Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Glucosylated nanoparticles for the oral delivery of antibiotics to the proximal small intestine protect mice from gut dysbiosis

Abstract

Orally delivered antibiotics can reach the caecum and colon, and induce gut dysbiosis. Here we show that the encapsulation of antibiotics in orally administered positively charged polymeric nanoparticles with a glucosylated surface enhances absorption by the proximal small intestine through specific interactions of glucose and the abundantly expressed sodium-dependent glucose transporter 1. This improves bioavailability of the antibiotics, and limits their exposure to flora in the large intestine and their accumulation in caecal and faecal contents. Compared with the standard administration of the same antibiotics, the oral administration of nanoparticle-encapsulated ampicillin, chloramphenicol or vancomycin in mice with bacterial infections in the lungs effectively eliminated the infections, decreased adverse effects on the intestinal microbiota by protecting the animals from dysbiosis-associated metabolic syndromes and from opportunistic pathogen infections, and reduced the accumulation of known antibiotic-resistance genes in commensal bacteria. Glucosylated nanocarriers may be suitable for the oral delivery of other drugs causing gut dysbiosis.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Glucosylated nanocarriers promote absorption specifically at the proximal small intestine.
Fig. 2: PGNPs reduce intestinal residues and enhance blood circulation.
Fig. 3: PGNPs-Abx effectively eliminates S. pneumoniae infection in lung.
Fig. 4: PGNPs delivery prevents antibiotic-induced dysbiosis.
Fig. 5: The SGLT1 inhibitor eliminates the protective effect of PGNPs-Abx.
Fig. 6: The PGNP-mediated delivery of antibiotics ameliorates dysbiosis-associated obesity.
Fig. 7: The PGNPs-mediated delivery of antibiotics via SGLT1 decreases dysbiosis-associated infection by C. rodentium.
Fig. 8: The PGNP-mediated delivery of antibiotics reduces antibiotic-resistance genes in gut microbiota.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. All sequence data generated in this study are available from the SRA database with accession numbers PRJNA666621 and PRJNA666612. Source data are provided with this paper.

References

  1. Lynch, S. V. & Pedersen, O. The human intestinal microbiome in health and disease. N. Engl. J. Med. 375, 2369–2379 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Levison, M. E. Pharmacokinetics and pharmacodynamics of antibacterial agents. Infect. Dis. Clin. North Am. 23, 791–815 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Leffler, D. A. & Lamont, J. T. Clostridium difficile infection. N. Engl. J. Med. 372, 1539–1548 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Brown, K. A., Khanafer, N., Daneman, N. & Fisman, D. N. Meta-analysis of antibiotics and the risk of community-associated Clostridium difficile infection. Antimicrob. Agents Chemother. 57, 2326–2332 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Baumler, A. J. & Sperandio, V. Interactions between the microbiota and pathogenic bacteria in the gut. Nature 535, 85–93 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kim, Y. G. et al. Gut dysbiosis promotes M2 macrophage polarization and allergic airway inflammation via fungi-induced PGE2. Cell Host Microbe 15, 95–102 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cho, I. et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature 488, 621–626 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cox, L. M. et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 158, 705–721 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sommer, M. O. A., Dantas, G. & Church, G. M. Functional characterization of the antibiotic resistance reservoir in the human microflora. Science 325, 1128–1131 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Salyers, A. A., Gupta, A. & Wang, Y. P. Human intestinal bacteria as reservoirs for antibiotic resistance genes. Trends Microbiol. 12, 412–416 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Stiefel, U. et al. Oral administration of beta-lactamase preserves colonization resistance of piperacillin-treated mice. J. Infect. Dis. 188, 1605–1609 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Léonard, F., Andremont, A., Leclerq, B., Labia, R. & Tancrède, C. Use of beta-lactamase-producing anaerobes to prevent ceftriaxone from degrading intestinal resistance to colonization. J. Infect. Dis. 160, 274–280 (1989).

    Article  PubMed  Google Scholar 

  13. Khoder, M., Tsapis, N., Domergue-Dupont, V., Gueutin, C. & Fattal, E. Removal of residual colonic ciprofloxacin in the rat by activated charcoal entrapped within zinc-pectinate beads. Eur. J. Pharm. Sci. 41, 281–288 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. De Gunzburg, J. et al. Targeted adsorption of molecules in the colon with the novel adsorbent-based medicinal product, dav132: a proof of concept study in healthy subjects. J. Clin. Pharmacol. 55, 10–16 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Zhang, L., Huang, Y., Zhou, Y., Buckley, T. & Wang, H. H. Antibiotic administration routes significantly influence the levels of antibiotic resistance in gut microbiota. Antimicrob. Agents Chemother. 57, 3659–3666 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mowat, A. M. & Agace, W. W. Regional specialization within the intestinal immune system. Nat. Rev. Immunol. 14, 667–685 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Chen, L., Tuo, B. & Dong, H. Regulation of intestinal glucose absorption by ion channels and transporters. Nutrients 8, 43 (2016).

    Article  PubMed Central  CAS  Google Scholar 

  18. Han, X. et al. Zwitterionic micelles efficiently deliver oral insulin without opening tight junctions. Nat. Nanotechnol. 15, 605–614 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cherazard, R. et al. Antimicrobial resistant Streptococcus pneumoniae: prevalence, mechanisms, and clinical implications. Am. J. Ther. 24, 361–369 (2017).

    Article  Google Scholar 

  20. Palleja, A. et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat. Microbiol. 3, 1255–1265 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Suez, J. et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell 174, 1406–1423 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Vangay, P., Ward, T., Gerber, J. S. & Knights, D. Antibiotics, pediatric dysbiosis, and disease. Cell Host Microbe 17, 553–564 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Turnbaugh, P. J. et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031 (2006).

    Article  PubMed  Google Scholar 

  24. Thomas, C., Pellicciari, R., Pruzanski, M., Auwerx, J. & Schoonjans, K. Targeting bile-acid signalling for metabolic diseases. Nat. Rev. Drug. Discov. 7, 678–693 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Zhao, L. The gut microbiota and obesity: from correlation to causality. Nat. Rev. Microbiol. 11, 639–647 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. McGlone, E. R. & Bloom, S. R. Bile acids and the metabolic syndrome. Ann. Clin. Biochem. 56, 326–337 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Wahlström, A. et al. Induction of farnesoid X receptor signaling in germ-free mice colonized with a human microbiota. J. Lipid Res. 58, 412–419 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Wlodarska, M. et al. Antibiotic treatment alters the colonic mucus layer and predisposes the host to exacerbated Citrobacter rodentium-induced colitis. Infect. Immun. 79, 1536–1545 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Buffie, C. G. et al. Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis. Infect. Immun. 80, 62–73 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Peng, Z. et al. Update on antimicrobial resistance in Clostridium difficile: resistance mechanisms and antimicrobial susceptibility testing. J. Clin. Microbiol. 55, 1998–2008 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Li, J. J. et al. Gold nanoparticles cure bacterial infection with benefit to intestinal microflora. ACS Nano 13, 5002–5014 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Van den Brule, S. et al. Dietary silver nanoparticles can disturb the gut microbiota in mice. Part. Fibre Toxicol. 13, 38–54 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Sun, C. Y. et al. Facile generation of tumor-pH-labile linkage-bridged block copolymers for chemotherapeutic delivery. Angew. Chem. Int. Ed. Engl. 55, 1010–1014 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Zhao, J., Pahovnik, D., Gnanou, Y. & Hadjichristidis, N. Sequential polymerization of ethylene oxide, ε-caprolactone and l-lactide: a one-pot metal-free route to tri- and pentablock terpolymers. Polym. Chem. 5, 3750–3753 (2014).

    Article  CAS  Google Scholar 

  35. Ramos, M. et al. Determination of chloramphenicol residues in shrimps by liquid chromatography-mass spectrometry. J. Chromatogr. B 791, 31–38 (2003).

    Article  CAS  Google Scholar 

  36. Gonçalves, T. M. et al. Determination of ampicillin in human plasma by solid-phase extraction-liquid chromatography-tandem mass spectrometry (SPE-LC-MS/MS) and its use in bioequivalence studies. Arzneim. Forsch. 58, 91–96 (2008).

    Google Scholar 

  37. Nalbant, D. et al. Development and validation of a simple and sensitive LC-MS/MS method for quantification of ampicillin and sulbactam in human plasma and its application to a clinical pharmacokinetic study. J. Pharm. Biomed. Anal. 196, 113899 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Schuijt, T. J. et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut 65, 575–583 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Pernigoni, N. et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science 374, 216–224 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Böckelmann, U. et al. Quantitative PCR monitoring of antibiotic resistance genes and bacterial pathogens in three European artificial groundwater recharge systems. Appl. Environ. Microbiol. 75, 154–163 (2009).

    Article  PubMed  CAS  Google Scholar 

  41. Fernando, D. M. et al. Detection of antibiotic resistance genes in source and drinking water samples from a first nations community in Canada. Appl. Environ. Microbiol. 82, 4767–4775 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Smith, K. M. et al. Prolonged partial liquid ventilation using conventional and high-frequency ventilatory techniques: gas exchange and lung pathology in an animal model of respiratory distress syndrome. Crit. Care Med. 25, 1888–1897 (1997).

    Article  CAS  PubMed  Google Scholar 

  43. Kleiner, D. E. et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 41, 1313–1321 (2005).

    Article  PubMed  Google Scholar 

  44. Dieleman, L. A. et al. Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines. Clin. Exp. Immunol. 114, 385–391 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Y. Yang for analysing the metagenomic sequencing results, J. Wang for helping with animal experiments, H. Tan for helping with microbial experiments, Q. Liang, H. Pan and C. Zeng for helping with the material experiments; Z. Tian, R. Zhou, W. Pan, R. Flavell, X. Song, K. Zhang, H. Ma and W. Wen for helpful discussion and comments on this patent-pending work. This work was supported by grants from the National Key R&D Program of China (2017YFA0205600 to Y.W. and 2018YFA0508000 to S.Z.), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDB29030101 to S.Z.), the National Natural Science Foundation of China (52025036 to Y.W., 82061148013 and 81821001 to S.Z. and 51903105 to W.J.) and the Shanghai Municipal Science and Technology Major Project (S.Z.).

Author information

Authors and Affiliations

Authors

Contributions

G.Z. and Q.W. designed, performed and interpreted experiments. W.T. designed and analysed the 16S rRNA and metagenomic sequencing results. W.J. designed and synthesized the polymers. W.J. helped with animal experiments and cellular experiments. E.E. provided analytical tools, critical comments and suggestions; S.Z., Y.W., G.Z. and Q.W. wrote the manuscript. E.E. edited the manuscript. Y.W. and S.Z. supervised the project.

Corresponding authors

Correspondence to Yucai Wang or Shu Zhu.

Ethics declarations

Competing interests

E.E. is a consultant at Daytwo and BiomX in topics unrelated to the subject of this work. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Jian-Dong Jiang, Xian-Zheng Zhang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 A proposed model for how PGNPs for oral delivery of antibiotics resolve antibiotic-associated dysbiosis.

The different segments of the intestine have quite distinct appearances. Distribution of gut microbiota is increasing along the length of the intestine while the expression of glucose transporter SGLT1 along small intestine is decreasing and no detection in large intestine. When oral free antibiotics to cure bacterial infection induced damages to the gut microbiota and related adverse effects in normal mice. But, when oral nanocarrier antibiotics alleviates disruptions to the gut microbiota and keep it homeostasis.

Extended Data Fig. 2 SGLT1 aids the uptake of PGNPs into cells.

a, Representative confocal images showing SGLT1-mediated endocytosis of PGNPs in SGLT1-expressing HEK293T cells. PNPs or PGNPs (red), SGLT1 (green), and nucleus (blue). Scale bars, 5 μm (left panel), 1 μm (middle and right panels). b, Quantification of the percentage of PNPs or PGNPs co-localized with SGLT1 during the internalization in confocal images. n = 3. All values are expressed as mean ± s.e.m. Statistical significance was determined using two-tailed Student’s t-test in b. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. a and b are representative of two independent experiments.

Source data

Extended Data Fig. 3 PGNPs co-localized with recycling endosome.

a, Representative confocal images showing the co-localization of PGNPs-DiD with Rab11 in SGLT1-expressing HEK293T cells. PGNPs-DiD (red), Rab11 (green), nucleus (blue). Scale bars, 10 μm. b, The percentage of co-localized PGNPs-DiD and Rab11 was quantified in (a). n = 18 images from three biologically independent samples. c, Representative confocal images showing the co-localization of PGNPs with Rab11 in proximal small intestine. PGNPs-DiD (purple), Rab11 (green), and nucleus (blue). Scale bars, 10 μm. d, The percentage of co-localized PGNPs-DiD and Rab11 was quantified in (c). n = 9 images from three mice. e, Schematic representation showing that PGNPs can be taken up and transported through SGLT1-mediated endocytosis and transcytosis in intestinal epithelial cells. All values are expressed as mean ± s.e.m. ad are representative of two independent experiments.

Source data

Extended Data Fig. 4 PGNPs-Amp effectively eliminate Listeria monocytogenes in a bacteremia model.

a, Study design: mice were received an intravenous injection with 2 × 106 CFUs of Listeria monocytogenes. Free Amp (40 mg kg−1) or Amp (40 mg kg−1)-loaded PGNPs was orally administered into mice. Control mice did not receive antibiotic treatment. Bacterial loads in blood, liver, spleen were determined at 24 h post infection. b, The appearance of the infected liver and spleen in mice with indicated treatments at 24 h post infection. ce, Quantification of bacteria CFUs in the blood (c), liver (d), and spleen (e) at 24 h post infection. n = 6 mice. All values are expressed as mean ± s.e.m. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons test in (c, d and e). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. be are representative of two independent experiments.

Source data

Extended Data Fig. 5 PGNPs delivery of antibiotics show minimized alteration of the microbiota.

PCoA analysis of microbiota community composition in mice treated with free Abx or Abx-loaded NPs and the recovery trajectory are marked by analyzing unweighted UniFrac distances.

Source data

Extended Data Fig. 6 PGNPs delivery of antibiotics reduces the microbiota alteration in small intestine.

C57BL/6 mice were orally administered with free or ampicillin (20 mg kg−1) and vancomycin (20 mg kg−1)-loaded PGNPs for 5d. Control mice had no antibiotic exposure. Both SI-P and SI-D content were collected for 16 s rRNA sequencing at the end of treatment. a,d, Alpha diversity quantified as observed species in SI-P (a) or SI-D (d) in indicated groups. b,e, Unweighted UniFrac principal-coordinates analysis (PCoA) of samples with indicated treatments in SI-P (b) and SI-D (e). c,f, Relative abundance of family-level taxonomy in fecal microbiota was presented as a percentage of the total detected sequences in SI-P (c) and SI-D (f). n ≥ 3 mice. All values are expressed as mean ± s.e.m. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons test in a and d. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Source data

Extended Data Fig. 7 Microbiota alterations upon Antibiotics (Abx) and HFD treatments.

Three-week-old C57BL/6 J male mice were administrated with free antibiotics or ampicillin (20 mg kg−1) and vancomycin (20 mg kg−1)-loaded nanoparticles for 5 successive days. Control mice had no antibiotics exposure. All mice were treated with HFD from age of 7 weeks. Fecal pellets were collected on the day end of the HFD treatment for 16S rRNA sequencing. a, The observed species number of gut microbiota. b, PCA plot generated from unweighted UniFrac distance matrix displaying the distinct clustering pattern of gut microbiota. c, Relative abundances of the gut commensal microorganisms at the family level. n = 5 mice. All values are expressed as the mean ± s.e.m. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons test in a. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Source data

Extended Data Fig. 8 Fecal bile acids levels after short-time antibiotics treatment following with a high-fat diet exposure.

Bile acids levels altered by free Abx or PGNPs-Abx were analyzed by mass spectrometer. n = 5 mice. All values are expressed as mean ± s.e.m. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. NS, no significance.

Source data

Extended Data Fig. 9 Fecal microbiota transplantation (FMT) from free Abx- but not PGNPs-Abx- treated mice results in metabolic alteration.

Donor mice were given Water, PGNPs, free Abx, or PGNPs-Abx daily for 5 days. Fecal pellets were collected for FMT. Fecal bacteria were transplanted into recipient mice after Abx treatment and followed by HFD feeding to induce obesity. Serum and liver cholesterol levels were measured after 13 weeks HFD. n = 5 mice. All values are expressed as mean ± s.e.m. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. NS, no significance. Data is representative of two independent experiments.

Source data

Extended Data Fig. 10 Quantification of pathological scores of cecum sections from the C. rodentium infected mice.

n = 6 mice. All values are expressed as mean ± s.e.m. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. NS, no significance.

Source data

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, G., Wang, Q., Tao, W. et al. Glucosylated nanoparticles for the oral delivery of antibiotics to the proximal small intestine protect mice from gut dysbiosis. Nat. Biomed. Eng 6, 867–881 (2022). https://doi.org/10.1038/s41551-022-00903-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-022-00903-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing