Skip to main content

Advertisement

Log in

The novel insight into anti-inflammatory and anxiolytic effects of psychobiotics in diabetic rats: possible link between gut microbiota and brain regions

  • Original Contribution
  • Published:
European Journal of Nutrition Aims and scope Submit manuscript

A Correction to this article was published on 06 September 2019

This article has been updated

Abstract

Purpose

Type 2 diabetes mellitus (T2DM) was associated with gut microbial impairment (dysbiosis) and neurological and behavioral disorders. The role of the gut–brain axis in the management of many diseases including T2DM has been the focus of much research activity in the recent years. However, a wide knowledge gap exists about the gut microbial effects on the function of glia cells. Hence, the present study was aimed to examine the effects of psychobatics on dysbiosis and glia cells function in enteric and central nervous system with an inflammatory insight in T2DM.

Methods

Thirty rats were treated by Lactobacillus (L.) plantarum, inulin, or their combination (synbiotic) for 8 weeks after inducing T2DM. Fecal sample was collected to evaluate gut microbial composition. Then, the rats were sacrificed, and the colon, amygdala, and prefrontal cortex (PFC) were studied.

Results

T2DM resulted in dysbiosis and increased levels of glial cell-derived neurotrophic factor (GDNF), glial fibrillary acidic protein (GFAP), and inflammatory markers (IL-17, IL-6, and TLR-2) in the colon and brain. However, concurrent supplementation of L. plantarum and inulin could improve the gut microbial composition as well as reduce the levels of inflammatory cytokines. While the administration of L. plantarum led to a significant decrease in TLR-2 as well as GDNF and GFAP only in the amygdala, the synbiotic intake could make such changes in the colon, amygdala, and PFC.

Conclusions

Our findings demonstrated an innovative approach to the beneficial effects of psychobiotics in neuroinflammation and behavioral performance through gut microbiota changes, focusing on possible role of glial cells in gut–brain axis.

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

Access this article

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

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11
Fig. 12

Similar content being viewed by others

Change history

  • 06 September 2019

    The original version of this article unfortunately contained a mistake in the order of the author list.

Abbreviations

GDNF:

Glial cell-derived neurotrophic factor

GFAP:

Glial fibrillary acidic protein

IL:

Interleukin

HFD:

High-fat diet

LPS:

Lipopolysaccharides

EPM:

Elevated plus maze

TLRs:

Toll-like receptors

References

  1. Abrahamian H, Endler G, Exner M, Mauler H, Raith M, Endler L, Rumpold H, Gerdov M, Mannhalter C, Prager R (2007) Association of low-grade inflammation with nephropathy in type 2 diabetic patients: role of elevated CRP-levels and 2 different gene-polymorphisms of proinflammatory cytokines. Exp Clin Endocrinol Diabetes 115(01):38–41

    CAS  PubMed  Google Scholar 

  2. Alipour M, Salehi I, Soufi FG (2012) Effect of exercise on diabetes-induced oxidative stress in the rat hippocampus. Iran Red Crescent Med J 14(4):222

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Egede LE, Ellis C (2010) Diabetes and depression: global perspectives. Diabetes Res Clin Pract 87(3):302–312

    PubMed  Google Scholar 

  4. Naseer IM, Bibi F, Alqahtani HM, Chaudhary GA, Azhar IE, Kamal AM, Yasir M (2014) Role of gut microbiota in obesity, type 2 diabetes and Alzheimer’s disease. CNS Neurol Disord Drug Targets 13(2):305–311

    CAS  PubMed  Google Scholar 

  5. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505(7484):559

    CAS  PubMed  Google Scholar 

  6. Shreiner AB, Kao JY, Young VB (2015) The gut microbiome in health and in disease. Curr Opin Gastroenterol 31(1):69

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Li X, Wang E, Yin B, Fang D, Chen P, Wang G, Zhao J, Zhang H, Chen W (2017) Effects of Lactobacillus casei CCFM419 on insulin resistance and gut microbiota in type 2 diabetic mice. Benef Microbes 8(3):421–432

    CAS  PubMed  Google Scholar 

  8. Sherwin E, Sandhu KV, Dinan TG, Cryan JF (2016) May the force be with you: the light and dark sides of the microbiota–gut–brain axis in neuropsychiatry. CNS Drugs 30(11):1019–1041

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Kanai T, Mikami Y, Hayashi A (2015) A breakthrough in probiotics: Clostridium butyricum regulates gut homeostasis and anti-inflammatory response in inflammatory bowel disease. J Gastroenterol 50(9):928–939

    CAS  PubMed  Google Scholar 

  10. Bates JM, Akerlund J, Mittge E, Guillemin K (2007) Intestinal alkaline phosphatase detoxifies lipopolysaccharide and prevents inflammation in zebrafish in response to the gut microbiota. Cell Host Microbe 2(6):371–382

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Sun J, Zhang S, Zhang X, Zhang X, Dong H, Qian Y (2015) IL-17A is implicated in lipopolysaccharide-induced neuroinflammation and cognitive impairment in aged rats via microglial activation. J Neuroinflamm 12(1):165

    Google Scholar 

  12. Burcelin R, Serino M, Chabo C, Blasco-Baque V, Amar J (2011) Gut microbiota and diabetes: from pathogenesis to therapeutic perspective. Acta Diabetol 48(4):257–273

    PubMed  PubMed Central  Google Scholar 

  13. Nagayach A, Patro N, Patro I (2014) Astrocytic and microglial response in experimentally induced diabetic rat brain. Metab Brain Dis 29(3):747–761

    CAS  PubMed  Google Scholar 

  14. da Cunha Franceschi R, Nardin P, Machado CV, Tortorelli LS, Martinez-Pereira MA, Zanotto C, Gonçalves C-A, Zancan DM (2017) Enteric glial reactivity to systemic LPS administration: changes in GFAP and S100B protein. Neurosci Res 119:15–23

    PubMed  Google Scholar 

  15. Hol EM, Pekny M (2015) Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system. Curr Opin Cell Biol 32:121–130

    CAS  PubMed  Google Scholar 

  16. Wang X, Hou Z, Yuan Y, Hou G, Liu Y, Li H, Zhang Z (2011) Association study between plasma GDNF and cognitive function in late-onset depression. J Affect Disord 132(3):418–421

    CAS  PubMed  Google Scholar 

  17. Zhang DK, He FQ, Li TK, Pang XH, Cui DJ, Xie Q, Huang XL, Gan HT (2010) Glial-derived neurotrophic factor regulates intestinal epithelial barrier function and inflammation and is therapeutic for murine colitis. J Pathol 222(2):213–222

    CAS  PubMed  Google Scholar 

  18. Jin Y, Wang G, Han S-S, He M-Y, Cheng X, Ma Z-L, Wu X, Yang X, Liu G-S (2016) Effects of oxidative stress on hyperglycaemia-induced brain malformations in a diabetes mouse model. Exp Cell Res 347(1):201–211

    CAS  PubMed  Google Scholar 

  19. Liu W, Yue W, Wu R (2010) Effects of diabetes on expression of glial fibrillary acidic protein and neurotrophins in rat colon. Auton Neurosci 154(1):79–83

    CAS  PubMed  Google Scholar 

  20. den Heijer T, Vermeer S, Van Dijk E, Prins N, Koudstaal PJ, Hofman A, Breteler M (2003) Type 2 diabetes and atrophy of medial temporal lobe structures on brain MRI. Diabetologia 46(12):1604–1610

    Google Scholar 

  21. Abdul-Rahman O, Sasvari-Szekely M, Ver A, Rosta K, Szasz BK, Kereszturi E, Keszler G (2012) Altered gene expression profiles in the hippocampus and prefrontal cortex of type 2 diabetic rats. BMC Genom 13(1):81

    CAS  Google Scholar 

  22. Morshedi M, Valenlia KB, Hosseinifard ES, Shahabi P, Abbasi MM, Ghorbani M, Barzegari A, Sadigh-Eteghad S, Saghafi-Asl M (2018) Beneficial psychological effects of novel psychobiotics in diabetic rats: the interaction among the gut, blood, and amygdala. J Nutr Biochem 57:145–152

    CAS  PubMed  Google Scholar 

  23. Robinson OJ, Charney DR, Overstreet C, Vytal K, Grillon C (2012) The adaptive threat bias in anxiety: amygdala–dorsomedial prefrontal cortex coupling and aversive amplification. Neuroimage 60(1):523–529

    PubMed  Google Scholar 

  24. Davidson RJ (2002) Anxiety and affective style: role of prefrontal cortex and amygdala. Biol Psychiatry 51(1):68–80

    PubMed  Google Scholar 

  25. Alard J, Lehrter V, Rhimi M, Mangin I, Peucelle V, Abraham AL, Mariadassou M, Maguin E, Waligora-Dupriet AJ, Pot B (2016) Beneficial metabolic effects of selected probiotics on diet-induced obesity and insulin resistance in mice are associated with improvement of dysbiotic gut microbiota. Environ Microbiol 18(5):1484–1497

    CAS  PubMed  Google Scholar 

  26. Lavasani S, Dzhambazov B, Nouri M, Fåk F, Buske S, Molin G, Thorlacius H, Alenfall J, Jeppsson B, Weström B (2010) A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PLoS One 5(2):e9009

    PubMed  PubMed Central  Google Scholar 

  27. Savignac HM, Couch Y, Stratford M, Bannerman DM, Tzortzis G, Anthony DC, Burnet PW (2016) Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical 5-HT2A receptor and IL1-β levels in male mice. Brain Behav Immun 52:120–131

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Jia S, Lu Z, Gao Z, An J, Wu X, Li X, Dai X, Zheng Q, Sun Y (2016) Chitosan oligosaccharides alleviate cognitive deficits in an amyloid-β1–42-induced rat model of Alzheimer’s disease. Int J Biol Macromol 83:416–425

    CAS  PubMed  Google Scholar 

  29. Asemi Z, Khorrami-Rad A, Alizadeh SA, Shakeri H, Esmaillzadeh A (2014) Effects of synbiotic food consumption on metabolic status of diabetic patients: a double-blind randomized cross-over controlled clinical trial. Clin Nutr 33(2):198–203. https://doi.org/10.1016/j.clnu.2013.05.015

    Article  PubMed  Google Scholar 

  30. Srinivasan K, Viswanad B, Asrat L, Kaul C, Ramarao P (2005) Combination of high-fat diet-fed and low-dose streptozotocin-treated rat: a model for type 2 diabetes and pharmacological screening. Pharmacol Res 52(4):313–320

    CAS  PubMed  Google Scholar 

  31. Pellow S, Chopin P, File SE, Briley M (1985) Validation of open: closed arm entries in an elevated plus-maze as a measure of anxiety in the rat. J Neurosci Methods 14(3):149–167

    CAS  PubMed  Google Scholar 

  32. Graeber MB, Li W, Rodriguez ML (2011) Role of microglia in CNS inflammation. FEBS Lett 585(23):3798–3805

    CAS  PubMed  Google Scholar 

  33. Lomax AE, Linden DR, Mawe GM, Sharkey KA (2006) Effects of gastrointestinal inflammation on enteroendocrine cells and enteric neural reflex circuits. Auton Neurosci 126:250–257

    PubMed  Google Scholar 

  34. Sampson TR, Mazmanian SK (2015) Control of brain development, function, and behavior by the microbiome. Cell Host Microbe 17(5):565–576

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Rizzo A, Losacco A, Carratelli CR, Di Domenico M, Bevilacqua N (2013) Lactobacillus plantarum reduces Streptococcus pyogenes virulence by modulating the IL-17, IL-23 and Toll-like receptor 2/4 expressions in human epithelial cells. Int Immunopharmacol 17(2):453–461

    CAS  PubMed  Google Scholar 

  36. Valenlia KB, Morshedi M, Saghafi-Asl M, Shahabi P, Abbasi MM (2018) Beneficial impacts of Lactobacillus plantarum and inulin on hypothalamic levels of insulin, leptin, and oxidative markers in diabetic rats. J Funct Foods 46:529–537

    CAS  Google Scholar 

  37. Brun P, Giron MC, Qesari M, Porzionato A, Caputi V, Zoppellaro C, Banzato S, Grillo AR, Spagnol L, De Caro R (2013) Toll-like receptor 2 regulates intestinal inflammation by controlling integrity of the enteric nervous system. Gastroenterology 145(6):1323–1333

    CAS  PubMed  Google Scholar 

  38. Matsunaga W, Isobe K, Shirokawa T (2006) Involvement of neurotrophic factors in aging of noradrenergic innervations in hippocampus and frontal cortex. Neurosci Res 54(4):313–318

    CAS  PubMed  Google Scholar 

  39. Straten G, Saur R, Laske C, Gasser T, Annas P, Basun H, Leyhe T (2011) Influence of lithium treatment on GDNF serum and CSF concentrations in patients with early Alzheimer’s disease. Curr Alzheimer Res 8(8):853–859

    CAS  PubMed  Google Scholar 

  40. Rémy S, Naveilhan P, Paillé V, Brachet P, Neveu I (2003) Lipopolysaccharide and TNFα regulate the expression of GDNF, neurturin and their receptors. Neuroreport 14(11):1529–1534

    PubMed  Google Scholar 

  41. Ushakova G, Fed’kiv O, Prykhod’ko O, Pierzynowski S, Kruszewska D (2009) The effect of long-term lactobacilli (lactic acid bacteria) enteral treatment on the central nervous system of growing rats. J Nutr Biochem 20(9):677–684

    CAS  PubMed  Google Scholar 

  42. Burokas A, Arboleya S, Moloney RD, Peterson VL, Murphy K, Clarke G, Stanton C, Dinan TG, Cryan JF (2017) Targeting the microbiota–gut–brain axis: prebiotics have anxiolytic and antidepressant-like effects and reverse the impact of chronic stress in mice. Biol Psychiatry 82(7):472–487

    CAS  PubMed  Google Scholar 

  43. Savignac HM, Corona G, Mills H, Chen L, Spencer JP, Tzortzis G, Burnet PW (2013) Prebiotic feeding elevates central brain derived neurotrophic factor, N-methyl-D-aspartate receptor subunits and D-serine. Neurochem Int 63(8):756–764

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Vogelzangs N, De Jonge P, Smit J, Bahn S, Penninx B (2016) Cytokine production capacity in depression and anxiety. Transl Psychiatry 6(5):e825

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Special thanks to Applied Research Center, Nutrition Research Center, and Laboratory Animal Center of Tabriz University of Medical Sciences, Tabriz, Iran.

Funding

This study was supported by Drug Applied Research Center, Tabriz University of Medical Sciences Tabriz, Iran. The results of this paper were extracted from M.Sc. thesis of Elaheh Sadat Hosseinifard (Grant number: 5/D/32825), registered at Tabriz University of Medical Sciences, Tabriz, Iran.

Author information

Authors and Affiliations

Authors

Contributions

MSA and ESH wrote the study design and protocol. MM and KBV helped with preparation of inulin and bacterial solutions and performing intervention phases. MM and ESH analyzed and interpreted the data and drew graphs. ESH and MM helped with keeping rats and intervening. ESH and MM performed behavioral tests and analyzed and interpreted the related data. ESH and MM and MSA were involved in drafting the manuscript or revising it critically for content. All authors have given the final approval of the version to be published.

Corresponding author

Correspondence to Maryam Saghafi-Asl.

Ethics declarations

Conflict of interest

There is nothing to declare.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 26 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hosseinifard, ES., Morshedi, M., Bavafa-Valenlia, K. et al. The novel insight into anti-inflammatory and anxiolytic effects of psychobiotics in diabetic rats: possible link between gut microbiota and brain regions. Eur J Nutr 58, 3361–3375 (2019). https://doi.org/10.1007/s00394-019-01924-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00394-019-01924-7

Keywords

Navigation