Your new experience awaits. Try the new design now and help us make it even better

OPINION article

Front. Neurosci., 26 November 2025

Sec. Neuropharmacology

Volume 19 - 2025 | https://doi.org/10.3389/fnins.2025.1711846

This article is part of the Research TopicNovel Pharmacological Interventions in Emotional and Neuropsychiatric DisordersView all 9 articles

Transforming mental health: the future of personalized psychobiotics in anxiety and depression therapy

  • 1Programa de Estancias Posdoctorales por México, Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno, Jalisco, Mexico
  • 2Departamento de Ciencias de la Tierra y de la Vida, Centro Universitario de Los Lagos, Universidad de Guadalajara, Lagos de Moreno, Jalisco, Mexico

1 Introduction

The incidence and prevalence of anxiety and depression continue to rise, and conventional pharmacotherapies offer only incomplete solutions for millions of patients around the world. At the same time, scientific insights into the microbiota-gut-brain axis have opened up unprecedented opportunities for therapeutic innovation. Recent literature suggests a positive role for probiotics in the treatment of depression and anxiety (Merkouris et al., 2024); however, the heterogeneity of therapeutic responses suggests that current approaches do not account for individual differences in microbial ecology and host-microbe interactions (Alberdi et al., 2022). The concept of psychobiotics—probiotics with positive effects on mental health—has evolved from empirical observations to a mechanistic understanding (Sarkar et al., 2016; Sharma and Bajwa, 2021). However, the field is at a critical juncture where generic interventions must give way to personalized approaches (Erikainen and Chan, 2019). The rapidly growing field of microbiome-directed therapeutics has attracted significant attention due to its potential to revolutionize healthcare (Yaqub et al., 2025) and represents a fundamental shift from population-based to precision medicine paradigms. This transition is particularly urgent as understanding how broad host-microbiome associations are maintained across populations reveals individualized host-microbiome phenotypes that can be integrated with other “omics” datasets to improve precision medicine (Bashiardes et al., 2018; Lloyd-Price et al., 2019). This opinion piece will address the potential of probiotics and psychobiotics as precision medicine tools in the treatment of anxiety disorders and depression. The current state of scientific evidence, the neurobiological mechanisms involved, limitations of the field, and future prospects for the design of individualized interventions will be discussed, with an emphasis on the integration of “omics” approaches and bioinformatics technologies.

2 Mechanisms beyond traditional neurotransmission

2.1 Neurotransmitter production and modulation

Research demonstrates substantial evidence for probiotic production and modulation of neurotransmitters through the gut-brain axis, the mechanistic basis for the efficacy of psychobiotics goes far beyond traditional psychiatric pharmacology (Rosas-Sánchez et al., 2025). Gut bacteria can influence the production and regulation of neurotransmitters such as dopamine, acetylcholine, gamma-aminobutyric acid (GABA) and serotonin and affect their availability in the brain (Rosas-Sánchez et al., 2025; Kyei-Baffour et al., 2025). Certain bacterial strains show a remarkable ability to directly synthesize neurotransmitters, with several species being among the documented producers of neuroactive compounds (Oroojzadeh et al., 2022). Recent discoveries in neuromicrobiology have elucidated sophisticated signaling networks (Jameson et al., 2020). GABA signaling networks in the brain-gut-microbiome axis involve multiple pathways, including activation of GABA receptors in the gut nervous system, activation of the immune system, and GABA-stimulated exosome-mediated signaling (Belelli et al., 2025). This multi-target approach distinguishes the psychobiotic mechanisms from conventional antidepressants, which generally modulate single neurotransmitter systems. The clinical relevance of these mechanisms is demonstrated by direct interventions showing that targeted microbial modulation can effectively influence central nervous system function via peripheral signaling pathways (Cocean and Vodnar, 2024), providing tangible evidence for the therapeutic potential of precision microbiome interventions (Figure 1).

Figure 1
Diagram illustrating the microbiota-gut-brain axis (MGB axis) and its impact on mental health. The brain section highlights benefits such as reduced stress, anxiety, and depression. Gut bacteria influence neurotransmitter production. Psychobiotics and intestinal microbiota affect central nervous system function. Probiotics alter CNS function by influencing serotonin and dopamine levels. Neural, immune, endocrine, and metabolic pathways are involved. The diagram also shows vagus nerve signaling, complex immunomodulatory mechanisms, and neuroinflammation reduction through microbial metabolites in the gut lumen.

Figure 1. Neurochemical and anti-inflammatory mechanisms of psychobiotics through the gut-brain axis. Psychobiotics act beyond traditional psychiatric pharmacology by modulating neurotransmitter production (dopamine, acetylcholine, GABA, serotonin) and regulating immunological and inflammatory processes through the gut-brain axis. These mechanisms include the direct synthesis of neuroactive compounds, the activation of GABA signaling networks in the enteric and immune nervous systems, and the reduction of proinflammatory cytokines (IL-1β, TNF-α, and IL-6) along with the increase of anti-inflammatory mediators (IL-10), generating a comprehensive and synergistic therapeutic effect. Figure created with BioRender.com.

2.2 Neuroinflammatory pathways and systemic effects

In addition to the production of neurotransmitters, psychobiotics influence neuroinflammation through complex immunomodulatory mechanisms reducing the production of proinflammatory cytokines such as IL-1β, TNF-α, and IL-6, while promoting anti-inflammatory mediators such as IL-10 (Rosas-Sánchez et al., 2025; Oleskin and Shenderov, 2019). The gut-brain axis is a bidirectional communication network that connects the enteric and the central nervous system (CNS) and also includes endocrine, humoral, metabolic and immunological communication pathways (Carabotti et al., 2015). This comprehensive network enables the simultaneous influence of inflammation, neurotransmission and neuroendocrine function. Probiotics indirectly alter CNS function by producing metabolites that influence serotonin levels, brain-derived neurotrophic factor (BDNF), dopamine and GABA (Dziedzic et al., 2024; Kimse et al., 2024). The integration of these mechanisms leads to synergistic therapeutic effects that go beyond individual signaling pathways and create a comprehensive neurochemical network suitable for microbial interventions (Figure 1).

3 Meta-analytical insights from recent literature

The clinical evidence for the efficacy of psychobiotics has evolved considerably, with recent meta-analysis providing nuanced perspectives on therapeutic potential (Vasiliu, 2023; Mosquera et al., 2024). A random-effects meta-analysis of 34 controlled clinical trials investigating the effects of prebiotics and probiotics on depression and anxiety found heterogeneous but promising results (Liu et al., 2019; Zandifar et al., 2025). In particular, taking probiotics for up to 4, 8, and 12 weeks is effective in reducing depressive and anxiety symptoms in clinically diagnosed patients compared to placebo (Lew et al., 2019; Asad et al., 2025). However, the evidence also has important limitations. It may be premature to conclude clinical efficacy in relieving anxiety, as the effect size is small and there is no consensus on the optimal dose, treatment duration or specific formulations (Zhao et al., 2023). These findings highlight the need for a personalized approach that takes into account individual variability in response to treatment. Recent analyses indicate that specific strains—such as Lacticaseibacillus casei Shirota, Lactobacillus gasseri CP2305, Lactiplantibacillus plantarum PS128, Lacticaseibacillus rhamnosus LR06, and Bifidobacterium longum subsp. longum R0175 have shown efficacy in alleviating anxiety and depression symptoms (Ross, 2023; Rahmannia et al., 2024). This strain-level activity supports the development of precision psychobiotic interventions.

3.1 Individual variability and response prediction

The heterogeneity in treatment responses across studies reflects fundamental individual differences in microbiome composition, host genetics, and environmental factors (Schupack et al., 2022). The current review suggests that probiotics may improve symptoms of depression and anxiety in clinical patients; however, given the limitations in the included studies, randomized controlled trials (RCTs) with long-term follow-up and large sample sizes are needed (Minayo et al., 2021). This variability represents both a challenge and an opportunity for precision medicine approaches. Understanding the biological basis for differential responses will enable stratification of patients based on predictive biomarkers (Yang et al., 2024), optimizing therapeutic outcomes while minimizing unnecessary interventions. It is essential to recognize that, despite advances, the heterogeneity in clinical study results reflects the complexity of the microbiome and its interaction with genetic and environmental factors. The lack of standardized protocols, in addition to variability in individual microbial composition, limits the reproducibility and clinical applicability of probiotic interventions. These aspects should be considered in future research to optimize their clinical utility.

4 Individual microbiome signatures and response prediction

The transition to precision psychobiotics requires a fundamental reconceptualization of therapeutic approaches based on individual biological profiles (Slykerman et al., 2025). The concept of the “precision microbiome” involves the precise analysis and typing of the microbiota in specific hosts using advanced tools such as high-throughput sequencing, genomics and artificial intelligence. This technological foundation enables comprehensive characterization of individual microbiome signatures that could predict treatment response. The inherent individuality of the composition of the microbiome requires personalized therapeutic strategies rather than one-size-fits-all solutions (Fang et al., 2025). The genomic revolution promises to transform our approach to treating patients by individualizing treatments, reducing side effects and lowering healthcare costs (Huang et al., 2023), with microbiome analysis being a critical component of this transformation.

4.1 Technological integration and therapeutic optimization

Advanced analytical approaches are revolutionizing the personalization of treatments by integrating multiple data streams (D'Urso and Broccolo, 2024). Precise reconstitution of the microbiome using high-throughput sequencing and artificial intelligence tools enables real-time monitoring and optimization of therapeutic interventions based on the dynamics of the individual microbiome (Fang et al., 2025; Huo and Wang, 2024). The application of these technologies facilitates unprecedented precision in the selection and monitoring of therapies. A new approach to treating disease through personalized probiotic therapies—also known as precision medicine—has been proposed by researchers (D'Urso and Broccolo, 2024; Huo and Wang, 2024), demonstrating the feasibility of individualized microbial interventions in various therapeutic areas. The integration of ‘omics' technologies—such as genomics, metabolomics, and microbiomics—allows for the identification of specific microbial profiles associated with positive or adverse responses to probiotics. These strategies pave the way for personalized therapies, where the selection of specific strains and combinations is tailored to a patient's unique microbial signature. Recent studies have exemplified how the analysis of these signatures can predict treatment efficacy, reaffirming the importance of a personalized medicine approach.

5 Future perspectives: toward psychobiotic medicine

The future of precision psychobiotics requires a deeper mechanistic understanding of host-microbe interactions in mental health (Slykerman et al., 2025). Psychobiotics are probiotic strains capable of influencing the gut-brain axis and have been shown to be effective in several neurological disorders (Oroojzadeh et al., 2022). To make progress in this field, a comprehensive mapping of microbial metabolic pathways, host genetic variants and environmental factors influencing therapeutic outcomes is required (Kwok et al., 2021). In psychobiotics, the potential of probiotics to influence the nervous system and mental health is being investigated through a comprehensive analysis of their effects on mood, cognition and stress response (Cocean and Vodnar, 2024). This mechanistic foundation will enable the rational design of personalized treatment regimens optimized for individual patients. The transition from research to clinical practice requires addressing multiple challenges while taking advantage of the opportunities that arise. Standardization of analytical protocols, validation of predictive biomarkers and integration into existing psychiatric care are crucial prerequisites for implementation (Kyei-Baffour et al., 2025; Liu et al., 2025). For example, in a recent pilot study, patients with major depressive disorder showed significant improvements after receiving a personalized probiotic regimen based on their baseline microbiome profile (Kreuzer et al., 2022; Gawlik-Kotelnicka et al., 2023; Johnson et al., 2025). These experiences, although preliminary, illustrate the feasibility and potential of personalized microbiome-centered therapies and underscore the need for larger-scale controlled trials. The development of precision psychobiotics requires interdisciplinary collaboration between microbiologists, psychiatrists, bioinformaticians and regulatory specialists. This collaborative approach will accelerate the translation of mechanistic insights into clinically applicable interventions to improve patient outcomes. The development of personalized microbiome therapies faces significant challenges, including regulatory hurdles, the costs of omics analysis, and ethical issues related to microbiome modification. Furthermore, it is essential to define reliable predictive biomarkers and establish clear clinical criteria for their use. Multidisciplinary collaboration, integrating microbiology, psychiatry, bioinformatics, and regulation, will be key to overcoming these barriers and bringing these innovations into routine clinical practice.

6 Discussion

The emergence of precision psychobiotics represents a convergence of multiple scientific advances that collectively challenge traditional approaches to mental health treatment. The evidence synthesized in this review reveals both unprecedented opportunities and significant challenges that must be addressed to realize the full therapeutic potential of personalized microbiome interventions. The mechanistic foundations for psychobiotic efficacy are increasingly well-established, with robust evidence for neurotransmitter production (Sarkar et al., 2016; Belelli et al., 2025), neuroinflammatory modulation (Carabotti et al., 2015; Kimse et al., 2024), and multi-pathway signaling networks (Cocean and Vodnar, 2024). However, translating these mechanistic insights into clinically effective interventions remains complex. The heterogeneity observed in meta-analysis (Liu et al., 2019; Zandifar et al., 2025; Zhao et al., 2023) suggests that individual differences in microbiome composition, host genetics, and environmental factors significantly influence treatment outcomes. This variability underscores the necessity for precision medicine approaches that can stratify patients based on predictive biomarkers (Yang et al., 2024; Fang et al., 2025). The challenge lies in identifying which combinations of microbial, genetic, and clinical factors best predict treatment response, requiring sophisticated analytical approaches that integrate multi-omics data (Bashiardes et al., 2018; Lloyd-Price et al., 2019).

Several critical limitations constrain current progress toward precision psychobiotics. The complexity of microbiome-brain interactions creates analytical challenges that exceed current technological capabilities (Lloyd-Price et al., 2019; Turnbaugh et al., 2007). Additionally, the temporal dynamics of microbial communities introduce variability that complicates therapeutic monitoring and optimization. RTCs have shown that probiotic treatment for 4 and 12 weeks does not pose a relative risk of treatment-associated adverse events, even in patients with irritable bowel syndrome (Mosquera et al., 2024; Hempel et al., 2011). The preponderance of evidence, including the long history of safe use of probiotics, as well as data from clinical trials and animal and in vitro studies, supports the assumption that probiotics are generally safe for most populations (Pinto-Sanchez et al., 2017). Future research should address heterogeneity in diagnosis and intervention types to better understand their efficacy (Moshfeghinia et al., 2025). The field requires rigorous adherence to evidence-based standards while resisting pressure for rapid commercialization of insufficiently validated interventions. Long-term safety considerations remain largely unexplored, particularly regarding potential unintended consequences of targeted microbial manipulation (Merkouris et al., 2024). Comprehensive safety assessment protocols must accompany therapeutic development to ensure patient welfare throughout treatment courses.

The transition to precision psychobiotics will require fundamental changes in clinical practice patterns, including integration of microbiome analysis into psychiatric assessment protocols (D'Urso and Broccolo, 2024; Kreuzer et al., 2022). This transformation necessitates substantial investment in diagnostic infrastructure, clinician training, and regulatory frameworks that can accommodate personalized interventions while maintaining safety standards. The economic implications are equally significant, as precision medicine approaches typically involve higher upfront costs for diagnostic testing and personalized formulation development. However, the potential for improved treatment outcomes and reduced healthcare utilization costs may justify these investments over the long term.

The evolution from population-based to precision-based psychobiotic interventions represents a paradigmatic shift in mental health therapeutics. Recent studies investigating the effects of probiotics on depression, anxiety and psychological stress provide the foundation for this transformation (Reis et al., 2020), but realizing the full therapeutic potential requires embracing personalized approaches that account for individual microbial signatures and host-microbe interactions. The convergence of advanced analytical technologies, mechanistic understanding, and clinical need creates unprecedented opportunities for therapeutic innovation. Success requires sustained commitment to rigorous scientific standards, collaborative research approaches, and patient-centered care models that prioritize safety and efficacy over commercial expediency. The future of mental health therapeutics lies not in generic interventions but in sophisticated, personalized approaches that leverage individual biological signatures to optimize therapeutic outcomes. Precision psychobiotics represent the next evolutionary step in this journey, promising more effective, safer, and more individualized treatments for anxiety and depression.

As we advance toward this future, the mental health community must embrace the complexity inherent in personalized medicine while maintaining focus on the ultimate goal: improving patient outcomes through scientifically rigorous, individually tailored therapeutic interventions. The transformation of probiotics from generic supplements to precision medicine tools represents both a scientific achievement and a moral imperative to provide the most effective possible care for patients suffering from anxiety and depression. This article proposes an innovative approach that combines recent scientific evidence, critical analysis, and a forward-looking perspective that emphasizes the convergence of microbiology, neuroscience, and personalized medicine. Its originality lies in contextualizing probiotic therapy within the precision medicine paradigm, offering both conceptual and practical insights, and proposing concrete pathways for its integration into clinical care.

Author contributions

GR-S: Conceptualization, Formal analysis, Investigation, Methodology, Resources, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing. JM-C: Investigation, Supervision, Validation, Visualization, Writing – review & editing. CS-F: Investigation, Supervision, Validation, Visualization, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Acknowledgments

Gilberto Uriel Rosas-Sánchez (CVU: 714866) and Jose Luis Muñoz-Carrillo (CVU: 545580) thanks the Secretaria de Ciencia, Humanidades, Tecnología e Innovacion (SECIHTI) for the support through the scholarship of Postdoctoral Stays in Mexico.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Gen AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Alberdi, A., Andersen, S. B., Limborg, M. T., Dunn, R. R., and Gilbert, M. T. P. (2022). Disentangling host-microbiota complexity through hologenomics. Nat. Rev. Genet. 23, 281–297. doi: 10.1038/s41576-021-00421-0

PubMed Abstract | Crossref Full Text | Google Scholar

Asad, A., Kirk, M., Zhu, S., Dong, X., and Gao, M. (2025). Effects of prebiotics and probiotics on symptoms of depression and anxiety in clinically diagnosed samples: systematic review and meta-analysis of randomized controlled trials. Nutr. Rev. 83, e1504–e1520. doi: 10.1093/nutrit/nuae177

Crossref Full Text | Google Scholar

Bashiardes, S., Godneva, A., Elinav, E., and Segal, E. (2018). Towards utilization of the human genome and microbiome for personalized nutrition. Curr. Opin. Biotechnol. 51, 57–63. doi: 10.1016/j.copbio.2017.11.013

PubMed Abstract | Crossref Full Text | Google Scholar

Belelli, D., Lambert, J. J., Wan, M. L. Y., Monteiro, A. R., Nutt, D. J., Swinny, J. D., et al. (2025). From bugs to brain: unravelling the GABA signalling networks in the brain-gut-microbiome axis. Brain 148, 1479–1506. doi: 10.1093/brain/awae413

PubMed Abstract | Crossref Full Text | Google Scholar

Carabotti, M., Scirocco, A., Maselli, M. A., and Severi, C. (2015). The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 28, 203–209.

PubMed Abstract | Google Scholar

Cocean, A. M., and Vodnar, D. C. (2024). Exploring the gut-brain axis: potential therapeutic impact of psychobiotics on mental health. Prog. Neuro-Psychopharmacol. Biol Psychiatry 134:111073. doi: 10.1016/j.pnpbp.2024.111073

PubMed Abstract | Crossref Full Text | Google Scholar

D'Urso, F., and Broccolo, F. (2024). Applications of artificial intelligence in microbiome analysis and probiotic interventions—an overview and perspective based on the current state of the art. Appl. Sci. 14:8627. doi: 10.3390/app14198627

Crossref Full Text | Google Scholar

Dziedzic, A., Maciak, K., Blizniewska-Kowalska, K., Gałecka, M., Kobierecka, W., Saluk, J., et al. (2024). The power of psychobiotics in depression: a modern approach through the microbiota-gut-brain axis: a literature review. Nutrients 16:1054. doi: 10.3390/nu16071054

PubMed Abstract | Crossref Full Text | Google Scholar

Erikainen, S., and Chan, S. (2019). Contested futures: envisioning “personalized,” “stratified,” and “precision” medicine. New Genet. Soc. 38, 308–330. doi: 10.1080/14636778.2019.1637720

PubMed Abstract | Crossref Full Text | Google Scholar

Fang, X., Wang, Y., Wei, H., and Huang, Y. (2025). Precision microbiome: a new era of targeted therapy with core probiotics. Research 8:0658. doi: 10.34133/research.0658

PubMed Abstract | Crossref Full Text | Google Scholar

Gawlik-Kotelnicka, O., Margulska, A., Skowrońska, A., and Strzelecki, D. (2023). PRO-DEMET randomized controlled trial on probiotics in depression-pilot study results. Nutrients 15:1400. doi: 10.3390/nu15061400

PubMed Abstract | Crossref Full Text | Google Scholar

Hempel, S., Newberry, S., Ruelaz, A., Wang, Z., Miles, J. N., Suttorp, M. J., et al. (2011). Safety of probiotics used to reduce risk and prevent or treat disease. Evid. Rep. Technol. Assess. 200:1–645.

PubMed Abstract | Google Scholar

Huang, G., Khan, R., Zheng, Y., Lee, P. C., Li, Q., Khan, I., et al. (2023). Exploring the role of gut microbiota in advancing personalized medicine. Front. Microbiol. 14, 1274925. doi: 10.3389/fmicb.2023.1274925

PubMed Abstract | Crossref Full Text | Google Scholar

Huo, D., and Wang, X. (2024). A new era in healthcare: the integration of artificial intelligence and microbial. Med. Nov. Technol. Devices 23:100319. doi: 10.1016/j.medntd.2024.100319

Crossref Full Text | Google Scholar

Jameson, K. G., Olson, C. A., Kazmi, S. A., and Hsiao, E. Y. (2020). Toward understanding microbiome-neuronal signaling. Mol. Cell 78, 577–583. doi: 10.1016/j.molcel.2020.03.006

PubMed Abstract | Crossref Full Text | Google Scholar

Johnson, D., Chua, K. O., Selvadurai, J., Niap, C. P. S., Thurairajasingam, S., Lee, L. H., et al. (2025). Pilot trial of probiotics in major depressive disorder: a randomized, double-blind, placebo-controlled approach. Prog. Microbes Mol. Biol. 8:a0000462. doi: 10.36877/pmmb.a0000462

Crossref Full Text | Google Scholar

Kimse, L., Reinis, A., Mikelsone-Jansone, L., Gintere, S., and Krumina, A. (2024). A narrative review of psychobiotics: probiotics that influence the gut-brain axis. Medicina 60:601. doi: 10.3390/medicina60040601

PubMed Abstract | Crossref Full Text | Google Scholar

Kreuzer, K., Reiter, A., Birkl-Töglhofer, A. M., Dalkner, N., Mörkl, S., Mairinger, M., et al. (2022). The PROVIT study-effects of multispecies probiotic add-on treatment on metabolomics in major depressive disorder-a randomized, placebo-controlled trial. Metabolites 12:770. doi: 10.3390/metabo12080770

PubMed Abstract | Crossref Full Text | Google Scholar

Kwok, A. J., Mentzer, A., and Knight, J. C. (2021). Host genetics and infectious disease: new tools, insights and translational opportunities. Nat. Rev. Genet. 22, 137–153. doi: 10.1038/s41576-020-00297-6

PubMed Abstract | Crossref Full Text | Google Scholar

Kyei-Baffour, V. O., Vijaya, A. K., Burokas, A., and Daliri, E. B. M. (2025). Psychobiotics and the gut-brain axis: advances in metabolite quantification and their implications for mental health. Crit. Rev. Food Sci. Nutr. 65, 7085–7104. doi: 10.1080/10408398.2025.2459341

PubMed Abstract | Crossref Full Text | Google Scholar

Lew, L. C., Hor, Y. Y., Yusoff, N. A. A., Choi, S. B., Yusoff, M. S. B., Roslan, N. S., et al. (2019). Probiotic Lactobacillus plantarum P8 alleviated stress and anxiety while enhancing memory and cognition in stressed adults: a randomised, double-blind, placebo-controlled study. Clin. Nutr. 38, 2053–2064. doi: 10.1016/j.clnu.2018.09.010

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, R. T., Walsh, R. F. L., and Sheehan, A. E. (2019). Prebiotics and probiotics for depression and anxiety: a systematic review and meta-analysis of controlled clinical trials. Neurosci. Biobehav. Rev. 102, 13–23. doi: 10.1016/j.neubiorev.2019.03.023

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, X., Xu, M., Wang, H., and Zhu, L. (2025). Integrating precision medicine and digital health in personalized weight management: the central role of nutrition. Nutrients 17:2695. doi: 10.3390/nu17162695

PubMed Abstract | Crossref Full Text | Google Scholar

Lloyd-Price, J., Arze, C., Ananthakrishnan, A. N., Schirmer, M., Avila-Pacheco, J., Poon, T. W., et al. (2019). Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662. doi: 10.1038/s41586-019-1237-9

PubMed Abstract | Crossref Full Text | Google Scholar

Merkouris, E., Mavroudi, T., Miliotas, D., Tsiptsios, D., Serdari, A., Christidi, F., et al. (2024). Probiotics' effects in the treatment of anxiety and depression: a comprehensive review of 2014-2023 clinical trials. Microorganisms 12:411. doi: 10.3390/microorganisms12020411

PubMed Abstract | Crossref Full Text | Google Scholar

Minayo, M. S., Miranda, I., and Telhado, R. S. (2021). A systematic review of the effects of probiotics on depression and anxiety: an alternative therapy? Cien. Saude Colet. 26, 4087–4099. doi: 10.1590/1413-81232021269.21342020

PubMed Abstract | Crossref Full Text | Google Scholar

Moshfeghinia, R., Nemati, H., Ebrahimi, A., Shekouh, D., Karami, S., Eraghi, M. M., et al. (2025). The impact of probiotics, prebiotics, and synbiotics on depression and anxiety symptoms of patients with depression: a systematic review and meta-analysis. J. Psychiatr. Res. 188, 104–116. doi: 10.1016/j.jpsychires.2025.05.053

PubMed Abstract | Crossref Full Text | Google Scholar

Mosquera, F. E. C., Lizcano Martinez, S., and Liscano, Y. (2024). Effectiveness of psychobiotics in the treatment of psychiatric and cognitive disorders: a systematic review of randomized clinical trials. Nutrients 16:1352. doi: 10.3390/nu16091352

PubMed Abstract | Crossref Full Text | Google Scholar

Oleskin, A. V., and Shenderov, B. A. (2019). Probiotics and psychobiotics: the role of microbial neurochemicals. Probiotics Antimicrob. Proteins 11, 1071–1085. doi: 10.1007/s12602-019-09583-0

PubMed Abstract | Crossref Full Text | Google Scholar

Oroojzadeh, P., Bostanabad, S. Y., and Lotfi, H. (2022). Psychobiotics: the tnfluence of Gut Microbiota on the gut-brain axis in neurological disorders. J. Mol. Neurosci. 72, 1952–1964. doi: 10.1007/s12031-022-02053-3

Crossref Full Text | Google Scholar

Pinto-Sanchez, M. I., Hall, G. B., Ghajar, K., Nardelli, A., Bolino, C., Lau, J. T., et al. (2017). Probiotic Bifidobacterium longum NCC3001 reduces depression scores and alters brain activity: a pilot study in patients with irritable bowel syndrome. Gastroenterology 153, 448–459. doi: 10.1053/j.gastro.2017.05.003

PubMed Abstract | Crossref Full Text | Google Scholar

Rahmannia, M., Poudineh, M., Mirzaei, R., Aalipour, M. A., Shahidi Bonjar, A. H., Goudarzi, M., et al. (2024). Strain-specific effects of probiotics on depression and anxiety: a meta-analysis. Gut Pathog. 16:46. doi: 10.1186/s13099-024-00634-8

PubMed Abstract | Crossref Full Text | Google Scholar

Reis, D. J., Ilardi, S. S., Namekata, M. S., Wing, E. K., and Fowler, C. H. (2020). The depressogenic potential of added dietary sugars. Med. Hypotheses 134:109421. doi: 10.1016/j.mehy.2019.109421

PubMed Abstract | Crossref Full Text | Google Scholar

Rosas-Sánchez, G. U., Germán-Ponciano, L. J., Puga-Olguín, A., Soto, M. E. F., Medina, A. Y. N., Muñoz-Carillo, J. L., et al. (2025). Gut–brain axis in mood disorders: a narrative review of neurobiological insights and probiotic interventions. Biomedicines 13:1831. doi: 10.3390/biomedicines13081831

PubMed Abstract | Crossref Full Text | Google Scholar

Ross, K. (2023). Psychobiotics: are they the future intervention for managing depression and anxiety? A literature review. Explore 19, 669–680. doi: 10.1016/j.explore.2023.02.007

PubMed Abstract | Crossref Full Text | Google Scholar

Sarkar, A., Lehto, S. M., Harty, S., Dinan, T. G., Cryan, J. F., Burnet, P. W. J., et al. (2016). Psychobiotics and the manipulation of bacteria–gut–brain signals. Trends Neurosci. 39, 763–781. doi: 10.1016/j.tins.2016.09.002

PubMed Abstract | Crossref Full Text | Google Scholar

Schupack, D. A., Mars, R. A. T., Voelker, D. H., Abeykoon, J. P., and Kashyap, P. C. (2022). The promise of the gut microbiome as part of individualized treatment strategies. Nat. Rev. Gastroenterol. Hepatol. 19, 7–25. doi: 10.1038/s41575-021-00499-1

PubMed Abstract | Crossref Full Text | Google Scholar

Sharma, H., and Bajwa, J. (2021). Approach of probiotics in mental health as a psychobiotics. Arch. Microbiol. 204:30. doi: 10.1007/s00203-021-02622-x

PubMed Abstract | Crossref Full Text | Google Scholar

Slykerman, R. F., Davies, N., Vlckova, K., O'Riordan, K. J., Bassett, S. A., Dekker, J., et al. (2025). Precision psychobiotics for gut-brain axis health: advancing the discovery pipelines to deliver mechanistic pathways and proven health efficacy. Microb. Biotechnol. 18:e70079. doi: 10.1111/1751-7915.70079

PubMed Abstract | Crossref Full Text | Google Scholar

Turnbaugh, P. J., Ley, R. E., Hamady, M., Fraser-Liggett, C. M., Knight, R., Gordon, J. I., et al. (2007). The human microbiome project: exploring the microbial part of ourselves in a changing world. Nature 449:804. doi: 10.1038/nature06244

Crossref Full Text | Google Scholar

Vasiliu, O. (2023). The current state of research for psychobiotics use in the management of psychiatric disorders–asystematic literature review. Front. Psychiatry 14:1074736. doi: 10.3389/fpsyt.2023.1074736

Crossref Full Text | Google Scholar

Yang, Y., Yang, L., Wan, M., Pan, D., Sun, G., Yang, C., et al. (2024). Assessment of optimal combinations of therapeutic probiotics for depression, anxiety, and stress. Psychol. Med. 54, 2547–2561. doi: 10.1017/S0033291724000679

PubMed Abstract | Crossref Full Text | Google Scholar

Yaqub, M. O., Jain, A., Joseph, C. E., and Edison, L. K. (2025). Microbiome-driven therapeutics: from gut health to precision medicine. Gastrointest. Disord. 7:7. doi: 10.3390/gidisord7010007

Crossref Full Text | Google Scholar

Zandifar, A., Badrfam, R., Mohammaditabar, M., Kargar, B., Goodarzi, S., Hajialigol, A., et al. (2025). The effect of prebiotics and probiotics on levels of depression, anxiety, and cognitive function: a meta-analysis of randomized clinical trials. Brain Behav. 15:e70401. doi: 10.1002/brb3.70401

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, Z., Xiao, G., Xia, J., Guo, H., Yang, X., Jiang, Q., et al. (2023). Effectiveness of probiotic/prebiotic/synbiotic treatments on anxiety: a systematic review and meta-analysis of randomized controlled trials. J. Affect. Disord. 343, 9–21. doi: 10.1016/j.jad.2023.09.018

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: anxiety, depression, gut-brain axis, probiotics, psychobiotics

Citation: Rosas-Sánchez GU, Muñoz-Carrillo JL and Soria-Fregozo C (2025) Transforming mental health: the future of personalized psychobiotics in anxiety and depression therapy. Front. Neurosci. 19:1711846. doi: 10.3389/fnins.2025.1711846

Received: 23 September 2025; Revised: 11 November 2025;
Accepted: 12 November 2025; Published: 26 November 2025.

Edited by:

Elvira Maria Hebert, National Scientific and Technical Research Council (CONICET), Argentina

Reviewed by:

Kim Ross, Sonoran University of Health Sciences, United States

Copyright © 2025 Rosas-Sánchez, Muñoz-Carrillo and Soria-Fregozo. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Gilberto Uriel Rosas-Sánchez, Z2lsYmVydG8ucm9zYXNAbGFnb3MudWRnLm14

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.