Skip to main content
Log in

Lactobacillus Helveticus Improves Controlled Cortical Impact Injury-Generated Neurological Aberrations by Remodeling of Gut-Brain Axis Mediators

  • Research
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Considerable studies augured the potential of gut microbiota-based interventions in brain injury-associated complications. Based on our earlier study results, we envisaged the sex-specific neuroprotective effect of Lactobacillus helveticus by remodeling of gut-brain axis. In this study, we investigated the effect of L. helveticus on neurological complications in a mouse model of controlled cortical impact (CCI). Adult, male and female, C57BL/6 mice underwent CCI surgery and received L. helveticus treatment for six weeks. Sensorimotor function was evaluated via neurological severity score and rotarod test. Long-term effects on anxiety-like behavior and cognition were assessed using the elevated-zero maze (EZM) and novel object recognition test (NORT). Brain perilesional area, blood, colon, and fecal samples were collected post-CCI for molecular biology analysis. CCI-operated mice displayed significant neurological impairments at 1-, 3-, 5-, and 7-days post-injury (dpi) and exhibited altered behavior in EZM and NORT compared to sham-operated mice. However, these behavioral changes were ameliorated in mice receiving L. helveticus. GFAP, Iba-1, TNF-α, and IL-1β expressions and corticotrophin-releasing hormone (CRH) levels were elevated in the perilesional cortex of CCI-operated male/female mice. These elevated biomarkers and decreased BDNF levels in both male/female mice were modified by L. helveticus treatment. Additionally, L. helveticus treatment restored altered short-chain fatty acids (SCFAs) levels in fecal samples and improved intestinal integrity but did not affect decreased plasma levels of progesterone and testosterone in CCI mice. These results indicate that L. helveticus exerts beneficial effects in the CCI mouse model by mitigating inflammation and remodeling of gut microbiota-brain mediators.

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

Access this article

Subscribe and save

Springer+
from $39.99 /Month
  • Starting from 10 chapters or articles per month
  • Access and download chapters and articles from more than 300k books and 2,500 journals
  • Cancel anytime
View plans

Buy Now

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

Similar content being viewed by others

Data Availability

No datasets were generated or analysed during the current study.

References

  1. Neufeld K-A, Foster JA (2009) Effects of gut microbiota on the brain: implications for psychiatry. J Psychiatry Neuroscience: JPN 34(3):230

    Google Scholar 

  2. Gao C et al (2017) A three-day consecutive fingolimod administration improves neurological functions and modulates multiple immune responses of CCI mice. Mol Neurobiol 54:8348–8360

    CAS  PubMed  Google Scholar 

  3. Chen X et al (2018) Omega-3 polyunsaturated fatty acid attenuates the inflammatory response by modulating microglia polarization through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway following experimental traumatic brain injury. J Neuroinflamm 15(1):1–15

    Google Scholar 

  4. Feighery L et al (2008) Increased intestinal permeability in rats subjected to traumatic frontal lobe percussion brain injury. J Trauma Acute Care Surg 64(1):131–138

    Google Scholar 

  5. Hang C-H et al (2003) Alterations of intestinal mucosa structure and barrier function following traumatic brain injury in rats. World J Gastroenterology: WJG 9(12):2776

    PubMed Central  Google Scholar 

  6. Sun B et al (2015) The effects of Lactobacillus acidophilus on the intestinal smooth muscle contraction through PKC/MLCK/MLC signaling pathway in TBI mouse model. PLoS ONE 10(6):e0128214

    PubMed  PubMed Central  Google Scholar 

  7. Sundman MH et al (2017) The bidirectional gut-brain-microbiota axis as a potential nexus between traumatic brain injury, inflammation, and disease. Brain Behav Immun 66:31–44

    CAS  PubMed  Google Scholar 

  8. Jang S et al (2018) Gastrointestinal inflammation by gut microbiota disturbance induces memory impairment in mice. Mucosal Immunol 11(2):369–379

    CAS  PubMed  Google Scholar 

  9. Erny D, Hrabě de Angelis AL, Prinz M (2017) Communicating systems in the body: how microbiota and microglia cooperate. Immunology 150(1):7–15

    CAS  PubMed  Google Scholar 

  10. Sharon G et al (2016) The central nervous system and the gut microbiome. Cell 167(4):915–932

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Rahman Z, Dandekar MP (2021) Crosstalk between gut microbiome and immunology in the management of ischemic brain injury. J Neuroimmunol 353:577498

    CAS  PubMed  Google Scholar 

  12. Benakis C et al (2016) Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med 22(5):516–523

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Houlden A et al (2016) Brain injury induces specific changes in the caecal microbiota of mice via altered autonomic activity and mucoprotein production. Brain Behav Immun 57:10–20

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Nicholson SE et al (2019) Moderate traumatic brain injury alters the gastrointestinal microbiome in a time-dependent manner. Shock 52(2):240–248

    CAS  PubMed  Google Scholar 

  15. Rice MW, Pandya JD, Shear DA (2019) Gut microbiota as a therapeutic target to ameliorate the biochemical, neuroanatomical, and behavioral effects of traumatic brain injuries. Front Neurol 10:875

    PubMed  PubMed Central  Google Scholar 

  16. Hanscom M, Loane DJ, Shea-Donohue T (2021) Brain-gut axis dysfunction in the pathogenesis of traumatic brain injury. J Clin Investig, 131(12)

  17. Ng SY, Lee AYW (2019) Traumatic brain injuries: pathophysiology and potential therapeutic targets. Front Cell Neurosci 13:528

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Huynh U, Zastrow ML (2023) Metallobiology of Lactobacillaceae in the gut microbiome. J Inorg Biochem 238:112023

    CAS  PubMed  Google Scholar 

  19. George AK et al (2021) Rebuilding microbiome for mitigating traumatic brain injury: importance of restructuring the gut-microbiome-brain axis. Mol Neurobiol 58:3614–3627

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Bruce-Keller AJ, Salbaum JM, Berthoud H-R (2018) Harnessing gut microbes for mental health: getting from here to there. Biol Psychiatry 83(3):214–223

    PubMed  Google Scholar 

  21. Shen NT et al (2017) Timely use of probiotics in hospitalized adults prevents Clostridium difficile infection: a systematic review with meta-regression analysis. Gastroenterology 152(8):1889–1900e9

    PubMed  Google Scholar 

  22. Castelli V et al (2021) The emerging role of probiotics in neurodegenerative diseases: new hope for Parkinson’s disease? Neural Regeneration Res 16(4):628

    CAS  Google Scholar 

  23. Mishra V et al (2023) A review on the Protective effects of Probiotics against Alzheimer’s Disease. Biology 13(1):8

    PubMed  PubMed Central  Google Scholar 

  24. Wallace CJ, Milev R (2017) The effects of probiotics on depressive symptoms in humans: a systematic review. Ann Gen Psychiatry 16(1):1–10

    Google Scholar 

  25. Zhong D-Y et al (2021) The effect of probiotics in stroke treatment. Evidence-based Complementary and Alternative Medicine, 2021

  26. Lépine AF et al (2018) Lactobacillus acidophilus attenuates Salmonella-induced stress of epithelial cells by modulating tight-junction genes and cytokine responses. Front Microbiol 9:1439

    PubMed  PubMed Central  Google Scholar 

  27. Wu D et al (2019) Nutritional modulation of immune function: analysis of evidence, mechanisms, and clinical relevance. Front Immunol 9:3160

    PubMed  PubMed Central  Google Scholar 

  28. Athari Nik Azm S et al (2018) Lactobacilli and bifidobacteria ameliorate memory and learning deficits and oxidative stress in β-amyloid (1–42) injected rats. Applied Physiology, Nutrition, and Metabolism, 43(7): pp. 718–726

  29. Abdrabou AM, Osman EY, Aboubakr OA (2018) Comparative therapeutic efficacy study of Lactobacilli probiotics and citalopram in treatment of acute stress-induced depression in lab murine models. Hum Microbiome J 10:33–36

    Google Scholar 

  30. O’Hagan C et al (2017) Long-term multi-species Lactobacillus and Bifidobacterium dietary supplement enhances memory and changes regional brain metabolites in middle-aged rats. Neurobiol Learn Mem 144:36–47

    PubMed  Google Scholar 

  31. Bravo JA et al (2011) Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci 108(38):16050–16055

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Corpuz HM et al (2018) Long-term diet supplementation with Lactobacillus paracasei K71 prevents age-related cognitive decline in senescence-accelerated mouse prone 8. Nutrients 10(6):762

    PubMed  PubMed Central  Google Scholar 

  33. Hwang Y-H et al (2019) Efficacy and safety of Lactobacillus plantarum C29-fermented soybean (DW2009) in individuals with mild cognitive impairment: a 12-week, multi-center, randomized, double-blind, placebo-controlled clinical trial. Nutrients 11(2):305

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Partrick KA et al (2021) Ingestion of probiotic (Lactobacillus helveticus and Bifidobacterium longum) alters intestinal microbial structure and behavioral expression following social defeat stress. Sci Rep 11(1):3763

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Rode J et al (2022) Probiotic mixture containing lactobacillus helveticus, bifidobacterium longum and lactiplantibacillus plantarum affects brain responses toward an emotional task in healthy subjects: a randomized clinical trial. Front Nutr 9:827182

    PubMed  PubMed Central  Google Scholar 

  36. Messaoudi M et al (2011) Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br J Nutr 105(5):755–764

    CAS  PubMed  Google Scholar 

  37. Messaoudi M et al (2011) Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human volunteers. Gut Microbes 2(4):256–261

    PubMed  Google Scholar 

  38. Benton D, Williams C, Brown A (2007) Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur J Clin Nutr 61(3):355–361

    CAS  PubMed  Google Scholar 

  39. Akkasheh G et al (2016) Clinical and metabolic response to probiotic administration in patients with major depressive disorder: a randomized, double-blind, placebo-controlled trial. Nutrition 32(3):315–320

    CAS  PubMed  Google Scholar 

  40. Chung Y-C et al (2014) Fermented milk of Lactobacillus helveticus IDCC3801 improves cognitive functioning during cognitive fatigue tests in healthy older adults. J Funct Foods 10:465–474

    CAS  Google Scholar 

  41. Holcomb M et al (2024) Probiotic treatment causes sex-specific neuroprotection after traumatic brain injury in mice. bioRxiv, : p. 2024.04. 01.587652

  42. Pasam T, Dandekar MP (2024) Fecal microbiota transplantation unveils sex-specific differences in a controlled cortical impact injury mouse model. Front Microbiol 14:1336537

    PubMed  PubMed Central  Google Scholar 

  43. Ahmed ME et al (2020) Glia maturation factor (GMF) regulates microglial expression phenotypes and the associated neurological deficits in a mouse model of traumatic brain injury. Mol Neurobiol 57(11):4438–4450

    CAS  PubMed  Google Scholar 

  44. Akamatsu Y, Hanafy KA (2020) Cell death and recovery in traumatic brain injury. Neurotherapeutics 17(2):446–456

    PubMed  PubMed Central  Google Scholar 

  45. Raikwar SP et al (2021) Real-time noninvasive bioluminescence, ultrasound and photoacoustic imaging in NFκB-RE-Luc transgenic mice reveal glia maturation factor-mediated immediate and sustained spatio-temporal activation of NFκB signaling post-traumatic brain injury in a gender-specific manner. Cell Mol Neurobiol 41:1687–1706

    CAS  PubMed  Google Scholar 

  46. Karve IP, Taylor JM, Crack PJ (2016) The contribution of astrocytes and microglia to traumatic brain injury. Br J Pharmacol 173(4):692–702

    CAS  PubMed  Google Scholar 

  47. Sakai K et al (2021) Reactive pericytes in early phase are involved in glial activation and late-onset hypersusceptibility to pilocarpine-induced seizures in traumatic brain injury model mice. J Pharmacol Sci 145(1):155–165

    CAS  PubMed  Google Scholar 

  48. Taylor AN et al (2008) Injury severity differentially affects short-and long-term neuroendocrine outcomes of traumatic brain injury. J Neurotrauma 25(4):311–323

    PubMed  Google Scholar 

  49. Griesbach GS et al (2011) Heightening of the stress response during the first weeks after a mild traumatic brain injury. Neuroscience 178:147–158

    CAS  PubMed  Google Scholar 

  50. Taylor AN et al (2006) Lasting neuroendocrine-immune effects of traumatic brain injury in rats. J Neurotrauma 23(12):1802–1813

    PubMed  Google Scholar 

  51. Neese SL et al (2010) Z-Bisdehydrodoisynolic acid (Z-BDDA): an estrogenic seco-steroid that enhances behavioral recovery following moderate fluid percussion brain injury in male rats. Brain Res 1362:93–101

    CAS  PubMed  Google Scholar 

  52. Brotfain E et al (2016) Neuroprotection by estrogen and progesterone in traumatic brain injury and spinal cord injury. Curr Neuropharmacol 14(6):641–653

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Roof RL, HALL ED (2000) Gender differences in acute CNS trauma and stroke: neuroprotective effects of estrogen and progesterone. J Neurotrauma 17(5):367–388

    CAS  PubMed  Google Scholar 

  54. Meltser I et al (2008) Estrogen receptor β protects against acoustic trauma in mice. J Clin Investig 118(4):1563–1570

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Singh M, Su C (2013) Progesterone and neuroprotection. Horm Behav 63(2):284–290

    CAS  PubMed  Google Scholar 

  56. Stein DG (2013) A clinical/translational perspective: can a developmental hormone play a role in the treatment of traumatic brain injury? Horm Behav 63(2):291–300

    CAS  PubMed  Google Scholar 

  57. Mechanick JI, Nierman DM (2006) Gonadal steroids in critical illness. Crit Care Clin 22(1):87–103

    CAS  PubMed  Google Scholar 

  58. WOOLF PD et al (1985) Transient hypogonadotropic hypogonadism caused by critical illness. J Clin Endocrinol Metabolism 60(3):444–450

    CAS  Google Scholar 

  59. Spratt DI et al (1992) Both hyper-and hypogonadotropic hypogonadism occur transiently in acute illness: bio-and immunoactive gonadotropins. J Clin Endocrinol Metabolism 75(6):1562–1570

    CAS  Google Scholar 

  60. Dimopoulou I et al (2004) Endocrine abnormalities in critical care patients with moderate-to-severe head trauma: incidence, pattern and predisposing factors. Intensive Care Med 30:1051–1057

    PubMed  Google Scholar 

  61. Du D et al (2021) Fecal microbiota transplantation is a promising method to restore gut microbiota dysbiosis and relieve neurological deficits after traumatic brain injury. Oxidative Medicine and Cellular Longevity, 2021

  62. Blixt J et al (2015) Aquaporins and blood–brain barrier permeability in early edema development after traumatic brain injury. Brain Res 1611:18–28

    CAS  PubMed  Google Scholar 

  63. Liu Y-l et al (2017) Sesamin alleviates blood-brain barrier disruption in mice with experimental traumatic brain injury. Acta Pharmacol Sin 38(11):1445–1455

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Koh A et al (2016) From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 165(6):1332–1345

    CAS  PubMed  Google Scholar 

  65. Opeyemi OM et al (2021) Sustained dysbiosis and decreased fecal short-chain fatty acids after traumatic brain injury and impact on neurologic outcome. J Neurotrauma 38(18):2610–2621

    PubMed  PubMed Central  Google Scholar 

  66. Rahman Z et al (2022) Binary classification model of machine learning detected altered gut integrity in controlled-cortical impact model of traumatic brain injury. Int J Neurosci, : p. 1–12

  67. Tentu PM et al (2023) Oxyberberine an oxoderivative of berberine confers neuroprotective effects in controlled-cortical impact mouse model of traumatic brain injury. Int J Neurosci, : p. 1–16

  68. Pasam T, Dandekar MP (2024) Insights from Rodent models for improving bench-to-Bedside translation in traumatic brain Injury. Method and Protocols, Neuroprotection, pp 599–622

    Google Scholar 

  69. Wang G et al (2013) Microglia/macrophage polarization dynamics in white matter after traumatic brain injury. J Cereb Blood Flow Metabolism 33(12):1864–1874

    CAS  Google Scholar 

  70. Zago M et al (2021) Functional characterization and immunomodulatory properties of Lactobacillus helveticus strains isolated from Italian hard cheeses. PLoS ONE 16(1):e0245903

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Liang S et al (2015) Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience 310:561–577

    CAS  PubMed  Google Scholar 

  72. Cowan CS, Callaghan BL, Richardson R (2016) The effects of a probiotic formulation (Lactobacillus rhamnosus and L. Helveticus) on developmental trajectories of emotional learning in stressed infant rats. Translational Psychiatry 6(5):e823–e823

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Minelli EB, Benini A (2008) Relationship between number of bacteria and their probiotic effects. Microb Ecol Health Disease 20(4):180–183

    Google Scholar 

  74. Tillmann S et al (2018) Probiotics affect one-carbon metabolites and catecholamines in a genetic rat model of depression, vol 62. Molecular nutrition & food research, p 1701070. 7

  75. Deuis JR, Dvorakova LS, Vetter I (2017) Methods used to evaluate pain behaviors in rodents. Front Mol Neurosci 10:284

    PubMed  PubMed Central  Google Scholar 

  76. Menéndez L et al (2002) Unilateral hot plate test: a simple and sensitive method for detecting central and peripheral hyperalgesia in mice. J Neurosci Methods 113(1):91–97

    PubMed  Google Scholar 

  77. Hamm RJ (2001) Neurobehavioral assessment of outcome following traumatic brain injury in rats: an evaluation of selected measures. J Neurotrauma 18(11):1207–1216

    CAS  PubMed  Google Scholar 

  78. Yarnell AM et al (2016) The revised neurobehavioral severity scale (NSS-R) for rodents. Curr Protoc Neurosci, 75(1): p. 9.52. 1-9.52. 16.

  79. Tucker LB, McCabe JT (2017) Behavior of male and female C57BL/6J mice is more consistent with repeated trials in the elevated zero maze than in the elevated plus maze. Front Behav Neurosci 11:13

    PubMed  PubMed Central  Google Scholar 

  80. Tucker LB, Fu AH, McCabe JT (2016) Performance of male and female C57BL/6J mice on motor and cognitive tasks commonly used in pre-clinical traumatic brain injury research. J Neurotrauma 33(9):880–894

    PubMed  PubMed Central  Google Scholar 

  81. Zhao Z et al (2012) Comparing the predictive value of multiple cognitive, affective, and motor tasks after rodent traumatic brain injury. J Neurotrauma 29(15):2475–2489

    PubMed  PubMed Central  Google Scholar 

  82. Clarke JR et al (2010) Plastic modifications induced by object recognition memory processing. Proceedings of the National Academy of Sciences, 107(6): pp. 2652–2657

  83. Nanfaro F et al (2010) Pregnenolone sulfate infused in lateral septum of male rats impairs novel object recognition memory. Pharmacol Rep 62(2):265–272

    CAS  PubMed  Google Scholar 

  84. Sarkisyan G, Hedlund PB (2009) The 5-HT7 receptor is involved in allocentric spatial memory information processing. Behav Brain Res 202(1):26–31

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Antunes M, Biala G (2012) The novel object recognition memory: neurobiology, test procedure, and its modifications. Cogn Process 13:93–110

    CAS  PubMed  Google Scholar 

  86. Luvuno M, Khathi A, Mabandla MV (2020) The effects of exercise treatment on learning and memory ability, and cognitive performance in diet-induced prediabetes animals. Sci Rep 10(1):15048

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Palepu MSK, Gajula SNR, K M, Sonti R, Dandekar MP (2024). SCFAs supplementation rescues anxiety-and depression-like phenotypes generated by fecal engraftment of treatment-resistant depression rats. ACS Chemical Neuroscience 15(5):1010–1025

  88. Dandekar MP et al (2022) Multi-strain probiotic formulation reverses maternal separation and chronic unpredictable mild stress-generated anxiety-and depression-like phenotypes by modulating gut microbiome–brain activity in rats. ACS Chem Neurosci 13(13):1948–1965

    CAS  PubMed  Google Scholar 

  89. Balasubramanian R et al (2023) Involvement of microbiome gut–brain axis in neuroprotective effect of quercetin in mouse model of repeated mild traumatic brain injury. Neuromol Med 25(2):242–254

    CAS  Google Scholar 

  90. Coronado VG et al (2012) Trends in traumatic brain injury in the US and the public health response: 1995–2009. J Saf Res 43(4):299–307

    Google Scholar 

  91. Chen Y et al (1996) An experimental model of closed head injury in mice: pathophysiology, histopathology, and cognitive deficits. J Neurotrauma 13(10):557–568

    CAS  PubMed  Google Scholar 

  92. Bazaz MR, Asthana A, Dandekar MP (2024) Chitosan revokes controlled-cortical impact generated neurological aberrations in circadian disrupted mice via TLR4-NLRP3 axis. Eur J Pharmacol 969:176436

    CAS  PubMed  Google Scholar 

  93. Shultz SR et al (2020) Clinical relevance of behavior testing in animal models of traumatic brain injury. J Neurotrauma 37(22):2381–2400

    PubMed  Google Scholar 

  94. Malkesman O et al (2013) Traumatic brain injury–modeling neuropsychiatric symptoms in rodents. Front Neurol 4:157

    PubMed  PubMed Central  Google Scholar 

  95. Popovitz J, Mysore SP, Adwanikar H (2019) Long-term effects of traumatic brain injury on anxiety-like behaviors in mice: behavioral and neural correlates. Front Behav Neurosci 13:6

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Pandey DK et al (2009) Depression-like and anxiety-like behavioural aftermaths of impact accelerated traumatic brain injury in rats: a model of comorbid depression and anxiety? Behav Brain Res 205(2):436–442

    PubMed  Google Scholar 

  97. Wright DK et al (2017) Sex matters: repetitive mild traumatic brain injury in adolescent rats. Ann Clin Transl Neurol 4(9):640–654

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Wirth P et al (2017) New method to induce mild traumatic brain injury in rodents produces differential outcomes in female and male Sprague Dawley rats. J Neurosci Methods 290:133–144

    PubMed  PubMed Central  Google Scholar 

  99. Jullienne A et al (2018) Male and female mice exhibit divergent responses of the cortical vasculature to traumatic brain injury. J Neurotrauma 35(14):1646–1658

    PubMed  PubMed Central  Google Scholar 

  100. Emerson CS, Headrick JP, Vink R (1993) Estrogen improves biochemical and neurologic outcome following traumatic brain injury in male rats, but not in females. Brain Res 608(1):95–100

    CAS  PubMed  Google Scholar 

  101. Palepu MSK, Dandekar MP (2022) Remodeling of microbiota gut-brain axis using psychobiotics in depression. Eur J Pharmacol 931:175171

    CAS  PubMed  Google Scholar 

  102. Sun J et al (2016) Clostridium butyricum attenuates cerebral ischemia/reperfusion injury in diabetic mice via modulation of gut microbiota. Brain Res 1642:180–188

    CAS  PubMed  Google Scholar 

  103. Li H et al (2018) Clostridium butyricum exerts a neuroprotective effect in a mouse model of traumatic brain injury via the gut-brain axis, vol 30. Neurogastroenterology & Motility, p e13260. 5

  104. Negi A et al (2024) In-vitro and preclinical testing of bacillus subtilis UBBS-14 probiotic in rats shows no toxicity. Toxicol Res 13(1):tfae021

    Google Scholar 

  105. Davis IV (2021) Differential fecal microbiome dysbiosis after equivalent traumatic brain injury in aged versus young adult mice. J Experimental Neurol 2(3):120

    Google Scholar 

  106. Yeon S-W et al (2010) Fermented milk of Lactobacillus helveticus IDCC3801 reduces beta-amyloid and attenuates memory deficit. J Funct Foods 2(2):143–152

    CAS  Google Scholar 

  107. De Oliveira FL et al (2023) Exploring the potential of lactobacillus helveticus R0052 and Bifidobacterium longum R0175 as promising psychobiotics using SHIME. Nutrients 15(6):1521

    PubMed  PubMed Central  Google Scholar 

  108. Morganti-Kossmann MC et al (2001) Role of cerebral inflammation after traumatic brain injury: a revisited concept. Shock 16(3):165–177

    CAS  PubMed  Google Scholar 

  109. Shi H et al (2019) Role of toll-like receptor mediated signaling in traumatic brain injury. Neuropharmacology 145:259–267

    CAS  PubMed  Google Scholar 

  110. Woodcock T, Morganti-Kossmann C (2013) The role of markers of inflammation in traumatic brain injury. Front Neurol 4:41121

    Google Scholar 

  111. Förstner P et al (2018) Neuroinflammation after traumatic brain injury is enhanced in activating transcription factor 3 mutant mice. J Neurotrauma 35(19):2317–2329

    PubMed  Google Scholar 

  112. Semple BD et al (2010) Role of CCL2 (MCP-1) in traumatic brain injury (TBI): evidence from severe TBI patients and CCL2–/– mice. J Cereb Blood Flow Metabolism 30(4):769–782

    Google Scholar 

  113. Hellewell S, Semple BD, Morganti-Kossmann MC (2016) Therapies negating neuroinflammation after brain trauma. Brain Res 1640:36–56

    CAS  PubMed  Google Scholar 

  114. Eyolfson E et al (2020) Microglia dynamics in adolescent traumatic brain injury. J Neuroinflamm 17:1–19

    Google Scholar 

  115. Kempuraj D et al (2017) Brain and peripheral atypical inflammatory mediators potentiate neuroinflammation and neurodegeneration. Front Cell Neurosci 11:216

    PubMed  PubMed Central  Google Scholar 

  116. Liddelow SA et al (2017) Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541(7638):481–487

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Wang W-Y et al (2015) Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Annals Translational Med, 3(10)

  118. Kumar A, Loane DJ (2012) Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention. Brain Behav Immun 26(8):1191–1201

    PubMed  Google Scholar 

  119. Liu Y-W et al (2016) Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naïve adult mice. Brain Res 1631:1–12

    CAS  PubMed  Google Scholar 

  120. Luo J et al (2014) Ingestion of Lactobacillus strain reduces anxiety and improves cognitive function in the hyperammonemia rat. Sci China Life Sci 57:327–335

    PubMed  Google Scholar 

  121. Arseneault-Breard J et al (2012) Combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 reduces post-myocardial infarction depression symptoms and restores intestinal permeability in a rat model. Br J Nutr 107(12):1793–1799

    CAS  PubMed  Google Scholar 

  122. Desbonnet L et al (2008) The probiotic bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J Psychiatr Res 43(2):164–174

    PubMed  Google Scholar 

  123. D’Mello C et al (2015) Probiotics improve inflammation-associated sickness behavior by altering communication between the peripheral immune system and the brain. J Neurosci 35(30):10821–10830

    PubMed  PubMed Central  Google Scholar 

  124. Jeong J-J et al (2015) Probiotic mixture KF attenuates age-dependent memory deficit and lipidemia in Fischer 344 rats. J Microbiol Biotechnol 25(9):1532–1536

    CAS  PubMed  Google Scholar 

  125. Gareau MG et al (2011) Bacterial infection causes stress-induced memory dysfunction in mice. Gut 60(3):307–317

    PubMed  Google Scholar 

  126. Kawano M, Miyoshi M, Miyazaki T (2019) Lactobacillus helveticus SBT2171 induces A20 expression via toll-like receptor 2 signaling and inhibits the lipopolysaccharide-induced activation of nuclear factor-kappa B and mitogen-activated protein kinases in peritoneal macrophages. Front Immunol 10:845

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Tan C et al (2022) Neuroprotective effects of probiotic-supplemented diet on cognitive behavior of 3xTg-AD mice. Journal of Healthcare Engineering, 2022

  128. Xin Z et al (2024) Neuroprotective effect of a multistrain probiotic mixture in SOD1G93A mice by reducing SOD1 aggregation and targeting the microbiota-gut-brain axis. Mol Neurobiol, : p. 1–21

  129. Karalis K et al (1991) Autocrine or paracrine inflammatory actions of corticotropin-releasing hormone in vivo. Science 254(5030):421–423

    CAS  PubMed  Google Scholar 

  130. Linthorst AC et al (1997) Long-term intracerebroventricular infusion of corticotropin-releasing hormone alters neuroendocrine, neurochemical, autonomic, behavioral, and cytokine responses to a systemic inflammatory challenge. J Neurosci 17(11):4448–4460

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Roe SY, McGowan EM, Rothwell NJ (1998) Evidence for the involvement of corticotrophin-releasing hormone in the pathogenesis of traumatic brain injury. Eur J Neurosci 10(2):553–559

    CAS  PubMed  Google Scholar 

  132. De Souza EB (1987) Corticotropin-releasing factor receptors in the rat central nervous system: characterization and regional distribution. J Neurosci 7(1):88–100

    PubMed  PubMed Central  Google Scholar 

  133. Taylor AN et al (2010) Injury severity differentially alters sensitivity to dexamethasone after traumatic brain injury. J Neurotrauma 27(6):1081–1089

    PubMed  Google Scholar 

  134. Weil ZM et al (2022) The role of the stress system in recovery after traumatic brain injury: a tribute to Bruce S. McEwen. Neurobiol Stress 19:100467

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Zhang N et al (2020) Efficacy of probiotics on stress in healthy volunteers: a systematic review and meta-analysis based on randomized controlled trials. Brain Behav 10(9):e01699

    PubMed  PubMed Central  Google Scholar 

  136. Rehman MU et al (2022) Probiotics (Bacillus clausii and Lactobacillus fermentum NMCC-14) ameliorate stress behavior in mice by increasing monoamine levels and mRNA expression of dopamine receptors (D1 and D2) and synaptophysin. Front Pharmacol 13:915595

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Ma EL et al (2017) Bidirectional brain-gut interactions and chronic pathological changes after traumatic brain injury in mice. Brain Behav Immun 66:56–69

    CAS  PubMed  PubMed Central  Google Scholar 

  138. McCullers DL et al (2002) Traumatic brain injury regulates adrenocorticosteroid receptor mRNA levels in rat hippocampus. Brain Res 947(1):41–49

    CAS  PubMed  Google Scholar 

  139. Gottesfeld Z, Moore AN, Dash PK (2002) Acute ethanol intake attenuates inflammatory cytokines after brain injury in rats: a possible role for corticosterone. J Neurotrauma 19(3):317–326

    PubMed  Google Scholar 

  140. Kuo L-T, Lu H-Y, Chen Y-H (2024) Traumatic brain injury-induced disruption of the circadian clock. J Mol Med, : p. 1–12

  141. DSouza AA et al (2024) Mild repetitive TBI reduces brain-derived neurotrophic factor (BDNF) in the substantia nigra and hippocampus: a preclinical model for testing BDNF-targeted therapeutics. Exp Neurol 374:114696

    CAS  PubMed  Google Scholar 

  142. Raghupathi R (2004) Cell death mechanisms following traumatic brain injury. Brain Pathol 14(2):215–222

    PubMed  Google Scholar 

  143. Butler T, Kassed C, Pennypacker K (2003) Signal transduction and neurosurvival in experimental models of brain injury. Brain Res Bull 59(5):339–351

    CAS  PubMed  Google Scholar 

  144. Rothman MS et al (2007) The neuroendocrine effects of traumatic brain injury. J Neuropsychiatry Clin Neurosci 19(4):363–372

    CAS  PubMed  Google Scholar 

  145. Wright DW et al (2001) Serum progesterone levels correlate with decreased cerebral edema after traumatic brain injury in male rats. J Neurotrauma 18(9):901–909

    CAS  PubMed  Google Scholar 

  146. Lopez-Rodriguez AB et al (2015) Correlation of brain levels of progesterone and dehydroepiandrosterone with neurological recovery after traumatic brain injury in female mice. Psychoneuroendocrinology 56:1–11

    CAS  PubMed  Google Scholar 

  147. Si D et al (2014) Progesterone protects blood–brain barrier function and improves neurological outcome following traumatic brain injury in rats. Experimental Therapeutic Med 8(3):1010–1014

    CAS  Google Scholar 

  148. Geddes RI et al (2014) Progesterone treatment shows benefit in a pediatric model of moderate to severe bilateral brain injury. PLoS ONE 9(1):e87252

    PubMed  PubMed Central  Google Scholar 

  149. Pascual JL et al (2013) Neuroprotective effects of progesterone in traumatic brain injury: blunted in vivo neutrophil activation at the blood-brain barrier. Am J Surg 206(6):840–846

    PubMed  PubMed Central  Google Scholar 

  150. Lopez-Rodriguez AB et al (2016) Profiling neuroactive steroid levels after traumatic brain injury in male mice. Endocrinology 157(10):3983–3993

    CAS  PubMed  Google Scholar 

  151. Fanaei H et al (2014) Testosterone enhances functional recovery after stroke through promotion of antioxidant defenses, BDNF levels and neurogenesis in male rats. Brain Res 1558:74–83

    CAS  PubMed  Google Scholar 

  152. Carteri RB et al (2019) Testosterone administration after traumatic brain injury reduces mitochondrial dysfunction and neurodegeneration. J Neurotrauma 36(14):2246–2259

    PubMed  PubMed Central  Google Scholar 

  153. Zheng C et al (2022) Mechanism of progesterone in treatment of traumatic brain injury based on network pharmacology and molecular docking technology. Med Sci Monitor: Int Med J Experimental Clin Res 28:e937564–e937561

    CAS  Google Scholar 

  154. Mannix R et al (2014) Sex differences in the effect of progesterone after controlled cortical impact in adolescent mice: a preliminary study. J Neurosurg 121(6):1337–1341

    PubMed  PubMed Central  Google Scholar 

  155. Ljunggren L et al (2024) Effects of probiotic supplementation on testosterone levels in healthy ageing men: a 12-week double-blind, placebo-controlled randomized clinical trial. Contemp Clin Trials Commun, : p. 101300

  156. Stein DG (2001) Brain damage, sex hormones and recovery: a new role for progesterone and estrogen? Trends Neurosci 24(7):386–391

    CAS  PubMed  Google Scholar 

  157. Adembri C et al (2014) Minocycline but not tigecycline is neuroprotective and reduces the neuroinflammatory response induced by the superimposition of sepsis upon traumatic brain injury. Crit Care Med 42(8):e570–e582

    CAS  PubMed  Google Scholar 

  158. Zeissig S et al (2006) Changes in expression and distribution of claudin-2,-5 and-8 lead to discontinuous tight junctions and barrier dysfunction in active Crohn’s disease. Gut

  159. Cone RA (2009) Barrier properties of mucus. Adv Drug Deliv Rev 61(2):75–85

    CAS  PubMed  Google Scholar 

  160. Hollingsworth MA, Swanson BJ (2004) Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer 4(1):45–60

    CAS  PubMed  Google Scholar 

  161. Melhem H, Regan-Komito D, Niess JH (2021) Mucins dynamics in physiological and pathological conditions. Int J Mol Sci 22(24):13642

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Vereecke L, Beyaert R, van Loo G (2011) Enterocyte death and intestinal barrier maintenance in homeostasis and disease. Trends Mol Med 17(10):584–593

    CAS  PubMed  Google Scholar 

  163. Bergstrom KS et al (2010) Muc2 protects against lethal infectious colitis by disassociating pathogenic and commensal bacteria from the colonic mucosa. PLoS Pathog 6(5):e1000902

    PubMed  PubMed Central  Google Scholar 

  164. Enss M-L et al (2000) Proinflammatory cytokines trigger MUC gene expression and mucin release in the intestinal cancer cell line LS180. Inflamm Res 49:162–169

    CAS  PubMed  Google Scholar 

  165. Tawiah A et al (2018) High MUC2 mucin expression and misfolding induce cellular stress, reactive oxygen production, and apoptosis in goblet cells. Am J Pathol 188(6):1354–1373

    CAS  PubMed  Google Scholar 

  166. Pang C et al (2015) The effect of trans-resveratrol on post-stroke depression via regulation of hypothalamus–pituitary–adrenal axis. Neuropharmacology 97:447–456

    CAS  PubMed  Google Scholar 

  167. Saunders PR et al (2002) Physical and psychological stress in rats enhances colonic epithelial permeability via peripheral CRH. Dig Dis Sci 47:208–215

    PubMed  Google Scholar 

  168. Kumar M et al (2017) Probiotic mixture VSL# 3 reduces colonic inflammation and improves intestinal barrier function in Muc2 mucin-deficient mice. Am J Physiology-Gastrointestinal Liver Physiol 312(1):G34–G45

    Google Scholar 

  169. Grin’kina NM et al (2016) Righting reflex predicts long-term histological and behavioral outcomes in a closed head model of traumatic brain injury. PLoS ONE 11(9):e0161053

    PubMed  PubMed Central  Google Scholar 

  170. Lowing JL et al (2014) Experimental traumatic brain injury alters ethanol consumption and sensitivity. J Neurotrauma 31(20):1700–1710

    PubMed  PubMed Central  Google Scholar 

  171. den Besten G et al (2013) Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids. Am J Physiology-Gastrointestinal Liver Physiol 305(12):G900–G910

    Google Scholar 

  172. La Rue A et al (1997) Nutritional status and cognitive functioning in a normally aging sample: a 6-y reassessment. Am J Clin Nutr 65(1):20–29

    PubMed  Google Scholar 

  173. Clarke R et al (2010) Effects of lowering homocysteine levels with B vitamins on cardiovascular disease, cancer, and cause-specific mortality: meta-analysis of 8 randomized trials involving 37 485 individuals. Arch Intern Med 170(18):1622–1631

    CAS  PubMed  Google Scholar 

  174. Tsang CK, Kamei Y (2002) Novel effect of vitamin K1 (phylloquinone) and vitamin K2 (menaquinone) on promoting nerve growth factor-mediated neurite outgrowth from PC12D cells. Neurosci Lett 323(1):9–12

    CAS  PubMed  Google Scholar 

  175. Eltokhi A et al (2020) Dysregulation of synaptic pruning as a possible link between intestinal microbiota dysbiosis and neuropsychiatric disorders. J Neurosci Res 98(7):1335–1369

    CAS  PubMed  Google Scholar 

  176. Mahajan C et al (2023) Characteristics of gut microbiome after traumatic brain injury. J Neurosurg Anesthesiol 35(1):86–90

    PubMed  Google Scholar 

  177. Yuan B, Lu X-j, Wu Q (2021) Gut microbiota and acute central nervous system injury: a new target for therapeutic intervention. Front Immunol 12:800796

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Pathare N et al (2020) The impact of traumatic brain injury on microbiome composition: a systematic review. Biol Res Nurs 22(4):495–505

    PubMed  Google Scholar 

  179. Wang H et al (2016) Effect of probiotics on central nervous system functions in animals and humans: a systematic review. J Neurogastroenterol Motil 22(4):589

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Davis IV (2022) Fecal microbiota transfer attenuates gut dysbiosis and functional deficits after traumatic brain injury. Shock 57(6):251–259

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Liu S-X et al (2017) Fecal microbiota transplantation induces remission of infantile allergic colitis through gut microbiota re-establishment. World J Gastroenterol 23(48):8570

    PubMed  PubMed Central  Google Scholar 

  182. Treangen TJ et al (2018) Traumatic brain injury in mice induces acute bacterial dysbiosis within the fecal microbiome. Front Immunol 9:2757

    PubMed  PubMed Central  Google Scholar 

  183. Chavarro JE et al (2020) Association of birth by cesarean delivery with obesity and type 2 diabetes among adult women. JAMA Netw open 3(4):e202605–e202605

    PubMed  PubMed Central  Google Scholar 

  184. Matharu D et al (2019) Repeated mild traumatic brain injury affects microbial diversity in rat jejunum. J Biosci 44:1–12

    Google Scholar 

  185. Taraskina A et al (2022) Effects of traumatic brain injury on the gut microbiota composition and serum amino acid profile in rats. Cells 11(9):1409

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Villapol S, Loane DJ, Burns MP (2017) Sexual dimorphism in the inflammatory response to traumatic brain injury. Glia 65(9):1423–1438

    PubMed  PubMed Central  Google Scholar 

  187. Free KE et al (2017) Comparable impediment of cognitive function in female and male rats subsequent to daily administration of haloperidol after traumatic brain injury. Exp Neurol 296:62–68

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Geddes RI et al (2016) Progesterone treatment shows benefit in female rats in a pediatric model of controlled cortical impact injury. PLoS ONE 11(1):e0146419

    PubMed  PubMed Central  Google Scholar 

  189. Suzuki T, Bramlett HM, Dietrich WD (2003) The importance of gender on the beneficial effects of posttraumatic hypothermia. Exp Neurol 184(2):1017–1026

    PubMed  Google Scholar 

  190. Shultz SR et al (2017) The potential for animal models to provide insight into mild traumatic brain injury: translational challenges and strategies. Neurosci Biobehavioral Reviews 76:396–414

    Google Scholar 

Download references

Acknowledgements

The authors express their gratitude to NIPER, Hyderabad for providing the necessary resources.

Funding

This work is funded by the DST-SERB (SRG/2020/002439).

Author information

Authors and Affiliations

Authors

Contributions

TP conceptualized the study design and executed all the experiments, data analysis, and manuscript drafting. HP analyzed SCFAs using NMR, and MPD assisted in the conceptualization of the hypothesis, and supervised the study, and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Manoj P. Dandekar.

Ethics declarations

Competing Interests

The authors declare no competing interests.

Ethics Approval

The animal studies were approved by the Institutional Animal Ethics Committee of the NIPER, Hyderabad (Approval number: NIP/10/2020/PC/377a). The studies were conducted in accordance with the local legislation and institutional requirements.

Consent to Participate

Not applicable.

Consent to Publish

Not applicable.

Additional information

Publisher’s Note

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

Electronic Supplementary Material

Below is the link to the electronic supplementary material.

Supplementary Material 1

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

Pasam, T., Padhy, H.P. & Dandekar, M.P. Lactobacillus Helveticus Improves Controlled Cortical Impact Injury-Generated Neurological Aberrations by Remodeling of Gut-Brain Axis Mediators. Neurochem Res 50, 3 (2025). https://doi.org/10.1007/s11064-024-04251-4

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11064-024-04251-4

Keywords