#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Differential effects of synthetic psychoactive cathinones and amphetamine stimulants on the gut microbiome in mice


Autoři: Mariana Angoa-Pérez aff001;  Branislava Zagorac aff001;  Andrew D. Winters aff003;  Jonathan M. Greenberg aff003;  Madison Ahmad aff003;  Kevin R. Theis aff003;  Donald M. Kuhn aff001
Působiště autorů: Research and Development Service, John D. Dingell VA Medical Center, Detroit, Michigan, United States of America aff001;  Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, United States of America aff002;  Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, United States of America aff003;  Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, Michigan, United States of America aff004
Vyšlo v časopise: PLoS ONE 15(1)
Kategorie: Collection Review
doi: https://doi.org/10.1371/journal.pone.0227774

Souhrn

The list of pharmacological agents that can modify the gut microbiome or be modified by it continues to grow at a high rate. The greatest amount of attention on drug-gut microbiome interactions has been directed primarily at pharmaceuticals used to treat infection, diabetes, cardiovascular conditions and cancer. By comparison, drugs of abuse and addiction, which can powerfully and chronically worsen human health, have received relatively little attention in this regard. Therefore, the main objective of this study was to characterize how selected synthetic psychoactive cathinones (aka “Bath Salts”) and amphetamine stimulants modify the gut microbiome. Mice were treated with mephedrone (40 mg/kg), methcathinone (80 mg/kg), methamphetamine (5 mg/kg) or 4-methyl-methamphetamine (40 mg/kg), following a binge regimen consisting of 4 injections at 2h intervals. These drugs were selected for study because they are structural analogs that contain a β-keto substituent (methcathinone), a 4-methyl group (4-methyl-methamphetamine), both substituents (mephedrone) or neither (methamphetamine). Mice were sacrificed 1, 2 or 7 days after treatment and DNA from caecum contents was subjected to 16S rRNA sequencing. We found that all drugs caused significant time- and structure-dependent alterations in the diversity and taxonomic structure of the gut microbiome. The two phyla most changed by drug treatments were Firmicutes (methcathinone, 4-methyl-methamphetamine) and Bacteriodetes (methcathinone, 4-methyl-methamphetamine, methamphetamine, mephedrone). Across time, broad microbiome changes from the phylum to genus levels were characteristic of all drugs. The present results signify that these selected psychoactive drugs, which are thought to exert their primary effects within the CNS, can have profound effects on the gut microbiome. They also suggest new avenues of investigation into the possibility that gut-derived signals could modulate drug abuse and addiction via altered communication along the gut-brain axis.

Klíčová slova:

Drug abuse – Drug synthesis – Drug therapy – Microbial taxonomy – Microbiome – Ribosomal RNA – Simpson index – Amphetamines


Zdroje

1. Karila L, Billieux J, Benyamina A, Lancon C, Cottencin O (2016) The effects and risks associated to mephedrone and methylone in humans: A review of the preliminary evidences. Brain Res Bull 126: 61–67. doi: 10.1016/j.brainresbull.2016.03.005 26995278

2. Karila L, Lafaye G, Scocard A, Cottencin O, Benyamina A (2017) MDPV and alpha-PVP use in humans: The twisted sisters. Neuropharmacology 156: 48–55. doi: 10.1016/j.neuropharm.2017.10.007 29030166

3. White CM (2016) Mephedrone and 3,4-Methylenedioxypyrovalerone (MDPV): Synthetic cathinones with serious health implications. J Clin Pharmacol 56: 1319–1325. doi: 10.1002/jcph.742 27029951

4. Baumann MH, Volkow ND (2016) Abuse of new psychoactive substances: Threats and solutions. Neuropsychopharmacology 41: 663–665. doi: 10.1038/npp.2015.260 26303285

5. German CL, Fleckenstein AE, Hanson GR (2014) Bath salts and synthetic cathinones: an emerging designer drug phenomenon. Life Sci 97: 2–8. doi: 10.1016/j.lfs.2013.07.023 23911668

6. Baumann MH, Ayestas MA Jr., Partilla JS, Sink JR, Shulgin AT, Daley PF, et al. (2012) The designer methcathinone analogs, mephedrone and methylone, are substrates for monoamine transporters in brain tissue. Neuropsychopharmacology 37: 1192–1203. doi: 10.1038/npp.2011.304 22169943

7. Cameron K, Kolanos R, Vekariya R, De Felice L, Glennon RA (2013) Mephedrone and methylenedioxypyrovalerone (MDPV), major constituents of "bath salts," produce opposite effects at the human dopamine transporter. Psychopharmacology (Berl) 227: 493–499. doi: 10.1007/s00213-013-2967-2 23371489

8. Eshleman AJ, Wolfrum KM, Hatfield MG, Johnson RA, Murphy KV, Janowsky A (2013) Substituted methcathinones differ in transporter and receptor interactions. Biochem Pharmacol 85: 1803–1815. doi: 10.1016/j.bcp.2013.04.004 23583454

9. Fantegrossi WE, Gannon BM, Zimmerman SM, Rice KC (2013) In vivo effects of abused ‘bath salt’ constituent 3,4-methylenedioxypyrovalerone (MDPV) in mice: drug discrimination, thermoregulation, and locomotor activity. Neuropsychopharmacology 38: 563–573. doi: 10.1038/npp.2012.233 23212455

10. Shortall SE, Spicer CH, Ebling FJ, Green AR, Fone KC, King MV (2016) Contribution of serotonin and dopamine to changes in core body temperature and locomotor activity in rats following repeated administration of mephedrone. Addict Biol 21: 1127–1139. doi: 10.1111/adb.12283 26180025

11. Marusich JA, Grant KR, Blough BE, Wiley JL (2012) Effects of synthetic cathinones contained in "bath salts" on motor behavior and a functional observational battery in mice. Neurotoxicology 33: 1305–1313. doi: 10.1016/j.neuro.2012.08.003 22922498

12. Gatch MB, Dolan SB, Forster MJ (2017) Locomotor activity and discriminative stimulus effects of a novel series of synthetic cathinone analogs in mice and rats. Psychopharmacology (Berl) 234: 1237–1245. doi: 10.1007/s00213-017-4562-4 28210779

13. Harvey EL, Baker LE (2016) Differential effects of 3,4-methylenedioxypyrovalerone (MDPV) and 4-methylmethcathinone (mephedrone) in rats trained to discriminate MDMA or a d-amphetamine + MDMA mixture. Psychopharmacology (Berl) 233: 673–680. doi: 10.1007/s00213-015-4142-4 26558618

14. Motbey CP, Clemens KJ, Apetz N, Winstock AR, Ramsey J, Li KM, et al. (2013) High levels of intravenous mephedrone (4-methylmethcathinone) self-administration in rats: Neural consequences and comparison with methamphetamine. J Psychopharmacol (Oxf) 27: 823–836. doi: 10.1177/0269881113490325 23739178

15. Karlsson L, Andersson M, Kronstrand R, Kugelberg FC (2014) Mephedrone, methylone and 3,4-methylenedioxypyrovalerone (MDPV) induce conditioned place preference in mice. Basic Clin Pharmacol Toxicol 115: 411–416. doi: 10.1111/bcpt.12253 24739011

16. Bonano JS, Glennon RA, De Felice LJ, Banks ML, Negus SS (2014) Abuse-related and abuse-limiting effects of methcathinone and the synthetic "bath salts" cathinone analogs methylenedioxypyrovalerone (MDPV), methylone and mephedrone on intracranial self-stimulation in rats. Psychopharmacology (Berl) 231: 199–207. doi: 10.1007/s00213-013-3223-5 23949206

17. Sender R, Fuchs S, Milo R (2016) Revised estimates for the number of human and bacteria cells in the body. PLoS Biol 14: 1–14. doi: 10.1371/journal.pbio.1002533 27541692

18. Savage DC (1977) Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol 31: 107–133. doi: 10.1146/annurev.mi.31.100177.000543 334036

19. Hamady M, Knight R (2009) Microbial community profiling for human microbiome projects: Tools, techniques, and challenges. Genome Res 19: 1141–1152. doi: 10.1101/gr.085464.108 19383763

20. Shreiner AB, Kao JY, Young VB (2015) The gut microbiome in health and in disease. Curr Opin Gastroenterol 31: 69–75. doi: 10.1097/MOG.0000000000000139 25394236

21. Pflughoeft KJ, Versalovic J (2012) Human microbiome in health and disease. Annu Rev Pathol 7: 99–122. doi: 10.1146/annurev-pathol-011811-132421 21910623

22. Tremlett H, Bauer KC, Appel-Cresswell S, Finlay BB, Waubant E (2017) The gut microbiome in human neurological disease: A review. Ann Neurol 81: 369–382. doi: 10.1002/ana.24901 28220542

23. Foster JA, McVey Neufeld KA (2013) Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci 36: 305–312. doi: 10.1016/j.tins.2013.01.005 23384445

24. Dinan TG, Cryan JF (2017) The microbiome-gut-brain axis in health and disease. Gastroenterol Clin North Am 46: 77–89. doi: 10.1016/j.gtc.2016.09.007 28164854

25. Barr T, Sureshchandra S, Ruegger P, Zhang J, Ma W, Borneman J, et al. (2018) Concurrent gut transcriptome and microbiota profiling following chronic ethanol consumption in nonhuman primates. Gut Microbes 9: 338–356. doi: 10.1080/19490976.2018.1441663 29517944

26. Xiao HW, Ge C, Feng GX, Li Y, Luo D, Dong JL, et al. (2018) Gut microbiota modulates alcohol withdrawal-induced anxiety in mice. Toxicol Lett 287: 23–30. doi: 10.1016/j.toxlet.2018.01.021 29391279

27. Lee K, Vuong HE, Nusbaum DJ, Hsiao EY, Evans CJ, Taylor AMW (2018) The gut microbiota mediates reward and sensory responses associated with regimen-selective morphine dependence. Neuropsychopharmacology 43: 2606–2614. doi: 10.1038/s41386-018-0211-9 30258112

28. Zhang L, Meng J, Ban Y, Jalodia R, Chupikova I, Fernandez I, et al. (2019) Morphine tolerance is attenuated in germfree mice and reversed by probiotics, implicating the role of gut microbiome. Proc Natl Acad Sci U S A 116: 13523–13532. doi: 10.1073/pnas.1901182116 31209039

29. Chi L, Mahbub R, Gao B, Bian X, Tu P, Ru H, et al. (2017) Nicotine alters the gut microbiome and metabolites of gut-brain interactions in a sex-specific manner. Chem Res Toxicol 30: 2110–2119. doi: 10.1021/acs.chemrestox.7b00162 29035044

30. Kiraly DD, Walker DM, Calipari ES, Labonte B, Issler O, Pena CJ, et al. (2016) Alterations of the host microbiome affect behavioral responses to cocaine. Sci Rep 6: 35455. doi: 10.1038/srep35455 27752130

31. Ning T, Gong X, Xie L, Ma B (2017) Gut microbiota analysis in rats with methamphetamine-induced conditioned place preference. Front Microbiol 8: 1–9.

32. Xu Y, Xie Z, Wang H, Shen Z, Guo Y, Gao Y, et al. (2017) Bacterial diversity of intestinal microbiota in patients with substance use disorders revealed by 16S rRNA gene deep sequencing. Sci Rep 7: 3628. doi: 10.1038/s41598-017-03706-9 28620208

33. Eshleman AJ, Wolfrum KM, Reed JF, Kim SO, Swanson T, Johnson RA, et al. (2017) Structure-activity relationships of substituted cathinones, with transporter binding, uptake, and release. J Pharmacol Exp Ther 360: 33–47. doi: 10.1124/jpet.116.236349 27799294

34. Li Z, Caron MG, Blakely RD, Margolis KG, Gershon MD (2010) Dependence of serotonergic and other nonadrenergic enteric neurons on norepinephrine transporter expression. J Neurosci 30: 16730–16740. doi: 10.1523/JNEUROSCI.2276-10.2010 21148012

35. Li ZS, Pham TD, Tamir H, Chen JJ, Gershon MD (2004) Enteric dopaminergic neurons: definition, developmental lineage, and effects of extrinsic denervation. J Neurosci 24: 1330–1339. doi: 10.1523/JNEUROSCI.3982-03.2004 14960604

36. Wade PR, Chen J, Jaffe B, Kassem IS, Blakely RD, Gershon MD (1996) Localization and function of a 5-HT transporter in crypt epithelia of the gastrointestinal tract. J Neurosci 16: 2352–2364. doi: 10.1523/JNEUROSCI.16-07-02352.1996 8601815

37. El Aidy S, Ramsteijn AS, Dini-Andreote F, van Eijk R, Houwing DJ, Salles JF, et al. (2017) Serotonin transporter genotype modulates the gut microbiota composition in young rats, an effect augmented by early life stress. Front Cell Neurosci 11: 1–12.

38. Greig CJ, Gandotra N, Tackett JJ, Bamdad MC, Cowles RA (2016) Enhanced serotonin signaling increases intestinal neuroplasticity. J Surg Res 206: 151–158. doi: 10.1016/j.jss.2016.07.021 27916355

39. Tackett JJ, Gandotra N, Bamdad MC, Muise ED, Cowles RA (2017) Enhanced serotonin signaling stimulates ordered intestinal mucosal growth. J Surg Res 208: 198–203. doi: 10.1016/j.jss.2016.09.036 27993209

40. Mobley HL, Hausinger RP (1989) Microbial ureases: significance, regulation, and molecular characterization. Microbiol Rev 53: 85–108. 2651866

41. Attarzadeh-Yazdi G, Arezoomandan R, Haghparast A (2014) Minocycline, an antibiotic with inhibitory effect on microglial activation, attenuates the maintenance and reinstatement of methamphetamine-seeking behavior in rat. Prog Neuropsychopharmacol Biol Psychiatry 53: 142–148. doi: 10.1016/j.pnpbp.2014.04.008 24768984

42. Abulseoud OA, Miller JD, Wu J, Choi DS, Holschneider DP (2012) Ceftriaxone upregulates the glutamate transporter in medial prefrontal cortex and blocks reinstatement of methamphetamine seeking in a condition place preference paradigm. Brain Res 1456: 14–21. doi: 10.1016/j.brainres.2012.03.045 22521042

43. Gregg RA, Hicks C, Nayak SU, Tallarida CS, Nucero P, Smith GR, et al. (2016) Synthetic cathinone MDPV downregulates glutamate transporter subtype I (GLT-1) and produces rewarding and locomotor-activating effects that are reduced by a GLT-1 activator. Neuropharmacology 108: 111–119. doi: 10.1016/j.neuropharm.2016.04.014 27085607

44. Angoa-Perez M, Kane MJ, Briggs DI, Francescutti DM, Sykes CE, Shah MM, et al. (2013) Mephedrone does not damage dopamine nerve endings of the striatum, but enhances the neurotoxicity of methamphetamine, amphetamine, and MDMA. J Neurochem 125: 102–110. doi: 10.1111/jnc.12114 23205838

45. Angoa-Perez M, Kane MJ, Francescutti DM, Sykes KE, Shah MM, Mohammed AM, et al. (2012) Mephedrone, an abused psychoactive component of ‘bath salts’ and methamphetamine congener, does not cause neurotoxicity to dopamine nerve endings of the striatum. J Neurochem 120: 1097–1107. doi: 10.1111/j.1471-4159.2011.07632.x 22191803

46. Anneken JH, Angoa-Perez M, Kuhn DM (2015) 3,4-Methylenedioxypyrovalerone (MDPV) prevents while methylone enhances methamphetamine-induced damage to dopamine nerve endings: beta-ketoamphetamine modulation of neurotoxicity by the dopamine transporter. J Neurochem 133: 211–222. doi: 10.1111/jnc.13048 25626880

47. Anneken JH, Angoa-Perez M, Sati GC, Crich D, Kuhn DM (2017) Dissecting the influence of two structural substituents on the differential neurotoxic effects of acute methamphetamine and mephedrone treatment on dopamine nerve endings with the use of 4-methylmethamphetamine and methcathinone. J Pharmacol Exp Ther 360: 417–423. doi: 10.1124/jpet.116.237768 28039330

48. Albers DS, Sonsalla PK (1995) Methamphetamine-induced hyperthermia and dopaminergic neurotoxicity in mice: pharmacological profile of protective and nonprotective agents. J Pharmacol Exp Ther 275: 1104–1114. 8531070

49. Thomas DM, Walker PD, Benjamins JA, Geddes TJ, Kuhn DM (2004) Methamphetamine neurotoxicity in dopamine nerve endings of the striatum is associated with microglial activation. J Pharmacol Exp Ther 311: 1–7. doi: 10.1124/jpet.104.070961 15163680

50. Stephans SE, Yamamoto BY (1995) Effect of repeated methamphetamine administrations on dopamine and glutamate efflux in rat prefrontal cortex. Brain Res 700: 99–106. doi: 10.1016/0006-8993(95)00938-m 8624733

51. Gygi MP, Gygi SP, Johnson M, Wilkins DG, Gibb JW, Hanson GR (1996) Mechanisms for tolerance to methamphetamine effects. Neuropharmacology 35: 751–757. doi: 10.1016/0028-3908(96)84647-8 8887984

52. Sparago M, Wlos J, Yuan J, Hatzidimitriou G, Tolliver J, Dal Cason TA, et al. (1996) Neurotoxic and pharmacologic studies on enantiomers of the N-methylated analog of cathinone (methcathinone): a new drug of abuse. J Pharmacol Exp Ther 279: 1043–1052. 8930215

53. Gygi MP, Fleckenstein AE, Gibb JW, Hanson GR (1997) Role of endogenous dopamine in the neurochemical deficits induced by methcathinone. J Pharmacol Exp Ther 283: 1350–1355. 9400010

54. Gygi MP, Gibb JW, Hanson GR (1996) Methcathinone: an initial study of its effects on monoaminergic systems. J Pharmacol Exp Ther 276: 1066–1072. 8786536

55. Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD (2013) Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Microbiol 79: 5112–5120. doi: 10.1128/AEM.01043-13 23793624

56. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, et al. (2009) Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol 75: 7537–7541. doi: 10.1128/AEM.01541-09 19801464

57. Hammer O, Harper DAT, Ryan PD (2001) PAST: Paleontological statistics software package for education and data analysis. Palaeontologia Electonica 4: 1–9.

58. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. (2011) Metagenomic biomarker discovery and explanation. Genome Biol 12: 1–18. doi: 10.1186/gb-2011-12-6-r60 21702898

59. Chong J, Soufan O, Li C, Caraus I, Li S, Bourque G, et al. (2018) MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis. Nucleic Acids Res 46: W486–W494. doi: 10.1093/nar/gky310 29762782

60. Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI (2005) Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A 102: 11070–11075. doi: 10.1073/pnas.0504978102 16033867

61. Scorza C, Piccini C, Martinez Busi M, Abin Carriquiry JA, Zunino P (2019) Alterations in the Gut Microbiota of Rats Chronically Exposed to Volatilized Cocaine and Its Active Adulterants Caffeine and Phenacetin. Neurotox Res 35: 111–121. doi: 10.1007/s12640-018-9936-9 30066173

62. Hagiwara H, Iyo M, Hashimoto K (2009) Mithramycin protects against dopaminergic neurotoxicity in the mouse brain after administration of methamphetamine. Brain Res 1301: 189–196. doi: 10.1016/j.brainres.2009.09.010 19748494

63. Althobaiti YS, Almalki AH, Das SC, Alshehri FS, Sari Y (2016) Effects of repeated high-dose methamphetamine and ceftriaxone post-treatments on tissue content of dopamine and serotonin as well as glutamate and glutamine. Neurosci Lett 634: 25–31. doi: 10.1016/j.neulet.2016.09.058 27702628

64. Hashimoto K, Tsukada H, Nishiyama S, Fukumoto D, Kakiuchi T, Iyo M (2007) Protective effects of minocycline on the reduction of dopamine transporters in the striatum after administration of methamphetamine: a positron emission tomography study in conscious monkeys. Biol Psychiatry 61: 577–581. doi: 10.1016/j.biopsych.2006.03.019 16712806

65. Choi JG, Huh E, Kim N, Kim DH, Oh MS (2019) High-throughput 16S rRNA gene sequencing reveals that 6-hydroxydopamine affects gut microbial environment. PLoS One 14: e0217194. doi: 10.1371/journal.pone.0217194 31404072

66. Lai F, Jiang R, Xie W, Liu X, Tang Y, Xiao H, et al. (2018) Intestinal pathology and gut microbiota alterations in a methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson’s disease. Neurochem Res 43: 1986–1999. doi: 10.1007/s11064-018-2620-x 30171422

67. Choi JG, Kim N, Ju IG, Eo H, Lim SM, Jang SE, et al. (2018) Oral administration of Proteus mirabilis damages dopaminergic neurons and motor functions in mice. Sci Rep 8: 1275. doi: 10.1038/s41598-018-19646-x 29352191

68. Ridge EA, Pachhain S, Choudhury SR, Bodnar SR, Larsen RA, Phuntumart V, et al. (2019) The influence of the host microbiome on 3,4-methylenedioxymethamphetamine (MDMA)-induced hyperthermia and vice versa. Sci Rep 9: 4313. doi: 10.1038/s41598-019-40803-3 30867489

69. Li B, Li L, Li M, Lam SM, Wang G, Wu Y, et al. (2019) Microbiota Depletion Impairs Thermogenesis of Brown Adipose Tissue and Browning of White Adipose Tissue. Cell Rep 26: 2720–2737.e2725. doi: 10.1016/j.celrep.2019.02.015 30840893

70. Jones BD, Mobley HL (1988) Proteus mirabilis urease: genetic organization, regulation, and expression of structural genes. J Bacteriol 170: 3342–3349. doi: 10.1128/jb.170.8.3342-3349.1988 2841283

71. Halpin LE, Northrop NA, Yamamoto BK (2014) Ammonia mediates methamphetamine-induced increases in glutamate and excitotoxicity. Neuropsychopharmacology 39: 1031–1038. doi: 10.1038/npp.2013.306 24165886

72. Halpin LE, Yamamoto BK (2012) Peripheral ammonia as a mediator of methamphetamine neurotoxicity. J Neurosci 32: 13155–13163. doi: 10.1523/JNEUROSCI.2530-12.2012 22993432

73. Herr RD, Caravati EM (1991) Acute transient ischemic colitis after oral methamphetamine ingestion. Am J Emerg Med 9: 406–409.

74. Brannan TA, Soundararajan S, Houghton BL (2004) Methamphetamine-associated shock with intestinal infarction. MedGenMed 6: 6.

75. Carlson TL, Plackett TP, Gagliano RA Jr., Smith RR (2012) Methamphetamine-induced paralytic ileus. Hawaii J Med Public Health 71: 44–45. 22454809

76. Persons AL, Bradaric BD, Dodiya HB, Ohene-Nyako M, Forsyth CB, Keshavarzian A, et al. (2018) Colon dysregulation in methamphetamine self-administering HIV-1 transgenic rats. PLoS One 13: e0190078. doi: 10.1371/journal.pone.0190078 29293553

77. Flack A, Persons AL, Kousik SM, Celeste Napier T, Moszczynska A (2017) Self-administration of methamphetamine alters gut biomarkers of toxicity. Eur J Neurosci 46: 1918–1932. doi: 10.1111/ejn.13630 28661099

78. Baker JH, Mitchell KS, Neale MC, Kendler KS (2010) Eating disorder symptomatology and substance use disorders: prevalence and shared risk in a population based twin sample. Int J Eat Disord 43: 648–658. doi: 10.1002/eat.20856 20734312

79. Gregorowski C, Seedat S, Jordaan GP (2013) A clinical approach to the assessment and management of co-morbid eating disorders and substance use disorders. BMC Psychiatry 13: 289. doi: 10.1186/1471-244X-13-289 24200300

80. Li H, He J, Jia W (2016) The influence of gut microbiota on drug metabolism and toxicity. Expert Opin Drug Metab Toxicol 12: 31–40. doi: 10.1517/17425255.2016.1121234 26569070

81. Caldwell J, Hawksworth GM (1973) The demethylation of methamphetamine by intestinal microflora. J Pharm Pharmacol 25: 422–424. doi: 10.1111/j.2042-7158.1973.tb10043.x 4146404

82. Simon SL, Richardson K, Dacey J, Glynn S, Domier CP, Rawson RA, et al. (2002) A comparison of patterns of methamphetamine and cocaine use. J Addict Dis 21: 35–44.

83. Rawson R, Huber A, Brethen P, Obert J, Gulati V, Shoptaw S, et al. (2000) Methamphetamine and cocaine users: differences in characteristics and treatment retention. J Psychoactive Drugs 32: 233–238. doi: 10.1080/02791072.2000.10400234 10908013

84. Winstock AR, Mitcheson LR, Deluca P, Davey Z, Corazza O, Schifano F (2011) Mephedrone, new kid for the chop? Addiction 106: 154–161. doi: 10.1111/j.1360-0443.2010.03130.x 20735367


Článek vyšel v časopise

PLOS One


2020 Číslo 1
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#