Laux A, Muller AH, Miehe M, Dirrig-Grosch S, Deloulme JC, Delalande F, Stuber D, Sage D, Van Dorsselaer A, Poisbeau P, Aunis D, Goumon Y (2011) Mapping of endogenous morphinelike compounds in the adult mouse brain: evidence of their

localization in astrocytes and GABAergic cells. J Comp Neurol 519:2390–2416. Laux A, Delalande F, Mouheiche J, Stuber D, Van Dorsselaer A, Bianchi E, Bezard E, Poisbeau P, Goumon Y (2012) Localization of endogenous morphine-like compounds in the mouse spinal cord. J Comp Neurol 520:1547–1561. Laux-Biehlmann A, Grafe N, Mouheiche J, Stuber D, Welters ID, Delalande F, Poisbeau P, Garnero P, Metz-Boutigue MH, Schneider F, Goumon Y (2012) Comparison of serum and lithium–heparinate plasma for the accurate measurements of endogenous and exogenous morphine concentrations. Br J Clin Pharmacol 74:381–383. Lazarus LH, Ling N, Guillemin R (1976) Beta-lipotropin as a prohormone for the morphinomimetic peptides endorphins and enkephalins. Proc Natl Acad Sci U S A 73:2156–2159. Lee EJ, Facchini PJ (2011) Tyrosine aminotransferase contributes to benzylisoquinoline alkaloid biosynthesis in opium poppy. Plant Physiol 157:1067–1078. Lee CS, Spector S (1991) Changes of endogenous morphine and codeine contents in the fasting rat. J Pharmacol Exp Ther 257:647–650. Lenz R, Zenk MH (1995) Purification and properties of codeinone reductase (NADPH) from Papaver somniferum cell cultures and differentiated plants. Eur J Biochem 233:132–139. Lewis SS, Hutchinson MR, Rezvani N, Loram LC, Zhang Y, Maier SF, Rice KC, Watkins LR (2010) Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via tolllike receptor 4/MD-2 and interleukin-1beta. Neuroscience 165:569–583. Lewis SS, Loram LC, Hutchinson MR, Li CM, Zhang Y, Maier SF, Huang Y, Rice KC, Watkins LR (2012) (+)Naloxone, an opioid-inactive toll-like receptor 4 signaling inhibitor, reverses multiple models of chronic neuropathic pain in rats. J Pain 13:498–506. Li CH, Chung D (1976) Isolation and structure of an untriakontapeptide with opiate activity from camel pituitary glands. Proc Natl Acad Sci U S A 73:1145–1148. Li CH, Chung D, Doneen BA (1976) Isolation, characterization and opiate activity of beta-endorphin from human pituitary glands. Biochem Biophys Res Commun 72:1542–1547. Loeffler S, Deus-neumann B, Zenk MH (1995) S-adenosyl-Lmethionine: (S)-coclaurine-N-methyl-transferase from tinospora cardifolia. Phytochemistry 38:1387–1395. Lotsch J, Geisslinger G (2001) Morphine-6-glucuronide: an analgesic of the future? Clin Pharmacokinet 40:485–499. Lotsch J, Stockmann A, Kobal G, Brune K, Waibel R, Schmidt N, Geisslinger G (1996) Pharmacokinetics of morphine and its glucuronides after intravenous infusion of morphine and morphine-6-glucuronide in healthy volunteers. Clin Pharmacol Ther 60:316–325. Lowney LI, Gentleman SB, Goldstein A (1979) A pituitary endorphin with novel properties. Life Sci 24:2377–2384. Ludwig M, Pittman QJ (2003) Talking back: dendritic neurotransmitter release. Trends Neurosci 26:255–261. Mackenzie PI, Owens IS, Burchell B, Bock KW, Bairoch A, Belanger A, Fournel-Gigleux S, Green M, Hum DW, Iyanagi T, Lancet D, Louisot P, Magdalou J, Chowdhury JR, Ritter JK, Schachter H, Tephly TR, Tipton KF, Nebert DW (1997) The UDP glycosyltransferase gene superfamily: recommended nomenclature update based on evolutionary divergence. Pharmacogenetics 7:255–269. Mackenzie PI, Bock KW, Burchell B, Guillemette C, Ikushiro S, Iyanagi T, Miners JO, Owens IS, Nebert DW (2005) Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics 15:677–685. Madbouly KM, Senagore AJ, Delaney CP (2010) Endogenous morphine levels after laparoscopic versus open colectomy. Br J Surg 97:759–764. Mague SD, Blendy JA (2010) OPRM1 SNP (A118G): involvement in disease development, treatment response, and animal models. Drug Alcohol Depend 108:172–182.

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117 Mantione KJ, Cadet P, Zhu W, Kream RM, Sheehan M, Fricchione GL, Goumon Y, Esch T, Stefano GB (2008) Endogenous morphine signaling via nitric oxide regulates the expression of CYP2D6 and COMT: autocrine/paracrine feedback inhibition. Addict Biol 13:118–123. Marchitti SA, Deitrich RA, Vasiliou V (2007) Neurotoxicity and metabolism of the catecholamine-derived 3,4dihydroxyphenylacetaldehyde and 3,4dihydroxyphenylglycolaldehyde: the role of aldehyde dehydrogenase. Pharmacol Rev 59:125–150. Mavrojannis M (1903) Action cataleptique de la morphine chez les Rats. Contribution a la theorie toxique da la catalepsie. C R Soc Biol 55:1092–1094. McCoy JG, Strawbridge C, McMurtrey KD, Kane VB, Ward CP (2003) A re-evaluation of the role of tetrahydropapaveroline in ethanol consumption in rats. Brain Res Bull 60:59–65. McDonough PC, Levine B, Vorce S, Jufer RA, Fowler D (2008) The detection of hydromorphone in urine specimens with high morphine concentrations. J Forensic Sci 53:752–754. McNicholas LF, Martin WR (1984) New and experimental therapeutic roles for naloxone and related opioid antagonists. Drugs 27:81–93. Medeiros MA, Nunes XP, Barbosa-Filho JM, Lemos VS, Pinho JF, Roman-Campos D, de Medeiros IA, Araujo DA, Cruz JS (2009) (S)-reticuline induces vasorelaxation through the blockade of Ltype Ca(2+) channels. Naunyn Schmiedebergs Arch Pharmacol 379:115–125. Meijerink WJ, Molina PE, Abumrad NN (1999) Mammalian opiate alkaloid synthesis: lessons derived from plant biochemistry. Shock 12:165–173. Meissner WG, Frasier M, Gasser T, Goetz CG, Lozano A, Piccini P, Obeso JA, Rascol O, Schapira A, Voon V, Weiner DM, Tison F, Bezard E (2011) Priorities in Parkinson’s disease research. Nat Rev Drug Discov 10:377–393. Mellon RD, Bayer BM (1998a) Evidence for central opioid receptors in the immunomodulatory effects of morphine: review of potential mechanism(s) of action. J Neuroimmunol 83:19–28. Mellon RD, Bayer BM (1998b) Role of central opioid receptor subtypes in morphine-induced alterations in peripheral lymphocyte activity. Brain Res 789:56–67. Mellon RD, Bayer BM (1999) The effects of morphine, nicotine and epibatidine on lymphocyte activity and hypothalamic–pituitary– adrenal axis responses. J Pharmacol Exp Ther 288:635–642. Meunier JC, Mollereau C, Toll L, Suaudeau C, Moisand C, Alvinerie P, Butour JL, Guillemot JC, Ferrara P, Monsarrat B, et al (1995) Isolation and structure of the endogenous agonist of opioid receptor-like ORL1 receptor. Nature 377:532–535. Miksys S, Rao Y, Hoffmann E, Mash DC, Tyndale RF (2002) Regional and cellular expression of CYP2D6 in human brain: higher levels in alcoholics. J Neurochem 82:1376–1387. Misra AL, Pontani RB, Mule SJ (1974a) Pharmacokinetics and metabolism of (3H) thebaine. Xenobiotica 4:17–32. Misra AL, Vadlamani NL, Pontani RB, Mule SJ (1974b) Some physicochemical and pharmacological properties of morphine2,3-quinone, the morphine metabolite in the rat brain. J Pharm Pharmacol 26:990–992. Miyake N, Thompson J, Skinbjerg M, Abi-Dargham A (2011) Presynaptic dopamine in schizophrenia. CNS Neurosci Ther 17:104–109. Molina PE, Hashiguchi Y, Meijerink WJ, Naukam RJ, Boxer R, Abumrad NN (1995a) Modulation of endogenous opiate production: effect of fasting. Biochem Biophys Res Commun 207:312–317. Molina R, Sanchez M, Hidalgo A, Garcia de Boto MJ (1995b) Effects of preanesthetic and anesthetic drugs on endothelium-dependent responses in the rat aorta. Gen Pharmacol 26:169–175. Morais LC, Barbosa-Filho JM, Almeida RN (1998) Central depressant effects of reticuline extracted from Ocotea duckei in rats and mice. J Ethnopharmacol 62:57–61. Morishige T, Tsujita T, Yamada Y, Sato F (2000) Molecular characterization of the S-adenosyl-L-methionine:30 -hydroxy-N-

115

methylcoclaurine 40 -O-methyltransferase involved in isoquinoline alkaloid biosynthesis in Coptis japonica. J Biol Chem 275:23398–23405. Muller A, Glattard E, Taleb O, Kemmel V, Laux A, Miehe M, Delalande F, Roussel G, Van Dorsselaer A, Metz-Boutigue MH, Aunis D, Goumon Y (2008) Endogenous morphine in SH-SY5Y cells and the mouse cerebellum. PLoS One 3:e1641. Munjal ID, Minna JD, Manneckjee R, Bieck P, Spector S (1995) Possible role of endogenous morphine and codeine on growth regulation of lung tissue. Life Sci 57:517–521. Murray CJL, Lopez AD (1996) The global burden of disease: a comprehensive assessment of mortality and disability from diseases, injuries, and risk factors in 1990 and projected to 2020. Cambridge, MA: Harvard University Press. Nagano E, Yamada H, Oguri K (2000) Characteristic glucuronidation pattern of physiologic concentration of morphine in rat brain. Life Sci 67:2453–2464. Neri C, Guarna M, Bianchi E, Sonetti D, Matteucci G, Stefano GB (2004) Endogenous morphine and codeine in the brain of non human primate. Med Sci Monit 10:MS1–MS5. Neri C, Ghelardini C, Sotak B, Palmiter RD, Guarna M, Stefano G, Bianchi E (2008) Dopamine is necessary to endogenous morphine formation in mammalian brain in vivo. J Neurochem 106:2337–2344. Nikolaev VO, Boettcher C, Dees C, Bunemann M, Lohse MJ, Zenk MH (2007) Live cell monitoring of mu-opioid receptor-mediated G-protein activation reveals strong biological activity of close morphine biosynthetic precursors. J Biol Chem 282:27126–27132. Norris PJ, Hardwick JP, Emson PC (1996) Regional distribution of cytochrome P450 2D1 in the rat central nervous system. J Comp Neurol 366:244–258. Oka K, Kantrowitz JD, Spector S (1985) Isolation of morphine from toad skin. Proc Natl Acad Sci U S A 82:1852–1854. Okada T, Shimada S, Sato K, Kotake Y, Kawai H, Ohta S, Tohyama M, Nishimura T (1998) Tetrahydropapaveroline and its derivatives inhibit dopamine uptake through dopamine transporter expressed in HEK293 cells. Neurosci Res 30:87–90. Osborne R, Joel S, Trew D, Slevin M (1990) Morphine and metabolite behavior after different routes of morphine administration: demonstration of the importance of the active metabolite morphine-6-glucuronide. Clin Pharmacol Ther 47:12–19. Oyler JM, Cone EJ, Joseph Jr RE, Huestis MA (2000) Identification of hydrocodone in human urine following controlled codeine administration. J Anal Toxicol 24:530–535. Perea-Sasiaı´ n J (2008) Biosynthesis of morphine: its importance in Parkinson’s disease. Rev Fac Med Unal 56:161–189. Persson AI, Thorlin T, Bull C, Eriksson PS (2003a) Opioid-induced proliferation through the MAPK pathway in cultures of adult hippocampal progenitors. Mol Cell Neurosci 23:360–372. Persson AI, Thorlin T, Bull C, Zarnegar P, Ekman R, Terenius L, Eriksson PS (2003b) Mu- and delta-opioid receptor antagonists decrease proliferation and increase neurogenesis in cultures of rat adult hippocampal progenitors. Eur J Neurosci 17:1159–1172. Pert CB, Snyder SH (1973) Opiate receptor: demonstration in nervous tissue. Science 179:1011–1014. Pert CB, Pasternak G, Snyder SH (1973) Opiate agonists and antagonists discriminated by receptor binding in brain. Science 182:1359–1361. Pert CB, Pert A, Tallman JF (1976) Isolation of a novel endogenous opiate analgesic from human blood. Proc Natl Acad Sci U S A 73:2226–2230. Poeaknapo C, Schmidt J, Brandsch M, Drager B, Zenk MH (2004) Endogenous formation of morphine in human cells. Proc Natl Acad Sci U S A 101:14091–14096. Pryor SC, Elizee R (2000) Evidence of opiates and opioid neuropeptides and their immune effects in parasitic invertebrates representing three different phyla: Schistosoma mansoni, Theromyzon tessulatum, Trichinella spiralis. Acta Biol Hung 51:331–341.

116

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117

Rein G, Glover V, Sandler M (1982) Multiple forms of phenolsulphotransferase in human tissues: selective inhibition by dichloronitrophenol. Biochem Pharmacol 31:1893–1897. Reinscheid RK, Nothacker HP, Bourson A, Ardati A, Henningsen RA, Bunzow JR, Grandy DK, Langen H, Monsma Jr FJ, Civelli O (1995) Orphanin FQ: a neuropeptide that activates an opioidlike G protein-coupled receptor. Science 270:792–794. Robinson TE, Kolb B (2004) Structural plasticity associated with exposure to drugs of abuse. Neuropharmacology 47(Suppl. 1):33–46. Sagud M, Muck-Seler D, Mihaljevic-Peles A, Vuksan-Cusa B, Zivkovic M, Jakovljevic M, Pivac N (2010) Catechol-O-methyl transferase and schizophrenia. Psychiatr Danub 22:270–274. Samanani N, Facchini PJ (2002) Purification and characterization of norcoclaurine synthase. The first committed enzyme in benzylisoquinoline alkaloid biosynthesis in plants. J Biol Chem 277:33878–33883. Sango K, Maruyama W, Matsubara K, Dostert P, Minami C, Kawai M, Naoi M (2000) Enantio-selective occurrence of (S)tetrahydropapaveroline in human brain. Neurosci Lett 283:224–226. Sargeant TJ, Miller JH, Day DJ (2008) Opioidergic regulation of astroglial/neuronal proliferation: where are we now? J Neurochem 107:883–897. Schestatsky P, Kumru H, Valls-Sole J, Valldeoriola F, Marti MJ, Tolosa E, Chaves ML (2007) Neurophysiologic study of central pain in patients with Parkinson disease. Neurology 69:2162–2169. Schulz R, Wuster M, Herz A (1977) Detection of a long acting endogenous opioid in blood and small intestine. Life Sci 21:105–116. Seelbach MJ, Brooks TA, Egleton RD, Davis TP (2007) Peripheral inflammatory hyperalgesia modulates morphine delivery to the brain: a role for P-glycoprotein. J Neurochem 102:1677–1690. Sery O, Prikryl R, Castulik L, St’astny F (2010) A118G polymorphism of OPRM1 gene is associated with schizophrenia. J Mol Neurosci 41:219–222. Shorr J, Foley K, Spector S (1978) Presence of a non-peptide morphine-like compound in human cerebrospinal fluid. Life Sci 23:2057–2062. Siegle I, Fritz P, Eckhardt K, Zanger UM, Eichelbaum M (2001) Cellular localization and regional distribution of CYP2D6 mRNA and protein expression in human brain. Pharmacogenetics 11:237–245. Silber D, McLaughlin D, Sinoway L (1991) Leg exercise conditioning increases peak forearm blood flow. J Appl Physiol 71: 1568–1573. Simantov R, Snyder SH (1976) Morphine-like peptides, leucine enkephalin and methionine enkephalin: interactions with the opiate receptor. Mol Pharmacol 12:987–998. Smith MT, Watt JA, Cramond T (1990) Morphine-3-glucuronide – a potent antagonist of morphine analgesia. Life Sci 47:579–585. Somogyi P, Klausberger T (2005) Defined types of cortical interneurone structure space and spike timing in the hippocampus. J Physiol 562:9–26. Somogyi AA, Barratt DT, Coller JK (2007) Pharmacogenetics of opioids. Clin Pharmacol Ther 81:429–444. Sonetti D, Peruzzi E, Stefano GB (2005) Endogenous morphine and ACTH association in neural tissues. Med Sci Monit 11:MS22–MS30. Stefano GB, Digenis A, Spector S, Leung MK, Bilfinger TV, Makman MH, Scharrer B, Abumrad NN (1993) Opiate-like substances in an invertebrate, an opiate receptor on invertebrate and human immunocytes, and a role in immunosuppression. Proc Natl Acad Sci U S A 90:11099–11103. Stefano GB, Goumon Y, Casares F, Cadet P, Fricchione GL, Rialas C, Peter D, Sonetti D, Guarna M, Welters ID, Bianchi E (2000) Endogenous morphine. Trends Neurosci 23:436–442. Stefano GB, Bianchi E, Guarna M, Fricchione GL, Zhu W, Cadet P, Mantione KJ, Casares FM, Kream RM, Esch T (2007) Nicotine, alcohol and cocaine coupling to reward processes via

endogenous morphine signaling: the dopamine–morphine hypothesis. Med Sci Monit 13:RA91–RA102. Stevens CW, Aravind S, Das S, Davis RL (2012) Pharmacological characterization of LPS and opioid interactions at the toll-like receptor 4. Br J Pharmacol. in press. http://dx.doi.org/10.1111/ bph.12028. Stone AN, Mackenzie PI, Galetin A, Houston JB, Miners JO (2003) Isoform selectivity and kinetics of morphine 3- and 6glucuronidation by human udp-glucuronosyltransferases: evidence for atypical glucuronidation kinetics by UGT2B7. Drug Metab Dispos 31:1086–1089. Suleman FG, Abid A, Gradinaru D, Daval JL, Magdalou J, Minn A (1998) Identification of the uridine diphosphate glucuronosyltransferase isoform UGT1A6 in rat brain and in primary cultures of neurons and astrocytes. Arch Biochem Biophys 358:63–67. Sullivan AF, McQuay HJ, Bailey D, Dickenson AH (1989) The spinal antinociceptive actions of morphine metabolites morphine-6glucuronide and normorphine in the rat. Brain Res 482:219–224. Surh YJ, Kim HJ (2010) Neurotoxic effects of tetrahydroisoquinolines and underlying mechanisms. Exp Neurobiol 19:63–70. Svingos AL, Colago EE, Pickel VM (2001a) Vesicular acetylcholine transporter in the rat nucleus accumbens shell: subcellular distribution and association with mu-opioid receptors. Synapse 40:184–192. Svingos AL, Garzon M, Colago EE, Pickel VM (2001b) Mu-opioid receptors in the ventral tegmental area are targeted to presynaptically and directly modulate mesocortical projection neurons. Synapse 41:221–229. Takeda H, Inazu M, Matsumiya T (2002) Astroglial dopamine transport is mediated by norepinephrine transporter. Naunyn Schmiedebergs Arch Pharmacol 366:620–623. Tenconi B, Lesma E, DiGiulio AM, Gorio A (1996) High opioid doses inhibit whereas low doses enhance neuritogenesis in PC12 cells. Brain Res Dev Brain Res 94:175–181. Thompson MA, Moon E, Kim UJ, Xu J, Siciliano MJ, Weinshilboum RM (1999) Human indolethylamine N-methyltransferase: cDNA cloning and expression, gene cloning, and chromosomal localization. Genomics 61:285–297. Trescot AM, Datta S, Lee M, Hansen H (2008) Opioid pharmacology. Pain Physician 11:S133–153. van de Wetering K, Zelcer N, Kuil A, Feddema W, Hillebrand M, Vlaming ML, Schinkel AH, Beijnen JH, Borst P (2007) Multidrug resistance proteins 2 and 3 provide alternative routes for hepatic excretion of morphine–glucuronides. Mol Pharmacol 72:387–394. Volpe DA, McMahon Tobin GA, Mellon RD, Katki AG, Parker RJ, Colatsky T, Kropp TJ, Verbois SL (2011) Uniform assessment and ranking of opioid mu receptor binding constants for selected opioid drugs. Regul Toxicol Pharmacol 59:385–390. von Bohlen Und Halbach O (2007) Immunohistological markers for staging neurogenesis in adult hippocampus. Cell Tissue Res 329:409–420. Wachtel SR, Abercrombie ED (1994) L-3,4-dihydroxyphenylalanineinduced dopamine release in the striatum of intact and 6hydroxydopamine-treated rats: differential effects of monoamine oxidase A and B inhibitors. J Neurochem 63:108–117. Wasan AD, Michna E, Janfaza D, Greenfield S, Teter CJ, Jamison RN (2008) Interpreting urine drug tests: prevalence of morphine metabolism to hydromorphone in chronic pain patients treated with morphine. Pain Med 9:918–923. Wassenberg T, Willemsen MA, Geurtz PB, Lammens M, Verrijp K, Wilmer M, Lee WT, Wevers RA, Verbeek MM (2010) Urinary dopamine in aromatic L-amino acid decarboxylase deficiency: the unsolved paradox. Mol Genet Metab 101:349–356. Weinstein DD, Spector S, Nanney LB, King LE (2002) Endogenous opiate alkaloids and human skin. J Invest Dermatol 119:709–710. Weitz CJ, Lowney LI, Faull KF, Feistner G, Goldstein A (1986) Morphine and codeine from mammalian brain. Proc Natl Acad Sci U S A 83:9784–9788. Weitz CJ, Faull KF, Goldstein A (1987) Synthesis of the skeleton of the morphine molecule by mammalian liver. Nature 330:674–677.

A. Laux-Biehlmann et al. / Neuroscience 233 (2013) 95–117 Weitz CJ, Lowney LI, Faull KF, Feistner G, Goldstein A (1988) 6Acetylmorphine: a natural product present in mammalian brain. Proc Natl Acad Sci U S A 85:5335–5338. Welch EB, Thompson DF (1994) Opiate antagonists for the treatment of schizophrenia. J Clin Pharm Ther 19:279–283. Whitman SC, Hazen SL, Miller DB, Hegele RA, Heinecke JW, Huff MW (1999) Modification of type III VLDL, their remnants, and VLDL from ApoE-knockout mice by phydroxyphenylacetaldehyde, a product of myeloperoxidase activity, causes marked cholesteryl ester accumulation in macrophages. Arterioscler Thromb Vasc Biol 19:1238–1249. Wonodi I, Adami H, Sherr J, Avila M, Hong LE, Thaker GK (2004) Naltrexone treatment of tardive dyskinesia in patients with schizophrenia. J Clin Psychopharmacol 24:441–445. Wuster M, Loth P, Schulz R (1978) Characterization of opiate-like materials in blood and urine. Adv Biochem Psychopharmacol 18:313–319. Yamada H, Ishii K, Ishii Y, Ieiri I, Nishio S, Morioka T, Oguri K (2003) Formation of highly analgesic morphine-6-glucuronide following physiologic concentration of morphine in human brain. J Toxicol Sci 28:395–401. Yao L, Fan P, Arolfo M, Jiang Z, Olive MF, Zablocki J, Sun HL, Chu N, Lee J, Kim HY, Leung K, Shryock J, Blackburn B, Diamond I (2010) Inhibition of aldehyde dehydrogenase-2 suppresses cocaine seeking by generating THP, a cocaine use-dependent inhibitor of dopamine synthesis. Nat Med 16:1024–1028. Yeh SY (1974) Absence of evidence of biotransformation of morphine to codeine in man. Experientia 30:264–266. Yeh SY (1975) Urinary excretion of morphine and its metabolites in morphine-dependent subjects. J Pharmacol Exp Ther 192:201–210. Yeh SY (1981) Analgesic activity and toxicity of oripavine and phidihydrothebaine in the mouse and rat. Arch Int Pharmacodyn Ther 254:223–240. Yeh SY, Gorodetzky CW, Krebs HA (1977a) Isolation and identification of morphine 3- and 6-glucuronides, morphine 3,6diglucuronide, morphine 3-ethereal sulfate, normorphine, and normorphine 6-glucuronide as morphine metabolites in humans. J Pharm Sci 66:1288–1293. Yeh SY, McQuinn RL, Gorodetzky CW (1977b) Biotransformation of morphine to dihydromorphinone and normorphine in the mouse, rat, rabbit, guinea pig, cat, dog, and monkey. Drug Metab Dispos 5:335–342. Yeh SY, McQuinn RL, Gorodetzky CW (1977c) Identification of diacetylmorphine metabolites in humans. J Pharm Sci 66:201–204. Yeh SY, Krebs HA, Gorodetzky CW (1979) Isolation and identification of morphine n-oxide alpha- and beta-dihydromorphines, beta- or gamma-isomorphine, and hydroxylated morphine as morphine metabolites in several mammalian species. J Pharm Sci 68:133–140. Yoshida S, Ohta J, Yamasaki K, Kamei H, Harada Y, Yahara T, Kaibara A, Ozaki K, Tajiri T, Shirouzu K (2000) Effect of surgical stress on endogenous morphine and cytokine levels in the plasma

117

after laparoscopoic or open cholecystectomy. Surg Endosc 14:137–140. Zadina JE, Hackler L, Ge LJ, Kastin AJ (1997) A potent and selective endogenous agonist for the mu-opiate receptor. Nature 386:499–502. Zeng YS, Nie JH, Zhang W, Chen SJ, Wu W (2007) Morphine acts via mu-opioid receptors to enhance spinal regeneration and synaptic reconstruction of primary afferent fibers injured by sciatic nerve crush. Brain Res 1130:108–113. Zhang Z, Pan ZZ (2010) Synaptic mechanism for functional synergism between delta- and mu-opioid receptors. J Neurosci 30:4735–4745. Zhang Y, Wang Z, Cox DP, Civelli O (2012) Study on the activation of the opioid receptors by a set of morphine derivatives in a welldefined assay system. Neurochem Res 37:410–416. Zhu W (2008) CYP2D6: a key enzyme in morphine synthesis in animals. Med Sci Monit 14:SC15–SC18. Zhu W, Baggerman G, Goumon Y, Casares F, Brownawell B, Stefano GB (2001a) Presence of morphine and morphine-6-glucuronide in the marine mollusk Mytilus edulis ganglia determined by GC/MS and Q-TOF-MS. Starvation increases opiate alkaloid levels. Brain Res Mol Brain Res 88:155–160. Zhu W, Bilfinger TV, Baggerman G, Goumon Y, Stefano GB (2001b) Presence of endogenous morphine and morphine 6 glucuronide in human heart tissue. Int J Mol Med 7:419–422. Zhu W, Baggerman G, Secor WE, Casares F, Pryor SC, Fricchione GL, Ruiz-Tiben E, Eberhard ML, Bimi L, Stefano GB (2002) Dracunculus medinensis and Schistosoma mansoni contain opiate alkaloids. Ann Trop Med Parasitol 96:309–316. Zhu W, Cadet P, Baggerman G, Mantione KJ, Stefano GB (2005) Human white blood cells synthesize morphine: CYP2D6 modulation. J Immunol 175:7357–7362. Zhu W, Mantione KJ, Casares FM, Cadet P, Kim JW, Bilfinger TV, Kream RM, Khalill S, Singh S, Stefano GB (2006a) Alcohol-, nicotine-, and cocaine-evoked release of morphine from invertebrate ganglia: model system for screening drugs of abuse. Med Sci Monit 12:BR155–BR161. Zhu W, Mantione KJ, Casares FM, Sheehan MH, Kream RM, Stefano GB (2006b) Cholinergic regulation of endogenous morphine release from lobster nerve cord. Med Sci Monit 12:BR295–BR301. Ziegler J, Diaz-Chavez ML, Kramell R, Ammer C, Kutchan TM (2005) Comparative macroarray analysis of morphine containing Papaver somniferum and eight morphine free Papaver species identifies an O-methyltransferase involved in benzylisoquinoline biosynthesis. Planta 222:458–471. Ziegler J, Facchini PJ, Geissler R, Schmidt J, Ammer C, Kramell R, Voigtlander S, Gesell A, Pienkny S, Brandt W (2009) Evolution of morphine biosynthesis in opium poppy. Phytochemistry 70:1696–1707. Zuckerman A, Bolan E, de Paulis T, Schmidt D, Spector S, Pasternak GW (1999) Pharmacological characterization of morphine-6sulfate and codeine-6-sulfate. Brain Res 842:1–5.

(Accepted 7 December 2012) (Available online 21 December 2012)