Neuroinflammation in Alzheimer’s Disease: Microbiome Mechanisms and Innovative Therapeutic Approaches

Alzheimer’s disease represents a complex neurodegenerative disorder characterized by progressive cognitive decline, amyloid-beta accumulation, and persistent neuroinflammation. Recent groundbreaking research has revealed the gut microbiome as a critical orchestrator of neuroinflammatory processes, fundamentally altering our understanding of Alzheimer’s pathogenesis. The microbiota-gut-brain axis serves as a bidirectional communication network wherein microbial dysbiosis triggers cascading inflammatory responses that propagate to the central nervous system. This comprehensive analysis examines the intricate molecular mechanisms by which gut microorganisms modulate neuroinflammation through metabolite production, immune system activation, blood-brain barrier disruption, and neurotransmitter regulation. Furthermore, we explore revolutionary therapeutic interventions targeting the gut microbiome, including precision probiotic therapies, metabolite-based treatments, and personalized microbiome restoration protocols. Understanding these complex interactions opens unprecedented opportunities for developing targeted interventions that could prevent, slow, or potentially reverse Alzheimer’s disease progression through microbiome-mediated neuroprotection.

Introduction

The emergence of Alzheimer’s disease as a global health crisis has necessitated a paradigm shift in our understanding of neurodegenerative pathophysiology. While traditional research has focused primarily on brain-centric mechanisms involving amyloid-beta plaques and neurofibrillary tangles, mounting evidence reveals a more comprehensive picture wherein peripheral factors, particularly the gut microbiome, play pivotal roles in disease initiation and progression. The concept of neuroinflammation as a driving force in Alzheimer’s pathogenesis has gained substantial traction, with chronic microglial activation and astrocytic dysfunction emerging as central pathological features that precede and accelerate cognitive decline.

The human gut microbiome, comprising trillions of microorganisms including bacteria, archaea, fungi, and viruses, functions as a metabolically active organ that profoundly influences host physiology. The discovery of the microbiota-gut-brain axis has revolutionized neuroscience research, revealing bidirectional communication pathways that link intestinal microbial communities with brain function and behavior. This axis operates through multiple interconnected mechanisms including vagal nerve signaling, immune system modulation, metabolite production, and neuroendocrine pathways.

In Alzheimer’s disease, the gut microbiome undergoes significant compositional changes characterized by reduced diversity, altered metabolic output, and dysregulated immune interactions. These perturbations create a pro-inflammatory environment that extends beyond the intestinal tract, ultimately contributing to neuroinflammation and neurodegeneration. The recognition of this microbiome-mediated pathophysiology has opened exciting avenues for therapeutic intervention, offering hope for treatments that address Alzheimer’s disease from a systemic rather than purely neurological perspective.

The Microbiome-Neuroinflammation Nexus

The relationship between gut microbiome composition and neuroinflammatory processes in Alzheimer’s disease represents a complex interplay of molecular mechanisms that operate across multiple biological systems. The healthy gut microbiome maintains immune homeostasis through the production of anti-inflammatory mediators, preservation of intestinal barrier integrity, and regulation of peripheral immune cell populations. However, in Alzheimer’s disease, this delicate balance becomes disrupted, leading to a state of chronic low-grade inflammation that perpetuates neurodegeneration.

Microbial dysbiosis in Alzheimer’s patients is characterized by specific taxonomic alterations including decreased abundance of beneficial bacteria such as Bifidobacterium and Lactobacillus, coupled with increased populations of potentially pathogenic organisms including Escherichia coli and other gram-negative bacteria. These compositional changes have profound functional consequences, as the altered microbial community produces different metabolic products that can either promote or inhibit inflammatory responses.

The transition from a healthy, diverse microbiome to a dysbiotic state triggers multiple pathological cascades that ultimately converge on neuroinflammatory pathways. Beneficial microorganisms typically produce short-chain fatty acids, neurotransmitter precursors, and other bioactive compounds that support neural health and immune regulation. Conversely, dysbiotic communities generate pro-inflammatory metabolites, endotoxins, and neurotoxic compounds that can breach the intestinal barrier and access systemic circulation.

This shift in microbial metabolism creates a permissive environment for systemic inflammation, which subsequently impacts the central nervous system through multiple routes. The blood-brain barrier, normally a protective interface that restricts the passage of potentially harmful substances, becomes compromised under conditions of chronic inflammation, allowing inflammatory mediators and microbial products to access brain tissue and activate resident immune cells.

Molecular Pathways of Microbiome-Mediated Neuroinflammation

Short-Chain Fatty Acid Signaling Networks

Short-chain fatty acids, primarily acetate, propionate, and butyrate, represent the most extensively studied microbiome-derived metabolites with direct neuroprotective properties. These compounds are produced through bacterial fermentation of dietary fiber and serve as signaling molecules that modulate immune function, maintain barrier integrity, and regulate gene expression. In healthy individuals, adequate SCFA production supports anti-inflammatory responses and promotes microglial homeostasis.

Butyrate, in particular, demonstrates remarkable neuroprotective capabilities through its dual mechanisms of action as a histone deacetylase inhibitor and G-protein coupled receptor agonist. By inhibiting HDAC activity, butyrate promotes the expression of anti-inflammatory genes while suppressing pro-inflammatory transcriptional programs in microglia and astrocytes. This epigenetic modulation creates a favorable environment for neuronal survival and synaptic maintenance.

The SCFA-mediated activation of GPR41 and GPR43 receptors triggers downstream signaling cascades that ultimately suppress NF-κB activation, reduce inflammatory cytokine production, and enhance the expression of tight junction proteins that maintain blood-brain barrier integrity. These effects collectively contribute to the resolution of neuroinflammation and the preservation of cognitive function.

In Alzheimer’s disease, the capacity for SCFA production becomes significantly impaired due to the loss of fiber-fermenting bacterial species and alterations in microbial metabolic pathways. This deficiency in protective metabolites removes a critical neuroprotective mechanism and allows pro-inflammatory processes to predominate, accelerating disease progression.

Tryptophan Metabolism and Neuromodulation

The amino acid tryptophan serves as a precursor for multiple bioactive compounds including serotonin, kynurenine, and indole derivatives, each of which can profoundly influence neuroinflammatory processes. The gut microbiome plays a central role in tryptophan metabolism, directing its conversion toward either neuroprotective or neurotoxic pathways depending on the composition and functional capacity of the microbial community.

Under healthy conditions, specific bacterial species promote the conversion of tryptophan to beneficial metabolites such as indole-3-propionic acid and other indole derivatives that activate the aryl hydrocarbon receptor pathway. This activation results in the production of anti-inflammatory cytokines, enhanced barrier function, and reduced microglial activation. Additionally, some microorganisms can directly synthesize serotonin or provide the enzymatic machinery necessary for its production by host cells.

The kynurenine pathway represents an alternative route of tryptophan metabolism that can have either neuroprotective or neurotoxic effects depending on the specific metabolites produced. In neuroinflammatory conditions, tryptophan is preferentially shunted toward the production of quinolinic acid and other neurotoxic kynurenine metabolites that can exacerbate neuronal damage and promote further inflammation.

Alzheimer’s disease is associated with dysregulated tryptophan metabolism characterized by increased production of neurotoxic metabolites and decreased synthesis of protective compounds. This metabolic imbalance contributes to the maintenance of chronic neuroinflammation and may directly contribute to neuronal dysfunction and death.

Trimethylamine N-Oxide and Neurodegeneration

Trimethylamine N-oxide represents a concerning example of how dysbiotic microbial communities can produce metabolites that directly promote neurodegeneration. TMAO is generated through the hepatic oxidation of trimethylamine, which is produced by specific bacterial species during the metabolism of choline, carnitine, and other quaternary ammonium compounds present in the diet.

Elevated TMAO levels have been consistently associated with increased risk of cardiovascular disease, and recent research has revealed significant associations between TMAO concentrations and Alzheimer’s disease severity. TMAO can cross the blood-brain barrier and directly activate microglia and astrocytes, promoting the release of inflammatory mediators and contributing to neuronal damage.

The molecular mechanisms underlying TMAO-induced neuroinflammation involve the activation of multiple signaling pathways including the NLRP3 inflammasome, NF-κB transcriptional program, and oxidative stress response systems. These pathways converge to create a pro-inflammatory environment that accelerates amyloid-beta accumulation, tau phosphorylation, and synaptic dysfunction.

Therapeutic interventions targeting TMAO production have shown promise in preclinical models, suggesting that modulation of specific microbial metabolic pathways could provide novel approaches for Alzheimer’s prevention and treatment.

Immune System Dysregulation and Peripheral Inflammation

The gut microbiome serves as a primary regulator of immune system development and function, with approximately seventy percent of the body’s immune cells residing within gut-associated lymphoid tissue. This intimate relationship between microorganisms and immune cells creates opportunities for both beneficial immune education and pathological immune activation depending on the composition and behavior of the microbial community.

In healthy individuals, the gut microbiome promotes immune tolerance through the induction of regulatory T cell populations, the production of anti-inflammatory mediators, and the maintenance of appropriate immune surveillance mechanisms. These processes help prevent excessive inflammatory responses while maintaining the capacity to respond to genuine threats.

Alzheimer’s disease is associated with significant perturbations in peripheral immune function that appear to precede and contribute to neuroinflammation. Dysbiotic microbial communities promote the expansion of pro-inflammatory immune cell populations including Th17 cells, which produce interleukin-17 and other inflammatory cytokines that can access the central nervous system and activate resident immune cells.

The loss of beneficial bacterial species also reduces the production of immune-regulatory metabolites and removes important signals that normally promote anti-inflammatory responses. This creates a permissive environment for chronic inflammation that extends beyond the gut and contributes to systemic immune dysfunction.

The concept of “inflammaging” describes the age-related increase in inflammatory mediators that contributes to multiple chronic diseases including Alzheimer’s disease. The gut microbiome appears to be a central driver of inflammaging, with age-related changes in microbial composition promoting increased inflammatory signaling and reduced immune regulation.

Blood-Brain Barrier Disruption and Neuroinvasion

The blood-brain barrier represents a critical protective interface that normally prevents the passage of potentially harmful substances from the systemic circulation into brain tissue. This selective barrier is maintained through tight junctions between endothelial cells, specialized transport systems, and active efflux mechanisms that work together to preserve the unique environment required for optimal brain function.

Neuroinflammation, whether initiated within the brain or triggered by peripheral inflammatory signals, can significantly compromise blood-brain barrier integrity through multiple mechanisms. Inflammatory mediators such as tumor necrosis factor-alpha, interleukin-1beta, and interferon-gamma can disrupt tight junction proteins, increase vascular permeability, and alter transport system function.

The gut microbiome influences blood-brain barrier function through both direct and indirect mechanisms. Beneficial microorganisms produce metabolites such as short-chain fatty acids that help maintain barrier integrity, while pathogenic species can generate endotoxins and other compounds that promote barrier disruption. Additionally, systemic inflammation triggered by intestinal dysbiosis can create conditions that compromise the blood-brain barrier even in the absence of direct microbial products.

In Alzheimer’s disease, blood-brain barrier dysfunction appears to be an early pathological feature that precedes significant cognitive decline. This barrier disruption allows the infiltration of peripheral immune cells, inflammatory mediators, and potentially toxic substances that can contribute to neuroinflammation and accelerate disease progression.

The recognition of blood-brain barrier dysfunction as both a consequence and contributor to Alzheimer’s pathophysiology has important therapeutic implications, suggesting that interventions aimed at preserving or restoring barrier function could provide significant clinical benefits.

Vagal Nerve Modulation and Neuroimmune Communication

The vagus nerve represents a major bidirectional communication pathway between the gut and brain that plays crucial roles in both physiological homeostasis and pathological processes. This extensive neural network carries both sensory information from the gut to the brain and motor commands from the brain to various organs including the digestive system.

Recent research has revealed that the vagus nerve serves as an important conduit for microbiome-brain communication, with specific bacterial species capable of producing neurotransmitters and other signaling molecules that can influence vagal signaling. This microbial-neural interface provides a direct mechanism through which gut microorganisms can influence brain function and behavior.

The vagus nerve also plays important roles in immune regulation through the so-called “cholinergic anti-inflammatory pathway.” Activation of this pathway leads to the release of acetylcholine, which binds to nicotinic receptors on macrophages and other immune cells, resulting in the suppression of inflammatory cytokine production and the promotion of anti-inflammatory responses.

In Alzheimer’s disease, vagal function appears to be compromised, potentially contributing to both peripheral immune dysfunction and central nervous system pathology. Reduced vagal tone has been associated with increased inflammatory signaling, suggesting that interventions aimed at enhancing vagal function could provide therapeutic benefits.

Emerging therapeutic approaches including vagal nerve stimulation and targeted interventions designed to enhance vagal signaling show promise for modulating neuroinflammation and potentially slowing Alzheimer’s disease progression.

Innovative Therapeutic Approaches

Precision Probiotic Interventions

The development of precision probiotic therapies represents a significant advancement beyond traditional broad-spectrum probiotic approaches. These targeted interventions involve the selection of specific bacterial strains based on their demonstrated ability to produce beneficial metabolites, modulate immune responses, or address particular aspects of Alzheimer’s pathophysiology.

Recent clinical trials have demonstrated the potential efficacy of targeted probiotic interventions in improving cognitive function and reducing inflammatory markers in individuals with mild cognitive impairment and early-stage Alzheimer’s disease. These studies have identified specific bacterial strains that show particular promise for neurotherapeutic applications.

Lactobacillus helveticus and Bifidobacterium longum have demonstrated ability to produce gamma-aminobutyric acid and other neurotransmitters that can influence brain function and behavior. Additionally, these organisms produce metabolites that support blood-brain barrier integrity and reduce neuroinflammation.

The future of probiotic therapy lies in the development of personalized approaches based on individual microbiome profiles, genetic factors, and specific disease characteristics. This precision medicine approach could maximize therapeutic efficacy while minimizing potential adverse effects.

Metabolite-Based Therapeutics

The direct administration of beneficial microbial metabolites represents an innovative therapeutic strategy that bypasses the need to establish specific microbial populations within the gut. This approach offers several advantages including precise dosing, consistent bioavailability, and the ability to target specific molecular pathways.

Short-chain fatty acid supplementation has shown promise in preclinical studies for reducing neuroinflammation and preserving cognitive function. Sodium butyrate and other SCFA derivatives can cross the blood-brain barrier and directly modulate microglial activation, gene expression, and inflammatory signaling pathways.

Indole-3-propionic acid and other tryptophan-derived metabolites represent another class of potentially therapeutic compounds that can be administered directly rather than relying on microbial production. These metabolites have demonstrated neuroprotective effects through their ability to activate beneficial signaling pathways and reduce oxidative stress.

The development of novel delivery systems including nanoparticle formulations and sustained-release preparations could enhance the therapeutic potential of metabolite-based interventions by improving bioavailability and extending duration of action.

Fecal Microbiota Transplantation and Beyond

Fecal microbiota transplantation represents the most comprehensive approach to microbiome restoration, involving the transfer of entire microbial communities from healthy donors to recipients with dysbiotic conditions. While primarily developed for the treatment of recurrent Clostridium difficile infections, FMT is now being investigated for a wide range of conditions including neurological disorders.

Early clinical reports have described improvements in cognitive function and behavioral symptoms following FMT in Alzheimer’s patients, suggesting that comprehensive microbiome restoration may provide therapeutic benefits. However, the mechanisms underlying these improvements remain incompletely understood, and standardized protocols for neurological applications are still under development.

The future evolution of FMT may involve the development of defined microbial consortia that contain specific combinations of beneficial organisms selected for their neurotherapeutic properties. These standardized preparations could provide the benefits of comprehensive microbiome restoration while offering greater safety and reproducibility than traditional FMT approaches.

Advanced techniques including encapsulated microbial preparations and targeted delivery systems may further enhance the therapeutic potential of microbiome restoration interventions.

Advanced Mechanistic Insights

Epigenetic Regulation of Neuroinflammation

The microbiome’s influence on neuroinflammation extends beyond direct metabolic effects to include epigenetic modifications that can have long-lasting impacts on gene expression and cellular function. Microbial metabolites such as short-chain fatty acids serve as histone deacetylase inhibitors, directly modulating chromatin structure and transcriptional activity in neural and immune cells.

These epigenetic modifications can influence the expression of genes involved in inflammatory signaling, microglial activation, and neuronal survival. The ability of microbial metabolites to create lasting changes in gene expression patterns suggests that microbiome-based interventions could have sustained therapeutic effects that persist beyond the period of active treatment.

Recent research has revealed that specific bacterial species can influence DNA methylation patterns in host cells, providing another mechanism through which the microbiome can modulate gene expression and cellular behavior. These findings suggest that the relationship between microbiome and host extends to fundamental aspects of cellular regulation and identity.

The recognition of microbiome-mediated epigenetic regulation has important implications for understanding both disease pathogenesis and therapeutic intervention, suggesting that targeting these pathways could provide novel approaches for modulating neuroinflammation and preserving cognitive function.

Mitochondrial Function and Metabolic Integration

The gut microbiome influences mitochondrial function through multiple mechanisms including the production of metabolites that serve as energy substrates, the modulation of oxidative stress responses, and the regulation of mitochondrial biogenesis. These effects have particular relevance for Alzheimer’s disease, where mitochondrial dysfunction is recognized as a key pathological feature.

Short-chain fatty acids produced by beneficial bacteria can serve as alternative energy sources for brain cells, potentially compensating for glucose metabolism deficits that occur in Alzheimer’s disease. Additionally, these metabolites can enhance mitochondrial function and promote the generation of new mitochondria through activation of specific transcriptional programs.

The microbiome also influences the production and metabolism of key cofactors required for optimal mitochondrial function, including B vitamins, coenzyme Q10 precursors, and other essential nutrients. Dysbiotic conditions may compromise the availability of these cofactors, contributing to mitochondrial dysfunction and cellular energy deficits.

Therapeutic interventions targeting microbiome-mitochondria interactions represent an emerging area of research that could provide novel approaches for addressing the metabolic aspects of Alzheimer’s disease pathophysiology.

Synaptic Plasticity and Neurotransmitter Systems

The gut microbiome influences synaptic function and plasticity through the production of neurotransmitters, neurotransmitter precursors, and modulators of synaptic signaling. Many bacterial species are capable of producing gamma-aminobutyric acid, serotonin, dopamine, and other neurotransmitters that can influence brain function when they reach the central nervous system.

Additionally, microbial metabolites can modulate the expression and function of neurotransmitter receptors, transporters, and synthetic enzymes, creating indirect effects on synaptic transmission and plasticity. These effects may be particularly important in Alzheimer’s disease, where synaptic dysfunction occurs early in the disease process and contributes significantly to cognitive decline.

The microbiome also influences the production of brain-derived neurotrophic factor and other growth factors that support synaptic health and neuronal survival. Dysbiotic conditions may reduce the availability of these protective factors, contributing to synaptic loss and cognitive impairment.

Understanding the microbiome’s role in synaptic function provides additional rationale for microbiome-based therapeutic interventions and suggests that these approaches could provide benefits beyond their anti-inflammatory effects.

Clinical Translation and Future Directions

Biomarker Development and Precision Medicine

The translation of microbiome research into clinical practice requires the development of reliable biomarkers that can predict treatment response, monitor therapeutic efficacy, and guide personalized intervention strategies. Current research efforts are focused on identifying specific microbial signatures, metabolite profiles, and immune markers that correlate with disease progression and treatment outcomes.

Advanced analytical techniques including metabolomics, proteomics, and multi-omics integration are providing new insights into the complex relationships between microbiome composition, metabolic function, and host physiology. These approaches may ultimately enable the development of personalized treatment protocols based on individual microbiome profiles and specific disease characteristics.

The integration of artificial intelligence and machine learning approaches with microbiome data analysis holds promise for identifying complex patterns and relationships that may not be apparent through traditional analytical methods. These computational approaches could accelerate biomarker discovery and enhance the precision of therapeutic interventions.

Regulatory Considerations and Safety Profiles

The clinical development of microbiome-based therapeutics faces unique regulatory challenges related to the complexity and variability of microbial products. Traditional pharmaceutical development paradigms may not be directly applicable to interventions involving live microorganisms or complex microbial metabolite mixtures.

Safety considerations for microbiome interventions include the potential for unintended ecological effects, the risk of transferring antibiotic resistance genes, and the possibility of promoting the growth of potentially pathogenic organisms. Comprehensive safety assessment protocols are being developed to address these concerns and ensure the safe clinical application of microbiome-based therapies.

The establishment of standardized protocols for microbiome intervention development, including good manufacturing practices for microbial products and standardized efficacy assessment methods, will be essential for advancing the field and ensuring reproducible clinical outcomes.

Combination Therapies and Integrated Approaches

The future of Alzheimer’s treatment likely lies in combination approaches that address multiple aspects of disease pathophysiology simultaneously. Microbiome-based interventions could be integrated with existing pharmaceutical treatments, lifestyle modifications, and other therapeutic modalities to provide comprehensive disease management.

Combination strategies might include the simultaneous administration of probiotics with anti-inflammatory drugs, the integration of dietary interventions with targeted metabolite supplementation, or the coordination of microbiome restoration with cognitive training programs. These integrated approaches could provide synergistic benefits that exceed the effects of individual interventions.

The development of personalized combination therapy protocols based on individual patient characteristics, disease stage, and therapeutic response profiles represents an important future direction that could maximize treatment efficacy while minimizing adverse effects.

Comparative Analysis of Therapeutic Modalities

Intervention TypeMechanism of ActionClinical EvidenceAdvantagesLimitations
Precision ProbioticsTargeted bacterial strain administrationPhase II trials showing cognitive improvementSpecific strain selection, measurable outcomesIndividual variability in colonization
Metabolite TherapyDirect administration of beneficial compoundsPreclinical studies, early human trialsPrecise dosing, consistent bioavailabilityLimited long-term data, delivery challenges
Fecal Microbiota TransplantationComprehensive microbiome restorationCase reports, ongoing clinical trialsRapid microbiome restructuringSafety concerns, standardization issues
Dietary InterventionsMicrobiome modulation through nutritionMultiple observational studiesNon-invasive, cost-effectiveSlow onset, compliance challenges
Prebiotic SupplementationSelective promotion of beneficial bacteriaMixed clinical resultsSafe profile, targeted approachVariable efficacy, individual responses

Mechanistic Pathways and Therapeutic Targets

PathwayKey MediatorsTherapeutic PotentialCurrent Interventions
SCFA SignalingButyrate, Propionate, AcetateHigh – direct neuroprotectionFiber supplementation, targeted probiotics
Tryptophan MetabolismIndole derivatives, KynurenineModerate – complex regulationProbiotic strains, dietary modification
Immune ModulationRegulatory T cells, CytokinesHigh – systemic effectsAnti-inflammatory probiotics, prebiotics
Vagal StimulationAcetylcholine, Neural signalingEmerging – direct brain communicationVagal nerve stimulation, specific bacterial strains
Barrier FunctionTight junction proteins, PermeabilityHigh – foundational protectionSCFA therapy, beneficial bacteria

Conclusion

The emerging understanding of neuroinflammation in Alzheimer’s disease as a microbiome-mediated phenomenon represents a fundamental shift in our conceptualization of neurodegenerative pathophysiology. The gut microbiome’s role as a central orchestrator of inflammatory processes, immune system function, and neural communication provides unprecedented opportunities for therapeutic intervention that extend far beyond traditional brain-focused approaches.

The complex molecular mechanisms underlying microbiome-brain interactions involve multiple interconnected pathways including metabolite production, immune system modulation, barrier function regulation, and direct neural communication. These pathways operate synergistically to influence neuroinflammation and ultimately determine the trajectory of cognitive decline in Alzheimer’s disease.

Innovative therapeutic approaches targeting the gut microbiome offer hope for more effective treatments that address the root causes of neuroinflammation rather than merely managing its consequences. The development of precision interventions based on individual microbiome profiles, specific metabolite pathways, and personalized therapeutic protocols represents the future of Alzheimer’s treatment.

The translation of these scientific advances into clinical practice will require continued research efforts, innovative regulatory frameworks, and collaborative approaches that integrate multiple disciplines and therapeutic modalities. The potential for microbiome-based interventions to prevent, slow, or potentially reverse Alzheimer’s disease progression represents one of the most promising developments in neurodegenerative disease research.

As our understanding of the microbiome-brain axis continues to evolve, we can anticipate the emergence of increasingly sophisticated therapeutic strategies that harness the power of microbial communities to promote brain health and cognitive resilience. The future of Alzheimer’s treatment may ultimately depend on our ability to restore and maintain the delicate balance between host and microbiome that is essential for optimal neural function and lifelong cognitive health.

The integration of microbiome science with traditional neuroscience approaches promises to yield transformative insights that could fundamentally change how we prevent, diagnose, and treat Alzheimer’s disease. This paradigm shift from brain-centric to systems-based thinking represents not just a scientific advancement but a potential revolution in our approach to neurodegenerative disease that could benefit millions of individuals worldwide.

 

Inline Feedbacks
View all comments
guest