Skip to main content
Log in

Characterization of Gut Microbiota in Rats and Rhesus Monkeys After Methamphetamine Self-administration

  • Research
  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Methamphetamine (MA) is one of the most abused drugs globally, but the mechanism of its addiction remains unclear. Several animal studies have shown that the gut microbiota (GM) influences addictive behaviors, but the pattern of GM changes during addiction in animals of different species remains unclear. The aim of this study was to explore the association between dynamic changes in GM and MA self-administration acquisition among two classical mammals, rhesus monkeys (Macaca mulatta) and rats, MA self-administration models. Male Sprague–Dawley rats and male rhesus monkeys were subjected to classical MA self-administration training, and fecal samples were collected before and after MA self-administration training, respectively. 16S rRNA sequencing was used for GM analyses. We found that GM changes were more pronounced in rats than in rhesus monkeys, as evidenced by more GM taxa producing significant differences before and after MA self-administration training in rats than in monkeys. We also found that the expression of the genus Clostridia_vadinBB60_group significantly decreased after MA self-administration training in both rats and rhesus monkeys. Lactobacillus changes were significantly negatively correlated with total MA uptake in rats (Pearson R =  − 0.666, p = 0.035; Spearman R =  − 0.721, p = 0.023), whereas its change was also highly negatively correlated with total MA uptake in rhesus monkeys (Pearson R =  − 0.882, p = 0.118; Spearman R =  − 1.000, p = 0.083), although this was not significant. These findings suggest that MA causes significant alterations in GM in both rhesus monkeys and rats and that the genus Lactobacillus might be a common therapeutic target for MA uptake prevention across the species.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Data Availability

All sequencing data is available at PRJNA1026554.

References

  1. UNODC (2022) World drug report 2022. United Nations publication, Vienna. https://www.unodc.org/unodc/en/data-and-analysis/world-drug-report-2022.html

  2. Panlilio LV, Goldberg SR (2007) Self-administration of drugs in animals and humans as a model and an investigative tool. Addiction (Abingdon, England) 102:1863–1870. https://doi.org/10.1111/j.1360-0443.2007.02011.x

    Article  PubMed  Google Scholar 

  3. Rudebeck PH, Izquierdo A (2022) Foraging with the frontal cortex: a cross-species evaluation of reward-guided behavior. 47:134-146.https://doi.org/10.1038/s41386-021-01140-0

  4. Striedter GF (2019) Variation across species and levels: implications for model species research. Brain Behav Evol 93:57–69. https://doi.org/10.1159/000499664

    Article  PubMed  Google Scholar 

  5. Fan Y, Pedersen O (2021) Gut microbiota in human metabolic health and disease. 19:55-71.https://doi.org/10.1038/s41579-020-0433-9

  6. Thaiss CA, Zmora N, Levy M, Elinav E (2016) The microbiome and innate immunity. Nature 535:65–74. https://doi.org/10.1038/nature18847

    Article  CAS  PubMed  Google Scholar 

  7. Sorboni SG, Moghaddam HS, Jafarzadeh-Esfehani R, Soleimanpour S (2022) A comprehensive review on the role of the gut microbiome in human neurological disorders. 35:e0033820. https://doi.org/10.1128/cmr.00338-20

  8. Dubinkina VB, Tyakht AV, Odintsova VY et al (2017) Links of gut microbiota composition with alcohol dependence syndrome and alcoholic liver disease. Microbiome 5:141. https://doi.org/10.1186/s40168-017-0359-2

    Article  PubMed  PubMed Central  Google Scholar 

  9. Lee K, Vuong HE, Nusbaum DJ (2018) The gut microbiota mediates reward and sensory responses associated with regimen-selective morphine dependence. 43:2606-2614.https://doi.org/10.1038/s41386-018-0211-9

  10. Salavrakos M, Leclercq S, De Timary P, Dom G (2021) Microbiome and substances of abuse. Prog Neuropsychopharmacol Biol Psychiatry 105:110113. https://doi.org/10.1016/j.pnpbp.2020.110113

    Article  CAS  PubMed  Google Scholar 

  11. Martino C, Zaramela LS (2022) Acetate reprograms gut microbiota during alcohol consumption. 13:4630.https://doi.org/10.1038/s41467-022-31973-2

  12. Thomas KR, Watt J, Wu CMJ et al (2022) Pain and opioid-induced gut microbial dysbiosis. 10. https://doi.org/10.3390/biomedicines10081815

  13. Chivero ET, Ahmad R, Thangaraj A, Periyasamy P (2019) Cocaine induces inflammatory gut milieu by compromising the mucosal barrier integrity and altering the gut microbiota colonization. 9:12187.https://doi.org/10.1038/s41598-019-48428-2

  14. Forouzan S, Hoffman KL, Kosten TA (2021) Methamphetamine exposure and its cessation alter gut microbiota and induce depressive-like behavioral effects on rats. 238:281-292.https://doi.org/10.1007/s00213-020-05681-y

  15. Yang Y, Yu X, Liu X et al (2021) Altered fecal microbiota composition in individuals who abuse methamphetamine. Sci Rep 11:18178. https://doi.org/10.1038/s41598-021-97548-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Deng D, Su H, Song Y et al (2021) Altered fecal microbiota correlated with systemic inflammation in male subjects with methamphetamine use disorder. Front Cell Infect Microbiol 11:783917. https://doi.org/10.3389/fcimb.2021.783917

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ning T, Gong X, Xie L, Ma B (2017) Gut microbiota analysis in rats with methamphetamine-induced conditioned place preference. Front Microbiol 8:1620. https://doi.org/10.3389/fmicb.2017.01620

    Article  PubMed  PubMed Central  Google Scholar 

  18. Chen LJ, Zhi X, Zhang KK et al (2021) Escalating dose-multiple binge methamphetamine treatment elicits neurotoxicity, altering gut microbiota and fecal metabolites in mice. Food Chem Toxicol: Int J Publ Br Ind Biol Res Assoc 148:111946. https://doi.org/10.1016/j.fct.2020.111946

    Article  CAS  Google Scholar 

  19. Wang LB, Xu LL, Chen LJ et al (2022) Methamphetamine induces intestinal injury by altering gut microbiota and promoting inflammation in mice. Toxicol Appl Pharmacol 443:116011. https://doi.org/10.1016/j.taap.2022.116011

    Article  CAS  PubMed  Google Scholar 

  20. Lai S, Wang J, Wang B et al (2022) Alterations in gut microbiota affect behavioral and inflammatory responses to methamphetamine in mice. Psychopharmacology 239:1–16. https://doi.org/10.1007/s00213-022-06154-0

    Article  CAS  PubMed  Google Scholar 

  21. Yang J, Zhang Z, Xie Z et al (2022) Metformin modulates microbiota-derived inosine and ameliorates methamphetamine-induced anxiety and depression-like withdrawal symptoms in mice. Biomed Pharmacother=Biomedecine & Pharmacotherapie 149:112837. https://doi.org/10.1016/j.biopha.2022.112837

    Article  CAS  Google Scholar 

  22. Wu Y, Dong Z, Jiang X et al (2023) Gut microbiota taxon-dependent transformation of microglial M1/M2 phenotypes underlying mechanisms of spatial learning and memory impairment after chronic methamphetamine exposure. 11:e0030223. https://doi.org/10.1128/spectrum.00302-23.

  23. Wang Y, Zhang J (2021) Differential perturbations of gut microbial profiles and co-occurrence networks among phases of methamphetamine-induced conditioned place preference. 99:2860-2873.https://doi.org/10.1002/jnr.24963

  24. Wang Y, Guo Z, Li J et al (2023) Unraveling the differential perturbations of species-level functional profiling of gut microbiota among phases of methamphetamine-induced conditioned place preference. Prog Neuropsychopharmacol Biol Psychiatry 127:110828. https://doi.org/10.1016/j.pnpbp.2023.110828

    Article  CAS  PubMed  Google Scholar 

  25. Yang C, Fu X (2021) Gut dysbiosis associated with the rats’ responses in methamphetamine-induced conditioned place preference. 26:e12975. https://doi.org/10.1111/adb.12975

  26. Zhang W, Zhou J, Su H et al (2023) Repeated methamphetamine exposure decreases plasma brain-derived neurotrophic factor levels in rhesus monkeys. 36:e101127. https://doi.org/10.1136/gpsych-2023-101127

  27. Czachowski CL, Samson HH (1999) Breakpoint determination and ethanol self-administration using an across-session progressive ratio procedure in the rat. Alcohol Clin Exp Res 23:1580–1586

    Article  CAS  PubMed  Google Scholar 

  28. Bolyen E, Rideout JR (2019) Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. 37:852-857.https://doi.org/10.1038/s41587-019-0209-9

  29. Quast C, Pruesse E, Yilmaz P et al (2013) The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res 41:D590-596. https://doi.org/10.1093/nar/gks1219

    Article  CAS  PubMed  Google Scholar 

  30. Leshner AI (1997) Addiction is a brain disease, and it matters. Science (New York, N.Y.) 278:45–47. https://doi.org/10.1126/science.278.5335.45

    Article  CAS  PubMed  Google Scholar 

  31. Socała K, Doboszewska U, Szopa A et al (2021) The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res 172:105840. https://doi.org/10.1016/j.phrs.2021.105840

    Article  CAS  PubMed  Google Scholar 

  32. Tian P, Chen Y, Zhu H et al (2022) Bifidobacterium breve CCFM1025 attenuates major depression disorder via regulating gut microbiome and tryptophan metabolism: a randomized clinical trial. Brain Behav Immun 100:233–241. https://doi.org/10.1016/j.bbi.2021.11.023

    Article  CAS  PubMed  Google Scholar 

  33. Schaub AC, Schneider E, Vazquez-Castellanos JF et al (2022) Clinical, gut microbial and neural effects of a probiotic add-on therapy in depressed patients: a randomized controlled trial. 12:227https://doi.org/10.1038/s41398-022-01977-z

  34. Hegelmaier T, Lebbing M, Duscha A et al (2020) Interventional influence of the intestinal microbiome through dietary intervention and bowel cleansing might improve motor symptoms in Parkinson’s disease. Cells 9. https://doi.org/10.3390/cells9020376.

  35. Ibrahim A, Ali RAR, Manaf MRA et al (2020) Multi-strain probiotics (Hexbio) containing MCP BCMC strains improved constipation and gut motility in Parkinson’s disease: a randomised controlled trial. 15:e0244680. https://doi.org/10.1371/journal.pone.0244680.

  36. Tamtaji OR, Heidari-Soureshjani R, Mirhosseini N et al (2019) Probiotic and selenium co-supplementation, and the effects on clinical, metabolic and genetic status in Alzheimer’s disease: a randomized, double-blind, controlled trial. Clin Nutr (Edinburgh, Scotland) 38:2569–2575. https://doi.org/10.1016/j.clnu.2018.11.034

    Article  CAS  Google Scholar 

  37. Vatsalya V, Feng W, Kong M et al (2023) The beneficial effects of Lactobacillus GG therapy on liver and drinking assessments in patients with moderate alcohol-associated hepatitis. 118:1457–1460. https://doi.org/10.14309/ajg.0000000000002283

  38. Bajaj JS, Gavis EA, Fagan A et al (2021) A randomized clinical trial of fecal microbiota transplant for alcohol use disorder. 73:1688-1700. https://doi.org/10.1002/hep.31496

  39. Arout CA, Waters AJ, MacLean RR et al (2019) Minocycline does not affect experimental pain or addiction-related outcomes in opioid maintained patients. Psychopharmacology 236:2857–2866. https://doi.org/10.1007/s00213-018-5146-7

    Article  CAS  PubMed  Google Scholar 

  40. Molavi N, Rasouli-Azad M (2022) The effects of probiotic supplementation on opioid-related disorder in patients under methadone maintenance treatment programs. 2022:1206914. https://doi.org/10.1155/2022/1206914

  41. Long-Smith C, O’Riordan KJ, Clarke G et al (2020) Microbiota-gut-brain axis: new therapeutic opportunities. Annu Rev Pharmacol Toxicol 60:477–502. https://doi.org/10.1146/annurev-pharmtox-010919-023628

    Article  CAS  PubMed  Google Scholar 

  42. Beam A, Clinger E, Hao L (2021) Effect of diet and dietary components on the composition of the gut microbiota. Nutrients 13.https://doi.org/10.3390/nu13082795

  43. Zmora N, Suez J, Elinav E (2019) You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol 16:35–56. https://doi.org/10.1038/s41575-018-0061-2

    Article  CAS  PubMed  Google Scholar 

  44. Jeynes KD, Gibson EL (2017) The importance of nutrition in aiding recovery from substance use disorders: a review. Drug Alcohol Depend 179:229–239. https://doi.org/10.1016/j.drugalcdep.2017.07.006

    Article  PubMed  Google Scholar 

  45. Lewis MJ (2020) Alcoholism and nutrition: a review of vitamin supplementation and treatment. Curr Opin Clin Nutr Metab Care 23:138–144. https://doi.org/10.1097/mco.0000000000000622

    Article  PubMed  Google Scholar 

  46. Ramamoorthy K, Sabui S, Srinivasan P et al (2021) Effect of chronic alcohol exposure on gut vitamin B7 uptake: involvement of epigenetic mechanisms and effect of alcohol metabolites. Am J Physiol Gastrointest Liver Physiol 321:G123-g133. https://doi.org/10.1152/ajpgi.00144.2021

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hwang IC, Vasquez R, Song JH et al (2023) Alterations in the gut microbiome and its metabolites are associated with the immune response to mucosal immunization with Lactiplantibacillus plantarum-displaying recombinant SARS-CoV-2 spike epitopes in mice. Front Cell Infect Microbiol 13:1242681. https://doi.org/10.3389/fcimb.2023.1242681

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Deng L, Wojciech L, Png CW et al (2022) Experimental colonization with Blastocystis ST4 is associated with protective immune responses and modulation of gut microbiome in a DSS-induced colitis mouse model. 79:245. https://doi.org/10.1007/s00018-022-04271-9

  49. Liu Y, Chen LJ, Li XW et al (2023) Gut microbiota contribute to methamphetamine-induced cardiotoxicity in mouse model. Chem Biol Interact 379:110512. https://doi.org/10.1016/j.cbi.2023.110512

    Article  CAS  PubMed  Google Scholar 

  50. Leclercq S, Matamoros S, Cani PD et al (2014) Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proc Natl Acad Sci USA 111:E4485-4493. https://doi.org/10.1073/pnas.1415174111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Forsyth CB, Farhadi A, Jakate SM et al (2009) Lactobacillus GG treatment ameliorates alcohol-induced intestinal oxidative stress, gut leakiness, and liver injury in a rat model of alcoholic steatohepatitis. Alcohol (Fayetteville, N.Y.) 43:163–172. https://doi.org/10.1016/j.alcohol.2008.12.009

    Article  CAS  PubMed  Google Scholar 

  52. Bruch-Bertani JP, Uribe-Cruz C, Pasqualotto A et al (2020) Hepatoprotective effect of probiotic Lactobacillus rhamnosus GG Through the modulation of gut permeability and inflammasomes in a model of alcoholic liver disease in zebrafish. J Am Coll Nutr 39:163–170. https://doi.org/10.1080/07315724.2019.1627955

    Article  CAS  PubMed  Google Scholar 

  53. Wang Y, Kirpich I, Liu Y et al (2011) Lactobacillus rhamnosus GG treatment potentiates intestinal hypoxia-inducible factor, promotes intestinal integrity and ameliorates alcohol-induced liver injury. Am J Pathol 179:2866–2875. https://doi.org/10.1016/j.ajpath.2011.08.039

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Persons AL, Bradaric BD, Dodiya HB et al (2018) Colon dysregulation in methamphetamine self-administering HIV-1 transgenic rats. PLoS One 13:e0190078. https://doi.org/10.1371/journal.pone.0190078

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by the National Natural Science Foundation of China(82271535), the Science and Technology Innovation Program of Hunan Province (2022RC1008), the Hunan Provincial Natural Science Foundation (2021JJ40894), the Changsha Natural Science Foundation Project (kq2014239), the Scientific Research Project of Hunan Provincial Health Commission (202103090528), the Innovation of Science and Technology 2030-Major Project “platform of non-human primate models” (2021ZD0200900), grants from the Lingang Lab (LG202106-03), the Brain Science and Brain-Like Intelligence Technology (2021ZD0202105), Shanghai “the Hospital Garden Star” Funding for training of young medical professionals (20224Z0017), and the National Natural Science Foundation of China (82171483). The authors report no biomedical financial interests or potential conflicts of interest.

Author information

Authors and Affiliations

Authors

Contributions

All the authors have read and approved the final manuscript. Jia-hui Zhou and Wen-lei Zhang conducted the monkey MA self-administration trials. Huan Li conducted the rat MA self-administration trials. Li He analyzed the data and drafted the manuscript. Jia-hui Zhou, Hai-feng Jiang, and Tie-qiao Liu reviewed and edited the manuscript. Rong-wei Zhai and Xiao-jie Zhang were responsible for the conceptualization, design, and review.

Corresponding authors

Correspondence to Rong-wei Zhai or Xiao-jie Zhang.

Ethics declarations

Ethics Approval

The use and care of rhesus monkeys (Macaca mulatta) complied with the guidelines of the Animal Advisory Committee at the Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences. The ethics application entitled “Cognitive-behavioral and neural circuitry of non-human primate models of schizophrenia, novel gene editing techniques and animal models of brain disorders in non-human primates, cortical developmental mechanisms and non-human primate models of related diseases” (#ION-2017006) was approved by the Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences.

Consent to Participate

Not applicable.

Consent for Publication

Not applicable.

Competing Interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

He, L., Zhou, Jh., Li, H. et al. Characterization of Gut Microbiota in Rats and Rhesus Monkeys After Methamphetamine Self-administration. Mol Neurobiol (2024). https://doi.org/10.1007/s12035-024-04318-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s12035-024-04318-x

Keywords

Navigation