Mu-type opioid receptor

Details

Name
Mu-type opioid receptor
Synonyms
  • hMOP
  • M-OR-1
  • MOP
  • MOR1
  • Mu opiate receptor
  • Mu opioid receptor
Gene Name
OPRM1
Organism
Humans
Amino acid sequence
>lcl|BSEQ0001536|Mu-type opioid receptor
MDSSAAPTNASNCTDALAYSSCSPAPSPGSWVNLSHLDGNLSDPCGPNRTDLGGRDSLCP
PTGSPSMITAITIMALYSIVCVVGLFGNFLVMYVIVRYTKMKTATNIYIFNLALADALAT
STLPFQSVNYLMGTWPFGTILCKIVISIDYYNMFTSIFTLCTMSVDRYIAVCHPVKALDF
RTPRNAKIINVCNWILSSAIGLPVMFMATTKYRQGSIDCTLTFSHPTWYWENLLKICVFI
FAFIMPVLIITVCYGLMILRLKSVRMLSGSKEKDRNLRRITRMVLVVVAVFIVCWTPIHI
YVIIKALVTIPETTFQTVSWHFCIALGYTNSCLNPVLYAFLDENFKRCFREFCIPTSSNI
EQQNSTRIRQNTRDHPSTANTVDRTNHQLENLEAETAPLP
Number of residues
400
Molecular Weight
44778.855
Theoretical pI
8.29
GO Classification
Functions
beta-endorphin receptor activity / G-protein alpha-subunit binding / G-protein beta-subunit binding / G-protein coupled receptor activity / morphine receptor activity / neuropeptide binding / voltage-gated calcium channel activity
Processes
adenylate cyclase-activating dopamine receptor signaling pathway / adenylate cyclase-inhibiting G-protein coupled receptor signaling pathway / behavioral response to ethanol / calcium ion transmembrane transport / cellular response to morphine / cellular response to stress / G-protein coupled receptor signaling pathway, coupled to cyclic nucleotide second messenger / locomotory behavior / negative regulation of adenylate cyclase activity / negative regulation of cAMP-mediated signaling / negative regulation of cell proliferation / negative regulation of cytosolic calcium ion concentration / negative regulation of nitric oxide biosynthetic process / negative regulation of Wnt protein secretion / neuropeptide signaling pathway / opioid receptor signaling pathway / phospholipase C-activating G-protein coupled receptor signaling pathway / positive regulation of cAMP-mediated signaling / positive regulation of cytosolic calcium ion concentration / positive regulation of ERK1 and ERK2 cascade / positive regulation of neurogenesis / positive regulation of nitric oxide biosynthetic process / regulation of N-methyl-D-aspartate selective glutamate receptor activity / sensory perception / sensory perception of pain / synaptic transmission
Components
cytosol / endoplasmic reticulum / Golgi apparatus / integral component of plasma membrane / neuron projection / plasma membrane
General Function
Voltage-gated calcium channel activity
Specific Function
Receptor for endogenous opioids such as beta-endorphin and endomorphin. Receptor for natural and synthetic opioids including morphine, heroin, DAMGO, fentanyl, etorphine, buprenorphin and methadone. Agonist binding to the receptor induces coupling to an inactive GDP-bound heterotrimeric G-protein complex and subsequent exchange of GDP for GTP in the G-protein alpha subunit leading to dissociation of the G-protein complex with the free GTP-bound G-protein alpha and the G-protein beta-gamma dimer activating downstream cellular effectors. The agonist- and cell type-specific activity is predominantly coupled to pertussis toxin-sensitive G(i) and G(o) G alpha proteins, GNAI1, GNAI2, GNAI3 and GNAO1 isoforms Alpha-1 and Alpha-2, and to a lesser extend to pertussis toxin-insensitive G alpha proteins GNAZ and GNA15. They mediate an array of downstream cellular responses, including inhibition of adenylate cyclase activity and both N-type and L-type calcium channels, activation of inward rectifying potassium channels, mitogen-activated protein kinase (MAPK), phospholipase C (PLC), phosphoinositide/protein kinase (PKC), phosphoinositide 3-kinase (PI3K) and regulation of NF-kappa-B. Also couples to adenylate cyclase stimulatory G alpha proteins. The selective temporal coupling to G-proteins and subsequent signaling can be regulated by RGSZ proteins, such as RGS9, RGS17 and RGS4. Phosphorylation by members of the GPRK subfamily of Ser/Thr protein kinases and association with beta-arrestins is involved in short-term receptor desensitization. Beta-arrestins associate with the GPRK-phosphorylated receptor and uncouple it from the G-protein thus terminating signal transduction. The phosphorylated receptor is internalized through endocytosis via clathrin-coated pits which involves beta-arrestins. The activation of the ERK pathway occurs either in a G-protein-dependent or a beta-arrestin-dependent manner and is regulated by agonist-specific receptor phosphorylation. Acts as a class A G-protein coupled receptor (GPCR) which dissociates from beta-arrestin at or near the plasma membrane and undergoes rapid recycling. Receptor down-regulation pathways are varying with the agonist and occur dependent or independent of G-protein coupling. Endogenous ligands induce rapid desensitization, endocytosis and recycling whereas morphine induces only low desensitization and endocytosis. Heterooligomerization with other GPCRs can modulate agonist binding, signaling and trafficking properties. Involved in neurogenesis. Isoform 12 couples to GNAS and is proposed to be involved in excitatory effects. Isoform 16 and isoform 17 do not bind agonists but may act through oligomerization with binding-competent OPRM1 isoforms and reduce their ligand binding activity.
Pfam Domain Function
Transmembrane Regions
69-93 107-131 143-165 186-207 231-255 284-307 315-338
Cellular Location
Cell membrane
Gene sequence
>lcl|BSEQ0021649|Mu-type opioid receptor (OPRM1)
ATGGACAGCAGCGCTGCCCCCACGAACGCCAGCAATTGCACTGATGCCTTGGCGTACTCA
AGTTGCTCCCCAGCACCCAGCCCCGGTTCCTGGGTCAACTTGTCCCACTTAGATGGCAAC
CTGTCCGACCCATGCGGTCCGAACCGCACCGACCTGGGCGGGAGAGACAGCCTGTGCCCT
CCGACCGGCAGTCCCTCCATGATCACGGCCATCACGATCATGGCCCTCTACTCCATCGTG
TGCGTGGTGGGGCTCTTCGGAAACTTCCTGGTCATGTATGTGATTGTCAGATACACCAAG
ATGAAGACTGCCACCAACATCTACATTTTCAACCTTGCTCTGGCAGATGCCTTAGCCACC
AGTACCCTGCCCTTCCAGAGTGTGAATTACCTAATGGGAACATGGCCATTTGGAACCATC
CTTTGCAAGATAGTGATCTCCATAGATTACTATAACATGTTCACCAGCATATTCACCCTC
TGCACCATGAGTGTTGATCGATACATTGCAGTCTGCCACCCTGTCAAGGCCTTAGATTTC
CGTACTCCCCGAAATGCCAAAATTATCAATGTCTGCAACTGGATCCTCTCTTCAGCCATT
GGTCTTCCTGTAATGTTCATGGCTACAACAAAATACAGGCAAGGTTCCATAGATTGTACA
CTAACATTCTCTCATCCAACCTGGTACTGGGAAAACCTGCTGAAGATCTGTGTTTTCATC
TTCGCCTTCATTATGCCAGTGCTCATCATTACCGTGTGCTATGGACTGATGATCTTGCGC
CTCAAGAGTGTCCGCATGCTCTCTGGCTCCAAAGAAAAGGACAGGAATCTTCGAAGGATC
ACCAGGATGGTGCTGGTGGTGGTGGCTGTGTTCATCGTCTGCTGGACTCCCATTCACATT
TACGTCATCATTAAAGCCTTGGTTACAATCCCAGAAACTACGTTCCAGACTGTTTCTTGG
CACTTCTGCATTGCTCTAGGTTACACAAACAGCTGCCTCAACCCAGTCCTTTATGCATTT
CTGGATGAAAACTTCAAACGATGCTTCAGAGAGTTCTGTATCCCAACCTCTTCCAACATT
GAGCAACAAAACTCCACTCGAATTCGTCAGAACACTAGAGACCACCCCTCCACGGCCAAT
ACAGTGGATAGAACTAATCATCAGCTAGAAAATCTGGAAGCAGAAACTGCTCCGTTGCCC
TAA
Chromosome Location
6
Locus
6q24-q25
External Identifiers
ResourceLink
UniProtKB IDP35372
UniProtKB Entry NameOPRM_HUMAN
GenBank Protein ID452073
GenBank Gene IDL25119
GenAtlas IDOPRM1
HGNC IDHGNC:8156
General References
  1. Wang JB, Johnson PS, Persico AM, Hawkins AL, Griffin CA, Uhl GR: Human mu opiate receptor. cDNA and genomic clones, pharmacologic characterization and chromosomal assignment. FEBS Lett. 1994 Jan 31;338(2):217-22. [Article]
  2. Bare LA, Mansson E, Yang D: Expression of two variants of the human mu opioid receptor mRNA in SK-N-SH cells and human brain. FEBS Lett. 1994 Nov 7;354(2):213-6. [Article]
  3. Mestek A, Hurley JH, Bye LS, Campbell AD, Chen Y, Tian M, Liu J, Schulman H, Yu L: The human mu opioid receptor: modulation of functional desensitization by calcium/calmodulin-dependent protein kinase and protein kinase C. J Neurosci. 1995 Mar;15(3 Pt 2):2396-406. [Article]
  4. Pan YX, Xu J, Mahurter L, Xu M, Gilbert AK, Pasternak GW: Identification and characterization of two new human mu opioid receptor splice variants, hMOR-1O and hMOR-1X. Biochem Biophys Res Commun. 2003 Feb 21;301(4):1057-61. [Article]
  5. Cadet P, Mantione KJ, Stefano GB: Molecular identification and functional expression of mu 3, a novel alternatively spliced variant of the human mu opiate receptor gene. J Immunol. 2003 May 15;170(10):5118-23. [Article]
  6. Pan L, Xu J, Yu R, Xu MM, Pan YX, Pasternak GW: Identification and characterization of six new alternatively spliced variants of the human mu opioid receptor gene, Oprm. Neuroscience. 2005;133(1):209-20. [Article]
  7. Xu J, Xu M, Hurd YL, Pasternak GW, Pan YX: Isolation and characterization of new exon 11-associated N-terminal splice variants of the human mu opioid receptor gene. J Neurochem. 2009 Feb;108(4):962-72. doi: 10.1111/j.1471-4159.2008.05833.x. Epub 2008 Dec 10. [Article]
  8. Shabalina SA, Zaykin DV, Gris P, Ogurtsov AY, Gauthier J, Shibata K, Tchivileva IE, Belfer I, Mishra B, Kiselycznyk C, Wallace MR, Staud R, Spiridonov NA, Max MB, Goldman D, Fillingim RB, Maixner W, Diatchenko L: Expansion of the human mu-opioid receptor gene architecture: novel functional variants. Hum Mol Genet. 2009 Mar 15;18(6):1037-51. doi: 10.1093/hmg/ddn439. Epub 2008 Dec 22. [Article]
  9. Ota T, Suzuki Y, Nishikawa T, Otsuki T, Sugiyama T, Irie R, Wakamatsu A, Hayashi K, Sato H, Nagai K, Kimura K, Makita H, Sekine M, Obayashi M, Nishi T, Shibahara T, Tanaka T, Ishii S, Yamamoto J, Saito K, Kawai Y, Isono Y, Nakamura Y, Nagahari K, Murakami K, Yasuda T, Iwayanagi T, Wagatsuma M, Shiratori A, Sudo H, Hosoiri T, Kaku Y, Kodaira H, Kondo H, Sugawara M, Takahashi M, Kanda K, Yokoi T, Furuya T, Kikkawa E, Omura Y, Abe K, Kamihara K, Katsuta N, Sato K, Tanikawa M, Yamazaki M, Ninomiya K, Ishibashi T, Yamashita H, Murakawa K, Fujimori K, Tanai H, Kimata M, Watanabe M, Hiraoka S, Chiba Y, Ishida S, Ono Y, Takiguchi S, Watanabe S, Yosida M, Hotuta T, Kusano J, Kanehori K, Takahashi-Fujii A, Hara H, Tanase TO, Nomura Y, Togiya S, Komai F, Hara R, Takeuchi K, Arita M, Imose N, Musashino K, Yuuki H, Oshima A, Sasaki N, Aotsuka S, Yoshikawa Y, Matsunawa H, Ichihara T, Shiohata N, Sano S, Moriya S, Momiyama H, Satoh N, Takami S, Terashima Y, Suzuki O, Nakagawa S, Senoh A, Mizoguchi H, Goto Y, Shimizu F, Wakebe H, Hishigaki H, Watanabe T, Sugiyama A, Takemoto M, Kawakami B, Yamazaki M, Watanabe K, Kumagai A, Itakura S, Fukuzumi Y, Fujimori Y, Komiyama M, Tashiro H, Tanigami A, Fujiwara T, Ono T, Yamada K, Fujii Y, Ozaki K, Hirao M, Ohmori Y, Kawabata A, Hikiji T, Kobatake N, Inagaki H, Ikema Y, Okamoto S, Okitani R, Kawakami T, Noguchi S, Itoh T, Shigeta K, Senba T, Matsumura K, Nakajima Y, Mizuno T, Morinaga M, Sasaki M, Togashi T, Oyama M, Hata H, Watanabe M, Komatsu T, Mizushima-Sugano J, Satoh T, Shirai Y, Takahashi Y, Nakagawa K, Okumura K, Nagase T, Nomura N, Kikuchi H, Masuho Y, Yamashita R, Nakai K, Yada T, Nakamura Y, Ohara O, Isogai T, Sugano S: Complete sequencing and characterization of 21,243 full-length human cDNAs. Nat Genet. 2004 Jan;36(1):40-5. Epub 2003 Dec 21. [Article]
  10. Mungall AJ, Palmer SA, Sims SK, Edwards CA, Ashurst JL, Wilming L, Jones MC, Horton R, Hunt SE, Scott CE, Gilbert JG, Clamp ME, Bethel G, Milne S, Ainscough R, Almeida JP, Ambrose KD, Andrews TD, Ashwell RI, Babbage AK, Bagguley CL, Bailey J, Banerjee R, Barker DJ, Barlow KF, Bates K, Beare DM, Beasley H, Beasley O, Bird CP, Blakey S, Bray-Allen S, Brook J, Brown AJ, Brown JY, Burford DC, Burrill W, Burton J, Carder C, Carter NP, Chapman JC, Clark SY, Clark G, Clee CM, Clegg S, Cobley V, Collier RE, Collins JE, Colman LK, Corby NR, Coville GJ, Culley KM, Dhami P, Davies J, Dunn M, Earthrowl ME, Ellington AE, Evans KA, Faulkner L, Francis MD, Frankish A, Frankland J, French L, Garner P, Garnett J, Ghori MJ, Gilby LM, Gillson CJ, Glithero RJ, Grafham DV, Grant M, Gribble S, Griffiths C, Griffiths M, Hall R, Halls KS, Hammond S, Harley JL, Hart EA, Heath PD, Heathcott R, Holmes SJ, Howden PJ, Howe KL, Howell GR, Huckle E, Humphray SJ, Humphries MD, Hunt AR, Johnson CM, Joy AA, Kay M, Keenan SJ, Kimberley AM, King A, Laird GK, Langford C, Lawlor S, Leongamornlert DA, Leversha M, Lloyd CR, Lloyd DM, Loveland JE, Lovell J, Martin S, Mashreghi-Mohammadi M, Maslen GL, Matthews L, McCann OT, McLaren SJ, McLay K, McMurray A, Moore MJ, Mullikin JC, Niblett D, Nickerson T, Novik KL, Oliver K, Overton-Larty EK, Parker A, Patel R, Pearce AV, Peck AI, Phillimore B, Phillips S, Plumb RW, Porter KM, Ramsey Y, Ranby SA, Rice CM, Ross MT, Searle SM, Sehra HK, Sheridan E, Skuce CD, Smith S, Smith M, Spraggon L, Squares SL, Steward CA, Sycamore N, Tamlyn-Hall G, Tester J, Theaker AJ, Thomas DW, Thorpe A, Tracey A, Tromans A, Tubby B, Wall M, Wallis JM, West AP, White SS, Whitehead SL, Whittaker H, Wild A, Willey DJ, Wilmer TE, Wood JM, Wray PW, Wyatt JC, Young L, Younger RM, Bentley DR, Coulson A, Durbin R, Hubbard T, Sulston JE, Dunham I, Rogers J, Beck S: The DNA sequence and analysis of human chromosome 6. Nature. 2003 Oct 23;425(6960):805-11. [Article]
  11. Gerhard DS, Wagner L, Feingold EA, Shenmen CM, Grouse LH, Schuler G, Klein SL, Old S, Rasooly R, Good P, Guyer M, Peck AM, Derge JG, Lipman D, Collins FS, Jang W, Sherry S, Feolo M, Misquitta L, Lee E, Rotmistrovsky K, Greenhut SF, Schaefer CF, Buetow K, Bonner TI, Haussler D, Kent J, Kiekhaus M, Furey T, Brent M, Prange C, Schreiber K, Shapiro N, Bhat NK, Hopkins RF, Hsie F, Driscoll T, Soares MB, Casavant TL, Scheetz TE, Brown-stein MJ, Usdin TB, Toshiyuki S, Carninci P, Piao Y, Dudekula DB, Ko MS, Kawakami K, Suzuki Y, Sugano S, Gruber CE, Smith MR, Simmons B, Moore T, Waterman R, Johnson SL, Ruan Y, Wei CL, Mathavan S, Gunaratne PH, Wu J, Garcia AM, Hulyk SW, Fuh E, Yuan Y, Sneed A, Kowis C, Hodgson A, Muzny DM, McPherson J, Gibbs RA, Fahey J, Helton E, Ketteman M, Madan A, Rodrigues S, Sanchez A, Whiting M, Madari A, Young AC, Wetherby KD, Granite SJ, Kwong PN, Brinkley CP, Pearson RL, Bouffard GG, Blakesly RW, Green ED, Dickson MC, Rodriguez AC, Grimwood J, Schmutz J, Myers RM, Butterfield YS, Griffith M, Griffith OL, Krzywinski MI, Liao N, Morin R, Palmquist D, Petrescu AS, Skalska U, Smailus DE, Stott JM, Schnerch A, Schein JE, Jones SJ, Holt RA, Baross A, Marra MA, Clifton S, Makowski KA, Bosak S, Malek J: The status, quality, and expansion of the NIH full-length cDNA project: the Mammalian Gene Collection (MGC). Genome Res. 2004 Oct;14(10B):2121-7. [Article]
  12. Uhl GR, Sora I, Wang Z: The mu opiate receptor as a candidate gene for pain: polymorphisms, variations in expression, nociception, and opiate responses. Proc Natl Acad Sci U S A. 1999 Jul 6;96(14):7752-5. [Article]
  13. Chuang TK, Killam KF Jr, Chuang LF, Kung HF, Sheng WS, Chao CC, Yu L, Chuang RY: Mu opioid receptor gene expression in immune cells. Biochem Biophys Res Commun. 1995 Nov 22;216(3):922-30. [Article]
  14. Koch T, Brandenburg LO, Schulz S, Liang Y, Klein J, Hollt V: ADP-ribosylation factor-dependent phospholipase D2 activation is required for agonist-induced mu-opioid receptor endocytosis. J Biol Chem. 2003 Mar 14;278(11):9979-85. Epub 2003 Jan 7. [Article]
  15. Zhang P, Johnson PS, Zollner C, Wang W, Wang Z, Montes AE, Seidleck BK, Blaschak CJ, Surratt CK: Mutation of human mu opioid receptor extracellular "disulfide cysteine" residues alters ligand binding but does not prevent receptor targeting to the cell plasma membrane. Brain Res Mol Brain Res. 1999 Oct 1;72(2):195-204. [Article]
  16. Wang D, Sadee W, Quillan JM: Calmodulin binding to G protein-coupling domain of opioid receptors. J Biol Chem. 1999 Jul 30;274(31):22081-8. [Article]
  17. Law PY, Wong YH, Loh HH: Molecular mechanisms and regulation of opioid receptor signaling. Annu Rev Pharmacol Toxicol. 2000;40:389-430. [Article]
  18. Wang D, Surratt CK, Sadee W: Calmodulin regulation of basal and agonist-stimulated G protein coupling by the mu-opioid receptor (OP(3)) in morphine-pretreated cell. J Neurochem. 2000 Aug;75(2):763-71. [Article]
  19. Suzuki S, Chuang LF, Yau P, Doi RH, Chuang RY: Interactions of opioid and chemokine receptors: oligomerization of mu, kappa, and delta with CCR5 on immune cells. Exp Cell Res. 2002 Nov 1;280(2):192-200. [Article]
  20. Massotte D, Brillet K, Kieffer B, Milligan G: Agonists activate Gi1 alpha or Gi2 alpha fused to the human mu opioid receptor differently. J Neurochem. 2002 Jun;81(6):1372-82. [Article]
  21. Feng GJ, Kellett E, Scorer CA, Wilde J, White JH, Milligan G: Selective interactions between helix VIII of the human mu-opioid receptors and the C terminus of periplakin disrupt G protein activation. J Biol Chem. 2003 Aug 29;278(35):33400-7. Epub 2003 Jun 16. [Article]
  22. Onoprishvili I, Andria ML, Kramer HK, Ancevska-Taneva N, Hiller JM, Simon EJ: Interaction between the mu opioid receptor and filamin A is involved in receptor regulation and trafficking. Mol Pharmacol. 2003 Nov;64(5):1092-100. [Article]
  23. Wang D, Sun X, Bohn LM, Sadee W: Opioid receptor homo- and heterodimerization in living cells by quantitative bioluminescence resonance energy transfer. Mol Pharmacol. 2005 Jun;67(6):2173-84. Epub 2005 Mar 18. [Article]
  24. Pascal G, Milligan G: Functional complementation and the analysis of opioid receptor homodimerization. Mol Pharmacol. 2005 Sep;68(3):905-15. Epub 2005 Jun 20. [Article]
  25. Choi HS, Kim CS, Hwang CK, Song KY, Wang W, Qiu Y, Law PY, Wei LN, Loh HH: The opioid ligand binding of human mu-opioid receptor is modulated by novel splice variants of the receptor. Biochem Biophys Res Commun. 2006 May 19;343(4):1132-40. Epub 2006 Mar 23. [Article]
  26. Ancevska-Taneva N, Onoprishvili I, Andria ML, Hiller JM, Simon EJ: A member of the heat shock protein 40 family, hlj1, binds to the carboxyl tail of the human mu opioid receptor. Brain Res. 2006 Apr 7;1081(1):28-33. Epub 2006 Mar 20. [Article]
  27. Roumy M, Lorenzo C, Mazeres S, Bouchet S, Zajac JM, Mollereau C: Physical association between neuropeptide FF and micro-opioid receptors as a possible molecular basis for anti-opioid activity. J Biol Chem. 2007 Mar 16;282(11):8332-42. Epub 2007 Jan 15. [Article]
  28. Lopez A, Salome L: Membrane functional organisation and dynamic of mu-opioid receptors. Cell Mol Life Sci. 2009 Jul;66(13):2093-108. doi: 10.1007/s00018-009-0008-4. Epub 2009 Mar 20. [Article]
  29. Talbot JN, Skifter DA, Bianchi E, Monaghan DT, Toews ML, Murrin LC: Regulation of mu opioid receptor internalization by the scaffold protein RanBPM. Neurosci Lett. 2009 Dec 11;466(3):154-8. doi: 10.1016/j.neulet.2009.09.048. Epub 2009 Sep 27. [Article]
  30. Jin J, Kittanakom S, Wong V, Reyes BA, Van Bockstaele EJ, Stagljar I, Berrettini W, Levenson R: Interaction of the mu-opioid receptor with GPR177 (Wntless) inhibits Wnt secretion: potential implications for opioid dependence. BMC Neurosci. 2010 Mar 9;11:33. doi: 10.1186/1471-2202-11-33. [Article]
  31. Gris P, Gauthier J, Cheng P, Gibson DG, Gris D, Laur O, Pierson J, Wentworth S, Nackley AG, Maixner W, Diatchenko L: A novel alternatively spliced isoform of the mu-opioid receptor: functional antagonism. Mol Pain. 2010 Jun 2;6:33. doi: 10.1186/1744-8069-6-33. [Article]
  32. Bergen AW, Kokoszka J, Peterson R, Long JC, Virkkunen M, Linnoila M, Goldman D: Mu opioid receptor gene variants: lack of association with alcohol dependence. Mol Psychiatry. 1997 Oct-Nov;2(6):490-4. [Article]
  33. Bond C, LaForge KS, Tian M, Melia D, Zhang S, Borg L, Gong J, Schluger J, Strong JA, Leal SM, Tischfield JA, Kreek MJ, Yu L: Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci U S A. 1998 Aug 4;95(16):9608-13. [Article]

Drug Relations

Drug Relations
DrugBank IDNameDrug groupPharmacological action?ActionsDetails
DB00193Tramadolapproved, investigationalyesagonistDetails
DB00295Morphineapproved, investigationalyesagonistregulatorDetails
DB00318Codeineapproved, illicityesagonistregulatorDetails
DB00327Hydromorphoneapproved, illicityesagonistDetails
DB00504Levallorphanapprovedyespartial agonistDetails
DB00647Dextropropoxypheneapproved, illicit, investigational, withdrawnyesagonistDetails
DB00652Pentazocineapproved, vet_approvedyesantagonistDetails
DB00704Naltrexoneapproved, investigational, vet_approvedyesantagonistDetails
DB00708Sufentanilapproved, investigationalyesagonistDetails
DB00802Alfentanilapproved, illicityesagonistDetails
DB00813Fentanylapproved, illicit, investigational, vet_approvedyesagonistDetails
DB00854LevorphanolapprovedyesagonistDetails
DB00899RemifentanilapprovedyesagonistDetails
DB00913Anileridineapproved, illicityesagonistDetails
DB00956Hydrocodoneapproved, illicit, investigationalyesagonistDetails
DB01081Diphenoxylateapproved, illicityesagonistDetails
DB01183Naloxoneapproved, vet_approvedyesantagonistDetails
DB01192Oxymorphoneapproved, investigational, vet_approvedyesagonistDetails
DB01209Dezocineapproved, investigationalyesagonistDetails
DB01227Levacetylmethadolapproved, investigational, withdrawnyesagonistDetails
DB00333MethadoneapprovedyesagonistDetails
DB00611Butorphanolapproved, illicit, vet_approvedyesantagonistDetails
DB00836LoperamideapprovedyesagonistDetails
DB00844NalbuphineapprovedyesantagonistDetails
DB00921Buprenorphineapproved, illicit, investigational, vet_approvedyespartial agonistDetails
DB01497Etorphineillicit, vet_approvedyesagonistDetails
DB01535Carfentanilillicit, investigational, vet_approvedyesagonistDetails
DB014393-Methylthiofentanylexperimental, illicityesagonistDetails
DB01444Dimethylthiambuteneexperimental, illicityesagonistDetails
DB01452Diamorphineapproved, illicit, investigationalyesagonistDetails
DB015713-MethylfentanylillicityesagonistDetails
DB05046V1003investigationalunknownDetails
DB01433Methadyl acetateexperimental, illicityesagonistDetails
DB00497Oxycodoneapproved, illicit, investigationalyesagonistDetails
DB05492Epicept NP-1investigationalunknownDetails
DB05509LI-301investigationalunknownDetails
DB06204TapentadolapprovedunknownagonistDetails
DB06230Nalmefeneapproved, investigational, withdrawnyesantagonistDetails
DB06409Morphine glucuronideinvestigationalunknownDetails
DB06274Alvimopanapproved, investigationalyesantagonistDetails
DB01466Ethylmorphineexperimental, illicityesagonistDetails
DB01565Dihydromorphineexperimental, illicityesagonistDetails
DB01548Diprenorphineillicit, vet_approvedyesantagonistDetails
DB00904Ondansetronapproved, withdrawnunknownother/unknownDetails
DB06800MethylnaltrexoneapprovedyesantagonistDetails
DB06738KetobemidoneinvestigationalyesagonistDetails
DB00514DextromethorphanapprovedunknownagonistregulatorDetails
DB00454MeperidineapprovedunknownagonistDetails
DB00321AmitriptylineapprovedunknownbinderDetails
DB01221Ketamineapproved, vet_approvedunknownbinderDetails
DB09049NaloxegolapprovedyesantagonistDetails
DB09272Eluxadolineapproved, investigationalyesagonistDetails
DB09089TrimebutineapprovedyesagonistDetails
DB11691Naldemedineapproved, investigationalyesantagonistDetails
DB09061Cannabidiolapproved, investigationalunknownDetails
DB09209Pholcodineapproved, illicityesantagonistDetails
DB11130Opiumapproved, illicityesagonistDetails
DB14011NabiximolsinvestigationalunknownDetails
DB14009Medical Cannabisexperimental, investigationalunknownDetails
DB11186Pentoxyverineapproved, investigational, withdrawnunknownantagonistDetails
DB09289TianeptineinvestigationalyesagonistDetails
DB14146LoxicodegolinvestigationalyesagonistDetails
DB09173ButyrfentanylillicityesagonistDetails
DB01238Aripiprazoleapproved, investigationalunknownligandDetails
DB12668MetenkefalininvestigationalunknownagonistDetails
DB14881Oliceridineapproved, investigationalyesagonistDetails
DB06288Amisulprideapproved, investigationalnoagonistDetails
DB12543Samidorphanapproved, investigationalyesantagonistDetails