The Microbiota-Gut-Brain Axis: From Mechanistic Understanding to Precision Medicine Applications in Metabolic and Neuropsychiatric Disorders

Introduction

The gut-brain axis has emerged as one of the most significant discoveries in modern medicine, fundamentally changing our understanding of how peripheral organs communicate with the central nervous system [1]. This bidirectional communication network enables the central nervous system to modulate gastrointestinal functions in response to psychological and physiological stressors, while simultaneously allowing the gut microbiota to regulate brain activity through immune, neuroendocrine, and vagal pathways [1].

The concept of gut-brain communication traces back to the 1840s when Beaumont first demonstrated that emotional states influenced digestion rates [2]. However, the modern understanding has evolved to encompass the “microbiota-gut-brain axis,” recognizing the crucial role of gut microorganisms as active mediators rather than passive bystanders [3, 4]. This paradigm shift has profound implications for understanding the pathophysiology of numerous conditions, from metabolic disorders to neuropsychiatric diseases.

Recent research has revealed that the gut microbiota can influence brain function through multiple mechanisms, including the production of neurotransmitters, modulation of inflammatory responses, and synthesis of bioactive metabolites [5]. These discoveries have opened new therapeutic avenues for treating conditions previously thought to be primarily neurological or psychiatric in nature.

Anatomical and Physiological Foundations

Neural Communication Pathways

The vagus nerve serves as the primary neural conduit in the gut-brain axis, comprising approximately 80% afferent and 20% efferent fibers [6, 7]. This mixed nerve acts essentially as a sensory pathway, continuously monitoring the gut environment and transmitting signals to the brainstem. Recent studies have demonstrated that vagal afferent fibers can detect changes in gut microbiota metabolites through enteroendocrine cells, which express toll-like receptors and metabolite-specific receptors [3].

The enteric nervous system, often called the “second brain,” contains approximately 100 million neurons—more than the human spinal cord [8]. This extensive neural network can function independently but maintains constant communication with the central nervous system through the vagus nerve and spinal afferents [4].

Endocrine and Immune Signaling

The hypothalamic-pituitary-adrenal (HPA) axis plays a central role in gut-brain communication, mediating stress responses that directly affect gut function. Stress-induced cortisol release increases intestinal permeability and alters microbial composition, creating a cycle of inflammation and dysbiosis [9, 10]. Conversely, gut microbiota can modulate HPA axis activity through microbial metabolites and inflammatory mediators.

The immune system forms another critical communication pathway, with gut-associated lymphoid tissue representing the largest immune organ in the body [11]. Disruptions in gut barrier function allow bacterial components to enter systemic circulation, triggering inflammatory responses that can affect brain function.

Microbial Metabolite Production

Gut bacteria produce numerous bioactive compounds that influence brain function, with short-chain fatty acids (SCFAs) being among the most significant. Acetate, propionate, and butyrate—the three major SCFAs—are produced through bacterial fermentation of dietary fiber and can cross the blood-brain barrier to exert direct effects on neural function [12–14].

These metabolites influence brain function through multiple mechanisms: they enhance blood-brain barrier integrity, reduce neuroinflammation, promote neurogenesis, and modulate neurotransmitter synthesis [13, 15]. Recent studies have shown that SCFA supplementation can improve cognitive function and reduce depressive behaviors in both animal models and human trials [12–15].

The Microbiota-Gut-Brain Axis in Health and Disease

Neurotransmitter Production by Gut Bacteria

The gut microbiota serves as a significant source of neurotransmitters and their precursors. Specific bacterial genera have been identified as producers of key neurotransmitters: Lactobacillus and Bifidobacterium species produce γ-aminobutyric acid (GABA) [16,17], while Enterococcus and Streptococcus produce serotonin [18, 19].

GABA, the primary inhibitory neurotransmitter in the central nervous system, is particularly significant in the gut-brain axis. Imbalances in GABA are associated with neurological diseases including Alzheimer’s and Parkinson’s disease, as well as psychological disorders such as anxiety and depression. Recent research has identified GABA as a potential postbiotic mediator, with specific bacterial strains capable of producing therapeutic levels of this neurotransmitter. [16]

Microbiome Diversity and Metabolic Function

Healthy gut microbiota diversity is essential for optimal metabolic function and brain health. Dysbiosis—an imbalance in microbial composition—has been linked to insulin resistance, cognitive decline, and neuroinflammation [20]. It is also shown that individuals with metabolic syndrome exhibit reduced microbial diversity and altered SCFA production patterns [20].

The relationship between microbiome diversity and cognitive function is particularly striking in aging populations. Research using germ-free mice and fecal microbiota transplantation has demonstrated that microbiome composition can directly influence neuroplasticity, with implications for neurodevelopment and neurodegeneration [21].

Metabolic Implications of the Gut-Brain Axis

Insulin Resistance and Brain Metabolism

The concept of “brain insulin resistance” has emerged as a critical link between metabolic dysfunction and neurodegeneration. Insulin receptors are widely distributed throughout the brain, particularly in regions involved in memory and executive function [20]. Impaired brain insulin signaling contributes to decreased glucose uptake, increased oxidative stress, and neuroinflammation [20].

Gut microbiota dysbiosis contributes to systemic insulin resistance through multiple mechanisms [22], including increased intestinal permeability [23], chronic low-grade inflammation [24], and altered production of metabolites that influence glucose metabolism [20]. Studies have demonstrated that specific bacterial strains can improve insulin sensitivity through SCFA production and anti-inflammatory effects [20, 25].

Obesity and Energy Homeostasis

The gut-brain axis plays a crucial role in energy homeostasis through regulation of appetite hormones and reward pathways. Ghrelin, the “hunger hormone” produced primarily in the stomach, stimulates food intake by activating NPY/AgRP neurons in the hypothalamus. In obesity, ghrelin levels are paradoxically reduced, yet food intake remains elevated, suggesting complex dysregulation of appetite control [26, 27].

Leptin, produced by adipose tissue, normally signals satiety to the hypothalamus. However, obesity is characterized by leptin resistance, where elevated leptin levels fail to suppress appetite. Recent research has shown that gut microbiota can influence leptin sensitivity through modulation of inflammatory pathways and blood-brain barrier function [26, 27].

Metabolic Endotoxemia and Inflammation

Chronic low-grade inflammation, often termed “metabolic endotoxemia,” represents a key mechanism linking gut dysfunction to metabolic disease. Increased intestinal permeability allows bacterial lipopolysaccharides to enter systemic circulation, triggering inflammatory responses that contribute to insulin resistance and neuroinflammation [24, 28].

This inflammatory cascade can compromise blood-brain barrier integrity, allowing peripheral inflammatory mediators to enter the brain and activate microglia. The resulting neuroinflammation has been implicated in cognitive decline, depression, and neurodegenerative diseases [28, 29].

Mental Health and Neuropsychiatric Disorders

Depression and Anxiety

The relationship between gut microbiota and mood disorders has been extensively documented in both preclinical and clinical studies. Patients with depression often exhibit reduced microbial diversity, altered SCFA production, and elevated inflammatory markers [30]. Clinical trials of psychobiotics—probiotics with mental health benefits—have shown significant improvements in depression and anxiety scores [31].

Specific bacterial strains have demonstrated particular efficacy in treating mood disorders. Lactobacillus helveticus and Bifidobacterium longum combinations have shown consistent benefits across multiple trials, with effects on depression, anxiety, and stress measures [31]. The mechanisms appear to involve modulation of the HPA axis, enhancement of GABA production, and reduction of systemic inflammation [31].

Neurodevelopmental Disorders

Autism spectrum disorder (ASD) represents one of the most intensively studied conditions in gut-brain axis research. Children with ASD frequently experience gastrointestinal symptoms and exhibit distinct gut microbiota patterns compared to neurotypical peers [32]. Studies have identified increased levels of ProteobacteriaActinobacteria, and Sutterella in ASD patients [33].

The mechanisms linking gut dysfunction to ASD symptoms involve immune dysregulation, altered neurotransmitter production, and compromised blood-brain barrier function [34]. Therapeutic interventions targeting the gut microbiota, including probiotics and dietary modifications, have shown promise in improving both gastrointestinal and behavioral symptoms [35, 36].

Neurodegenerative Diseases

The “gut-first” hypothesis of Parkinson’s disease has gained significant support from recent research. This theory proposes that α-synuclein aggregation begins in the enteric nervous system and spreads to the brain via the vagus nerve. Supporting evidence includes the observation that many Parkinson’s patients experience constipation years before motor symptoms appear [37].

Recent studies have demonstrated that autoimmune reactions to α-synuclein in the gut can produce gastrointestinal symptoms resembling early Parkinson’s disease. This research suggests that targeting gut inflammation early in the disease process might prevent progression to brain pathology [37].

Alzheimer’s disease has also been linked to gut microbiota alterations. Studies using germ-free mice have shown that microbiota from Alzheimer’s patients can exacerbate amyloid plaque formation and neuroinflammation when transplanted into disease models. Conversely, beneficial bacteria can reduce neuroinflammation and protect against cognitive decline [38].

Therapeutic Interventions and Clinical Applications

Psychobiotics and Probiotic Interventions

Psychobiotics represent a promising therapeutic approach for mental health disorders, with growing evidence supporting their efficacy. A systematic review of randomized clinical trials involving 3,353 patients found notable effectiveness in treating depression symptoms, with most studies utilizing Lactobacillus and Bifidobacterium strains over 4-24 week treatment periods [39].

Specific probiotic formulations have demonstrated particular promise. The combination of L. helveticus Rosell-52® and B. longum Rosell-175® showed significant results in depression, anxiety, and stress measures across multiple studies. B. longum 1714 and L. paracasei Lpc-37® also demonstrated efficacy in subsets of psychological outcomes [40].

Dietary Interventions

The Mediterranean diet has emerged as a particularly effective intervention for gut-brain axis health. This dietary pattern, characterized by high intake of plant-based foods, monounsaturated fats, and polyphenols, promotes beneficial gut bacteria growth and increases SCFA production [41].

Recent studies have shown that Mediterranean diet adherence is associated with improved cognitive function, reduced neuroinflammation, and enhanced gut microbiota diversity. A recent Tulane University study demonstrated that rats following a Mediterranean diet developed distinct bacterial patterns that correlated with better memory and cognitive performance compared to those on a Western diet [42].

Intermittent fasting has also shown promise as a gut-brain axis intervention. This approach enhances gut microbiota diversity, reduces inflammation, and improves insulin sensitivity while promoting neurogenesis and protecting against cognitive decline [43].

Fecal Microbiota Transplantation

Fecal microbiota transplantation (FMT) represents the most direct approach to modifying gut microbiota composition. While clinical applications have primarily focused on C. difficile infections, emerging research explores FMT’s potential in neurological disorders [44].

Clinical trials have yielded mixed results. A recent randomized clinical trial in Parkinson’s disease found that while FMT was safe, it did not provide clinically meaningful improvements over placebo. However, the study revealed important insights about the complex relationships between donor microbiota composition, dysbiosis resolution, and clinical outcomes [45].

Studies in autism spectrum disorder have shown more promising results, with improvements reported in both gastrointestinal and behavioral symptoms. However, methodological limitations and contradictory findings highlight the need for larger, well-designed trials [35].

Precision Medicine Approaches

The future of gut-brain axis therapeutics lies in personalized medicine approaches that consider individual microbiome profiles, genetic factors, and clinical presentations. Recent advances in microbiome analysis, including AI-powered predictive models, are enabling more precise therapeutic targeting [46–49].

Biomarker development is crucial for clinical implementation. Potential markers include microbial composition profiles, SCFA levels, inflammatory mediators, and gut permeability measures. These biomarkers could guide treatment selection and monitor therapeutic response [49–54].

Mechanisms of Blood-Brain Barrier Modulation

SCFA-Mediated Barrier Protection

Short-chain fatty acids play a critical role in maintaining blood-brain barrier integrity. Recent research has shown that propionate, in particular, can reverse antibiotic-induced blood-brain barrier permeability through free fatty acid receptor 2-dependent mechanisms. This finding has significant implications for understanding how gut dysfunction contributes to neuroinflammation and cognitive decline [13].

The blood-brain barrier’s selective vulnerability to peripheral inflammation varies across brain regions, with some areas being more susceptible to dysfunction than others. This regional heterogeneity necessitates targeted therapeutic approaches that consider specific brain region vulnerabilities [29].

Inflammatory Pathways and Barrier Dysfunction

Chronic peripheral inflammation, often originating from gut dysfunction, can compromise blood-brain barrier integrity through multiple pathways. Inflammatory cytokines can alter tight junction protein expression, increase endothelial permeability, and facilitate immune cell infiltration into the brain [55].

The size of molecules attempting to cross the blood-brain barrier influences the detection of permeability changes. Smaller molecules, including cytokines, may cross more readily than larger proteins, highlighting the importance of using appropriate markers when assessing barrier function in clinical studies [55].

Future Research Directions and Clinical Translation

Mechanistic Understanding

Despite significant progress, several key mechanistic questions remain unanswered. Future research must focus on identifying specific microbial species and metabolites responsible for therapeutic effects, understanding individual variability in response to interventions, and elucidating the temporal dynamics of gut-brain communication [56].

Longitudinal studies are particularly needed to establish causal relationships between microbiome changes and clinical outcomes. Advanced technologies including single-cell transcriptomics, metabolomics, and organ-on-chip models will be crucial for translating preclinical findings into effective clinical applications [57].

Standardization and Regulatory Challenges

The clinical translation of gut-brain axis therapeutics faces significant challenges in standardization and regulation. Current limitations include inter-individual variability in microbiota composition, lack of standardized diagnostic tools, and regulatory frameworks that are not well-adapted to microbiome-based therapies [58].

Addressing these challenges will require collaborative efforts between researchers, clinicians, and regulatory agencies to develop appropriate guidelines for microbiome diagnostics and therapeutics. Standardization of protocols, outcome measures, and safety assessments is essential for advancing the field [58].

Personalized Medicine Integration

The integration of gut-brain axis research into personalized medicine frameworks represents a major opportunity for improving treatment outcomes. Future approaches should incorporate microbiome profiling, genetic analysis, and clinical phenotyping to develop individualized treatment strategies [59].

Artificial intelligence and machine learning will play increasingly important roles in analyzing complex microbiome data and predicting treatment responses. These technologies may enable the identification of optimal therapeutic combinations and dosing regimens for individual patients [59].

Conclusions

The gut-brain axis represents a fundamental communication network that profoundly influences both metabolic and mental health. The bidirectional nature of this system, mediated by neural, immune, endocrine, and metabolic pathways, provides multiple targets for therapeutic intervention. The expanding understanding of how gut microbiota influence brain function through neurotransmitter production, SCFA synthesis, and immune modulation has opened new avenues for treating previously intractable conditions.

Current evidence strongly supports the role of gut dysfunction in metabolic disorders, including insulin resistance and obesity, as well as neuropsychiatric conditions such as depression, anxiety, and neurodevelopmental disorders. The emerging concept of “brain insulin resistance” and the “gut-first” hypothesis of neurodegenerative diseases highlight the importance of peripheral factors in central nervous system pathology.

Therapeutic interventions targeting the gut-brain axis, including psychobiotics, dietary modifications, and fecal microbiota transplantation, show considerable promise but require further refinement. The future success of these approaches will depend on developing personalized medicine strategies that account for individual microbiome profiles and clinical characteristics.

As research in this field advances, the integration of gut-brain axis considerations into standard clinical practice will likely transform the management of metabolic and neuropsychiatric disorders. The conceptualization of the gut as the “second brain” is increasingly supported by empirical evidence, highlighting the profound impact of microbial regulation on brain function and behavior. Continued interdisciplinary collaboration will be essential for translating these scientific discoveries into effective therapeutic strategies that improve patient outcomes and quality of life.

The gut-brain axis represents not merely a research curiosity but a fundamental aspect of human physiology with immediate clinical relevance. Understanding and therapeutically targeting this system may provide the key to addressing some of the most challenging health conditions of our time, from the global obesity epidemic to the rising prevalence of neurodegenerative diseases and mental health disorders.

References

  1. Doenyas C, Clarke G, Cserjési R. Gut–brain axis and neuropsychiatric health: recent advances. Sci Rep 2025; 15: 3415.
  2. Margolis KG, Cryan JF, Mayer EA. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology 2021; 160: 1486–1501.
  3. Bonaz B, Bazin T, Pellissier S. The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis. Front Neurosci; 12. Epub ahead of print 7 February 2018. DOI: 10.3389/fnins.2018.00049.
  4. Mishra C, Mewani A. Exploring Gut Microbiome, Gut-Brain Axis and Relationship with Exercise; Exploring the Impact of Exercise on Metabolic and Mental Well-Being. International Journal of Horticulture, Agriculture and Food science 2025; 9: 17–25.
  5. Chen Y, Xu J, Chen Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021; 13: 2099.
  6. Bonaz B, Sinniger V, Pellissier S. Vagus Nerve Stimulation at the Interface of Brain-Gut Interactions. Cold Spring Harb Perspect Med; 9. Epub ahead of print 1 August 2019. DOI: 10.1101/cshperspect.a034199.
  7. Han Y, Wang B, Gao H, et al. Vagus Nerve and Underlying Impact on the Gut Microbiota-Brain Axis in Behavior and Neurodegenerative Diseases. J Inflamm Res 2022; Volume 15: 6213–6230.
  8. Goldstein A, Hofstra R, Burns A. Building a brain in the gut: development of the enteric nervous system. Clin Genet 2013; 83: 307–316.
  9. Bertollo AG, Santos CF, Bagatini MD, et al. Hypothalamus-pituitary-adrenal and gut-brain axes in biological interaction pathway of the depression. Front Neurosci 2025; 19: 1541075.
  10. Rusch JA, Layden BT, Dugas LR. Signalling cognition: the gut microbiota and hypothalamic-pituitary-adrenal axis. Front Endocrinol (Lausanne) 2023; 14: 1130689.
  11. Appleton J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr Med (Encinitas) 2018; 17: 28–32.
  12. Ni Y, Tong Q, Xu M, et al. Gut Microbiota-Induced Modulation of the Central Nervous System Function in Parkinson’s Disease Through the Gut-Brain Axis and Short-Chain Fatty Acids. Mol Neurobiol 2025; 62: 2480–2492.
  13. Chenghan M, Wanxin L, Bangcheng Z, et al. Short‐chain fatty acids mediate gut microbiota–brain communication and protect the blood–brain barrier integrity. Ann N Y Acad Sci 2025; 1545: 116–131.
  14. Petakh P, Duve K, Oksenych V, et al. Molecular mechanisms and therapeutic possibilities of short-chain fatty acids in posttraumatic stress disorder patients: a mini-review. Front Neurosci; 18. Epub ahead of print 3 June 2024. DOI: 10.3389/fnins.2024.1394953.
  15. Silva YP, Bernardi A, Frozza RL. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front Endocrinol (Lausanne); 11. Epub ahead of print 31 January 2020. DOI: 10.3389/fendo.2020.00025.
  16. Braga JD, Thongngam M, Kumrungsee T. Gamma-aminobutyric acid as a potential postbiotic mediator in the gut–brain axis. NPJ Sci Food 2024; 8: 16.
  17. Loh JS, Mak WQ, Tan LKS, et al. Microbiota–gut–brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther 2024; 9: 37.
  18. Mhanna A, Martini N, Hmaydoosh G, et al. The correlation between gut microbiota and both neurotransmitters and mental disorders: A narrative review. Medicine 2024; 103: e37114.
  19. Aggarwal S, Ranjha R, Paul J. Neuroimmunomodulation by gut bacteria: Focus on inflammatory bowel diseases. World J Gastrointest Pathophysiol 2021; 12: 25–39.
  20. Abildinova GZ, Benberin V V, Vochshenkova TA, et al. The gut-brain-metabolic axis: exploring the role of microbiota in insulin resistance and cognitive function. Front Microbiol 2024; 15: 1463958.
  21. Yassin LK, Nakhal MM, Alderei A, et al. Exploring the microbiota-gut-brain axis: impact on brain structure and function. Front Neuroanat; 19. Epub ahead of print 12 February 2025. DOI: 10.3389/fnana.2025.1504065.
  22. Utzschneider KM, Kratz M, Damman CJ, et al. Mechanisms Linking the Gut Microbiome and Glucose Metabolism. J Clin Endocrinol Metab 2016; 101: 1445–54.
  23. Mishra SP, Wang B, Jain S, et al. A mechanism by which gut microbiota elevates permeability and inflammation in obese/diabetic mice and human gut. Gut 2023; 72: 1848.
  24. Sadagopan A, Mahmoud A, Begg M, et al. Understanding the Role of the Gut Microbiome in Diabetes and Therapeutics Targeting Leaky Gut: A Systematic Review. Cureus 2023; 15: e41559.
  25. Carvalho BM, Saad MJA. Influence of gut microbiota on subclinical inflammation and insulin resistance. Mediators Inflamm 2013; 2013: 986734.
  26. van Son J, Koekkoek LL, La Fleur SE, et al. The Role of the Gut Microbiota in the Gut-Brain Axis in Obesity: Mechanisms and Future Implications. Int J Mol Sci; 22. Epub ahead of print 15 March 2021. DOI: 10.3390/ijms22062993.
  27. Kuśmierska M, Kuśmierski J, Janik I, et al. Significance of the Gut-Brain Axis in the Development of Overweight and Obesity. Journal of Education, Health and Sport 2024; 70: 49434.
  28. Mou Y, Du Y, Zhou L, et al. Gut Microbiota Interact With the Brain Through Systemic Chronic Inflammation: Implications on Neuroinflammation, Neurodegeneration, and Aging. Front Immunol; 13. Epub ahead of print 7 April 2022. DOI: 10.3389/fimmu.2022.796288.
  29. Beltran-Velasco AI, Clemente-Suárez VJ. Impact of Peripheral Inflammation on Blood–Brain Barrier Dysfunction and Its Role in Neurodegenerative Diseases. Int J Mol Sci 2025; 26: 2440.
  30. Cheng J, Hu H, Ju Y, et al. Gut microbiota-derived short-chain fatty acids and depression: deep insight into biological mechanisms and potential applications. Gen Psychiatr 2024; 37: e101374.
  31. Mosquera FEC, Lizcano Martinez S, Liscano Y. Effectiveness of Psychobiotics in the Treatment of Psychiatric and Cognitive Disorders: A Systematic Review of Randomized Clinical Trials. Nutrients; 16. Epub ahead of print 30 April 2024. DOI: 10.3390/nu16091352.
  32. Korteniemi J, Karlsson L, Aatsinki A. Systematic review: Autism spectrum disorder and the gut microbiota. Acta Psychiatr Scand 2023; 148: 242–254.
  33. Abuljadayel D, Alotibi A, Alqothmi K, et al. Gut microbiota of children with autism spectrum disorder and healthy siblings: A comparative study. Exp Ther Med 2024; 28: 430.
  34. Erbescu A, Papuc SM, Budisteanu M, et al. Re-emerging concepts of immune dysregulation in autism spectrum disorders. Front Psychiatry 2022; 13: 1006612.
  35. Maniscalco I, Bartochowski P, Priori V, et al. The Effects of Fecal Microbial Transplantation on the Symptoms in Autism Spectrum Disorder, Gut Microbiota and Metabolites: A Scoping Review. Microorganisms 2025; 13: 1290.
  36. Feng P, Zhao S, Zhang Y, et al. A review of probiotics in the treatment of autism spectrum disorders: Perspectives from the gut-brain axis. Front Microbiol 2023; 14: 1123462.
  37. Garretti F, Monahan C, Sloan N, et al. Interaction of an α-synuclein epitope with HLA-DRB1∗15:01 triggers enteric features in mice reminiscent of prodromal Parkinson’s disease. Neuron 2023; 111: 3397-3413.e5.
  38. Jamerlan AM, An SSA, Hulme JP. Microbial diversity and fitness in the gut–brain axis: influences on developmental risk for Alzheimer’s disease. Gut Microbes; 17. Epub ahead of print 31 December 2025. DOI: 10.1080/19490976.2025.2486518.
  39. Lima AA de, Santos SS, Araújo MA de, et al. Probiotics as technological innovations in psychiatric disorders: patents and research reviews. Front Nutr; 12. Epub ahead of print 24 April 2025. DOI: 10.3389/fnut.2025.1567097.
  40. Slykerman RF, Davies N, Donohoe R. Evaluating the scientific evidence to support mental health and well-being claims made on probiotic products. Nutr Health. Epub ahead of print 19 January 2025. DOI: 10.1177/02601060241305682.
  41. Khavandegar A, Heidarzadeh A, Angoorani P, et al. Adherence to the Mediterranean diet can beneficially affect the gut microbiota composition: a systematic review. BMC Med Genomics 2024; 17: 91.
  42. Solch-Ottaiano RJ, Engler-Chiurazzi EB, Harper C, et al. Comparison between two divergent diets, Mediterranean and Western, on gut microbiota and cognitive function in young sprague dawley rats. Gut Microbes Reports; 1. Epub ahead of print 31 December 2024. DOI: 10.1080/29933935.2024.2439490.
  43. Hein ZM, Arbain MFF, Kumar S, et al. Intermittent Fasting as a Neuroprotective Strategy: Gut–Brain Axis Modulation and Metabolic Reprogramming in Neurodegenerative Disorders. Nutrients 2025; 17: 2266.
  44. Vendrik KEW, Ooijevaar RE, de Jong PRC, et al. Fecal Microbiota Transplantation in Neurological Disorders. Front Cell Infect Microbiol 2020; 10: 98.
  45. Scheperjans F, Levo R, Bosch B, et al. Fecal Microbiota Transplantation for Treatment of Parkinson Disease. JAMA Neurol 2024; 81: 925.
  46. Chunduri A, Reddy SDM, Jahanavi M, et al. Gut-Brain Axis, Neurodegeneration and Mental Health: A Personalized Medicine Perspective. Indian J Microbiol 2022; 62: 505–515.
  47. Shoji F, Yamashita T, Kinoshita F, et al. Artificial intelligence-derived gut microbiome as a predictive biomarker for therapeutic response to immunotherapy in lung cancer: protocol for a multicentre, prospective, observational study. BMJ Open 2022; 12: e061674.
  48. Ceccon M, Kantsjö JB, Ronchi F. Personalized Paths: Unlocking Alzheimer’s via the Gut-Brain Axis. Visc Med 2024; 40: 194–209.
  49. Hemmati MA, Monemi M, Asli S, et al. Using New Technologies to Analyze Gut Microbiota and Predict Cancer Risk. Cells; 13. Epub ahead of print 1 December 2024. DOI: 10.3390/cells13231987.
  50. O’Riordan KJ, Collins MK, Moloney GM, et al. Short chain fatty acids: Microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol 2022; 546: 111572.
  51. Severance EG, Yolken RH. Tracking a dysregulated gut-brain axis with biomarkers of the microbiome. Biomark Neuropsychiatry 2020; 2: 100009.
  52. Feng W, Liu J, Ao H, et al. Targeting gut microbiota for precision medicine: Focusing on the efficacy and toxicity of drugs. Theranostics 2020; 10: 11278–11301.
  53. Di Vincenzo F, Del Gaudio A, Petito V, et al. Gut microbiota, intestinal permeability, and systemic inflammation: a narrative review. Intern Emerg Med 2024; 19: 275–293.
  54. Iacucci M, Santacroce G, Majumder S, et al. Opening the doors of precision medicine: novel tools to assess intestinal barrier in inflammatory bowel disease and colitis-associated neoplasia. Gut 2024; 73: 1749.
  55. Galea I. The blood–brain barrier in systemic infection and inflammation. Cell Mol Immunol 2021; 18: 2489–2501.
  56. Franco CD, Sagar RS, Bokhari SFH. From Microbes to Memories: Challenges and Future Perspectives Regarding the Gut-Brain Axis for Improved Cognitive Health in Alzheimer’s. Cureus 2024; 16: e52795.
  57. Arneth B. Gut-Brain Axis and Brain Microbiome Interactions from a Medical Perspective. Brain Sci; 15. Epub ahead of print 8 February 2025. DOI: 10.3390/brainsci15020167.
  58. Chen Z, Zhang F. The Role of Gut Microbiota in Personalized Medicine: Implications for Disease Prevention and Treatment. J Neonatal Surg 2025; 14: 723–727.
  59. Slykerman RF, Davies N, Vlckova K, et al. Precision Psychobiotics for Gut-Brain Axis Health: Advancing the Discovery Pipelines to Deliver Mechanistic Pathways and Proven Health Efficacy. Microb Biotechnol 2025; 18: e70079.