Gut colonization by proteobacteria modulates mice’s neurobehavioral response to cocaine

NewsGuard 100/100 Score

A recent study published in Cell Host & Microbe revealed that gut colonization by proteobacteria modulates cocaine neurobehavioral response in mice.

Study: Gut colonization by Proteobacteria alters host metabolism and modulates cocaine neurobehavioral responses. Image Credit: Rost9/Shutterstock
Study: Gut colonization by Proteobacteria alters host metabolism and modulates cocaine neurobehavioral responses. Image Credit: Rost9/Shutterstock

Background

The human microbiota is composed of bacteria of numerous phyla, such as Firmicutes, Proteobacteria, and Bacteroides. Distinct microbiota compositions impact host physiology, homeostasis, metabolic profile, and disease vulnerability. Likewise, host environmental and genetic factors shape the establishment of microbiota enterotypes. This suggests strong bidirectional crosstalk between the microbiome and the host.

Recent studies have demonstrated that some gut microbes can modulate different host behaviors. The altered gut microbiota composition has adverse neurologic implications in humans and animals. However, it remains unknown whether the observed changes in gut microbiota cause, promote, or enhance disease or are consequences of an unrelated pathology.

Similar scenarios are noted in the role of gut microbiota on substance use disorders (SUDs) and addiction. Psychostimulants cause changes in gut microbiota composition, and microbiota depletion affects cocaine responses. However, the underlying molecular mechanisms of these interactions are less explicit.

The study and findings

In the present study, researchers investigated whether and how cocaine affects intestinal microbial colonization in mice. First, mice were infected with Citrobacter rodentium and treated with cocaine or saline, which showed an increased pathogen burden in cecum tissues in mice treated with cocaine. Cocaine treatment also elevated intestinal disease markers in response to infection.

Further investigation indicated that cocaine exposure was the pro-virulence mediator for Citrobacter rodentium by increasing the levels of norepinephrine in cecal tissues, which activated the bacterial adrenergic receptor (QseC) signaling and promoted the subsequent expression of the locus of the enterocyte effacement (LEE) and pathogen colonization.

Next, the researchers assessed if C. rodentium infection altered behavioral plastic changes observed after repeated/multiple exposure(s) to cocaine. The authors noted that infected mice significantly increased locomotor response to repeated cocaine exposure compared to mock-infected mice. This effect was not observed in infected mice with saline administration. Metagenomic analysis revealed significant changes in the gut microbiota of mice infected with C. rodentium.

Infected mice showed a marked increase in γ-proteobacteria, primarily due to C. rodentium, whereas non-infected animals were free from γ-proteobacteria. The authors confirmed that this bloom in γ-proteobacteria driven by the colonization of C. rodentium was responsible for the alterations in cocaine-induced behavioral plasticity rather than the inflammation triggered by the pathogen.

Next, the research team tested if changes in behavioral responses to cocaine were related to metabolite alterations in mice infected with the avirulent C. rodentium escN mutant or Escherichia coli HS. Liquid chromatography-mass spectrometry was performed for untargeted comparative metabolomics of cerebrospinal fluid (CSF) from antibiotic-treated mice reconstituted with C. rodentium escN mutant, E. coli HS, or fecal microbiota transplant (FMT) and treated with cocaine.

Integrated pathway analysis of CSF metabolites between mice reconstituted with FMT and those reconstituted with γ-proteobacteria revealed several affected pathways; the glycine, serine, and threonine metabolism were one of the most significantly altered. Seven of the 11 hits identified in the pathway analysis were downregulated in mice reconstituted with γ-proteobacteria.

Moreover, glycine levels were significantly reduced in the CSF of mice reconstituted with E. coli HS or C. rodentium escN mutant relative to those reconstituted with FMT. Besides, CSF glycine levels were negatively correlated with cocaine-induced behavioral responses. Glycine levels were decreased in the cecal tissues of mice reconstituted with γ-proteobacteria compared with FMT.

Furthermore, genome-wide transcriptional profiling was performed in a brain area related to cocaine-elicited synaptic plasticity, the nucleus accumbens (NAcc). The transcriptomic analysis of NAcc of the FMT- and γ-proteobacteria-reconstituted mice revealed multiple differentially expressed genes (DEGs) after cocaine exposure. There were substantial alterations in pathways involved in glutamatergic synapse and those regulating dopaminergic synapse between FMT- and γ-proteobacteria-reconstituted mice.

Glycine transport uptake is mediated by a permease encoded by the cycA gene. Therefore, the researchers reasoned if cycA deletion may decrease bacteria-induced glycine depletion. Strikingly, the cocaine-induced locomotor behavioral response was no longer detected in mice reconstituted with E. coli HS with a cycA deletion.

Finally, using a conditioned place preference (CPP), the authors demonstrated that antibiotic-treated mice reconstituted with FMT lacked drug-seeking behavior to cocaine. In contrast, significant drug-seeking behavior was observed in mice infected with E. coli HS or C. rodentium escN mutant. Furthermore, mice infected with E. coli HS cycA mutant that cannot uptake glycine did not exhibit significant drug-seeking behavior.

Conclusions

The findings suggest that γ-proteobacteria sense cocaine-induced alterations in host catecholamine levels. In addition, they also indicate that such a microbiota compositional shift toward γ-proteobacteria could deplete host metabolites from the CSF and gut, thereby influencing cocaine-elicited NAcc plasticity and addiction-like behavior. γ-proteobacteria colonization of the gut caused a significant decrease in glycine levels in the blood, CSF, and the gut leading to an exacerbated response to cocaine.

Journal reference:
Tarun Sai Lomte

Written by

Tarun Sai Lomte

Tarun is a writer based in Hyderabad, India. He has a Master’s degree in Biotechnology from the University of Hyderabad and is enthusiastic about scientific research. He enjoys reading research papers and literature reviews and is passionate about writing.

Citations

Please use one of the following formats to cite this article in your essay, paper or report:

  • APA

    Sai Lomte, Tarun. (2022, November 03). Gut colonization by proteobacteria modulates mice’s neurobehavioral response to cocaine. News-Medical. Retrieved on April 24, 2024 from https://www.news-medical.net/news/20221103/Gut-colonization-by-proteobacteria-modulates-micee28099s-neurobehavioral-response-to-cocaine.aspx.

  • MLA

    Sai Lomte, Tarun. "Gut colonization by proteobacteria modulates mice’s neurobehavioral response to cocaine". News-Medical. 24 April 2024. <https://www.news-medical.net/news/20221103/Gut-colonization-by-proteobacteria-modulates-micee28099s-neurobehavioral-response-to-cocaine.aspx>.

  • Chicago

    Sai Lomte, Tarun. "Gut colonization by proteobacteria modulates mice’s neurobehavioral response to cocaine". News-Medical. https://www.news-medical.net/news/20221103/Gut-colonization-by-proteobacteria-modulates-micee28099s-neurobehavioral-response-to-cocaine.aspx. (accessed April 24, 2024).

  • Harvard

    Sai Lomte, Tarun. 2022. Gut colonization by proteobacteria modulates mice’s neurobehavioral response to cocaine. News-Medical, viewed 24 April 2024, https://www.news-medical.net/news/20221103/Gut-colonization-by-proteobacteria-modulates-micee28099s-neurobehavioral-response-to-cocaine.aspx.

Comments

The opinions expressed here are the views of the writer and do not necessarily reflect the views and opinions of News Medical.
Post a new comment
Post

While we only use edited and approved content for Azthena answers, it may on occasions provide incorrect responses. Please confirm any data provided with the related suppliers or authors. We do not provide medical advice, if you search for medical information you must always consult a medical professional before acting on any information provided.

Your questions, but not your email details will be shared with OpenAI and retained for 30 days in accordance with their privacy principles.

Please do not ask questions that use sensitive or confidential information.

Read the full Terms & Conditions.

You might also like...
Study unlocks genetic secrets in APOEε4 carriers that could defend against Alzheimer's