Microbial Bioactives | Online ISSN 2209-2161
REVIEWS   (Open Access)

Alzheimer’s and the Gut Microbiota: Impacts on Brain Health, Memory, and Inflammation

Md. Al Noor Hossain1*, Salma jahan1, Anik Barman1, Sneha Sengupta2, Badhan Mojumder2

+ Author Affiliations

Microbial Bioactives 8 (1) 1-8 https://doi.org/10.25163/microbbioacts.8110303

Submitted: 01 January 2025 Revised: 11 February 2025  Published: 12 February 2025 


Abstract

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder marked by cognitive decline, memory loss, and neuronal degeneration. While genetic and environmental factors are well-established contributors, growing evidence highlights the gut microbiome as a potential modifiable risk factor in AD pathogenesis. The gut-brain axis, a complex communication network between the gastrointestinal tract and the central nervous system, plays a vital role in regulating inflammation, immune responses, and neurochemical signaling. Dysbiosis—an imbalance in gut microbial composition—has been associated with neuroinflammation, amyloid-beta accumulation, blood-brain barrier disruption, and cognitive dysfunction. Recent studies suggest that pathogenic bacteria such as Escherichia coli and Bacteroides fragilis may exacerbate amyloid-beta deposition and inflammation, while beneficial microbes like Lactobacillus and Bifidobacterium offer neuroprotective effects. Microbial metabolites, particularly short-chain fatty acids (SCFAs), are critical for maintaining neuronal health; diminished SCFA production correlates with increased inflammation and AD progression. Additionally, gut-derived lipopolysaccharides (LPS) can trigger systemic immune responses and chronic neuroinflammation via increased gut permeability (“leaky gut”). Probiotic and prebiotic interventions have shown promise in restoring microbial balance and improving cognitive outcomes, though clinical validation remains limited. Understanding the gut microbiome's influence on AD not only offers novel insights into disease mechanisms but also opens avenues for early diagnosis and microbiome-based therapeutic strategies. Continued research is essential to clarify causal relationships and develop effective interventions.

Keywords: Alzheimer’s disease, Gut microbiome, Neuroinflammation, Gut-brain axis, Microbial metabolites.

1. Introduction


Alzheimer’s disease (AD) is one of the most devastating neurodegenerative disorders, affecting millions worldwide. Characterized by progressive memory loss, cognitive impairment, and neuronal degeneration, AD remains a major challenge for modern medicine. The disease is primarily marked by amyloid-beta (Aß) plaques and neurofibrillary tangles composed of hyperphosphorylated tau protein, leading to synaptic dysfunction and neuronal loss (Acosta et al.,2017). While genetic predisposition, such as the presence of the APOE e4 allele, and environmental factors contribute to disease onset, a growing body of evidence suggests that gut microbiota may play a crucial role in AD pathogenesis. The gut-brain axis, a bidirectional communication network linking the gut microbiome and the central nervous system, has gained significant attention in recent years. Research indicates that gut microbiota influences brain health through immune regulation, neurotransmitter production, and metabolic pathways. Dysbiosis, or an imbalance in gut microbial composition, has been observed in AD patients, suggesting that microbial alterations may contribute to neurodegeneration. The presence of pathogenic bacteria and a reduction in beneficial microbes have been linked to increased inflammation, blood-brain barrier (BBB) disruption, and amyloid deposition—key mechanisms in AD progression (Adak et al.,2019).One of the primary ways gut microbes influence AD is through inflammation. Chronic systemic inflammation is a recognized factor in neurodegeneration, and gut-derived inflammatory markers such as lipopolysaccharides (LPS) have been found in the brains of AD patients. LPS, components of Gram-negative bacterial cell walls, can cross the BBB and activate microglial cells, leading to chronic neuroinflammation and neuronal damage. Additionally, microbial metabolites, including short-chain fatty acids (SCFAs), play a vital role in brain health. SCFAs, such as butyrate and acetate, exert anti-inflammatory effects and support neuronal function. Reduced SCFA levels in AD patients further implicate gut microbiota in disease progression (Agirman et al.,2021). Moreover, studies have demonstrated that gut microbial imbalances can exacerbate amyloid-beta accumulation. Bacteria such as Escherichia coli and Bacteroides fragilis have been associated with increased amyloid aggregation, while probiotic strains like Lactobacillus and Bifidobacterium exhibit neuroprotective properties. These findings highlight the potential of microbiome-targeted interventions in mitigating AD pathology.Despite promising evidence, the exact mechanisms linking gut microbes to Alzheimer’s remain incompletely understood. However, the growing recognition of the gut-brain connection offers a paradigm shift in how neurodegenerative diseases are studied and managed. This review explores five key aspects of the gut microbiome’s role in AD: gut dysbiosis and neuroinflammation, microbial metabolites and brain function, gut permeability and endotoxemia, the impact of probiotics on cognitive health, and future therapeutic directions. By synthesizing existing research, this paper aims to provide a clearer understanding of how gut microbes influence Alzheimer’s disease and how these insights can inform novel treatment strategies (Agus et al.,2018).

2. Gut Dysbiosis and Neuroinflammation


The concept of gut dysbiosis—an imbalance in the gut microbial ecosystem—has gained prominence in Alzheimer’s disease (AD) research due to its role in promoting neuroinflammation (Figure 1). Neuroinflammation is a critical factor in AD progression, and gut microbes play a significant role in modulating immune responses that affect the brain. Dysbiosis has been observed in AD patients, with an increase in pathogenic bacteria and a decrease in beneficial species, leading to systemic inflammation that exacerbates neurodegeneration (Akhtar et al.,2020).

2.1 The Gut-Brain Axis and Neuroimmune Signaling

The gut-brain axis is a bidirectional communication system linking the gastrointestinal tract and the central nervous system (CNS) through neural, endocrine, and immune pathways. This axis allows gut microbes to influence brain function and vice versa. When gut dysbiosis occurs, pro-inflammatory signals from the gut can travel to the brain, triggering microglial activation (Figure 2). Microglia, the resident immune cells of the brain, play a protective role under normal conditions but can become overactive in response to chronic inflammation, leading to neuronal damage (Alexeev et al.,2018).

2.2 Lipopolysaccharides (LPS) and Chronic Inflammation

One of the most well-studied mechanisms linking gut dysbiosis to AD is the presence of lipopolysaccharides (LPS), toxic components found in the outer membranes of Gram-negative bacteria. LPS can enter the bloodstream through a compromised intestinal barrier, a condition often referred to as “leaky gut”. Once in circulation, LPS triggers a systemic inflammatory response by activating Toll-like receptors (TLRs), which, in turn, stimulate pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-a) and interleukin-6 (IL-6). Elevated levels of these cytokines have been found in the brains of AD patients, suggesting that chronic inflammation originating in the gut contributes to neurodegeneration. Postmortem studies have detected LPS in the hippocampus, the brain region most affected by Alzheimer’s (Alzheimer’s disease facts and figures,2023). Additionally, experimental models have shown that chronic LPS exposure leads to increased amyloid-beta deposition and cognitive decline, further supporting the role of gut-derived inflammation in AD pathology.

2.3 Reduction in Anti-Inflammatory Microbes

Healthy gut microbiota consists of beneficial bacteria that produce anti-inflammatory metabolites, helping to regulate immune responses. Studies have shown that Lactobacillus and Bifidobacterium species, which have neuroprotective properties, are significantly reduced in AD patients. These bacteria help maintain gut integrity, prevent pathogen overgrowth, and produce beneficial compounds such as short-chain fatty acids (SCFAs), which have been linked to reduced neuroinflammation (Ang et al.,2020).

2.4 Microglial Activation and Neurodegeneration

Microglial cells, when exposed to persistent inflammatory signals from the gut, become primed for hyperreactivity. This leads to the excessive release of reactive oxygen species (ROS) and nitric oxide, both of which contribute to oxidative stress and neuronal death. In AD patients, heightened microglial activation correlates with increased tau pathology and synaptic dysfunction (Arumugam et al.,2011). This suggests that controlling gut inflammation could be a potential strategy for modulating neuroinflammation and slowing disease progression.Gut dysbiosis and neuroinflammation represent a crucial link between the gastrointestinal tract and Alzheimer’s disease. An imbalance in gut microbiota leads to systemic inflammation, increased permeability of the blood-brain barrier, and heightened microglial activity—all of which contribute to neurodegeneration. Understanding how gut microbes regulate inflammation provides opportunities for therapeutic interventions that could mitigate AD progression. Strategies aimed at restoring microbial balance, such as probiotic supplementation and dietary modifications, are gaining attention as potential tools for reducing neuroinflammation in AD patients.

 

Figure 1. Gut microbial involvement in Alzheimer's disease pathogenesis. (Courtesy of image from Fan et al.,2021)

 

 

 

 

Figure 2.  Target Dysbiosis of Gut Microbes as a Future. (Courtesy of image from Grieco et al.,2019)

3.Microbial Metabolites and Brain Function


The gut microbiome exerts profound effects on brain function through the production of microbial metabolites, which influence neurotransmission, neuroinflammation, and cognitive processes. These metabolites include short-chain fatty acids (SCFAs), neurotransmitter precursors, and bacterial endotoxins, all of which have been implicated in Alzheimer’s disease (AD) pathology. Changes in gut microbial composition can disrupt the delicate balance of these metabolites, contributing to neurodegeneration and cognitive decline (Atanasov et al.,2021).

3.1 Short-Chain Fatty Acids (SCFAs) and Neuroprotection

SCFAs, including butyrate, acetate, and propionate, are metabolites produced by gut bacteria during the fermentation of dietary fibers. These compounds play a crucial role in maintaining gut barrier integrity, modulating immune responses, and influencing brain function through the gut-brain axis. Butyrate, in particular, has been shown to have anti-inflammatory and neuroprotective properties. It enhances the production of brain-derived neurotrophic factor (BDNF), a protein essential for neuronal survival and synaptic plasticity (Backhed et al.,2005).In AD patients, lower levels of SCFAs have been observed, suggesting a potential link between gut dysbiosis and neurodegeneration. Reduced butyrate levels have been associated with increased neuroinflammation, blood-brain barrier (BBB) dysfunction, and cognitive impairment. Animal studies have demonstrated that supplementing butyrate can mitigate cognitive decline and reduce amyloid-beta (Aß) deposition, further supporting its neuroprotective role.

3.2 Neurotransmitter Production by Gut Microbes

The gut microbiota significantly contributes to neurotransmitter synthesis, affecting mood, cognition, and neural signaling. Certain bacterial species produce gamma-aminobutyric acid (GABA), serotonin, and dopamine, all of which are crucial for brain function. Disruptions in microbial communities can lead to imbalances in these neurotransmitters, potentially contributing to cognitive dysfunction and neuropsychiatric symptoms seen in AD patients. For example, Lactobacillus and Bifidobacterium species are known to produce GABA, the primary inhibitory neurotransmitter in the brain. GABA deficiency has been implicated in heightened neuroexcitability, which may contribute to AD-related neuronal damage. Similarly, gut-derived serotonin plays a role in regulating neuroinflammation, and alterations in serotonin-producing bacteria have been linked to mood disorders and cognitive decline (Baloni et al.,2020).

3.3 Trimethylamine-N-Oxide (TMAO) and Alzheimer’s Pathology

Trimethylamine-N-oxide (TMAO) is a microbial metabolite derived from the bacterial metabolism of dietary choline and carnitine, found in red meat and dairy products. Elevated TMAO levels have been associated with cardiovascular disease, but emerging research suggests that it also plays a role in neurodegeneration. High TMAO levels have been correlated with increased oxidative stress and neuroinflammation, both of which contribute to AD pathology (Barka et al.,2016). Additionally, TMAO has been shown to promote amyloid aggregation and tau phosphorylation, key mechanisms underlying AD progression. Animal studies have demonstrated that reducing TMAO levels through dietary interventions or microbial modulation can protect against cognitive decline, highlighting its potential as a target for AD prevention.3.4 Bacterial Endotoxins and Blood-Brain Barrier DysfunctionThe integrity of the blood-brain barrier (BBB) is crucial for preventing harmful substances from entering the brain. However, microbial endotoxins, particularly lipopolysaccharides (LPS), can compromise BBB integrity, leading to neuroinflammation and increased permeability. LPS exposure triggers an inflammatory cascade that activates microglial cells, resulting in excessive production of pro-inflammatory cytokines and reactive oxygen species (Blair et al.,2015). Postmortem analyses of AD patients have revealed higher concentrations of LPS in brain tissues, suggesting a direct link between gut-derived endotoxins and neurodegeneration. The ability of LPS to stimulate amyloid-beta production further supports the hypothesis that microbial metabolites contribute to AD pathology.

3.5 Dietary Interventions to Modulate Microbial Metabolites

Given the role of microbial metabolites in AD progression, dietary interventions that promote beneficial gut bacteria may offer therapeutic potential. Fiber-rich diets support SCFA production, while reducing dietary choline and carnitine intake can lower TMAO levels. Probiotic and prebiotic supplementation has also shown promise in restoring microbial balance and enhancing the production of neuroprotective metabolites. For instance, dietary supplementation with prebiotic fibers, such as inulin and resistant starch, has been shown to enhance butyrate production, reduce inflammation, and improve cognitive function in animal models. Similarly, probiotic formulations containing Lactobacillus and Bifidobacterium strains have demonstrated potential in improving gut health and mitigating neuroinflammation in AD patients (Blazquez et al.,2014).Microbial metabolites play a critical role in brain health, influencing neuroinflammation, neurotransmitter balance, and cognitive function. The reduction of beneficial metabolites such as SCFAs and neurotransmitters, combined with the accumulation of harmful metabolites like TMAO and LPS, contributes to the progression of Alzheimer’s disease. Understanding these mechanisms opens avenues for targeted dietary and microbiome-based interventions to mitigate AD risk and slow cognitive decline. Future research should focus on clinical trials to validate these findings and develop microbiome-targeted therapies for Alzheimer’s prevention and treatment (Bonaz et al.,2017).

4. Gut Permeability and Endotoxemia in Alzheimer's Disease


The concept of gut permeability, commonly referred to as “leaky gut,” has gained significant attention in Alzheimer’s disease (AD) research due to its role in promoting systemic inflammation and neurodegeneration. The intestinal barrier is a crucial protective system that regulates the movement of nutrients, metabolites, and immune signals while preventing harmful pathogens and toxins from entering circulation (Bonfili et al.,2017). However, disruptions in this barrier can lead to endotoxemia, a condition characterized by the leakage of bacterial endotoxins into the bloodstream, ultimately contributing to AD pathology.

4.1The Intestinal Barrier and Its Role in Brain Health

The gut lining is composed of epithelial cells held together by tight junction proteins, which act as gatekeepers to control the passage of molecules. A healthy gut barrier prevents harmful substances, including bacterial toxins, undigested food particles, and inflammatory compounds, from entering circulation (Braniste et al.,2014). However, gut dysbiosis, poor diet, stress, and aging can weaken these tight junctions, leading to increased intestinal permeability. When the gut barrier is compromised, endotoxins such as lipopolysaccharides (LPS) can enter the bloodstream, triggering an immune response. LPS, a component of Gram-negative bacterial cell walls, is highly inflammatory and has been detected in elevated levels in AD patients. This systemic inflammation can cross the blood-brain barrier (BBB), activating microglia and astrocytes, which contribute to neurodegeneration and cognitive decline (Brunt et al.,2021).

4.2 Leaky Gut and Blood-Brain Barrier Dysfunction

Both the gut and the brain possess highly selective barriers that regulate the exchange of molecules. The gut barrier prevents the entry of harmful substances into circulation, while the BBB shields the brain from toxins and pathogens. Disruptions in gut integrity often coincide with BBB dysfunction, allowing inflammatory compounds to infiltrate the brain (Cai et al.,2018). Studies have demonstrated that increased intestinal permeability correlates with BBB breakdown in neurodegenerative diseases, including AD. LPS and pro-inflammatory cytokines can weaken BBB integrity, making it easier for toxic molecules to accumulate in the brain. This breakdown facilitates amyloid-beta (Aß) deposition, tau hyperphosphorylation, and neuroinflammation—hallmarks of AD progression. Animal models have shown that inducing gut permeability through a high-fat diet or antibiotic treatment leads to increased LPS levels in the brain, causing neuroinflammation and cognitive impairments similar to those seen in AD patients. Conversely, restoring gut integrity through dietary interventions and probiotic supplementation has been found to reduce BBB permeability and protect against cognitive decline (Cani et al.,2007).

4.3 Endotoxemia and Systemic Inflammation in AD

Endotoxemia, characterized by elevated LPS levels in circulation, is a key driver of chronic inflammation. The presence of LPS in the bloodstream stimulates Toll-like receptor 4 (TLR4) activation, leading to the release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-a), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6). These cytokines can cross the BBB and trigger neuroinflammatory responses, exacerbating AD pathology.Postmortem studies have revealed that AD patients exhibit significantly higher levels of LPS in the hippocampus, the brain region most affected by the disease (Capuron et al.,2018). Furthermore, elevated systemic inflammation has been linked to accelerated cognitive decline, suggesting that gut-derived endotoxins contribute to disease progression.

4.4 Dietary and Lifestyle Factors Influencing Gut Permeability

Several factors contribute to increased gut permeability, including diet, stress, aging, and the use of certain medications. Western-style diets high in processed foods, refined sugars, and saturated fats have been shown to disrupt gut integrity and promote inflammation. Conversely, fiber-rich diets rich in polyphenols and antioxidants support gut health by reinforcing tight junctions and promoting the growth of beneficial bacteria (Carabotti et al.,2015). Probiotics and prebiotics play a crucial role in maintaining gut integrity by enhancing the production of short-chain fatty acids (SCFAs), which strengthen tight junction proteins and reduce inflammation. Clinical trials have shown that probiotic supplementation can improve gut barrier function, lower systemic inflammation, and enhance cognitive performance in older adults. Lifestyle interventions such as regular exercise, stress reduction techniques, and adequate sleep have also been found to improve gut barrier integrity and reduce systemic inflammation. Stress, in particular, has been shown to compromise gut permeability by altering microbial composition and increasing cortisol levels, which negatively impact tight junction function (Casterline et al.,2017).

4.5 Targeting Gut Permeability for AD Prevention

Given the strong link between gut permeability and AD, therapeutic strategies aimed at restoring gut integrity could provide promising avenues for disease prevention and management. Emerging research suggests that targeting intestinal permeability through dietary modifications, probiotic supplementation, and gut microbiota modulation may help reduce neuroinflammation and slow cognitive decline. One potential approach is the use of gut-targeted anti-inflammatory compounds, such as polyphenols and omega-3 fatty acids, which have been shown to improve gut barrier function and reduce systemic inflammation (Cattaneo et al.,2017). Additionally, fecal microbiota transplantation (FMT) is being explored as a method to restore a healthy gut microbiome and mitigate gut-derived inflammation in neurodegenerative diseases.Gut permeability and endotoxemia represent critical links between the gastrointestinal tract and Alzheimer’s disease. A compromised gut barrier allows bacterial endotoxins to enter circulation, triggering systemic inflammation that ultimately affects brain health. Understanding the role of gut permeability in AD progression provides opportunities for targeted interventions that could help reduce neuroinflammation and improve cognitive function. Future research should focus on clinical trials to explore the efficacy of gut-targeted therapies in preventing and managing Alzheimer’s disease (Chang et al.,2014).

5. Probiotics and Their Potential Role in Alzheimer's Prevention


The gut microbiome has emerged as a key player in brain health, influencing inflammation, neurotransmission, and cognitive function. With growing evidence linking dysbiosis to Alzheimer’s disease (AD), researchers have turned their attention to probiotics—live microorganisms that confer health benefits when consumed in adequate amounts. Probiotics have been shown to restore microbial balance, enhance gut barrier function, and modulate neuroinflammation, making them a promising therapeutic option for AD prevention (Chaudhri et al.,2006).

5.1The Role of Probiotics in Restoring Gut Microbial Balance

Probiotics primarily consist of beneficial bacteria from the Lactobacillus and Bifidobacterium genera, which play a crucial role in maintaining gut homeostasis. These microbes help regulate immune function, produce neurotransmitters, and inhibit the growth of harmful bacteria. In AD patients, gut dysbiosis is often characterized by a reduction in beneficial bacteria and an overgrowth of pathogenic species that contribute to systemic inflammation and cognitive decline (Chen et al.,2017). Studies have demonstrated that probiotic supplementation can restore microbial balance and reduce markers of inflammation. In animal models of AD, probiotics have been shown to lower amyloid-beta (Aß) accumulation, improve synaptic plasticity, and enhance cognitive performance. Similarly, clinical trials in older adults have reported improvements in memory and executive function following probiotic administration (Choi et al.,2016).

5.2 Probiotics and the Gut-Brain Axis

The gut-brain axis serves as a bidirectional communication system that links the gastrointestinal tract and the central nervous system. Probiotics influence this axis through multiple mechanisms, including the production of neuroactive compounds, modulation of immune responses, and maintenance of gut barrier integrity (Chong et al.,2018). One key way probiotics impact brain health is through the production of short-chain fatty acids (SCFAs), such as butyrate, acetate, and propionate. SCFAs exert neuroprotective effects by reducing neuroinflammation, enhancing blood-brain barrier (BBB) integrity, and stimulating the release of brain-derived neurotrophic factor (BDNF), which supports neuronal survival and function. Additionally, certain probiotic strains, such as Lactobacillus rhamnosus and Bifidobacterium longum, have been shown to modulate the hypothalamic-pituitary-adrenal (HPA) axis, reducing stress-related inflammation and improving cognitive function. Stress is a known risk factor for AD, and interventions that lower stress-induced inflammation could help mitigate disease progression (Dai et al.,2022).

5.3Probiotics and Neuroinflammation in Alzheimer's Disease

Chronic neuroinflammation is a major driver of AD pathology, contributing to amyloid plaque formation, tau hyperphosphorylation, and neuronal damage (Bakar et al., 2025). Probiotics have demonstrated anti-inflammatory properties that may help counteract these effects. For instance, probiotic supplementation has been shown to reduce levels of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-a), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß). These cytokines are commonly elevated in AD patients and play a role in exacerbating neuronal damage (Den et al.,2020). By modulating immune responses, probiotics help create a more favorable environment for neuronal survival and cognitive function. A study involving elderly individuals with mild cognitive impairment (MCI) found that probiotic supplementation led to significant reductions in systemic inflammation and improved cognitive scores compared to a placebo group. These findings suggest that probiotics could serve as a preventive strategy for individuals at risk of developing AD (Desai et al.,2016).

5.4 Probiotics and Amyloid-Beta Clearance

Amyloid-beta (Aß) accumulation is a hallmark of AD, and recent research suggests that probiotics may play a role in its clearance. Gut bacteria can influence Aß metabolism by modulating bile acid production, immune signaling, and gut permeability. Animal studies have shown that probiotic supplementation enhances Aß clearance by increasing the expression of enzymes involved in its degradation (Dodiya et al.,2020). Additionally, probiotics help maintain a healthy gut barrier, preventing the leakage of lipopolysaccharides (LPS) and other endotoxins that promote Aß aggregation. A clinical study involving AD patients reported that a combination of Lactobacillus and Bifidobacterium strains reduced Aß levels in cerebrospinal fluid and improved cognitive function over a 12-week period. These findings highlight the potential of probiotics as a non-invasive intervention for AD management (Dodiya et al.,2019).

5.5 Dietary Sources and Practical Considerations

Probiotics are naturally present in fermented foods such as yogurt, kefir, sauerkraut, kimchi, and miso. Consuming a diet rich in these foods may help support gut health and reduce AD risk. However, probiotic supplements provide a more targeted approach, allowing for the administration of specific strains that have been studied for their cognitive benefits. When selecting probiotic supplements, it is important to consider factors such as strain specificity, dosage, and viability. Research suggests that multi-strain formulations containing Lactobacillus and Bifidobacterium species are most effective in supporting cognitive function and reducing inflammation (D'onofrio et al.,2012). Additionally, ensuring that probiotics are taken with prebiotic fibers, such as inulin and fructooligosaccharides (FOS), can enhance their efficacy by promoting bacterial growth.

5.6 Future Directions and Clinical Implications

While current research supports the role of probiotics in AD prevention, further studies are needed to establish optimal dosages, treatment durations, and strain combinations. Large-scale clinical trials are essential to confirm the long-term benefits of probiotics and determine their effectiveness in different populations (Amin et al., 2025). Moreover, the integration of probiotics into personalized medicine approaches could enhance their therapeutic potential. Advances in microbiome sequencing and artificial intelligence (AI)-based diagnostics may allow for the identification of individuals who would benefit most from probiotic interventions (Dupuis et al.,2023).Probiotics offer a promising avenue for Alzheimer’s prevention by restoring gut microbial balance, modulating neuroinflammation, and enhancing amyloid-beta clearance. The gut-brain axis plays a crucial role in cognitive function, and interventions that support gut health may help mitigate AD progression. While probiotic-rich diets and supplements show potential, further research is needed to optimize their use as part of a comprehensive strategy for brain health (Erny et al.,2020).

6.Conclusion


The gut microbiome is increasingly recognized as a key player in Alzheimer’s disease (AD), influencing neuroinflammation, amyloid-beta accumulation, and cognitive decline via the gut-brain axis. Gut dysbiosis may drive AD pathology by triggering inflammation, weakening the blood-brain barrier, and disrupting neuroprotective mechanisms. Promising interventions include probiotics, prebiotics, and dietary strategies that restore microbial balance, enhance neuroactive compounds, and support brain health. Probiotic strains like Lactobacillus and Bifidobacterium show neuroprotective potential, but more research is needed on formulation and efficacy. Ultimately, microbiome-targeted therapies offer a compelling, innovative direction for AD prevention and treatment, warranting deeper investigation through personalized, longitudinal studies.

References


Acosta C., Anderson H. D., Anderson C. M. (2017). Astrocyte dysfunction in Alzheimer disease. J. Neurosci. Res. 95, 2430–2447. doi: 10.1002/jnr.24075,

Adak A., Khan M. R. (2019). An insight into gut microbiota and its functionalities. Cell. Mol. Life Sci. 76, 473–493. doi: 10.1007/s00018-018-2943-4,

Agirman G., Hsiao E. Y. (2021). SnapShot: the microbiota-gut-brain axis. Cells 184:e2521. doi: 10.1016/j.cell.2021.03.022

Agus A., Planchais J., Sokol H. (2018). Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 23, 716–724. doi: 10.1016/j.chom.2018.05.003

Akhtar A., Sah S. P. (2020). Insulin signaling pathway and related molecules: role in neurodegeneration and Alzheimer's disease. Neurochem. Int. 135:104707. doi: 10.1016/j.neuint.2020.104707

Alexeev E. E., Lanis J. M., Kao D. J., Campbell E. L., Kelly C. J., Battista K. D., et al. (2018). Microbiota-derived indole metabolites promote human and murine intestinal homeostasis through regulation of Interleukin-10 receptor. Am. J. Pathol. 188, 1183–1194. doi: 10.1016/j.ajpath.2018.01.011

Alzheimer’s disease facts and figures (2023). Alzheimer’s disease facts and figures. Alzheimers Dement. 19, 1598–1695. doi: 10.1002/alz.13016

Amin, M. S., Rashid, M. J., Tufael, Rahman, A. (2025). "Probiotics as Emerging Neurotherapeutics in Spinal Cord Injury: Modulating Inflammation, Infection, and Regeneration", Microbial Bioactives, 8(1),1-8,10290. https://doi.org/10.25163/microbbioacts.8110290

Ang Q. Y., Alexander M., Newman J. C., Tian Y., Cai J., Upadhyay V., et al. (2020). Ketogenic diets Alter the gut microbiome resulting in decreased intestinal Th17 cells. Cells 181:e1216. doi: 10.1016/j.cell.2020.04.027

Arumugam M., Raes J., Pelletier E., Le Paslier D., Yamada T., Mende D. R., et al. (2011). Enterotypes of the human gut microbiome. Nature 473, 174–180. doi: 10.1038/nature09944

Atanasov A. G., Zotchev S. B., Dirsch V. M., The International Natural Product Sciences Taskforce. Supuran C. T. (2021). Natural products in drug discovery: advances and opportunities. Nat. Rev. Drug Discov. 20, 200–216. doi: 10.1038/s41573-020-00114-z

Backhed F., Ley R. E., Sonnenburg J. L., Peterson D. A., Gordon J. I. (2005). Host-bacterial mutualism in the human intestine. Science 307, 1915–1920. doi: 10.1126/science.1104816

Bakar, M. A., Sefaut, M., Sakil, M., Afroza, M., Rahman, M. A., Noor, M. S. B., ... & Hasan, M. M. (2025). Targeting p38 MAPK: Molecular Docking and Therapeutic Insights for Alzheimer’s Disease Management. Journal of Primeasia, 6(1), 1-11. https://doi.org/10.25163/primeasia.6110116

Bakar, M. A., Sefaut, M., Sakil, M., Afroza, M., Rahman, M. A., Noor, M. S. B., ... & Hasan, M. M. (2025). Targeting p38 MAPK: Molecular Docking and Therapeutic Insights for Alzheimer’s Disease Management. Journal of Primeasia, 6(1), 1-11. https://doi.org/10.25163/primeasia.6110116

Baloni P., Funk C. C., Yan J., Yurkovich J. T., Kueider-Paisley A., Nho K., et al. (2020). Metabolic network analysis reveals altered bile acid synthesis and metabolism in Alzheimer's disease. Cell Rep Med 1:100138. doi: 10.1016/j.xcrm.2020.100138

Barka E. A., Vatsa P., Sanchez L., Gaveau-Vaillant N., Jacquard C., Meier-Kolthoff J. P., et al. (2016). Taxonomy, physiology, and natural products of Actinobacteria. Microbiol. Mol. Biol. Rev. 80, 1–43. doi: 10.1128/MMBR.00019-15

Blair L. J., Frauen H. D., Zhang B., Nordhues B. A., Bijan S., Lin Y. C., et al. (2015). Tau depletion prevents progressive blood-brain barrier damage in a mouse model of tauopathy. Acta Neuropathol. Commun. 3:8. doi: 10.1186/s40478-015-0186-2

Blazquez E., Velazquez E., Hurtado-Carneiro V., Ruiz-Albusac J. M. (2014). Insulin in the brain: its pathophysiological implications for states related with central insulin resistance, type 2 diabetes and Alzheimer's disease. Front Endocrinol (Lausanne) 5:161. doi: 10.3389/fendo.2014.00161

Bonaz B., Sinniger V., Pellissier S. (2017). The Vagus nerve in the neuro-immune Axis: implications in the pathology of the gastrointestinal tract. Front. Immunol. 8:1452. doi: 10.3389/fimmu.2017.01452

Bonfili L., Cecarini V., Berardi S., Scarpona S., Suchodolski J. S., Nasuti C., et al. (2017). Microbiota modulation counteracts Alzheimer's disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci. Rep. 7:2426. doi: 10.1038/s41598-017-02587-2

Braniste V., Al-Asmakh M., Kowal C., Anuar F., Abbaspour A., Toth M., et al. (2014). The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 6:263ra158. doi: 10.1126/scitranslmed.3009759

Brunt V. E., Larocca T. J., Bazzoni A. E., Sapinsley Z. J., Miyamoto-Ditmon J., Gioscia-Ryan R. A., et al. (2021). The gut microbiome-derived metabolite trimethylamine N-oxide modulates neuroinflammation and cognitive function with aging. Geroscience 43, 377–394. doi: 10.1007/s11357-020-00257-2

Cai Z., Qiao P. F., Wan C. Q., Cai M., Zhou N. K., Li Q. (2018). Role of blood-brain barrier in Alzheimer's disease. J. Alzheimers Dis. 63, 1223–1234. doi: 10.3233/JAD-180098

Cani P. D., Neyrinck A. M., Fava F., Knauf C., Burcelin R. G., Tuohy K. M., et al. (2007). Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 50, 2374–2383. doi: 10.1007/s00125-007-0791-0

Capuron L., Miller A. H. (2011). Immune system to brain signaling: neuropsychopharmacological implications. Pharmacol. Ther. 130, 226–238. doi: 10.1016/j.pharmthera.2011.01.014

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

Casterline B. W., Hecht A. L., Choi V. M., Bubeck Wardenburg J. (2017). The Bacteroides fragilis pathogenicity island links virulence and strain competition. Gut Microbes 8, 374–383. doi: 10.1080/19490976.2017.1290758

Cattaneo A., Cattane N., Galluzzi S., Provasi S., Lopizzo N., Festari C., et al. (2017). Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 49, 60–68. doi: 10.1016/j.neurobiolaging.2016.08.019

Chang P. V., Hao L., Offermanns S., Medzhitov R. (2014). The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. U. S. A. 111, 2247–2252. doi: 10.1073/pnas.1322269111

Chaudhri O., Small C., Bloom S. (2006). Gastrointestinal hormones regulating appetite. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 361, 1187–1209. doi: 10.1098/rstb.2006.1856

Chen D., Yang X., Yang J., Lai G., Yong T., Tang X., et al. (2017). Prebiotic effect of Fructooligosaccharides from Morinda officinalis on Alzheimer's disease in rodent models by targeting the microbiota-gut-brain Axis. Front. Aging Neurosci. 9:403. doi: 10.3389/fnagi.2017.00403

Choi H. S., Cho J. Y., Jin M. R., Lee Y. G., Kim S. J., Ham K. S., et al. (2016). Phenolics, acyl galactopyranosyl glycerol, and lignan amides from Tetragonia tetragonioides (pall.) Kuntze. Food Sci. Biotechnol. 25, 1275–1281. doi: 10.1007/s10068-016-0201-9

Chong C. Y. L., Bloomfield F. H., O'sullivan J. M. (2018). Factors affecting gastrointestinal microbiome development in neonates. Nutrients 10:274. doi: 10.3390/nu10030274

Dai R., Sun Y., Su R., Gao H. (2022). Anti-Alzheimer's disease potential of traditional chinese medicinal herbs as inhibitors of BACE1 and AChE enzymes. Biomed. Pharmacother. 154:113576. doi: 10.1016/j.biopha.2022.113576

Den H., Dong X., Chen M., Zou Z. (2020). Efficacy of probiotics on cognition, and biomarkers of inflammation and oxidative stress in adults with Alzheimer's disease or mild cognitive impairment - a meta-analysis of randomized controlled trials. Aging (Albany NY) 12, 4010–4039. doi: 10.18632/aging.102810

Desai M. S., Seekatz A. M., Koropatkin N. M., Kamada N., Hickey C. A., Wolter M., et al. (2016). A dietary Fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cells 167, 1339–1353.e1321. doi: 10.1016/j.cell.2016.10.043

Dodiya H. B., Frith M., Sidebottom A., Cao Y., Koval J., Chang E., et al. (2020). Synergistic depletion of gut microbial consortia, but not individual antibiotics, reduces amyloidosis in APPPS1-21 Alzheimer's transgenic mice. Sci. Rep. 10:8183. doi: 10.1038/s41598-020-64797-5

Dodiya H. B., Kuntz T., Shaik S. M., Baufeld C., Leibowitz J., Zhang X., et al. (2019). Sex-specific effects of microbiome perturbations on cerebral Aβ amyloidosis and microglia phenotypes. J. Exp. Med. 216, 1542–1560. doi: 10.1084/jem.20182386

D'onofrio G., Sancarlo D., Panza F., Copetti M., Cascavilla L., Paris F., et al. (2012). Neuropsychiatric symptoms and functional status in Alzheimer's disease and vascular dementia patients. Curr. Alzheimer Res. 9, 759–771. doi: 10.2174/156720512801322582

Dupuis J. P., Nicole O., Groc L. (2023). NMDA receptor functions in health and disease: old actor, new dimensions. Neuron 111, 2312–2328. doi: 10.1016/j.neuron.2023.05.002

Erny D., Prinz M. (2020). How microbiota shape microglial phenotypes and epigenetics. Glia 68, 1655–1672. doi: 10.1002/glia.23822

Fan X., Liu B., Zhou J., Gu X., Zhou Y., Yang Y., et al. (2021). High-fat diet alleviates Neuroinflammation and metabolic disorders of APP/PS1 mice and the intervention with Chinese medicine. Front. Aging Neurosci. 13:658376. doi: 10.3389/fnagi.2021.658376

Grieco M., Giorgi A., Gentile M. C., D'erme M., Morano S., Maras B., et al. (2019). Glucagon-like Peptide-1: a focus on neurodegenerative diseases. Front. Neurosci. 13:1112. doi: 10.3389/fnins.2019.01112

Abstract
Export Citation

View Dimensions


View Plumx


View Altmetric



0
Save
0
Citation
32
View
0
Share