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:

Artificial-enzymes-armed Bifidobacterium longum probiotics for alleviating intestinal inflammation and microbiota dysbiosis

Abstract

Inflammatory bowel disease can be caused by the dysfunction of the intestinal mucosal barrier and dysregulation of gut microbiota. Traditional treatments use drugs to manage inflammation with possible probiotic therapy as an adjuvant. However, current standard practices often suffer from metabolic instability, limited targeting and result in unsatisfactory therapeutic outcomes. Here we report on artificial-enzyme-modified Bifidobacterium longum probiotics for reshaping a healthy immune system in inflammatory bowel disease. Probiotics can promote the targeting and retention of the biocompatible artificial enzymes to persistently scavenge elevated reactive oxygen species and alleviate inflammatory factors. The reduced inflammation caused by artificial enzymes improves bacterial viability to rapidly reshape the intestinal barrier functions and restore the gut microbiota. The therapeutic effects are demonstrated in murine and canine models and show superior outcomes to traditional clinical drugs.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Characterization of artificial-enzyme-armed probiotics.
Fig. 2: Multiple ROS-scavenging abilities in vitro.
Fig. 3: Stability, targeting and retention of BL@B-SA50 in an inflamed colon.
Fig. 4: Artificial-enzymes-armed BL probiotics ameliorate DSS-induced UC.
Fig. 5: Artificial-enzymes-armed BL probiotics modulating the gut microbiota of UC.
Fig. 6: UC therapy with BL@B-SA50 in beagle dogs.

Similar content being viewed by others

Data availability

The 16S ribosomal RNA gene sequencing data are deposited at NCBI (accession no. PRJNA917220). Source data for Figs. 16; Extended Data Figs. 1 and 2; and Supplementary Figs. 1, 3, 4, 814, 16, 18, 19, 2229, 32, 33 and 37 are provided with this paper.

References

  1. Alatab, S. et al. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol. 5, 17–30 (2020).

    Article  Google Scholar 

  2. Hoivik, M. L. et al. Health-related quality of life in patients with ulcerative colitis after a 10-year disease course: results from the IBSEN study. Inflamm. Bowel Dis. 18, 1540–1549 (2012).

    Article  Google Scholar 

  3. Citi, S. Intestinal barriers protect against disease. Science 359, 1097–1098 (2018).

    Article  CAS  Google Scholar 

  4. Turner, J. R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 9, 799–809 (2009).

    Article  CAS  Google Scholar 

  5. Chu, H. et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science 352, 1116–1120 (2016).

    Article  CAS  Google Scholar 

  6. Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019).

    Article  CAS  Google Scholar 

  7. Plichta, D. R., Graham, D. B., Subramanian, S. & Xavier, R. J. Therapeutic opportunities in inflammatory bowel disease: mechanistic dissection of host-microbiome relationships. Cell 178, 1041–1056 (2019).

    Article  CAS  Google Scholar 

  8. Jostins, L. et al. Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

    Article  CAS  Google Scholar 

  9. Grisham, M. B. Oxidants and free radicals in inflammatory bowel disease. Lancet 344, 859–861 (1994).

    Article  CAS  Google Scholar 

  10. Dickinson, B. C. & Chang, C. J. Chemistry and biology of reactive oxygen species in signaling or stress responses. Nat. Chem. Biol. 7, 504–511 (2011).

    Article  CAS  Google Scholar 

  11. Lee, Y. et al. Hyaluronic acid–bilirubin nanomedicine for targeted modulation of dysregulated intestinal barrier microbiome and immune responses in colitis. Nat. Mater. 19, 118–126 (2020).

    Article  CAS  Google Scholar 

  12. Scott, B. M. et al. Self-tunable engineered yeast probiotics for the treatment of inflammatory bowel disease. Nat. Med. 27, 1212–1222 (2021).

    Article  CAS  Google Scholar 

  13. Bernstein, C. N. et al. World Gastroenterology Organization practice guidelines for the diagnosis and management of IBD in 2010. Inflamm. Bowel Dis. 16, 112–124 (2010).

    Article  Google Scholar 

  14. Lautenschläger, C., Schmidt, C., Fischer, D. & Stallmach, A. Drug delivery strategies in the therapy of inflammatory bowel disease. Adv. Drug Deliv. Rev. 71, 58–76 (2014).

    Article  Google Scholar 

  15. Cader, M. Z. & Kaser, A. Finding the right target for drug-resistant inflammatory bowel disease. Nat. Med. 27, 1870–1871 (2021).

    Article  CAS  Google Scholar 

  16. Suez, J., Zmora, N., Segal, E. & Elinav, E. The pros, cons, and many unknowns of probiotics. Nat. Med. 25, 716–729 (2019).

    Article  CAS  Google Scholar 

  17. Motta, J.-P. et al. Food-grade bacteria expressing elafin protect against inflammation and restore colon homeostasis. Sci. Transl. Med. 4, 158ra144–158ra144 (2012).

    Article  Google Scholar 

  18. Brioukhanov, A. L. & Netrusov, A. I. Aerotolerance of strictly anaerobic microorganisms and factors of defense against oxidative stress: a review. Appl. Biochem. Microbiol. 43, 567–582 (2007).

    Article  CAS  Google Scholar 

  19. McCord, J. M., Keele, B. B. Jr. & Fridovich, I. An enzyme-based theory of obligate anaerobiosis: the physiological function of superoxide dismutase. Proc. Natl Acad. Sci. USA 68, 1024–1027 (1971).

    Article  CAS  Google Scholar 

  20. Imlay, J. A. How oxygen damages microbes: oxygen tolerance and obligate anaerobiosis. Adv. Microb. Physiol. 46, 111–153 (2002).

    Article  CAS  Google Scholar 

  21. Tally, F. P., Goldin, B. R., Jacobus, N. V. & Gorbach, S. L. Superoxide dismutase in anaerobic bacteria of clinical significance. Infect. Immun. 16, 20–25 (1977).

    Article  CAS  Google Scholar 

  22. Huang, Y., Ren, J. & Qu, X. Nanozymes: classification, catalytic mechanisms, activity regulation, and applications. Chem. Rev. 119, 4357–4412 (2019).

    Article  CAS  Google Scholar 

  23. Jiao, L. et al. When nanozymes meet single-atom catalysis. Angew. Chem. Int. Ed. 59, 2565–2576 (2020).

    Article  CAS  Google Scholar 

  24. Cao, F. et al. An enzyme-mimicking single-atom catalyst as an efficient multiple reactive oxygen and nitrogen species scavenger for sepsis management. Angew. Chem. Int. Ed. 32, 5108–5115 (2020).

    Article  Google Scholar 

  25. Zhang, C. et al. Colonization and probiotic function of Bifidobacterium longum. J. Funct. Foods 53, 157–165 (2019).

    Article  CAS  Google Scholar 

  26. Chen, Y. et al. Isolated single iron atoms anchored on N-doped porous carbon as an efficient electrocatalyst for the oxygen reduction reaction. Angew. Chem. Int. Ed. 56, 6937–6941 (2017).

    Article  CAS  Google Scholar 

  27. Zhang, H. et al. Single atomic iron catalysts for oxygen reduction in acidic media: particle size control and thermal activation. J. Am. Chem. Soc. 139, 14143–14149 (2017).

    Article  CAS  Google Scholar 

  28. Pan, Y. et al. Regulating the coordination structure of single-atom Fe-NxCy catalytic sites for benzene oxidation. Nat. Commun. 10, 4290 (2019).

    Article  Google Scholar 

  29. Bull, S. D. et al. Exploiting the reversible covalent bonding of boronic acids: recognition, sensing, and assembly. Acc. Chem. Res. 46, 312–326 (2013).

    Article  CAS  Google Scholar 

  30. Geng, W. et al. Click reaction for reversible encapsulation of single yeast cells. ACS Nano 13, 14459–14467 (2019).

    Article  CAS  Google Scholar 

  31. Bron, P. A., van Baarlen, P. & Kleerebezem, M. Emerging molecular insights into the interaction between probiotics and the host intestinal mucosa. Nat. Rev. Microbiol. 10, 66–78 (2012).

    Article  CAS  Google Scholar 

  32. Hua, S. Advances in oral drug delivery for regional targeting in the gastrointestinal tract-influence of physiological, pathophysiological and pharmaceutical factors. Front. Pharmacol. 11, 524 (2020).

    Article  CAS  Google Scholar 

  33. Stillhart, C. et al. Impact of gastrointestinal physiology on drug absorption in special populations––an UNGAP review. Eur. J. Pharm. Sci. 147, 105280 (2020).

    Article  CAS  Google Scholar 

  34. Pawar, V. K. et al. Gastroretentive dosage forms: a review with special emphasis on floating drug delivery systems. Drug Deliv. 18, 97–110 (2011).

    Article  CAS  Google Scholar 

  35. Wirtz, S., Neufert, C., Weigmann, B. & Neurath, M. F. Chemically induced mouse models of intestinal inflammation. Nat. Protoc. 2, 541–546 (2007).

    Article  CAS  Google Scholar 

  36. Liu, Y. et al. Integrated cascade nanozyme catalyzes in vivo ROS scavenging for anti-inflammatory therapy. Sci. Adv. 6, eabb2695 (2020).

    Article  CAS  Google Scholar 

  37. Sokol, H. et al. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm. Bowel Dis. 15, 1183–1189 (2009).

    Article  CAS  Google Scholar 

  38. Morgan, X. C. et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 13, R79 (2012).

    Article  CAS  Google Scholar 

  39. Manichanh, C. et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 55, 205–211 (2006).

    Article  Google Scholar 

  40. Wright, E. K. et al. Recent advances in characterizing the gastrointestinal microbiome in Crohn’s disease: a systematic review. Inflamm. Bowel Dis. 21, 1219–1228 (2015).

    Google Scholar 

  41. Shang, L. et al. Core altered microorganisms in colitis mouse model: a comprehensive time-point and fecal microbiota transplantation analysis. Antibiotics 10, 643 (2021).

    Article  CAS  Google Scholar 

  42. Salem, F. et al. Gut microbiome in chronic rheumatic and inflammatory bowel diseases: similarities and differences. United Eur. Gastroenterol. J. 7, 1008–1032 (2019).

    Article  CAS  Google Scholar 

  43. Baldelli, V., Scaldaferri, F., Putignani, L. & Del Chierico, F. The role of enterobacteriaceae in gut microbiota dysbiosis in inflammatory bowel diseases. Microorganisms 9, 697 (2021).

    Article  CAS  Google Scholar 

  44. Castellarin, M. et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 22, 299–306 (2012).

    Article  CAS  Google Scholar 

  45. Kostic, A. D. et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 22, 292–298 (2012).

    Article  CAS  Google Scholar 

  46. Ziegler, A., Gonzalez, L. & Blikslager, A. Large animal models: the key to translational discovery in digestive disease research. Cell. Mol. Gastroenterol. Hepatol. 2, 716–724 (2016).

    Article  Google Scholar 

  47. National Institute of Diabetes and Digestive and Kidney Diseases. Opportunities and Challenges in Digestive Diseases Research: Recommendations of the National Commission on Digestive Diseases. NIH Publication No. 08–6514 (National Institutes of Health, 2009).

  48. Chandra, L. et al. Derivation of adult canine intestinal organoids for translational research in gastroenterology. BMC Biol. 17, 33 (2019).

    Article  Google Scholar 

  49. Schaefer, K., Rensing, S., Hillen, H., Burkhardt, J. E. & Germann, P. G. Is science the only driver in species selection? An internal study to evaluate compound requirements in the minipig compared to the dog in preclinical studies. Toxicol. Pathol. 44, 474–479 (2016).

    Article  CAS  Google Scholar 

  50. Mehrabani, D. et al. The healing effect of Teucrium polium in acetic acid-induced ulcerative colitis in the dog as an animal model. Middle East J. Dig. Dis. 4, 40–47 (2012).

    Google Scholar 

  51. Ballal, S. A. et al. Host lysozyme-mediated lysis of Lactococcus lactis facilitates delivery of colitis-attenuating superoxide dismutase to inflamed colons. Proc. Natl Acad. Sci. USA 112, 7803–7808 (2015).

    Article  CAS  Google Scholar 

  52. Han, W. et al. Improvement of an experimental colitis in rats by lactic acid bacteria producing superoxide dismutase. Inflamm. Bowel Dis. 12, 1044–1052 (2006).

    Article  Google Scholar 

  53. de Moreno de LeBlanc, A. et al. Oral administration of a catalase-producing Lactococcus lactis can prevent a chemically induced colon cancer in mice. J. Med. Microbiol. 57, 100–105 (2008).

    Article  Google Scholar 

  54. Liu, M. et al. Oral engineered Bifidobacterium longum expressing rhMnSOD to suppress experimental colitis. Int. Immunopharmacol. 57, 25–32 (2018).

    Article  Google Scholar 

  55. LeBlanc, J. G. et al. Use of superoxide dismutase and catalase producing lactic acid bacteria in TNBS induced Crohn’s disease in mice. J. Biotechnol. 151, 287–293 (2011).

    Article  CAS  Google Scholar 

  56. Zhao, S. et al. An orally administered CeO2@montmorillonite nanozyme targets inflammation for inflammatory bowel disease therapy. Adv. Funct. Mater. 30, 2004692 (2020).

    Article  CAS  Google Scholar 

  57. Cheng, C., Zhao, S., Cheng, Y., Liu, Y. & Wei, H. Design of nanozymes for inflammatory bowel disease therapy. Sci. China Life Sci. 64, 1368–1371 (2021).

    Article  CAS  Google Scholar 

  58. Wei, H. & Wang, E. Nanomaterials with enzyme-like characteristics (nanozymes): next-generation artificial enzymes. Chem. Soc. Rev. 42, 6060–6093 (2013).

    Article  CAS  Google Scholar 

  59. Cheng, C. et al. Multifunctional nanozyme hydrogel with mucosal healing activity for single-dose ulcerative colitis therapy. Bioconjugate Chem. 33, 248–259 (2022).

    Article  CAS  Google Scholar 

  60. Lin, S. et al. Mucosal immunity-mediated modulation of the gut microbiome by oral delivery of probiotics into Peyer’s patches. Sci. Adv. 7, eabf0677 (2021).

    Article  CAS  Google Scholar 

  61. Anselmo, A. C., McHugh, K. J., Webster, J., Langer, R. & Jaklenec, A. Layer-by-layer encapsulation of probiotics for delivery to the microbiome. Adv. Mater. 28, 9486–9490 (2016).

    Article  CAS  Google Scholar 

  62. Zheng, D. W. et al. Prebiotics‐encapsulated probiotic spores regulate gut microbiota and suppress colon cancer. Adv. Mater. 32, 2004529 (2020).

    Article  CAS  Google Scholar 

  63. Centurion, F. et al. Nanoencapsulation for probiotic delivery. ACS Nano 15, 18653–18660 (2021).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (grant nos. 52273152, 51822306 and 22161132027 to Z.M.), Science Technology Department of Zhejiang Province (grant no. 2021C03121 to W.W. and Z.M.), Starry Night Science Fund of Zhejiang University Shanghai Institute for Advanced Study (grant no. SN-ZJU-SIAS-006 to Z.M.), Zhejiang High-Level Young Talent Special Support Plan (Z.M.), the National University of Singapore Start-Up Grant (NUHSRO/2020/133/Startup/08 to X.C.), NUS School of Medicine Nanomedicine Translational Research Programme (NUHSRO/2021/034/TRP/09/Nanomedicine to X.C.) and the Lee Foundation Microbiome Education Grant (X.C.). We cordially thank Q. He from the School of Chemical and Biological Engineering at Zhejiang University and L. Zheng from Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, for their support in the X-ray absorption spectroscopy measurements and analyses.

Author information

Authors and Affiliations

Authors

Contributions

Z.M., X.C., W.W. and F.C. conceived the study, designed the experiments and wrote the manuscript. F.C., L.J. and Y.G. prepared and characterized the platform. F.C. and Y.G. investigated the ROS-scavenging ability. F.C. and L.J. evaluated the bacterial experiments. F.C. and H.W. carried out the in vitro experiments. F.C., Z.Q., C.Z., Y.G. and J.Z. conducted the in vivo experiments on mice. F.C., Y.D., Z.Q., C.Z., Y.G., L.H., H.Y. and Z.T. conducted the in vivo experiments on dogs. All the authors reviewed the manuscript.

Corresponding authors

Correspondence to Weilin Wang, Xiaoyuan Chen or Zhengwei Mao.

Ethics declarations

Competing interests

X.C., Z.M. and F.C. are inventors on a patent application (international patent application no. PCT/CN2021/116214) based on the technology presented in this manuscript. All other authors declare no competing interests.

Peer review

Peer review information

Nature Nanotechnology thanks Alejandra De Moreno de LeBlanc, Kam Leong, Hui Wei 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 Comparison of UC therapy between BL@B-SA50 and clinical therapy.

(a) C57BL/6 mice were provided with water or 3% DSS-containing water for 4 days. On days 4, 5, 6 and 7, mice were orally administered with Medium, 5-ASA (30 mg kg–1), DEX (1 mg/kg), MPS (1 mg/kg) or BL@B-SA50 (BL: 2.5 × 108 CFU kg–1, B-SA: 1.25 mg/kg). (b) Daily body weight changes in each group for 9 days. Data were normalized as a percentage of the body weight at day 0. (c) Changes in DAI. (d) Lengths and (e) pictures of mice colon with indicated treatments on day 8. (f) C57BL/6 mice were provided with water or 3% DSS-containing water for 4 days. On days 4, 5, 6 and 7, mice were orally administered with Medium, BL (2.5 × 108 CFU/kg) + 5-ASA (30 mg/kg), BL (2.5 × 108 CFU/kg) + DEX (1 mg/kg), BL (2.5 × 108 CFU/kg) + MPS (1 mg/kg) or BL@B-SA50 (BL: 2.5 × 108 CFU/kg, B-SA: 1.25 mg/kg). (g) Daily body weight changes in each group for 9 days. Data were normalized as a percentage of the body weight at day 0. (h) Changes in DAI. (i) Lengths and (j) pictures of mice colon with indicated treatments on day 8. Data are presented as mean ± s. d. from n = 5 biological replicates. Statistical analysis was calculated by unpaired Student’s two-sided t test. 1P, 2P, 3P, 4P and 5P, in (b, c) denote the statistical significance of Health, BL@B-SA50, MPS, DEX and 5-ASA group relative to the Control group, while in (g, h) denotes the statistical significance of Health, BL@B-SA50, BL + MPS, BL + DEX and BL + 5-ASA group relative to the Control group, respectively. P in (d, i) denotes the statistical significance relative to the Control group. #P in (d, i) denotes the statistical significance of BL@B-SA50 relative to the Health group.

Extended Data Fig. 2 CD therapy with BL@B-SA50.

(a) C57BL/6 mice were pre-sensitized by absorption of TNBS solution through the skin. After one week, TNBS solutions were slowly administered into the colon lumen of mice to induce CD. Then, mice were orally administered with Medium or B-SA (1.25 mg/kg), BL (2.5 × 108 CFU/kg), BL (2.5 × 108 CFU kg−1) + B-SA (1.25 mg/kg), BL@B-SA50 (BL: 2.5 × 108 CFU/kg, B-SA: 1.25 mg/kg) for four days. (b) Daily bodyweight development after administering TNBS solution into the colon lumen. Data were normalized as a percentage of the bodyweight at day 8. (c) Changes in DAI. (d) Lengths and (e) picture of mice colon with indicated treatments on day 13. Data are presented as mean ± s. d. from n = 5 biological replicates. Statistical analysis was calculated by unpaired Student’s two-sided t test. 1P, 2P, 3P, 4P and 5P, in (b, c) denotes the statistical significance of Health, BL@B-SA50, BL + B-SA, BL and B-SA group relative to the Control group, respectively. P in (d) denotes the statistical significance relative to the Control group. #P in (d) denotes the statistical significance of BL@B-SA50 relative to the Health group.

Supplementary information

Supplementary Information

Supplementary Figs. 1–38, Table 1 and methods.

Reporting Summary

Supplementary Data 1

Supplementary source data.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

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

Cao, F., Jin, L., Gao, Y. et al. Artificial-enzymes-armed Bifidobacterium longum probiotics for alleviating intestinal inflammation and microbiota dysbiosis. Nat. Nanotechnol. 18, 617–627 (2023). https://doi.org/10.1038/s41565-023-01346-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-023-01346-x

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research