Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2157 word(s) 2157 2021-01-11 09:06:00 |
2 Format correct Meta information modification 2157 2021-01-16 12:37:51 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Huang, X. β-Endorphin. Encyclopedia. Available online: https://encyclopedia.pub/entry/6483 (accessed on 28 March 2024).
Huang X. β-Endorphin. Encyclopedia. Available at: https://encyclopedia.pub/entry/6483. Accessed March 28, 2024.
Huang, Xudong. "β-Endorphin" Encyclopedia, https://encyclopedia.pub/entry/6483 (accessed March 28, 2024).
Huang, X. (2021, January 15). β-Endorphin. In Encyclopedia. https://encyclopedia.pub/entry/6483
Huang, Xudong. "β-Endorphin." Encyclopedia. Web. 15 January, 2021.
β-Endorphin
Edit

β-Endorphins are peptides that exert a wide variety of effects throughout the body. Produced through the cleavage pro-opiomelanocortin (POMC), β-endorphins are the primarily agonist of mu opioid receptors, which can be found throughout the body, brain, and cells of the immune system that regulate a diverse set of systems. As an agonist of the body’s opioid receptors, β-endorphins are most noted for their potent analgesic effects, but they also have their involvement in reward-centric and homeostasis-restoring behaviors, among other effects. These effects have implicated the peptide in psychiatric and neurodegenerative disorders, making it a research target of interest.

β-Endorphin,Behavior

1. Introduction

β-Endorphins are peptides that exert effects throughout the body. In the brain, they are considered to be both neurotransmitters and neuromodulators, as they have the ability to elicit more stable and long-lasting effects on more distant targets than typical neurotransmitters [1]; β-endorphins exhibit a notably high degree of degradation-resistance in the brain [1]. β-Endorphins are produced primarily by both the anterior lobe of the pituitary gland [2][3], and in pro-opiomelanocortin (POMC) cells primarily located in the hypothalamus [3][4]. β-Endorphin and other cleavage products are produced in the multistage processing of POMC primarily involving prohormone convertases (PC) 1 and 2. PC-1 cleaves POMC into adrenocorticotrophic hormone biosynthetic intermediate and β-lipotropic hormone. PC-2 cleaves β-lipotropic hormone into β-endorphin and γ-lipotropic hormone [5]. Carboxypeptidase-E (CPE) is also involved in the processing of POMC and with the removal of C-terminal basic residues (arginine/lysine) which are left after PC cleavage. Notably, however, mice with inactive CPE (CPEfat/CPEfat) produce greater quantities of β-endorphin(1–31) than wild-type mice, indicating that CPE is not required for its synthesis [6]. Additional processing, such as the acetylation of some β-endorphins, and shortening, may also occur [7][8][9].

In the brain, the peptide and other related proteins are most prevalent in the hypothalamus, thalamus–midbrain, amygdala, hippocampus, and brainstem [1]. Though the primary source of peripheral β-endorphin is the pituitary gland [3], β-endorphins, POMC, and PCs 1 and 2 have been identified in the skin [10][11][12], as well as cells of the immune system [13], though transcripts of POMC are notably found only at very low levels in the latter [14]. Though there is evidence that peptides such as β-endorphins can penetrate the blood–brain barrier to a degree, based on studies of radiolabeled, intracarotidally injected peptides [15], and P-glycoprotein notably is involved in the efflux of β-endorphin from the brain [16], peripheral and central CSF levels of the peptide are not necessarily related [3].

There are multiple forms of endorphins, though β-endorphin(1–31) is the only form with a potent analgesic effect. The other, more inactive forms are shorter, such as β-endorphin(1–27); notably, β-endorphin(1–31) is the primary form found in the anterior pituitary gland and brain regions such as the hypothalamus, midbrain, and amygdala [17][18], while the shorter forms are more common in the intermediate pituitary and brain regions, such as the hippocampus, colliculae, and brain stem [17][18]. Typical antibodies against β-endorphin will recognize 1–31, 1–26, 1–27, and the acetylated forms [18]. β-Endorphin(1–27) has notably reported to be an antagonist of β-endorphin(1–31) in in vivo animal studies [19][20][21], though this has been refuted in more recent in vivo studies [22] and in vitro [8][23], indicating that the shorter forms of β-endorphin are full agonists, and the studies indicating antagonistic activity were somewhat flawed [8]. There is evidence that receptors with opiate activity that preferentially bind β-endorphin(1–31) are present [1].

β-Endorphins are part of the system of opioid receptor agonists. The endorphin family includes β-endorphin, α-neoendorphin, enkephalins, and dynorphins [24]. β-Endorphins exert an analgesic effect that is more potent than morphine [1][2], and act primarily on the mu family of opioid receptors [25], which are, like the two other opioid receptors, delta and kappa, G-protein coupled receptors [24]. Naloxone, a typical antagonist of other opiates, has been found to reduce β-endorphin binding as well, and is commonly used in opioid-related studies [1]. β-Endorphins, along with other opioids, appear to attenuate cyclic adenosine monophosphate levels, and decrease calcium uptake [2]. The peptide is typically released to the periphery in response to a painful or stressful event, where they inhibit somatosensory fibers, with a focus on nociceptors [2]. It should be noted that, while β-endorphin has the highest affinity for the mu receptor, it also acts on other opioid receptors [24], particularly the delta opioid receptor [26]. Other opioid receptor agonists, such as the enkephalins and endomorphins, can also activate the mu-receptors [24][26][27][28], so all mu-receptor activity cannot necessarily be attributed to β-endorphin. It should also be noted that endomorphins are peptides which have been isolated in brain and immune tissue that exhibit the highest affinity for the mu opioid receptor [29][30][31][32], but as no precursor has yet been identified, it is unclear if they are truly endogenous [33].

β-Endorphins are related to the hypothalamic–pituitary–adrenal (HPA) axis. The HPA axis, which governs a wide range of functions, including metabolic and immune responses [34][35], is heavily involved in the body’s stress-response, with stress being defined as the perception of a threat, real or imagined, to one’s well-being or homeostatic state [36]. The HPA is first stimulated when a stressful event induces production of corticotropin-releasing hormone (CRH). This results in the simultaneous release of Adrenocorticotropic hormone (ACTH) and β-endorphins, both of which are produced from the cleavage of POMC, and are stored and released together in secretory vesicles [37]. As the initial cleavage generates a β-endorphin precursor, β-lipotropic hormone (LPH), which requires further processing to form β-endorphin, the two are not necessarily present in a 1:1 ratio [4]. The effects of the HPA can be tied to a variety of behaviors, including exercise, drug-use, sexual behaviors, among others [38].

2. β-Endorphins and Behavior

Much of β-endorphin’s governance of behavior has to do with either brain reward-system pathways, among other changes in food-consumption, sexual behaviors, among others [39]. Though much focus on reward systems, and how it relates to many of the same behavioral changes as β-endorphin, is given to dopamine [40][41][42][43][44][45], β-endorphins are also active in various reward-system pathways. In fact, β-endorphins appear to exert regulatory effects on serotonin, inhibiting its release and modulating its turnover in a region-dependent manner that can be nullified with opioid antagonists [46][47][48]. Reciprocally, serotonin appears to regulate the secretion of β-endorphins in a similarly region-dependent way. Notably, secretion is increased in the hypothalamus and decreased in the hypothalamus in response to serotonin [49][50][51].

While not necessarily independent of other factors, the peptide is heavily involved in certain reward pathways. It is known that β-endorphins inhibit the release of gamma-aminobutyric acid (GABA) [52], the primary inhibitory neurotransmitter of the brain [53], which can lead to excess accumulation of dopamine, a key agent associated with feelings of pleasure [52]. Indeed, binding of opiates in general to their respective receptors is associated with feelings of well-being and euphoria [54]. Rats given access to levers that administer endorphins directly to their brain will self-administer the peptide to themselves regularly [55]. Injections of endorphins and exogenous opioids such as heroin can produce conditioned place preference, a common test for drug-reward [56], in rats, suggesting opioids’ involvement in reinforcement and reward [55][57]. Consumption of highly palatable foods in non-food-deprived rats has been observed to increase binding of β-endorphin in the hypothalamus, further reinforcing β-endorphin’s role in reward pathways [58].

2.1. Addiction

Much of the research on β-endorphin’s human effects has to do with addictions, particularly those involving drug use and alcohol abuse. It is integrally involved in producing the feelings of euphoria associated with certain drugs and has been implicated in the development of alcoholism [59]. Consumption of cocaine, for example, leads to an increase in plasma concentrations of β-endorphins [60]. Opioid antagonists have been found to alleviate cocaine-addiction behaviors, as cocaine-induced reinforcement of conditioned place preference (CPP) was reduced through the administration of naltrexone, and reinforced by methadone [61]. Interestingly, the rewarding action of cocaine does not appear to be mediated by the mu-opioid receptor; mu-receptor knockout mice exhibit cocaine induced CPP, but β-endorphin deficient mice do not [62]. β-Endorphin has been suggested to interact with the delta opioid receptor in cocaine addiction, which may not only mediate the direct reward, but also the incubation of craving that can induce relapses in abstinent recovering cocaine addicts [63]. Opioid drugs such as heroin directly interface with opioid receptors, inducing pain relief and stimulating reward centers [64]. Opioid antagonists, in particular naloxone, are frequently used in the treatment of heroin addiction, through tandem treatment with a low-risk opioid such as buprenorphine [65], and heroin overdoses [66]. Certain variations of the mu-opioid receptor have been found to influence the condition and treatment outcome of heroin addicts [67]. Alcohol consumption appears to stimulate the release of β-endorphin, but habitual consumption ultimately results in a reduction in β-endorphin levels [68]. Those with genetic deficiencies in β-endorphin levels are more likely to become alcoholics [68], and excessive alcohol consumption in rats can be curbed through the use of opiate antagonists [69], further cementing the peptide’s role in the development of alcoholism. β-endorphin has also been suggested to be related to the phenomenon of exercise addiction; increases in plasma levels of the peptide is often observed in strenuous exercise, and is associated with feelings of well-being and euphoria similar to that observed in drug addiction [70][71]. However, difficulties in relating circulating β-endorphins to brain levels of the peptide makes verification difficult [70].

2.2. Food Consumption

Many opioid receptors, which can be found throughout the brain, are localized in brain regions that pertain to food/energy homeostasis [72]. A 1979 study found that administration of the opioid antagonist naloxone suppresses eating even in food-deprived rats [73], and successive studies have gone on to confirm this, and note that opioid antagonists reduce overall food intake in mice, including those that are genetically obese [72]. Single nucleotide polymorphisms of the mu-receptor gene have been found to be correlated to BMI in humans [74]. Total knockout of the mu-receptor gene decreases motivation to eat in mice, while showing no significant impact on the hedonic processing of food intake [75]; this is somewhat contradictory to β-endorphin’s role in food reward-pathways, and could indicate an alternative reward path, or that the β-endorphin reward pathway is not mediated by the mu receptor. In support of this, while knocking out β-endorphin’s primary receptor appears to have little effect on food hedonism, rats ingesting oil were found to have significantly increased levels of β-endorphin in serum and CSF. Interestingly, overall intake of the oil was found to be lowest where β-endorphin levels were highest, suggesting an inverse relationship. Pretreatment with naloxone decreased initial affinity for the oil based on licking-tests, suggesting a possible hedonic disruption [76].

Though it would seem that β-endorphins promote food consumption, with disruptions limiting food intake, the longer-term implications are quite different. Mice lacking the ability to produce β-endorphin were found to weigh significantly less than their wild-type counterparts after several weeks with food provided freely, suggesting that the peptide simultaneously has appetitive and anorexic effects, limiting excessive food consumption [77]. β-Endorphins appear to have peripheral function on taste and overall gastrointestinal function, suggesting that CNS action is not the only route through which β-endorphins modulate food intake [78].

2.3. Sexual Behavior

Exogenous opioids are known to have an overall inhibitory effect on sexual behaviors [79], but the impact of opioid receptor agonists including β-endorphin are considerably more nuanced. In both male rats and humans, administration of the opiate antagonist naloxone has been shown to induce copulatory behavior and improve sexual performance [80][81][82]. It would seem, however, that β-endorphins still have their place in the mechanisms of sexual reward. Rats with access to a preferred and non-preferred chamber would, when allowed to copulate to ejaculation in a non-preferred chamber, shift their preference toward the originally non-preferred chamber even following castration. This shift in preference was reversible with naloxone, with the extent of this reverse increasing over time [83].

Females have been found to experience similar inhibition of sexual behavior by β-endorphin. Numerous studies involving the lordosis reflex of female rats note that it is diminished following infusions of β-endorphins [84][85][86][87]. However, Torii et al. found that the inhibitory effect becomes a facilitatory one if β-endorphin administration occurs within 6 h of priming with estrogen [86]. Pfaus et al. found that the inhibitive effect of β-endorphins on lordosis is dose dependent, noting that it is likely that high-affinity mu-receptor activity inhibits lordosis, whereas low-affinity receptor activity facilitates it [87]. While it is fair to say that β-endorphin follows the trend of exogenous opioids in inhibiting sexual behavior, interactions and the multiplicity of the pathways β-endorphins influence allow for notable exceptions to the rule.

2.4. Other Effects

Aside from these regulatory effects on behavior, β-endorphins have their hand in other actions as well. Injection of β-endorphins to the brain has been found to induce tremors and jerking movements of the head, ocular fixation on empty space, pupillary dilation, and overall excitation in cats. The duration and severity of these effects was dose dependent, with the effects of a 12.5 µg dose lasting approximately one hour [88]. Intraventricular injections have also been observed to induce wet-dog shakes in rats [89][90], with the frequency and occurrence of the shakes depending on the ambient temperature; correlation of the wet-dog shake behavior to increased body temperature suggests a possible role in thermoregulation [90]. However, levels of β-endorphins have not been found to increase in proportion to heat loss in cold water immersion tests [91].

References

  1. Smyth, D.G. 60 years of pomc: Lipotropin and beta-endorphin: A perspective. J. Mol. Endocrinol. 2016, 56, T13–T25.
  2. Hartwig, A.C. Peripheral beta-endorphin and pain modulation. Anesth. Prog. 1991, 38, 75–78.
  3. Veening, J.G.; Gerrits, P.O.; Barendregt, H.P. Volume transmission of beta-endorphin via the cerebrospinal fluid; a review. Fluids Barriers CNS 2012, 9, 16.
  4. Harno, E.; Gali Ramamoorthy, T.; Coll, A.P.; White, A. POMC: The Physiological Power of Hormone Processing. Physiol. Rev. 2018, 98, 2381–2430.
  5. Tanaka, S. Comparative aspects of intracellular proteolytic processing of peptide hormone precursors: Studies of proopiomelanocortin processing. Zool. Sci. 2003, 20, 1183–1198.
  6. Ji, L.; Wu, H.-T.; Qin, X.-Y.; Lan, R. Dissecting carboxypeptidase E: Properties, functions and pathophysiological roles in disease. Endocr. Connect. 2017, 6, R18–R38.
  7. Cawley, N.X.; Li, Z.; Loh, Y.P. 60 YEARS OF POMC: Biosynthesis, trafficking, and secretion of pro-opiomelanocortin-derived peptides. J. Mol. Endocrinol. 2016, 56, T77–T97.
  8. Gomes, I.; Sierra, S.; Lueptow, L.; Gupta, A.; Gouty, S.; Margolis, E.B.; Cox, B.M.; Devi, L.A. Biased signaling by endogenous opioid peptides. Proc. Natl. Acad. Sci. USA 2020, 117, 11820–11828.
  9. Fricker, L.D.; Margolis, E.B.; Gomes, I.; Devi, L.A. Five decades of research on opioid peptides: Current knowledge and unanswered questions. Mol. Pharmacol. 2020, 98, 96–108.
  10. Chakraborty, A.K.; Funasaka, Y.; Slominski, A.; Ermak, G.; Hwang, J.; Pawelek, J.M.; Ichihashi, M. Production and release of proopiomelanocortin (POMC) derived peptides by human melanocytes and keratinocytes in culture: Regulation by ultraviolet B. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 1996, 1313, 130–138.
  11. Slominski, A. Identification of β-endorphin, α-MSH and ACTH peptides in cultured human melanocytes, melanoma and squamous cell carcinoma cells by RP-HPLC. Exp. Dermatol. 1998, 7, 213–216.
  12. Slominski, A.; Wortsman, J.; Tuckey, R.C.; Paus, R. Differential expression of HPA axis homolog in the skin. Mol. Cell Endocrinol. 2007, 265–266, 143–149.
  13. Blalock, J. Proopiomelanocortin-derived peptides in the immune system. Clin. Endocrinol. 1985, 22, 823–827.
  14. Sharp, B.; Linner, K. What do we know about the expression of proopiomelanocortin transcripts and related peptides in lymphoid tissue? Endocrinology 1993, 133, 1921a–1921b.
  15. Ermisch, A.; Rühle, H.-J.; Landgraf, R.; Hess, J. Blood—Brain barrier and peptides. J. Cereb. Blood Flow Metab. 1985, 5, 350–357.
  16. King, M.; Su, W.; Chang, A.; Zuckerman, A.; Pasternak, G.W. Transport of opioids from the brain to the periphery by P-glycoprotein: Peripheral actions of central drugs. Nat. Neurosci. 2001, 4, 268–274.
  17. Mains, R.E.; Eipper, B.A. Differences in the post-translational processing of beta-endorphin in rat anterior and intermediate pituitary. J. Biol. Chem. 1981, 256, 5683–5688.
  18. Zakarian, S.; Smyth, D. Distribution of β-endorphin-related peptides in rat pituitary and brain. Biochem. J. 1982, 202, 561–571.
  19. Hammonds, R.G.; Nicolas, P.; Li, C.H. beta-endorphin-(1-27) is an antagonist of beta-endorphin analgesia. Proc. Natl. Acad. Sci. USA 1984, 81, 1389–1390.
  20. Suh, H.H.; Tseng, L.-F.; Li, C.H. Beta-endorphin-(1–27) antagonizes beta-endorphin-induced hypothermia in mice. Peptides 1987, 8, 123–126.
  21. Nicolas, P.; Li, C.H. Beta-endorphin-(1-27) is a naturally occurring antagonist to etorphine-induced analgesia. Proc. Natl. Acad. Sci. USA 1985, 82, 3178–3181.
  22. Hirsch, M.D.; Millington, W.R. Endoproteolytic conversion of β-endorphin-1-31 to β-endorphin-1-27 potentiates its central cardioregulatory activity. Brain Res. 1991, 550, 61–68.
  23. Alt, A.; Mansour, A.; Akil, H.; Medzihradsky, F.; Traynor, J.R.; Woods, J.H. Stimulation of guanosine-5′-O-(3-[35S]thio)triphosphate binding by endogenous opioids acting at a cloned mu receptor. J. Pharmacol. Exp. Ther. 1998, 286, 282–288.
  24. Koneru, A.; Satyanarayana, S.; Rizwan, S. Endogenous opioids: Their physiological role and receptors. Glob. J. Pharmacol. 2009, 3, 149–153.
  25. Facchinetti, F.; Petraglia, F.; Genazzani, A.R. Localization and expression of the three opioid systems. Semin. Reprod. Endocrinol. 1987, 5, 103–113.
  26. Raynor, K.; Kong, H.; Chen, Y.; Yasuda, K.; Yu, L.; Bell, G.I.; Reisine, T. Pharmacological characterization of the cloned kappa-, delta-, and mu-opioid receptors. Mol. Pharmacol. 1994, 45, 330–334.
  27. Gong, J.; Strong, J.A.; Zhang, S.; Yue, X.; DeHaven, R.N.; Daubert, J.D.; Cassel, J.A.; Yu, G.; Mansson, E.; Yu, L. Endomorphins fully activate a cloned human mu opioid receptor. FEBS Lett. 1998, 439, 152–156.
  28. Goldberg, I.E.; Rossi, G.C.; Letchworth, S.R.; Mathis, J.P.; Ryan-Moro, J.; Leventhal, L.; Su, W.; Emmel, D.; Bolan, E.A.; Pasternak, G.W. Pharmacological characterization of endomorphin-1 and endomorphin-2 in mouse brain. J. Pharmacol. Exp. Ther. 1998, 286, 1007–1013.
  29. Hackler, L.; Zadina, J.E.; Ge, L.-J.; Kastin, A.J. Isolation of relatively large amounts of endomorphin-1 and endomorphin-2 from human brain cortex. Peptides 1997, 18, 1635–1639.
  30. Zadina, J.E.; MARTIN-SCHILD, S.; Gerall, A.A.; Kastin, A.J.; Hackler, L.; GE, L.J.; Zhang, X. Endomorphins: Novel endogenous μ-opiate receptor agonists in regions of high μ-opiate receptor density. Ann. N. Y. Acad. Sci. 1999, 897, 136–144.
  31. Schreff, M.; Schulz, S.; Wiborny, D.; Höllt, V. Immunofluorescent identification of endomorphin-2-containing nerve fibers and terminals in the rat brain and spinal cord. Neuroreport 1998, 9, 1031–1034.
  32. Jessop, D.S.; Major, G.N.; Coventry, T.L.; Kaye, S.J.; Fulford, A.J.; Harbuz, M.S.; De Bree, F.M. Novel opioid peptides endomorphin-1 and endomorphin-2 are present in mammalian immune tissues. J. Neuroimmunol. 2000, 106, 53–59.
  33. Terskiy, A.; Wannemacher, K.M.; Yadav, P.N.; Tsai, M.; Tian, B.; Howells, R.D. Search of the human proteome for endomorphin-1 and endomorphin-2 precursor proteins. Life Sci. 2007, 81, 1593–1601.
  34. Pariante, C.M.; Lightman, S.L. The HPA axis in major depression: Classical theories and new developments. Trends Neurosci. 2008, 31, 464–468.
  35. Harbuz, M.; Lightman, S. Stress and the hypothalamo-pituitary-adrenal axis: Acute, chronic and immunological activation. J. Endocrinol. 1992, 134, 327–339.
  36. Smith, S.M.; Vale, W.W. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin. Neurosci. 2006, 8, 383–395.
  37. Gianoulakis, C. Alcohol-seeking behavior: The roles of the hypothalamic-pituitary-adrenal axis and the endogenous opioid system. Alcohol Health Res. World 1998, 22, 202–210.
  38. Spencer, R.L.; Deak, T. A users guide to HPA axis research. Physiol. Behav. 2017, 178, 43–65.
  39. Veening, J.G.; Barendregt, H.P. The effects of beta-endorphin: State change modification. Fluids Barriers CNS 2015, 12, 3.
  40. Hernandez, L.; Hoebel, B.G. Food reward and cocaine increase extracellular dopamine in the nucleus accumbens as measured by microdialysis. Life Sci. 1988, 42, 1705–1712.
  41. Söderpalm, B.; Löf, E.; Ericson, M. Mechanistic studies of ethanol’s interaction with the mesolimbic dopamine reward system. Pharmacopsychiatry 2009, 42, S87–S94.
  42. Taber, K.H.; Black, D.N.; Porrino, L.J.; Hurley, R.A. Neuroanatomy of dopamine: Reward and addiction. J. Neuropsychiatry Clin. Neurosci. 2012, 24, 1–4.
  43. Di Chiara, G.; Bassareo, V. Reward system and addiction: What dopamine does and doesn’t do. Curr. Opin. Pharmacol. 2007, 7, 69–76.
  44. Volkow, N.D.; Wang, G.-J.; Baler, R.D. Reward, dopamine and the control of food intake: Implications for obesity. Trends Cogn. Sci. 2011, 15, 37–46.
  45. Phillips, A.; Pfaus, J.; Blaha, C. Dopamine and motivated behavior: Insights provided by in vivo analyses. Mesolimbic Dopamine Syst. Motiv. Action 1991, 199–224.
  46. Loh, H.; Brase, D.; Sampath-Khanna, S.; Mar, J.; Way, E.; Li, C. beta-Endorphin in vitro inhibition of striatal dopamine release. Nature 1976, 264, 567.
  47. Deyo, S.; Swift, R.; Miller, R. Morphine and endorphins modulate dopamine turnover in rat median eminence. Proc. Natl. Acad. Sci. USA 1979, 76, 3006.
  48. van Loon, G.; Ho, D.; Kim, C. Beta-endorphin-induced decrease in hypothalamic dopamine turnover. Endocrinology 1980, 106, 76.
  49. Van Loon, G. Plasma dopamine: Regulation and significance. Fed. Proc. 1983, 42, 3012.
  50. Rasmussen, D.D.; Liu, J.H.; Wolf, P.L.; Yen, S.S. Neurosecretion of human hypothalamic immunoreactive beta-endorphin: In vitro regulation by dopamine. Neuroendocrinology 1987, 45, 197–200.
  51. Farah, J.M., Jr.; Malcolm, D.S.; Mueller, G.P. Dopaminergic inhibition of pituitary beta-endorphin-like immunoreactivity secretion in the rat. Endocrinology 1982, 110, 657–659.
  52. Sprouse-Blum, A.S.; Smith, G.; Sugai, D.; Parsa, F.D. Understanding endorphins and their importance in pain management. Hawaii Med. J. 2010, 69, 70–71.
  53. Cryan, J.F.; Kaupmann, K. Don’t worry ‘B’ happy!: A role for GABA(B) receptors in anxiety and depression. Trends Pharmacol. Sci. 2005, 26, 36–43.
  54. Froehlich, J.C. Opioid peptides. Alcohol Health Res. World 1997, 21, 132–136.
  55. Olive, M.F.; Koenig, H.N.; Nannini, M.A.; Hodge, C.W. Stimulation of endorphin neurotransmission in the nucleus accumbens by ethanol, cocaine, and amphetamine. J. Neurosci. 2001, 21, RC184.
  56. Bardo, M.T.; Bevins, R.A. Conditioned place preference: What does it add to our preclinical understanding of drug reward? Psychopharmacology 2000, 153, 31–43.
  57. Amalric, M.; Cline, E.J.; Martinez, J.L., Jr.; Bloom, F.E.; Koob, G.F. Rewarding properties of beta-endorphin as measured by conditioned place preference. Psychopharmacology 1987, 91, 14–19.
  58. Dum, J.; Gramsch, C.; Herz, A. Activation of hypothalamic beta-endorphin pools by reward induced by highly palatable food. Pharmacol. Biochem. Behav. 1983, 18, 443–447.
  59. Roth-Deri, I.; Green-Sadan, T.; Yadid, G. Beta-endorphin and drug-induced reward and reinforcement. Prog. Neurobiol. 2008, 86, 1–21.
  60. Moldow, R.L.; Fischman, A.J. Cocaine induced secretion of ACTH, beta-endorphin, and corticosterone. Peptides 1987, 8, 819–822.
  61. Bilsky, E.J.; Montegut, M.J.; Delong, C.L.; Reid, L.D. Opioidergic modulation of cocaine conditioned place preferences. Life Sci. 1992, 50, Pl85–P190.
  62. Nguyen, A.T.; Marquez, P.; Hamid, A.; Kieffer, B.; Friedman, T.C.; Lutfy, K. The rewarding action of acute cocaine is reduced in β-endorphin deficient but not in μ opioid receptor knockout mice. Eur. J. Pharmacol. 2012, 686, 50–54.
  63. Dikshtein, Y.; Barnea, R.; Kronfeld, N.; Lax, E.; Roth-Deri, I.; Friedman, A.; Gispan, I.; Elharrar, E.; Levy, S.; Ben-Tzion, M.; et al. β-endorphin via the delta opioid receptor is a major factor in the incubation of cocaine craving. Neuropsychopharmacology 2013, 38, 2508–2514.
  64. Dhaliwal, A.; Gupta, M. Physiology, Opioid Receptor. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2019.
  65. Weiss, R.D.; Rao, V. The Prescription Opioid Addiction Treatment Study: What have we learned. Drug Alcohol Depend. 2017, 173 (Suppl. S1), S48–S54.
  66. Dunne, R.B. Prescribing naloxone for opioid overdose intervention. Pain Manag. 2018, 8, 197–208.
  67. Woodcock, E.A.; Lundahl, L.H.; Burmeister, M.; Greenwald, M.K. Functional mu opioid receptor polymorphism (OPRM1 A(118) G) associated with heroin use outcomes in Caucasian males: A pilot study. Am. J. Addict. 2015, 24, 329–335.
  68. Zalewska-Kaszubska, J.; Czarnecka, E. Deficit in beta-endorphin peptide and tendency to alcohol abuse. Peptides 2005, 26, 701–705.
  69. Ulm, R.R.; Volpicelli, J.R.; Volpicelli, L.A. Opiates and alcohol self-administration in animals. J. Clin. Psychiatry 1995, 56 (Suppl. S7), 5–14.
  70. Berczik, K.; Griffiths, M.D.; Szabó, A.; Kurimay, T.; Urban, R.; Demetrovics, Z. Exercise addiction. In Behavioral Addictions; Elsevier: Amsterdam, The Netherlands, 2014; pp. 317–342.
  71. Nogueira, A.; Molinero, O.; Salguero, A.; Márquez, S. Exercise addiction in practitioners of endurance sports: A literature review. Front. Psychol. 2018, 9, 1484.
  72. Nogueiras, R.; Romero-Pico, A.; Vazquez, M.J.; Novelle, M.G.; Lopez, M.; Dieguez, C. The opioid system and food intake: Homeostatic and hedonic mechanisms. Obes. Facts 2012, 5, 196–207.
  73. Holtzman, S.G. Suppression of appetitive behavior in the rat by naloxone: Lack of effect of prior morphine dependence. Life Sci. 1979, 24, 219–226.
  74. Xu, L.; Zhang, F.; Zhang, D.D.; Chen, X.D.; Lu, M.; Lin, R.Y.; Wen, H.; Jin, L.; Wang, X.F. OPRM1 gene is associated with BMI in Uyghur population. Obesity (Silver Spring) 2009, 17, 121–125.
  75. Papaleo, F.; Kieffer, B.L.; Tabarin, A.; Contarino, A. Decreased motivation to eat in mu-opioid receptor-deficient mice. Eur. J. Neurosci. 2007, 25, 3398–3405.
  76. Mizushige, T.; Saitoh, K.; Manabe, Y.; Nishizuka, T.; Taka, Y.; Eguchi, A.; Yoneda, T.; Matsumura, S.; Tsuzuki, S.; Inoue, K.; et al. Preference for dietary fat induced by release of beta-endorphin in rats. Life Sci. 2009, 84, 760–765.
  77. Appleyard, S.M.; Hayward, M.; Young, J.I.; Butler, A.A.; Cone, R.D.; Rubinstein, M.; Low, M.J. A role for the endogenous opioid β-endorphin in energy homeostasis. Endocrinology 2003, 144, 1753–1760.
  78. Levine, A.S.; Morley, J.; Gosnell, B.A.; Billington, C.J.; Bartness, T. Opioids and consummatory behavior. Brain Res. Bull. 1985, 14, 663–672.
  79. Pfaus, J.G.; Gorzalka, B.B. Opioids and sexual behavior. Neurosci. Biobehav. Rev. 1987, 11, 1–34.
  80. Gessa, G.L.; Paglietti, E.; Quarantotti, B.P. Induction of copulatory behavior in sexually inactive rats by naloxine. Science 1979, 204, 203–205.
  81. Fabbri, A.; Jannini, E.A.; Gnessi, L.; Moretti, C.; Ulisse, S.; Franzese, A.; Lazzari, R.; Fraioli, F.; Frajese, G.; Isidori, A. Endorphins in male impotence: Evidence for naltrexone stimulation of erectile activity in patient therapy. Psychoneuroendocrinology 1989, 14, 103–111.
  82. McIntosh, T.K.; Vallano, M.L.; Barfield, R.J. Effects of morphine, beta-endorphin and naloxone on catecholamine levels and sexual behavior in the male rat. Pharmacol. Biochem. Behav. 1980, 13, 435–441.
  83. Miller, R.; Baum, M. Naloxone inhibits mating and conditioned place preference for an estrous female in male rats soon after castration. Pharmacol. Biochem. Behav. 1987, 26, 781–789.
  84. Wiesner, J.B.; Moss, R.L. Suppression of receptive and proceptive behavior in ovariectomized, estrogen-progesterone-primed rats by intraventricular beta-endorphin: Studies of behavioral specificity. Neuroendocrinology 1986, 43, 57–62.
  85. Sirinathsinghji, D.J. Regulation of lordosis behaviour in the female rat by corticotropin-releasing factor, beta-endorphin/corticotropin and luteinizing hormone-releasing hormone neuronal systems in the medial preoptic area. Brain Res. 1986, 375, 49–56.
  86. Torii, M.; Kubo, K.; Sasaki, T. Facilitatory and Inhibitory Effects of β-Endorphin on Lordosis in Female Rats: Relation to Time of Administration. Horm. Behav. 1999, 35, 271–278.
  87. Pfaus, J.G.; Gorzalka, B.B. Selective activation of opioid receptors differentially affects lordosis behavior in female rats. Peptides 1987, 8, 309–317.
  88. Meglio, M.; Hosobuchi, Y.; Loh, H.H.; Adams, J.E.; Li, C.H. beta-Endorphin: Behavioral and analgesic activity in cats. Proc. Natl. Acad. Sci. USA 1977, 74, 774–776.
  89. Bloom, F.; Segal, D.; Ling, N.; Guillemin, R. Endorphins: Profound behavioral effects in rats suggest new etiological factors in mental illness. Science (New York, N.Y.) 1976, 194, 630–632.
  90. Loh, H.H.; Li, C.H. Unique behavioral effects of β endorphin and their relationship to thermoregulation and hypothalamic function. Life Sci. 1978, 22, 1525–1535.
  91. Glickman-Weiss, E.L.; Nelson, A.G.; Hearon, C.M.; Goss, F.L.; Robertson, R.J. Are beta-endorphins and thermoregulation during cold-water immersion related? Undersea Hyperb. Med. 1993, 20, 205–213.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 1.3K
Revisions: 2 times (View History)
Update Date: 16 Apr 2021
1000/1000