Morphine induces changes in the gut microbiome and metabolome in a morphine dependence model

Fuyuan Wang, Jingjing Meng, Li Zhang, Timothy Johnson, Chi Chen, Sabita Roy, Fuyuan Wang, Jingjing Meng, Li Zhang, Timothy Johnson, Chi Chen, Sabita Roy

Abstract

Opioid analgesics are frequently prescribed in the United States and worldwide. However, serious comorbidities, such as dependence, tolerance, immunosuppression and gastrointestinal disorders limit their long-term use. In the current study, a morphine-murine model was used to investigate the role of the gut microbiome and metabolome as a potential mechanism contributing to the negative consequences associated with opioid use. Results reveal a significant shift in the gut microbiome and metabolome within one day following morphine treatment compared to that observed after placebo. Morphine-induced gut microbial dysbiosis exhibited distinct characteristic signatures, including significant increase in communities associated with pathogenic function, decrease in communities associated with stress tolerance and significant impairment in bile acids and morphine-3-glucuronide/morphine biotransformation in the gut. Moreover, expansion of Enterococcus faecalis was strongly correlated with gut dysbiosis following morphine treatment, and alterations in deoxycholic acid (DCA) and phosphatidylethanolamines (PEs) were associated with opioid-induced metabolomic changes. Collectively, these results indicate that morphine induced distinct alterations in the gut microbiome and metabolome, contributing to negative consequences associated with opioid use. Therapeutics directed at maintaining microbiome homeostasis during opioid use may reduce the comorbidities associated with opioid use for pain management.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Morphine treatment leads to temporal modulation of the gut microbiome in 3 days post-treatment. Beta diversity measures of the gut microbiome following treatment with placebo, morphine, naltrexone, or morphine plus naltrexone. Wild-type mice were implanted with placebo (P), 25 mg morphine (M), 30 mg naltrexone (N), or morphine and naltrexone (MN) pellets subcutaneously (n = 4 in each group). Fecal samples were taken for analysis at the following time points: day 0, day 1, day 2, and day 3 post-treatment. Principal coordinates analysis of samples from day 0 (A), day 1 (B), day 2 (C) and day 3 (D) using the UniFrac metric at the OTU level. In Fig. 1A, F value = 4.766, total degree of freedom (DF) = 27, DF(treatment, between columns) = 2, DF(residual, within columns) = 25. In Fig. 1B, F = 1.789, DF (total) = 27, DF(treatment, between columns) = 2, DF(residual, within columns) = 25. In Fig. 1C, F = 17.25, DF (total) = 27, DF(treatment, between columns) = 2, DF(residual, within columns) = 25. In Fig. 1D, F = 89.39, DF (total) = 27, DF(treatment, between columns) = 2, DF(residual, within columns) = 25. Mantel test is run over these distance classes versus the ecological distance matrix. Parametric p-value (Bonferroni-corrected) < 0.01.
Figure 2
Figure 2
Morphine treatment leads to sustained modulation of the gut microbiome for 6 days post-treatment. Beta diversity measures of the gut microbiome following treatment with placebo, morphine, naltrexone, or morphine plus naltrexone. Wild-type mice were implanted with placebo (P), 25 mg morphine (M), 30 mg naltrexone (N), or morphine and naltrexone (MN) pellets subcutaneously (n = 4 in each group). Fecal samples were taken for analysis at the following time points: day 0, day 4, day 5, and day 6 post-treatment. Principal coordinates analysis of samples from day 0, day 4, day 5, and day 6 post-treatment using the UniFrac metric at the OTU level. F value = 7.304. Total degree of freedom (DF) is 495, including DF of treatment (between columns), which is 35, and DF of residuals (within columns), which is 460. A Mantel test is run over these distance classes versus the ecological distance matrix. Parametric p-value (Bonferroni-corrected) 

Figure 3

Morphine treatment induces distinct changes…

Figure 3

Morphine treatment induces distinct changes in the gut microbiome. Wild-type mice were implanted…

Figure 3
Morphine treatment induces distinct changes in the gut microbiome. Wild-type mice were implanted with placebo or 25 mg morphine pellets subcutaneously. Fecal samples were taken for analysis at day 3 post-treatment. (A) Alpha diversity was assessed using the chao1 index. Morphine treatment (n = 8) results in decreased alpha diversity compared to that of controls (n = 8) measured by using the chao1 index. The OTU tables were rarefied at the cutoff value of 31000 sequences per sample. (B) t-test was conducted on the chao1 index. **Indicates a significant difference, P value = 0.0030. (C) Principal coordinates analysis (PCoA) of samples using the UniFrac metric at the OTU level. (D) UniFrac distance significant tests were performed using QIIME. The tests of significance were performed using a two-sided student’s t-test. *Parametric p-value (Bonferroni-corrected) < 0.05, **Parametric p-value (Bonferroni-corrected) < 0.01. ANOVA test indicated P < 0.0001, F value = 63.29. Total degree of freedom (DF) is 119, including DF of treatment (between columns), which is 2 and DF of residuals (within columns), which is 117. (E) Morphine treatment results in a significant increase in pathogenic bacteria. Multiple hypothesis test with the given threshold (FDR = 0.05) demonstrates that the relative abundance of potential pathogenic bacteria (genus level) increases significantly at day 3 post-treatment with morphine compared to that following placebo treatment. Increased (red color) representative pathogenic bacteria include Flavobacterium, Enterococcus, Fusobacterium, Sutterella, Clostridium.

Figure 4

Enterococcus faecalis is a biomarker…

Figure 4

Enterococcus faecalis is a biomarker of morphine-induced alteration of the gut microbiome. Real-time…
Figure 4
Enterococcus faecalis is a biomarker of morphine-induced alteration of the gut microbiome. Real-time PCR expression profiling of species-specific 16S-rRNA gene in gut microbiota. (A) The expression of species-specific 16S-rRNA gene was profiled in stool samples from mice s.c. implanted with placebo (control) or morphine using a qPCR assay (n = 8 in each group). The heat map was generated by a log transformation of the real-time PCR data presented as ΔCT (CT_species – CT_universal_16SrRNA). Red color indicates increased levels of amplification. (B) E. Faecalis 16S-rRNA gene amplification fold change due to treatments on day 3 post-treatment (n = 4 in each group). (C) E. Faecalis 16S rRNA genes amplification fold change due to treatments in a short-term study (n = 8 in each group).

Figure 5

Enterococcus faecalis infection accelerates morphine…

Figure 5

Enterococcus faecalis infection accelerates morphine induced analgesic tolerance. The mice were administrated with…
Figure 5
Enterococcus faecalis infection accelerates morphine induced analgesic tolerance. The mice were administrated with 5 g/L of streptomycin sulphate in the drinking water for 2 days and switch to normal drinking water for 24 hours before E. faecalis (EF) inoculation by oral gavage. The spectinomycin sulphate selective E. faecalis were diluted up to the concentration of 2 × 1010/mL in phosphate buffered saline (PBS). Each mouse was administered with 200ul spectinomycin solution by oral gavage daily. After 48 hours post gavage, the mice were treated with 250 mg/L spectinomycin sulfate (to prevent overgrowth of pathogenic gram-negative bacteria) in the drinking water during the behavior study. To maintain the population of E. faecalis in the mouse gut, mice were administered the same dose of E. faecalis and the same dose of spectinomycin sulphate by oral gavage daily during the behavior experiment for 8 days. (A) Analgesic effectiveness of morphine was evaluated by mouse reaction to heat. Analgesic tolerance, interpreted as percentage of maximum possible effect (MPE%), was determined by tail flick analgesic test. Mice were intraperitoneally injected with saline or 15 mg/kg morphine twice daily for 8 days with 12 hours interval. Behavioral assessment was performed before and 30 min after saline or morphine administration in the morning. All groups had a minimum of 10 mice/group. For positive controls, a new group of 10 mice treated with Placebo + PBS and a new group of 10 mice treated with Placebo + E. faecalis were administered with 15 mg/kg morphine at each time point to test analgesic reaction of these naive mice to morphine treatment. T-test analyses were used to compare MPE% in each group daily. P value of 0.05 or less was considered significant. (B) Daily nociceptive behavior and morphine antinociception throughout an 8-day chronic morphine schedule (15 mg/kg, intraperitoneally, twice daily): antinociceptive behavior by tail flick. Voltage to the light source was adjusted to achieve baseline latency between 2–3 seconds. The cut-off time is 10 seconds to avoid tissue damage. The mice were put on the tail flick assay for 5 min of habituation everyday for two days before the behavior test. The averages of each two measurements before and after morphine injection were recorded as baseline and response to morphine antinocipective effect at each time point daily during experimental period. The tests of significance were performed using a two-sided student’s t-test. *: morphine compared to morphine + E. faecalis 30 min after morphine injection, #: morphine compared to morphine + E. faecalis baseline. *p < 0.05, **p < 0.01 and ***p < 0.001, #### and ****p < 0.0001.

Figure 6

Metabolomic analysis of fecal matter…

Figure 6

Metabolomic analysis of fecal matter and identification of significant shifts of gut metabolome…

Figure 6
Metabolomic analysis of fecal matter and identification of significant shifts of gut metabolome following morphine treatment. (A) Mice were treated with 25 mg morphine or placebo pellet subcutaneously (n = 8 in each group). Fecal samples were taken for analysis at 3 days post-treatment. Scores scatter plot of the partial least square discriminant analysis (PLS-DA) model of fecal samples from wild-type mice (C57B6/J) with morphine (□) or placebo (Δ) treatment. The t[1] and t[2] values represent scores of each sample in principal components 1 and 2, respectively. (B) Loadings plot of the principal components (n = 8 in each group). Metabolites contributing to the differences in fecal samples from mice following morphine and placebo treatment were labeled, and their chemical identities were confirmed. (C) In a short-term study, fecal samples were taken from mice at the following time points: day 0, day 1, day 2, and day 3 post-morphine. Scores scatter plot of the partial least square discriminant analysis (PLS-DA) model of fecal samples from wild-type mice (C57B6/J) with morphine (□) or placebo (Δ) treatment at day 0, day 1, day 2, and day 3 post-treatment (n = 4 in each group). The t[1] and t[2] values represent scores of each sample in principal components 1 and 2, respectively. (D) Heat map plot of significant associations with morphine treatments and the loading of indicator metabolites in fecal matter from mice at day 0, day 1, day 2, and day 3 post-treatment (n = 4 in each group). All relative abundances are row z-score-normalized for visualization. (E) Metabolomic analysis of fecal samples and measurement of effects of naltrexone on morphine-induced gut metabolomic shifts at day 3 post-treatments. Mice were treated with placebo, 25 mg morphine, 30 mg naltrexone, or morphine + naltrexone pellets subcutaneously. (F) Relative abundance analysis of metabolites reveals naltrexone–an opioid receptor antagonist–reversed the effect of morphine on loading of deoxycholic acid (DCA), a secondary bile acid, and phosphatidylethanolamines (PEs), a class of phospholipids found in biological membranes. The tests of significance were performed using a two-sided student’s t-test (n = 4 in each group at day 3 post-treatments).

Figure 7

The M3G/MS concentration ratio in…

Figure 7

The M3G/MS concentration ratio in serum and fecal matter increases post-morphine treatment. LC-MS…

Figure 7
The M3G/MS concentration ratio in serum and fecal matter increases post-morphine treatment. LC-MS analysis identified MS and M3G concentrations. The M3G/MS concentration ratio increases following morphine treatment in both mouse serum and intestinal matter. Statistical significance tests were performed using a two-sided student’s t test (P 

Figure 8

Model of metabolism and biotransformation…

Figure 8

Model of metabolism and biotransformation of morphine and bile acids. In liver, cholesterol…

Figure 8
Model of metabolism and biotransformation of morphine and bile acids. In liver, cholesterol is transformed to primary bile acids, and morphine is conjugated to M3G. In the gut, intestinal bacteria transform primary bile acids and M3G into secondary bile acids and morphine, respectively. Bile acids and morphine are reabsorbed and recycled through enterohepatic circulation.
All figures (8)
Similar articles
Cited by
References
    1. Docherty MJ, Jones RCW, Wallace MS. Managing pain in inflammatory bowel disease. Gastroenterol. Hepatol. (N. Y). 2011;7:592–601. - PMC - PubMed
    1. Hilburger ME, et al. Morphine induces sepsis in mice. J. Infect. Dis. 1997;176:183–8. doi: 10.1086/514021. - DOI - PubMed
    1. Gomes T, et al. Trends in opioid use and dosing among socio-economically disadvantaged patients. Open Med. 2011;5:e13–22. - PMC - PubMed
    1. Leppert W. Emerging therapies for patients with symptoms of opioid-induced bowel dysfunction. Drug Des. Devel. Ther. 2015;9:2215–2231. doi: 10.2147/DDDT.S32684. - DOI - PMC - PubMed
    1. Buccigrossi V, Nicastro E, Guarino A. Functions of intestinal microflora in children. Curr. Opin. Gastroenterol. 2013;29:31–8. doi: 10.1097/MOG.0b013e32835a3500. - DOI - PubMed
Show all 60 references
Publication types
MeSH terms
LinkOut - more resources
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3
Figure 3
Morphine treatment induces distinct changes in the gut microbiome. Wild-type mice were implanted with placebo or 25 mg morphine pellets subcutaneously. Fecal samples were taken for analysis at day 3 post-treatment. (A) Alpha diversity was assessed using the chao1 index. Morphine treatment (n = 8) results in decreased alpha diversity compared to that of controls (n = 8) measured by using the chao1 index. The OTU tables were rarefied at the cutoff value of 31000 sequences per sample. (B) t-test was conducted on the chao1 index. **Indicates a significant difference, P value = 0.0030. (C) Principal coordinates analysis (PCoA) of samples using the UniFrac metric at the OTU level. (D) UniFrac distance significant tests were performed using QIIME. The tests of significance were performed using a two-sided student’s t-test. *Parametric p-value (Bonferroni-corrected) < 0.05, **Parametric p-value (Bonferroni-corrected) < 0.01. ANOVA test indicated P < 0.0001, F value = 63.29. Total degree of freedom (DF) is 119, including DF of treatment (between columns), which is 2 and DF of residuals (within columns), which is 117. (E) Morphine treatment results in a significant increase in pathogenic bacteria. Multiple hypothesis test with the given threshold (FDR = 0.05) demonstrates that the relative abundance of potential pathogenic bacteria (genus level) increases significantly at day 3 post-treatment with morphine compared to that following placebo treatment. Increased (red color) representative pathogenic bacteria include Flavobacterium, Enterococcus, Fusobacterium, Sutterella, Clostridium.
Figure 4
Figure 4
Enterococcus faecalis is a biomarker of morphine-induced alteration of the gut microbiome. Real-time PCR expression profiling of species-specific 16S-rRNA gene in gut microbiota. (A) The expression of species-specific 16S-rRNA gene was profiled in stool samples from mice s.c. implanted with placebo (control) or morphine using a qPCR assay (n = 8 in each group). The heat map was generated by a log transformation of the real-time PCR data presented as ΔCT (CT_species – CT_universal_16SrRNA). Red color indicates increased levels of amplification. (B) E. Faecalis 16S-rRNA gene amplification fold change due to treatments on day 3 post-treatment (n = 4 in each group). (C) E. Faecalis 16S rRNA genes amplification fold change due to treatments in a short-term study (n = 8 in each group).
Figure 5
Figure 5
Enterococcus faecalis infection accelerates morphine induced analgesic tolerance. The mice were administrated with 5 g/L of streptomycin sulphate in the drinking water for 2 days and switch to normal drinking water for 24 hours before E. faecalis (EF) inoculation by oral gavage. The spectinomycin sulphate selective E. faecalis were diluted up to the concentration of 2 × 1010/mL in phosphate buffered saline (PBS). Each mouse was administered with 200ul spectinomycin solution by oral gavage daily. After 48 hours post gavage, the mice were treated with 250 mg/L spectinomycin sulfate (to prevent overgrowth of pathogenic gram-negative bacteria) in the drinking water during the behavior study. To maintain the population of E. faecalis in the mouse gut, mice were administered the same dose of E. faecalis and the same dose of spectinomycin sulphate by oral gavage daily during the behavior experiment for 8 days. (A) Analgesic effectiveness of morphine was evaluated by mouse reaction to heat. Analgesic tolerance, interpreted as percentage of maximum possible effect (MPE%), was determined by tail flick analgesic test. Mice were intraperitoneally injected with saline or 15 mg/kg morphine twice daily for 8 days with 12 hours interval. Behavioral assessment was performed before and 30 min after saline or morphine administration in the morning. All groups had a minimum of 10 mice/group. For positive controls, a new group of 10 mice treated with Placebo + PBS and a new group of 10 mice treated with Placebo + E. faecalis were administered with 15 mg/kg morphine at each time point to test analgesic reaction of these naive mice to morphine treatment. T-test analyses were used to compare MPE% in each group daily. P value of 0.05 or less was considered significant. (B) Daily nociceptive behavior and morphine antinociception throughout an 8-day chronic morphine schedule (15 mg/kg, intraperitoneally, twice daily): antinociceptive behavior by tail flick. Voltage to the light source was adjusted to achieve baseline latency between 2–3 seconds. The cut-off time is 10 seconds to avoid tissue damage. The mice were put on the tail flick assay for 5 min of habituation everyday for two days before the behavior test. The averages of each two measurements before and after morphine injection were recorded as baseline and response to morphine antinocipective effect at each time point daily during experimental period. The tests of significance were performed using a two-sided student’s t-test. *: morphine compared to morphine + E. faecalis 30 min after morphine injection, #: morphine compared to morphine + E. faecalis baseline. *p < 0.05, **p < 0.01 and ***p < 0.001, #### and ****p < 0.0001.
Figure 6
Figure 6
Metabolomic analysis of fecal matter and identification of significant shifts of gut metabolome following morphine treatment. (A) Mice were treated with 25 mg morphine or placebo pellet subcutaneously (n = 8 in each group). Fecal samples were taken for analysis at 3 days post-treatment. Scores scatter plot of the partial least square discriminant analysis (PLS-DA) model of fecal samples from wild-type mice (C57B6/J) with morphine (□) or placebo (Δ) treatment. The t[1] and t[2] values represent scores of each sample in principal components 1 and 2, respectively. (B) Loadings plot of the principal components (n = 8 in each group). Metabolites contributing to the differences in fecal samples from mice following morphine and placebo treatment were labeled, and their chemical identities were confirmed. (C) In a short-term study, fecal samples were taken from mice at the following time points: day 0, day 1, day 2, and day 3 post-morphine. Scores scatter plot of the partial least square discriminant analysis (PLS-DA) model of fecal samples from wild-type mice (C57B6/J) with morphine (□) or placebo (Δ) treatment at day 0, day 1, day 2, and day 3 post-treatment (n = 4 in each group). The t[1] and t[2] values represent scores of each sample in principal components 1 and 2, respectively. (D) Heat map plot of significant associations with morphine treatments and the loading of indicator metabolites in fecal matter from mice at day 0, day 1, day 2, and day 3 post-treatment (n = 4 in each group). All relative abundances are row z-score-normalized for visualization. (E) Metabolomic analysis of fecal samples and measurement of effects of naltrexone on morphine-induced gut metabolomic shifts at day 3 post-treatments. Mice were treated with placebo, 25 mg morphine, 30 mg naltrexone, or morphine + naltrexone pellets subcutaneously. (F) Relative abundance analysis of metabolites reveals naltrexone–an opioid receptor antagonist–reversed the effect of morphine on loading of deoxycholic acid (DCA), a secondary bile acid, and phosphatidylethanolamines (PEs), a class of phospholipids found in biological membranes. The tests of significance were performed using a two-sided student’s t-test (n = 4 in each group at day 3 post-treatments).
Figure 7
Figure 7
The M3G/MS concentration ratio in serum and fecal matter increases post-morphine treatment. LC-MS analysis identified MS and M3G concentrations. The M3G/MS concentration ratio increases following morphine treatment in both mouse serum and intestinal matter. Statistical significance tests were performed using a two-sided student’s t test (P 

Figure 8

Model of metabolism and biotransformation…

Figure 8

Model of metabolism and biotransformation of morphine and bile acids. In liver, cholesterol…

Figure 8
Model of metabolism and biotransformation of morphine and bile acids. In liver, cholesterol is transformed to primary bile acids, and morphine is conjugated to M3G. In the gut, intestinal bacteria transform primary bile acids and M3G into secondary bile acids and morphine, respectively. Bile acids and morphine are reabsorbed and recycled through enterohepatic circulation.
All figures (8)
Figure 8
Figure 8
Model of metabolism and biotransformation of morphine and bile acids. In liver, cholesterol is transformed to primary bile acids, and morphine is conjugated to M3G. In the gut, intestinal bacteria transform primary bile acids and M3G into secondary bile acids and morphine, respectively. Bile acids and morphine are reabsorbed and recycled through enterohepatic circulation.

References

    1. Docherty MJ, Jones RCW, Wallace MS. Managing pain in inflammatory bowel disease. Gastroenterol. Hepatol. (N. Y). 2011;7:592–601.
    1. Hilburger ME, et al. Morphine induces sepsis in mice. J. Infect. Dis. 1997;176:183–8. doi: 10.1086/514021.
    1. Gomes T, et al. Trends in opioid use and dosing among socio-economically disadvantaged patients. Open Med. 2011;5:e13–22.
    1. Leppert W. Emerging therapies for patients with symptoms of opioid-induced bowel dysfunction. Drug Des. Devel. Ther. 2015;9:2215–2231. doi: 10.2147/DDDT.S32684.
    1. Buccigrossi V, Nicastro E, Guarino A. Functions of intestinal microflora in children. Curr. Opin. Gastroenterol. 2013;29:31–8. doi: 10.1097/MOG.0b013e32835a3500.
    1. Gordon JI. Honor Thy Gut Symbionts Redux. Science. 2012;336:1251–1253. doi: 10.1126/science.1224686.
    1. Wells JM, Rossi O, Meijerink M, van Baarlen P. Epithelial crosstalk at the microbiota-mucosal interface. Proc. Natl. Acad. Sci. USA. 2011;108(Suppl):4607–14. doi: 10.1073/pnas.1000092107.
    1. Stecher B, Hardt W-D. The role of microbiota in infectious disease. Trends Microbiol. 2008;16:107–14. doi: 10.1016/j.tim.2007.12.008.
    1. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489:220–30. doi: 10.1038/nature11550.
    1. Yatsunenko T, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–7.
    1. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Human gut microbes associated with obesity. Nature. 2006;444:1022–1023. doi: 10.1038/4441022a.
    1. Willing BP, et al. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology. 2010;139:1844–1854.e1. doi: 10.1053/j.gastro.2010.08.049.
    1. Chang JY, et al. Decreased diversity of the fecal Microbiome in recurrent Clostridium difficile-associated diarrhea. J. Infect. Dis. 2008;197:435–8. doi: 10.1086/525047.
    1. Meng J, et al. Morphine induces bacterial translocation in mice by compromising intestinal barrier function in a TLR-dependent manner. PLoS One. 2013;8:e54040. doi: 10.1371/journal.pone.0054040.
    1. Meng J, et al. Opioid Exacerbation of Gram-positive sepsis, induced by Gut Microbial Modulation, is Rescued by IL-17A Neutralization. Sci. Rep. 2015;5:10918. doi: 10.1038/srep10918.
    1. Mora AL, et al. Moderate to high use of opioid analgesics are associated with an increased risk of Clostridium difficile infection. Am. J. Med. Sci. 2012;343:277–80. doi: 10.1097/MAJ.0b013e31822f42eb.
    1. Banerjee S, et al. Morphine induced exacerbation of sepsis is mediated by tempering endotoxin tolerance through modulation of miR-146a. Sci. Rep. 2013;3:1977. doi: 10.1038/srep01977.
    1. Banerjee S, et al. Opioid-induced gut microbial disruption and bile dysregulation leads to gut barrier compromise and sustained systemic inflammation. Mucosal Immunol. 2016;9:1418–1428. doi: 10.1038/mi.2016.9.
    1. Nicholson JK, et al. Host-Gut Microbiota Metabolic Interactions. Science. 2012;336:1262–1268. doi: 10.1126/science.1223813.
    1. Lawley TD, Walker AW. Intestinal colonization resistance. Immunology. 2013;138:1–11. doi: 10.1111/j.1365-2567.2012.03616.x.
    1. Buffie CG, et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517:205–8. doi: 10.1038/nature13828.
    1. Venkatesh M, et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity. 2014;41:296–310. doi: 10.1016/j.immuni.2014.06.014.
    1. Heinken A, Thiele I. Systematic prediction of health-relevant human- microbial co-metabolism through a computational framework. Gut Microbes. 2015;6:120–130. doi: 10.1080/19490976.2015.1023494.
    1. Marcobal A, et al. A metabolomic view of how the human gut microbiota impacts the host metabolome using humanized and gnotobiotic mice. ISME J. 2013;7:1933–43. doi: 10.1038/ismej.2013.89.
    1. Clayton TA, Baker D, Lindon JC, Everett JR, Nicholson JK. Pharmacometabonomic identification of a significant host-microbiome metabolic interaction affecting human drug metabolism. Proc. Natl. Acad. Sci. USA. 2009;106:14728–33. doi: 10.1073/pnas.0904489106.
    1. Claus SP, et al. Colonization-Induced Host-Gut Microbial Metabolic Interaction. MBio. 2011;2:1–8. doi: 10.1128/mBio.00271-10.
    1. Pacifici GM, Säwe J, Kager L, Rane A. Morphine glucuronidation in human fetal and adult liver. Eur. J. Clin. Pharmacol. 1982;22:553–558. doi: 10.1007/BF00609630.
    1. Frances B, Gout R, Monsarrat B, Cros J, Zajac JM. Further evidence that morphine-6 beta-glucuronide is a more potent opioid agonist than morphine. J. Pharmacol. Exp. Ther. 1992;262:25–31.
    1. Koster AS, Frankhuijzen-Sierevogel AC, Noordhoek J. Distribution of glucuronidation capacity (1-naphthol and morphine) along the rat intestine. Biochem. Pharmacol. 1985;34:3527–3532. doi: 10.1016/0006-2952(85)90728-2.
    1. Hawksworth G, Drasar BS, Hill MJ. Intestinal bacteria and the hydrolysis of glycosidic bonds. J. Med. Microbiol. 1971;4:451–459. doi: 10.1099/00222615-4-4-451.
    1. Walsh CT, Levine RR. Studies of the enterohepatic circulation of morphine in the rat. J. Pharmacol. Exp. Ther. 1975;195:303–10.
    1. Das S, Kelschenbach J, Charboneau R, Barke RA, Roy S. Morphine Withdrawal Stress Modulates Lipopolysaccharide-induced Interleukin 12p40 (IL-12p40) Expression by Activating Extracellular Signal-regulated Kinase 1/2, Which Is Further Potentiated by Glucocorticoids. J. Biol. Chem. 2011;286:29806–29817. doi: 10.1074/jbc.M111.271460.
    1. Babrowski T, et al. Pseudomonas aeruginosa virulence expression is directly activated by morphine and is capable of causing lethal gut-derived sepsis in mice during chronic morphine administration. Ann. Surg. 2012;255:386–93. doi: 10.1097/SLA.0b013e3182331870.
    1. Pham TAN, Lawley TD. Emerging insights on intestinal dysbiosis during bacterial infections. Curr. Opin. Microbiol. 2014;17:67–74. doi: 10.1016/j.mib.2013.12.002.
    1. McNicol ED, Midbari A, Eisenberg E. Opioids for neuropathic pain. Cochrane database Syst. Rev. 2013;8:CD006146.
    1. Sommer F, Bäckhed F. The gut microbiota — masters of host development and physiology. Nat. Rev. Microbiol. 2013;11:227–238. doi: 10.1038/nrmicro2974.
    1. Ursell LK, et al. The intestinal metabolome: an intersection between microbiota and host. Gastroenterology. 2014;146:1470–6. doi: 10.1053/j.gastro.2014.03.001.
    1. Turnbaugh PJ, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–4. doi: 10.1038/nature07540.
    1. Lötsch J, Geisslinger G. Morphine-6-glucuronide: an analgesic of the future? Clin. Pharmacokinet. 2001;40:485–99. doi: 10.2165/00003088-200140070-00001.
    1. Wang F, Roy S. Gut Homeostasis, Microbial Dysbiosis, and Opioids. Toxicol. Pathol. 2017;45:150–156. doi: 10.1177/0192623316679898.
    1. Ouellet DM, Pollack GM. Biliary excretion and enterohepatic recirculation of morphine-3-glucuronide in rats. Drug Metab. Dispos. 1995;23:478–84.
    1. Wang J, Barke RA, Charboneau R, Roy S. Morphine impairs host innate immune response and increases susceptibility to Streptococcus pneumoniae lung infection. J. Immunol. 2005;174:426–34. doi: 10.4049/jimmunol.174.1.426.
    1. Degenhardt L, et al. Mortality among regular or dependent users of heroin and other opioids: A systematic review and meta-analysis of cohort studies. Addiction. 2011;106:32–51. doi: 10.1111/j.1360-0443.2010.03140.x.
    1. Bäckhed F, et al. Defining a healthy human gut microbiome: current concepts, future directions, and clinical applications. Cell Host Microbe. 2012;12:611–22. doi: 10.1016/j.chom.2012.10.012.
    1. Fite A, et al. Longitudinal analyses of gut mucosal microbiotas in ulcerative colitis in relation to patient age and disease severity and duration. J. Clin. Microbiol. 2013;51:849–56. doi: 10.1128/JCM.02574-12.
    1. Greiner T, Bäckhed F. Effects of the gut microbiota on obesity and glucose homeostasis. Trends Endocrinol. Metab. 2011;22:117–123. doi: 10.1016/j.tem.2011.01.002.
    1. Honda K, Littman DR. The microbiome in infectious disease and inflammation. Annu. Rev. Immunol. 2012;30:759–95. doi: 10.1146/annurev-immunol-020711-074937.
    1. Keller R, et al. The Escherichia coli Envelope Stress Sensor CpxA Responds to Changes in Lipid Bilayer Properties. Biochemistry. 2015;54:3670–6. doi: 10.1021/acs.biochem.5b00242.
    1. Pellock, S. J. & Redinbo, M. R. Glucuronides in the Gut: Sugar-Driven Symbioses Between Microbe and Host. J. Biol. Chem. 10.1074/jbc.R116.767434 (2017).
    1. Stain-Texier F, Sandouk P, Scherrmann JM. Intestinal absorption and stability of morphine 6-glucuronide in different physiological compartments of the rat. Drug Metab. Dispos. 1998;26:383–7.
    1. Atmanene C, et al. Characterization of human and bovine phosphatidylethanolamine-binding protein (PEBP/RKIP) interactions with morphine and morphine-glucuronides determined by noncovalent mass spectrometry. Med. Sci. Monit. 2009;15:BR178–R187.
    1. Goumon Y, et al. Identification of morphine-6-glucuronide in chromaffin cell secretory granules. J. Biol. Chem. 2006;281:8082–8089. doi: 10.1074/jbc.M502298200.
    1. Bryant HU, Yoburn BC, Inturrisi CE, Bernton EW, Holaday JW. Morphine-induced immunomodulation is not related to serum morphine concentrations. Eur. J. Pharmacol. 1988;149:165–9. doi: 10.1016/0014-2999(88)90057-X.
    1. Ross GR, Gabra BH, Dewey WL, Akbarali HI. Morphine tolerance in the mouse ileum and colon. J. Immunol. 2008;327:561–572.
    1. Gohl DM, et al. Systematic improvement of amplicon marker gene methods for increased accuracy in microbiome studies. Nat. Biotechnol. 2016;34:942–9. doi: 10.1038/nbt.3601.
    1. Gohl, D. et al. An optimized protocol for high-throughput amplicon-based microbiome profiling. Protoc. Exch. 10.1038/protex.2016.030 (2016).
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods. 2010;7:335–6. doi: 10.1038/nmeth.f.303.
    1. Weingarden AR, et al. Microbiota transplantation restores normal fecal bile acid composition in recurrent Clostridium difficile infection. Am. J. Physiol. Gastrointest. Liver Physiol. 2014;306:G310–9. doi: 10.1152/ajpgi.00282.2013.
    1. Lahti L, et al. Associations between the human intestinal microbiota, Lactobacillus rhamnosus GG and serum lipids indicated by integrated analysis of high-throughput profiling data. PeerJ. 2013;1:e32. doi: 10.7717/peerj.32.
    1. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat. Protoc. 2008;3:1101–1108. doi: 10.1038/nprot.2008.73.

Source: PubMed

3
Abonneren