- Department of Neurosurgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
- Department of Neurosurgery, Queen Mary Hospital, Pok Fu Lam, Hong Kong Hospital Authority, Hong Kong
Correspondence Address:
Benjamin W. Y. Lo, Department of Neurosurgery, Queen Mary Hospital, Hong Kong Hospital Authority, Hong Kong.
DOI:10.25259/SNI_703_2024
Copyright: © 2024 Surgical Neurology International This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, transform, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.How to cite this article: Brian D. Y. Ma1, Travis Y. H. Chan1, Benjamin W. Y. Lo2. Unveiling the hidden culprit: How the brain-gut axis fuels neuroinflammation in ischemic stroke. 01-Nov-2024;15:394
How to cite this URL: Brian D. Y. Ma1, Travis Y. H. Chan1, Benjamin W. Y. Lo2. Unveiling the hidden culprit: How the brain-gut axis fuels neuroinflammation in ischemic stroke. 01-Nov-2024;15:394. Available from: https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13198
Abstract
Background: The brain-gut axis represents a bidirectional communication network between the gut microbiome and the central nervous system that plays an important role in homeostasis. Compelling evidence now confirms that ischemic stroke disrupts this delicate balance by inducing gut dysbiosis.
Methods: A comprehensive literature search was performed in PubMed, Web of Science, and Google Scholar for articles published between January 2000 and January 2023 using relevant keywords. Studies were limited to English and included original studies, literature, and systematic reviewers from peer-reviewed journals which discussed gut microbiota composition in models/subjects with ischemic stroke or assessed stroke impact on gut microbiota. Comments, meeting abstracts, and case reports were excluded. From the 80 relevant articles, we summarized key findings related to gut microbiota changes after stroke and their association with stroke outcomes.
Results: Emerging preclinical evidence underscores the pivotal role of the gut microbiome in glial cell development and function. Germ-free models exhibit compromised microglial activation and impaired cellular debris clearance, exacerbating tissue damage following ischemic stroke. Targeted interventions, including prebiotics, probiotics, and fecal microbiota transplantation, have demonstrated efficacy in rescuing glial phenotypes in preclinical stroke models. Beyond its local effects, the gut microbiome significantly influences systemic immunity. Ischemic stroke polarizes pro-inflammatory phenotypes of neutrophils and T cells, amplifying neurovascular inflammation. Microbiota manipulation modulates leukocyte trafficking and metabolic signaling, offering potential avenues to mitigate infarct pathology.
Conclusion: Our review demonstrates that in preclinical stroke models, modulating the lipopolysaccharide, short-chain fatty acid, and trimethylamine N-oxide pathways through the gut-brain axis reduces infarct sizes and edema and improves functional recovery after ischemic stroke. Further exploration of this important axis may unveil additional adjunctive stroke therapies by elucidating the complex interplay between the microbiome and the brain. Rigorously controlled clinical studies are now warranted to translate these promising preclinical findings and investigate whether manipulating the microbiome-brain relationship can help improve outcomes for stroke patients. Overall, continued research on the gut-brain axis holds exciting possibilities for developing novel treatment strategies that may enhance recovery after stroke.
Keywords: Brain-gut axis, Ischemic stroke, Neuroinflammation, Neurovascular inflammation
INTRODUCTION
Ischemic strokes secondary to cerebral blood vessel occlusions deprive brain tissue of oxygen, potentially leading to a secondary injury cascade.[
Recently, increasing research has highlighted the potential role of the brain-gut axis, a bidirectional communication network linking the central nervous system (CNS) and gastrointestinal tract, in stroke pathophysiology.[
The gut microbiota is an essential component of the brain-gut axis involving trillions of microorganisms, including bacteria, viruses, fungi, and protozoa. The predominant population includes three main bacterial phyla, namely Firmicutes, Actinobacteria, and Bacteroidetes, accounting for over 90% of the total gut flora.[
In the symbiotic relationship with the host, the gut microbiota produces neuroactive metabolites, including neurotransmitters or their precursors and different microbial metabolites, which directly influence the brain-gut axis.[
Gut microbiota is closely associated with the regulation of various neural pathways. For instance, the ENS’s maturation, which controls gut processes such as mucosal immunity, peristalsis, and motility, is closely linked to microbiota.[
Another crucial pathway is the ANS. In the context of the brain-gut axis, the vagus nerve has been studied for its interoceptive awareness.[
The role of the gut-brain axis has been under investigation in other diseases, including depression, dementia, and Parkinson’s disease.[
MATERIALS AND METHODS
A comprehensive literature search was performed in PubMed, Web of Science and Google Scholar for articles published between January 2000 to January 2023 using relevant keywords. Studies were limited to English and included original studies, literature and systematic reviewers from peer-reviewed journals which discussed gut microbiota composition in models/subjects with ischemic stroke or assessed stroke impact on gut microbiota. Comments, meeting abstracts and case reports were excluded. From the 80 relevant articles, we summarized key findings related to gut microbiota changes after stroke and their association with stroke outcomes.
RESULTS
Emerging preclinical evidence underscores the pivotal role of the gut microbiome in glial cell development and function. Germ-free models exhibit compromised microglial activation and impaired cellular debris clearance, exacerbating tissue damage following ischemic stroke. Targeted interventions, including prebiotics, probiotics, and fecal microbiota transplantation have demonstrated efficacy in rescuing glial phenotypes in preclinical stroke models. Beyond its local effects, the gut microbiome significantly influences systemic immunity. Ischemic stroke polarizes pro-inflammatory phenotypes of neutrophils and T cells, amplifying neurovascular inflammation. Microbiota manipulation modulates leukocyte trafficking and metabolic signaling, offering potential avenues to mitigate infarct pathology.
DISCUSSION
Effect of the Brain-Gut Axis During Physiological Conditions
Under normal physiological conditions, the primary function of the gut-brain axis is to regulate and maintain the homeostasis of the digestive system and its associated organs. It also has secondary modulatory functions affecting the immune system, metabolism, and mood. Several key pathways involving molecules synthesized by gut microbiota are outlined below.
Lipopolysaccharide, simplified as LPS, is an endotoxin derived from the outer membranes of Gram-negative bacteria. Gut flora is the principal source of LPS in healthy people.[
Another key metabolite produced by the gut microbiota important in the gut-brain axis is SCFAs. SCFAs are bacterial fermentation byproducts of dietary fibers, particularly from Prevotella and Ruminococcus gut microbiota species. Acetate is the most abundant SCFA produced by the colon, making up 60–70% of the total SCFA population, while propionate comprises around 20–25%, and butyrate is the least at 15%.[
Finally, trimethylamine (TMA) is also important in the gut-brain axis. This metabolite is a product of digestion from dietary choline and L-carnitine by intestinal microbiota, mainly found in red meat and seafood. TMA is transported to the liver through the portal vein and is oxidized to trimethylamine-N-oxide (TMAO) by liver flavin monooxygenase 3.[
GUT-brain axis in stroke pathophysiology
Research has shed light on the significance of gut microbiota on the modulation of neuroimmunological functions modulated by metabolites such as LPS and SCFA. This immunomodulatory role of gut microbiota becomes particularly crucial during stroke, where the immune system can either exacerbate or ameliorate stroke outcomes. Recent studies have shown that respiratory or urinary tract infections increase the risk of ischemic stroke by 3.19 and 2.72, respectively.[
Central immune system
Stroke pathology in the hyperacute stage involves the extensive release of damage-associated molecular patterns (DAMPs) into systemic blood circulation, such as adenosine triphosphate, high mobility group box 1, nucleic acids, and peroxiredoxin family proteins.[
Figure 1:
The gut-brain axis’ major central immune system pathways which contribute to stroke outcomes. In germ-free mice, microglial activation pathways such as mitogen-activated protein kinase 8 and interleukin-1α are downregulated, which decreases the number of functional astrocytes, leading to worsened cognitive outcomes in stroke. On the other hand, short-chain fatty acid treated with nongerm-free mice causes reduced CSFR1 expression with bone marrow and spleen cells, improving microglial clearance.
Microglia
Under normal physiological conditions, diverse gut microbiota regulate microglial maturation and function at the mucosal surface. Under these circumstances, microglia play an important role in the hyperacute phase of stroke by removing cellular debris from infarcted tissues.[
SCFAs likely mediate the effects of gut microbiota on microglial maturation. Studies on GF mice have shown that administering SCFAs in drinking water for 4 weeks can normalize microglial density and morphology, induce a resting phenotype in microglia, and improve stroke prognosis.[
Astrocyte
Astrocytes are another important cell type involved in the neuroinflammatory cascade postacute ischemic stroke, interacting with microglia in a tightly regulated fashion.[
Gut microbiota can modulate astrocyte function through TLR and LPS signaling pathways.[
Histological studies conducted on GF mice and specific pathogen-free (SPF) mice showed a lower proportion of astrocytes in GF mice compared to SPF mice.[
Peripheral immune systems
Activation of the central immune system by DAMPs alters the BBB function. Instead of excluding peripheral immune cells, the BBB facilitates the infiltration of neutrophils, lymphocytes, and monocytes with the brain parenchyma, where they typically aggregate around the area of infarcted tissue [
Figure 2:
A complex interaction exists between the brain-gut axis and peripheral immune cell components. Germ-free mice models show a decrease in inflammatory pathway activations, reducing the level of neutrophil priming crucial for the clearance of cellular debris. The gut dysbiosis effect upregulates the presence of inflammatory neutrophils and T cells, further worsening the disrupted blood–brain barrier’s patency and adversely affecting stroke clinical outcomes. These effects are contrasted with nongerm-free mice or supplement-treated mice. Upregulation of neuroprotective cells and anti-inflammatory cytokines play important roles in limiting the extent of stroke damage.
Neutrophil
One of the principal recruited cell types is circulating neutrophils, which undergo several migration phases before ultimately crossing the endothelium through paracellular or transcellular mechanisms to travel to areas of necrotic tissue.[
Gut microbiota regulates neutrophil production under normal physiological conditions. Antibiotic-treated and GF models show a substantial decrease in neutrophil numbers in both adults and neonates. GF mice have decreased levels of circulating LPS within their bloodstream, reducing available LPS to bind with TLRs and nucleotide oligomerization domains (NODs) and downregulating TLR4/Myd88 signaling cascade responsible for producing granulocyte colony-stimulating factor.[
T cells
As one of the most implicated immune cell types in stroke pathophysiology, T lymphocytes infiltrate the BBB similarly to neutrophils through selectin-mediated adhesion, rolling, and transmigration of the endothelium.[
T cells can mediate poststroke inflammation through both antigen-dependent and antigen-independent mechanisms. Antigen-independent T-cell responses occur first, involving the peripheral infiltration of crucial stroke neuroinflammatory T cells, including CD3+, CD4+, γδ CD8-, and regulatory T (Treg) cells.[
As shown above, gut microbiota plays an important role in T-cell priming. Stroke-induced gut dysbiosis triggers T-cell polarization toward proinflammatory Th17 and Th1 cells in the Peyer’s patches of transplanted GF mice due to the upregulation of cytokines IL-17 and interferon-gamma.[
Treg cells form a critical subset of T cells that assume a key role in modulating the transition from inflammatory to neuroprotective effects. They account for 5–10% of the peripheral CD4+ T-cell population and are characterized by the expression of transcription factor forkhead box protein P3 (FoxP3). Depletion of Treg cells in animal studies leads to expansion of infarct size and uncontrolled neutrophil and T-cell activation. Conversely, fecal matter transplant (FMT) has improved clinical outcomes in stroke models by increasing the number of FoxP3+ Treg cells.[
In contrast, FMT improves clinical outcomes in the MCAO stroke model through an observed increase in the number of FoxP3+ Treg cells.[
Monocytes
After T cells, monocytes are the subsequent cell type mediating the acute inflammatory phase of ischemic stroke, entering the brain within the first 24 hours and reaching its maximum level within 3–5 days.[
Therapeutics
Therapeutic strategies modulate the gut-brain axis based on the 3 a forementioned metabolite pathways in the gut-brain axis: LPS, SCFA, and TMAO. Some of these interventions achieve protective effects by reducing ictal or recurrent ischemic events, while other interventions reduce neuroinflammatory cascades following ischemic stroke to improve poststroke prognosis [
Figure 3:
Lipopolysaccharide preconditions and dietary changes act as preventative measures, reducing the chance of ischemic stroke by reducing the overall level of systemic inflammation. Probiotics, short-chain fatty acid supplementation, and fecal matter transplant can be used in the acute and chronic poststroke phase by acting upon key gastrointestinal lymphoid tissue to reduce inflammatory pathways such as nuclear factor-κB and histone deacetylase and lower systemic levels of inflammatory cytokines. This reduces neuroinflammatory events and limits the severity of stroke outcomes.
LPS pathway
High LPS levels are associated with systemic inflammatory states that precipitate procoagulant states, increasing ischemic stroke risk.[
Probiotics, consumed live bacterial cultures, have therapeutic benefits for stroke patients associated with improved poststroke outcomes and shorter hospital stays, especially when combined with enteral nutrition.[
SCFA pathway
Another important therapeutic pathway is SCFAs, primary metabolites produced by the gut microbiota. SCFA supplements are beneficial in managing stroke outcomes in both early and later phases.[
Adequate fiber consumption, a quotidian dietary intervention, acts as a preventative measure to reduce the incidence of stroke by encouraging microbiota fermentation to produce SCFAs.[
Another major therapeutic strategy acting upon the SCFA pathway is FMT. Systematic reviews based upon preliminary animal studies have concluded that FMT, rich in butyric acid, has been effective in reducing infarct volume and associated cerebral edema, survival rates, and neurological and behavioral outcomes while also reducing blood lipid levels and risk of further thrombosis.[
Besides FMT, direct supplementation of SCFA compounds is an alternative strategy that could be used to manage the gut concentrations of SCFA compounds.[
Another effective SCFA supplementation that has shown promising results is liposome-encapsulated acetate (LIPA). LIPA was shown to modulate chronic neuroinflammatory events, preventing expansion of post infarct size.[
Both FMT and SCFA have shown clinical promise in the treatment of other conditions, but the effects of specific strains of microbiota remain unknown. To support its role in the clinical management of ischemic stroke, we suggest future studies to investigate its long-term effects, which would provide a foundation for its implementation in clinical trials.
Trimethylamine N-oxide (TMAO)
Elevated plasma TMAO levels from TMA-producing gut bacteria are seen in patients with moderate-to-severe ischemic stroke.[
Atherosclerosis is a major risk factor for stroke, as it can reduce cerebral blood flow and increase the risk of embolic events. As TMAO is a product formed by dietary choline and carnitine metabolism, modification of the levels of these two metabolites may lead to improved stroke outcomes.[
On the other hand, dietary interventions have seen success in reducing circulating TMAO levels. This includes a vegan diet, which showed a rapid decrease in TMAO levels from baseline within 8 weeks, and also with improvements seen in lipid profile markers.[
CONCLUSION
The gut-brain axis is a complex and dynamic system critical in stroke pathophysiology. The gut microbiota produces key metabolites such as LPS, SCFA, and TMAO, which are involved in maintaining normal homeostatic brain functions.
However, growing evidence suggests that disruption in the levels of these metabolites can lead to immune pathway dysregulation affecting both the central and peripheral immune systems, thereby exacerbating stroke outcomes. Interventions targeting gut microbiota composition and its metabolites offer promise in mitigating stroke-related damage. However, the exact importance of the gut-brain axis in these interventions is not yet fully understood. Further research is needed to elucidate the precise mechanisms associated with these therapies and to translate these findings into effective clinical interventions. In summary, the gut-brain axis represents a promising area of research for improving our understanding and treatment of stroke.
Ethical approval
The Institutional Review Board has waived the ethical approval for this study as it is retrospective and qualitative in nature without unique patient identifiers.
Declaration of patient consent
The authors certify that they have obtained all appropriate patient consent.
Financial support and sponsorship
Nil.
Conflicts of interest
There are no conflicts of interest.
Use of artificial intelligence (AI)-assisted technology for manuscript preparation
The authors confirm that there was no use of artificial intelligence (AI)-assisted technology for assisting in the writing or editing of the manuscript and no images were manipulated using AI.
Disclaimer
The views and opinions expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Journal or its management. The information contained in this article should not be considered to be medical advice; patients should consult their own physicians for advice as to their specific medical needs.
References
1. Allaire JM, Crowley SM, Law HT, Chang S, Ko H, Vallance BA. The intestinal epithelium: Central coordinator of mucosal immunity. Trends Immunol. 2018. 39: 677-96
2. Argyridou S, Davies MJ, Biddle GJ, Bernieh D, Suzuki T, Dawkins NP. Evaluation of an 8-week vegan diet on plasma trimethylamine-N-oxide and postchallenge glucose in adults with dysglycemia or obesity. J Nutr. 2021. 151: 1844-53
3. Asahi M, Wang X, Mori T, Sumii T, Jung JC, Moskowitz MA. Effects of matrix metalloproteinase-9 gene knock-out on the proteolysis of blood-brain barrier and white matter components after cerebral ischemia. J Neurosci. 2001. 21: 7724-32
4. Barroso A, Mahler JV, Fonseca-Castro PH, Quintana FJ. The aryl hydrocarbon receptor and the gut-brain axis. Cell Mol Immunol. 2021. 18: 259-68
5. Battaglini D, Pimentel-Coelho PM, Robba C, Dos Santos CC, Cruz FF, Pelosi P. Gut microbiota in acute ischemic stroke: From pathophysiology to therapeutic implications. Front Neurol. 2020. 11: 598
6. Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016. 22: 516-23
7. Benakis C, Liesz A. The gut-brain axis in ischemic stroke: Its relevance in pathology and as a therapeutic target. Neurol Res Pract. 2022. 4: 57
8. Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR, Cheng S. Heart disease and stroke statistics-2018 update: A report from the American Heart Association. Circulation. 2018. 137: e67-492
9. Bonaz B, Bazin T, Pellissier S. The vagus nerve at the interface of the microbiota-gut-brain axis. Front Neurosci. 2018. 12: 49
10. Buffone A, Anderson NR, Hammer DA. Human neutrophils will crawl upstream on ICAM-1 If Mac-1 is blocked. Biophys J. 2019. 117: 1393-404
11. Bugl S, Wirths S, Radsak MP, Schild H, Stein P, André MC. Steady-state neutrophil homeostasis is dependent on TLR4/TRIF signaling. Blood. 2013. 121: 723-33
12. Candelli M, Franza L, Pignataro G, Ojetti V, Covino M, Piccioni A. Interaction between lipopolysaccharide and gut microbiota in inflammatory bowel diseases. Int J Mol Sci. 2021. 22: 6242
13. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015. 28: 203-9
14. Chen R, Xu Y, Wu P, Zhou H, Lasanajak Y, Fang Y. Transplantation of fecal microbiota rich in short chain fatty acids and butyric acid treat cerebral ischemic stroke by regulating gut microbiota. Pharmacol Res. 2019. 148: 104403
15. Chen Y, Xu J, Chen Y. Regulation of neurotransmitters by the gut microbiota and effects on cognition in neurological disorders. Nutrients. 2021. 13: 2099
16. Cho YS. Fecal microbiota transplantation is effective for the treatment of partially treated Clostridioides difficile infection. Gut Liver. 2021. 15: 1-2
17. Congdon KL, Sanchez-Perez LA, Sampson JH. Effective effectors: How T cells access and infiltrate the central nervous system. Pharmacol Ther. 2019. 197: 52-60
18. De Filippis F, Pellegrini N, Vannini L, Jeffery IB, La Storia A, Laghi L. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2015. 65: 1812-21
19. Dinan TG, Cryan JF. The microbiome-gut-brain axis in health and disease. Gastroenterol Clin N Am. 2017. 46: 77-89
20. Dong R, Huang R, Wang J, Liu H, Xu Z. Effects of microglial activation and polarization on brain injury after stroke. Front Neurol. 2021. 12: 620948
21. Du J, Miao M, Lu Z, Chen H, Bao A, Che B. Plasma l-carnitine and risks of cardiovascular events and recurrent stroke after ischemic stroke: A nested case-control study. Nutr Metab Cardiovasc Dis. 2022. 32: 2579-87
22. Egerod KL, Petersen N, Timshel PN, Rekling JC, Wang Y, Liu Q. Profiling of G protein-coupled receptors in vagal afferents reveals novel gut-to-brain sensing mechanisms. Mol Metab. 2018. 12: 62-75
23. Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015. 18: 965-77
24. Feigin VL, Brainin M, Norrving B, Martins SC, Sacco RL, Hacke W. World Stroke Organization (WSO): Global stroke fact sheet 2022. Int J Stroke. 2022. 17: 18-29
25. Garcia-Bonilla L, Brea D, Benakis C, Lane DA, Murphy M, Moore J. Endogenous protection from ischemic brain injury by preconditioned monocytes. J Neurosci. 2018. 38: 6722-36
26. Geng ZH, Zhu Y, Li QL, Zhao C, Zhou PH. Enteric nervous system: The bridge between the gut microbiota and neurological disorders. Front Aging Neurosci. 2022. 14: 810483
27. Gelderblom M, Weymar A, Bernreuther C, Velden J, Arunachalam P, Steinbach K. Neutralization of the IL-17 axis diminishes neutrophil invasion and protects from ischemic stroke. Blood. 2012. 120: 3793-802
28. Griffin S, Clark JB, Canevari L. Astrocyte-neurone communication following oxygen-glucose deprivation. J Neurochem. 2005. 95: 1015-22
29. Hayakawa K, Miyamoto N, Seo JH, Pham LD, Kim KW, Lo EH. High-mobility group box 1 from reactive astrocytes enhances the accumulation of endothelial progenitor cells in damaged white matter. J Neurochem. 2013. 125: 273-80
30. Haghikia A, Li XS, Liman TG, Bledau N, Schmidt D, Zimmermann F. Gut microbiota-dependent trimethylamine N-oxide predicts risk of cardiovascular events in patients with stroke and is related to proinflammatory monocytes. Arterioscler Thromb Vasc Biol. 2018. 38: 2225-35
31. Hines DJ, Choi HB, Hines RM, Phillips AG, MacVicar BA. Prevention of LPS-induced microglia activation, cytokine production and sickness behavior with TLR4 receptor interfering peptides. PLoS One. 2013. 8: e60388
32. Huang Y, Wu J, Zhang H, Li Y, Wen L, Tan X. The gut microbiome modulates the transformation of microglial subtypes. Mol Psychiatry. 2023. 28: 1611-21
33. Jacob MA, Ekker MS, Allach Y, Cai M, Aarnio K, Arauz A. Global differences in risk factors, etiology, and outcome of ischemic stroke in young adults-a worldwide meta-analysis: The GOAL initiative. Neurology. 2022. 98: e573-88
34. Jing L, Zhang H, Xiang Q, Shen L, Guo X, Zhai C. Targeting trimethylamine N-Oxide: a new therapeutic strategy for alleviating atherosclerosis. Front Cardiovasc Med. 2022. 9: 864600
35. Kaito M, Araya S, Gondo Y, Fujita M, Minato N, Nakanishi M. Relevance of distinct monocyte subsets to clinical course of ischemic stroke patients. PLoS One. 2013. 8: e69409
36. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013. 19: 576-85
37. King CH, Desai H, Sylvetsky AC, LoTempio J, Ayanyan S, Carrie J. Baseline human gut microbiota profile in healthy people and standard reporting template. PLoS One. 2019. 14: e0206484
38. Krishnamurthi RV, Feigin VL, Forouzanfar MH, Mensah GA, Connor M, Bennett DA. Global and regional burden of first-ever ischaemic and haemorrhagic stroke during 1990-2010: findings from the Global Burden of Disease Study 2010. Lancet Glob Health. 2013. 1: e259-81
39. Krummel MF, Bartumeus F, Gérard A. T cell migration, search strategies and mechanisms. Nat Rev Immunol. 2016. 16: 193-201
40. Liao S, Yang Z, Sheng H, Liu M, Sun Q. Effects of long-term vagus nerve electrical stimulation therapy on acute cerebral infarction and neurological function recovery in post MCAO mice. Oxid Med Cell Longev. 2022. 2022: 8131391
41. Liesz A, Zhou W, Mracskó É, Karcher S, Bauer H, Schwarting S. Inhibition of lymphocyte trafficking shields the brain against deleterious neuroinflammation after stroke. Brain. 2011. 134: 704-20
42. Li T, Liesz A. Immunity in stroke: The next frontier. Thromb Haemost. 2022. 122: 1454-60
43. Li M, van Esch BC, Henricks PA, Folkerts G, Garssen J. The anti-inflammatory effects of short chain fatty acids on lipopolysaccharide-or tumor necrosis factor α-stimulated endothelial cells via activation of GPR41/43 and inhibition of HDACs. Front Pharmacol. 2018. 9: 533
44. Mei J, Liu Y, Dai N, Hoffmann C, Hudock KM, Zhang P. Cxcr2 and Cxcl5 regulate the IL-17/G-CSF axis and neutrophil homeostasis in mice. J Clin Invest. 2012. 122: 974-86
45. Mueller NT, Bakacs E, Combellick J, Grigoryan Z, Dominguez-Bello MG. The infant microbiome development: Mom matters. Trends Mol Med. 2015. 21: 109-17
46. Nanì S, Fumagalli L, Sinha U, Kamen L, Scapini P, Berton G. Src family kinases and Syk are required for neutrophil extracellular trap formation in response to β-glucan particles. J Innate Immun. 2015. 7: 59-73
47. Nogal A, Valdes AM, Menni C. The role of short-chain fatty acids in the interplay between gut microbiota and diet in cardio-metabolic health. Gut Microbes. 2021. 13: 1-24
48. Ohkubo T, Tsuda M, Tamura M, Yamamura M. Impaired superoxide production in peripheral blood neutrophils of germ-free rats. Scand J Immunol. 1990. 32: 727-9
49. Oliver A, Chase AB, Weihe C, Orchanian SB, Riedel SF, Hendrickson CL. High-fiber, whole-food dietary intervention alters the human gut microbiome but not fecal short-chain fatty acids. mSystems. 2021. 6: e00115-21
50. Paciorek A, Skora L. Vagus nerve stimulation as a gateway to interoception. Front Psychol. 2020. 11: 1659
51. Park MJ, Sohrabji F. The histone deacetylase inhibitor, sodium butyrate, exhibits neuroprotective effects for ischemic stroke in middle-aged female rats. J Neuroinflammation. 2016. 13: 300
52. Pasokh A, Farzipour M, Mahmoudi J, Sadigh-Eteghad S. The effect of fecal microbiota transplantation on stroke outcomes: A systematic review. J Stroke Cerebrovasc Dis. 2022. 31: 106727
53. Peh A, O’Donnell JA, Broughton BR, Marques FZ. Gut microbiota and their metabolites in stroke: A double-Edged Sword. Stroke. 2022. 53: 1788-801
54. Pluta R, Januszewski S. Gut microbiota neurotransmitters: Influence on risk and outcome of ischemic stroke. Neural Regen Res. 2023. 18: 1707-8
55. Pluta R, Januszewski S, Czuczwar SJ. The role of gut microbiota in an ischemic stroke. Int J Mol Sci. 2021. 22: 915
56. Ridet JL, Privat A, Malhotra SK, Gage FH. Reactive astrocytes: Cellular and molecular cues to biological function. Trends Neurosci. 1997. 20: 570-7
57. Rothhammer V, Borucki DM, Tjon EC, Takenaka MC, Chao CC, Ardura-Fabregat A. Microglial control of astrocytes in response to microbial metabolites. Nature. 2018. 557: 724-8
58. Rutsch A, Kantsjö JB, Ronchi F. The Gut-Brain axis: How microbiota and host inflammasome influence brain physiology and pathology. Front Immunol. 2020. 11: 604179
59. Shen XY, Gao ZK, Han Y, Yuan M, Guo YS, Bi X. Activation and role of astrocytes in ischemic stroke. Front Cell Neurosci. 2021. 15: 755955
60. Silva YP, Bernardi A, Frozza RL. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol (Lausanne). 2020. 11: 25
61. Singh V, Roth S, Llovera G, Sadler R, Garzetti D, Stecher B. Microbiota dysbiosis controls the neuroinflammatory response after stroke. J Neurosci. 2016. 36: 7428-40
62. Singh V, Sadler R, Heindl S, Llovera G, Roth S, Benakis C. The gut microbiome primes a cerebroprotective immune response after stroke. J Cereb Blood Flow Metab. 2018. 38: 1293-8
63. Smeeth L, Thomas SL, Hall AJ, Hubbard R, Farrington P, Vallance P. Risk of myocardial infarction and stroke after acute infection or vaccination. N Engl J Med. 2004. 351: 2611-8
64. Soliman ML, Puig KL, Combs CK, Rosenberger TA. Acetate reduces microglia inflammatory signaling in vitro. J Neurochem. 2012. 123: 555-67
65. Stanley D, Mason LJ, Mackin KE, Srikhanta YN, Lyras D, Prakash MD. Translocation and dissemination of commensal bacteria in post-stroke infection. Nat Med. 2016. 22: 1277-84
66. Stokum JA, Kurland DB, Gerzanich V, Simard JM. Mechanisms of astrocyte-mediated cerebral edema. Neurochem Res. 2015. 40: 317-28
67. Suganya K, Koo BS. Gut-brain axis: Role of gut microbiota on neurological disorders and how probiotics/prebiotics beneficially modulate microbial and immune pathways to improve brain functions. Int J Mol Sci. 2020. 21: 7551
68. Takata M, Nakagomi T, Kashiwamura S, Nakano-Doi A, Saino O, Nakagomi N. Glucocorticoid-induced TNF receptor-triggered T cells are key modulators for survival/death of neural stem/progenitor cells induced by ischemic stroke. Cell Death Differ. 2012. 19: 756-67
69. Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. 2017. 474: 1823-36
70. Tu R, Xia J. Stroke and vascular cognitive impairment: The role of intestinal microbiota metabolite TMAO. CNS Neurol Disord Drug Targets. 2023. 22: 102-21
71. Usami M, Kishimoto K, Ohata A, Miyoshi M, Aoyama M, Fueda Y. Butyrate and trichostatin A attenuate nuclear factor κB activation and tumor necrosis factor α secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr Res. 2008. 28: 321-8
72. Xia Y, Zhai Q. IL-1beta enhances the antibacterial activity of astrocytes by activation of NF-kappaB. Glia. 2010. 58: 244-52
73. Zera KA, Buckwalter MS. The local and peripheral immune responses to stroke: Implications for therapeutic development. Neurotherapeutics. 2020. 17: 414-35
74. Zhang D, Chen G, Manwani D, Mortha A, Xu C, Faith JJ. Neutrophil ageing is regulated by the microbiome. Nature. 2015. 525: 528-32
75. Zhen J, Zhou Z, He M, Han HX, Lv EH, Wen PB. The gut microbial metabolite trimethylamine N-oxide and cardiovascular diseases. Front Endocrinol (Lausanne). 2023. 14: 1085041
76. Zhong DY, Li L, Ma RM, Deng YH. The effect of probiotics in stroke treatment. Evid Based Complement Alternat Med. 2021. 2021: 4877311
77. Zhou SY, Guo ZN, Yang Y, Qu Y, Jin H. Gut-brain axis: Mechanisms and potential therapeutic strategies for ischemic stroke through immune functions. Front Neurosci. 2023. 17: 1081347
78. Zhou Z, Xu N, Matei N, McBride DW, Ding Y, Liang H. Sodium butyrate attenuated neuronal apoptosis via GPR41/Gβγ/PI3K/Akt pathway after MCAO in rats. J Cereb Blood Flow Metab. 2021. 41: 267-81
79. Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016. 165: 111-24
80. Zhu Y, Li Q, Jiang H. Gut microbiota in atherosclerosis: focus on trimethylamine N-oxide. APMIS. 2020. 128: 353-66