Next Article in Journal
Enamel Maturation as a Systems Physiology: Ion Transport and Pi Flux
Previous Article in Journal
The m6A Modification in Neurodegenerative Disease: A Cellular Perspective
Previous Article in Special Issue
Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain–Gut–Microbiome Axis: A Focus on Sex Differences
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Valeric Acid: A Gut-Derived Metabolite as a Potential Epigenetic Modulator of Neuroinflammation in the Gut–Brain Axis

by
Chiara Paciolla
1,†,
Michele Manganelli
1,†,
Mariagiovanna Di Chiano
1,
Francesca Montenegro
2,
Anna Gallone
1,
Fabio Sallustio
2 and
Gabriella Guida
1,*
1
Department of Traslational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, 70121 Bari, Italy
2
Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, 70121 Bari, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2025, 14(22), 1823; https://doi.org/10.3390/cells14221823
Submission received: 16 October 2025 / Revised: 12 November 2025 / Accepted: 18 November 2025 / Published: 20 November 2025
(This article belongs to the Special Issue Molecular and Cellular Advances in Gut-Brain Axis)

Highlights

What are the main findings?
  • Valeric Acid (VA), a gut-derived short-chain fatty acid (SCFA), acts as a selective inhibitor of Class I Histone Deacetylases (HDACs), particularly HDAC3.
  • VA modulates neuroinflammation and promotes neuroprotection by both epigenetic and GABAergic mechanisms.
What are the implications of the main findings?
  • VA offers a safer, physiological strategy to the non-selective pharmacological analogue, Valproic Acid (VPA), which is limited by significant systemic toxicity.
  • Elucidating VA’s role might promote microbiome-derived compounds for targeted epigenetic modulation of neurodegenerative disorders.

Abstract

The gut–brain axis (GBA) is a critical area of research for understanding the pathogenesis of neuroinflammatory and neurodegenerative diseases. Metabolites produced by the gut microbiota, particularly short-chain fatty acids (SCFAs), act as key mediators in this bidirectional communication. While the roles of acetate, propionate, and butyrate are well-established, valeric acid (VA), a five-carbon SCFA, is poorly understood. This comprehensive review explores VA as a gut-derived physiological epigenetic modulator, examining its microbial biosynthesis and systemic effects. This review discusses how VA acts as a selective histone deacetylase inhibitor (HDACi), particularly targeting Class I HDACs, to modulate gene expression and exert neuroprotective and anti-inflammatory effects. The analysis compares VA with its pharmacological analog, valproic acid (VPA), a well-known but non-selective HDACi. This comparison highlights how VA’s physiological nature may offer a more targeted and safer intervention. In conclusion, elucidating VA’s role as a microbiome-derived epigenetic regulator would open promising avenues for therapeutic strategies that directly connect gut and CNS health within the GBA.

1. Introduction

An increasingly relevant concept in modern medicine is the intricate bidirectional communication between the gut and the brain, known as the gut–brain axis (GBA) [1,2,3]. This complex network is profoundly influenced by the different and dynamic microbial communities residing in the gastrointestinal tract [4,5,6]. This network allows the gut to communicate with the central nervous system (CNS) through multiple channels, including spinal nerves (especially the vagus nerve) and the systemic circulation, which transports microbial-derived molecules and their precursors [7,8,9]. While a state of eubiosis supports host health through the synthesis of vital bioactive compounds, dysbiosis disrupts GBA signaling [10,11,12,13,14,15,16,17,18,19]. Dysbiosis contributes to pathology by compromising the intestinal epithelial barrier, often referred to as leaky gut [20,21]. This hyperpermeability allows microbial products and pro-inflammatory molecules to cross into the bloodstream, triggering chronic, low-grade systemic inflammation [22,23,24]. This, in turn, can compromise the integrity of the blood-brain barrier (BBB), causing the translocation of harmful substances into the CNS and triggering neuroinflammation [25,26].
Neuroinflammation represents a highly coordinated immune response within the CNS, involving the interplay of microglia, astrocytes, neurons, and endothelial cells of the BBB [27,28,29]. Although initially protective, its prolonged state would evolve into a chronic, damaging process characterized by the persistent release of pro-inflammatory cytokines and reactive oxygen species (ROS), leading to neuronal dysfunction and neurodegeneration [30]. Activated microglia [29], often adopting a pro-inflammatory M1 phenotype, release key cytokines such as TNF-α, IL-1β, IL-6, and COX-2, promoting neuronal injury [31]. Concurrently, reactive astrocytes amplify the cascade by secreting VEGF-A, MMP-9, and MCP-1, which increase BBB permeability, alongside IL-6 and oncostatin M that downregulate tight junction proteins such as claudins [32]. This BBB disruption allows the infiltration of peripheral immune cells and the entry of plasma proteins like fibrinogen, which primes microglial NLRP3-inflammasome, driving the sustained release of IL-1β and IL-6 and further inflammatory spread [33,34,35]. Dysbiosis-induced inflammation is a major contributor to this process [36,37]. The progression and severity of chronic neurodegenerative diseases like Alzheimer’s Disease (AD), Parkinson’s Disease (PD), Multiple Sclerosis (MS), and certain forms of Frontotemporal Dementia (FTD) also result from a complex interplay crossing genetic susceptibility variants and environmental factors that shape the epigenome [38,39]. Epigenetic modifications, such as DNA methylation and histone modifications (acetylation, methylation, phosphorylation), and the activity of non-coding RNA (ncRNAs) do not alter the DNA sequence but alter gene expression [40]. This leads to long-lasting modifications crucial to regulatory processes in the CNS, thereby modulating the activity of resident immune cells like microglia and astrocytes [41,42]. For instance, HDAC3 plays a particular pro-inflammatory role, controlling NF-κB activation and the resultant immuno-neuroinflammatory response [43]. Consistently, hypomethylation of NF-κB can lead to the abnormal activation of TNF-α, while histone hyperacetylation can open chromatin and favor the expression of neuroprotective genes, as BDNF [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62], or as for NLRP3, H3K9-acetylation represents a target for therapeutic modulation [63,64,65].
In this context, among the key players linking the GBA to this epigenetic regulation are Short-Chain Fatty Acids (SCFAs), bioactive molecules produced by gut bacteria through the fermentation of dietary fibers and resistant starches [66]. While the major SCFAs (acetate, propionate, and butyrate) are well known, Valeric Acid (VA)—though less abundant—is emerging as a molecule of significant interest [25]. VA is a microbial-derived SCFA, primarily known for its selective inhibition of Histone Deacetylases (HDACs), along with its neuroprotective and anti-inflammatory properties [67,68].
This review explores the role of VA as an emerging gut-derived mediator in the GBA. It investigates its mechanisms of action, including its influence on neuroinflammatory pathways and neurodegenerative diseases. This analysis provides an updated overview of how this small gut-derived molecule could have a profound impact on brain health and highlights its importance for future research.

2. The Gut Microbiota: Eubiosis and Dysbiosis

The gut microbiota refers to the complex and diverse community of bacteria, yeasts, fungi, viruses, and protozoa that predominantly resides in the gastrointestinal tract, harboring over 70% of all microbes within the human body [69]. In recent decades, the application of high-throughput “omics” technologies, such as metagenomics and metabolomics, has been crucial in elucidating the vast complexity of the gut microbiota ecosystem and its interactions with the host [70]. The microbial ecosystem is predominantly composed of five main bacterial phyla: Firmicutes, Bacteroidetes, Actinobacteria, Fusobacteria, and Proteobacteria, with the addition of Verrucomicrobia and Cyanobacteria. Among these, Firmicutes and Bacteroidetes account for over 90% of the total microbial population [71]. Approximately 99% of these microorganisms are anaerobic [4,72].
There is no universally accepted definition of a “healthy” gut microbiota, as it exists in a state of dynamic equilibrium unique to each individual, that ensures resilience against transient external perturbations, such as fluctuations in diet and lifestyle [73,74,75]. This balanced status, called eubiosis, contributes to host health—defined as the absence of disease—and supports normal physiological function. The relationship between the intestinal microbiota and the host is a mutualistic symbiosis, where the microbiota contributes to host homeostasis by synthesizing bioactive compounds such as vitamins, antimicrobial peptides, and SCFAs [76,77]. Conversely, an imbalance in microbial composition that negatively impacts gastrointestinal, immune, or neural function is termed dysbiosis [78]. Dysbiosis is often caused by factors such as an unhealthy diet, chronic stress, and antibiotic use [79,80]. Alterations in dietary habits represent one of the most influential factors shaping the composition of the individual microbial community [81]. Diets characterized by a low intake of microbiota-accessible carbohydrates, particularly soluble and insoluble fibers commonly present in fruits, vegetables, whole grains, legumes, and potatoes, significantly impair the growth of fermentative bacterial taxa, thereby reducing microbial diversity [82]. Such a condition directly compromises the mutualistic symbiosis essential for maintaining host health, giving rise to a stable dysbiotic configuration characterized by an overrepresentation of opportunistic species and an enhanced risk of disease [83,84]. Accordingly, an altered Firmicutes/Bacteroidetes ratio—specifically, an increase in Firmicutes and a decrease in Bacteroidetes—is a common feature in several disorders, including those involving inflammatory states as neurodegenerative conditions [85,86].
Clinical manifestations most frequently associated with intestinal dysbiosis include abdominal bloating, excessive gas production, and reduced appetite linked to irregular intestinal motility, together with extraintestinal symptoms such as fatigue, depressive states [87].
This loss of symbiosis directly impacts intestinal mucosal health, which is crucial for nutrient absorption and preventing pathogen passage [88,89]. The effectiveness of this epithelial barrier is controlled by tight junction proteins, including transmembrane proteins (occludins and claudins) and intracellular scaffold proteins (ZO-1, -2, and -3) [90]. A disruption in these proteins leads to leaky gut, allowing inflammatory molecules and microbial products to enter the bloodstream, triggering systemic inflammation. This systemic inflammation, as established, subsequently compromises the integrity of the BBB, whose permeability is similarly controlled by tight junction proteins [91].

3. The Role of Short-Chain Fatty Acids (SCFAs): Systemic Impact

SCFAs, including acetate (C2), propionate (C3), and butyrate (C4), typically in a 60:20:20 ratio, alongside lesser amounts of valeric (C5), isobutyric, and isovaleric acids, are produced alongside gases during the fermentation of dietary substrates like fiber and resistant starch—substrates that the host cannot digest due to the lack of a specific repertoire of carbohydrate-active enzymes (CAZymes) [66,92]. Minor SCFAs like isobutyric and isovaleric acids are generated from protein fermentation [90]. Reports consistently show that low-fiber diets are often associated with increased inflammatory disturbances [93], hence adding fibers can counteract negative effects by decreasing inflammatory mediators both in humans and rats [94,95,96,97,98].
The amount and pattern of SCFAs formed are dependent on the type of dietary fiber consumed [99,100]. Butyric acid serves as a primary energy source for colonocytes; in fact, it is promptly metabolized by colonocytes via mitochondrial β-oxidation. By contrast, acetate serves as a substrate for cholesterol biosynthesis, and propionic acid is primarily utilized by hepatocytes for glucose production, which plays a key role in the maintenance of blood glucose levels [66]. Once formed, most SCFAs are rapidly absorbed through the colonic mucosa, but some reach the systemic circulation, where they can have effects on peripheral organs like the liver, heart, and brain (Figure 1) [101]. As weak acids, SCFAs can exist in either protonated or deprotonated form. The protonated SCFAs are easily absorbed via passive diffusion, whereas the deprotonated form requires transporters, including H+-coupled monocarboxylate transporters (MCTs) and sodium-coupled monocarboxylate transporters (SMCTs) [25]. These transporters are expressed in a variety of tissues, including the gut, liver, and brain [101,102]. The regulatory effects of SCFAs on metabolic and immune disorders are mediated by their binding to G-protein coupled receptors (GPRs), such as GPR41 and GPR43 [103]. GPR41 activation is preferred with longer chain lengths (C3–C5), including valeric acid (VA) [104]. Furthermore, SCFAs can regulate the expression of inflammatory mediators by inhibiting histone deacetylases (HDACs), proteins expressed mainly in immune system-mediating cells such as Tregs, antigen-presenting cells (APCs), and effector T cells [105].

4. Valeric Acid: Biosynthesis, Metabolism, and Local Effects

Valeric acid (pentanoic acid, VA) is a naturally occurring SCFA with a five-carbon (C5) straight-chain structure, conferring it moderate lipophilicity and efficient membrane permeability [106]. It was named after its origin in the perennial flowering plant Valeriana officinalis [107,108], whose extracts have long been used for their sedative effects to treat pain, epilepsy, insomnia, anxiety, and depression [109]. VA is mainly produced by anaerobic gut bacteria through a chain elongation process. One key route involves the condensation of ethanol and propionate (Figure 2). Ethanol is first oxidized to acetyl-CoA (Wood–Ljungdahl pathway), while propionate is activated to propionyl-CoA. These two CoA derivatives undergo thiolase-mediated condensation to form 3-ketopentanoyl-CoA, which is then reduced and converted to VA through CoA-thioester intermediates [110]. This pathway shares similarities with butyrate synthesis and reflects metabolic plasticity based on substrate availability. Notably, the balance of ethanol and propionate in the gut influences whether microbes produce butyrate (C4) or valerate (C5) [110].
Once produced in the colon, VA is absorbed as its ionized form, valerate. Locally, VA contributes to the acidic environment in the colon, which inhibits the growth of pathogenic bacteria (e.g., E. coli and Salmonella) and promotes beneficial strains (e.g., Lactobacillus and Bifidobacterium) [111,112]. It is transported by the portal vein to the liver, where it can be metabolized through β-oxidation, and then enters systemic circulation to exert its effects on peripheral organs and the brain [101]. Its physiological role extends beyond energy metabolism, as it acts as a potent signaling molecule. The dietary input, therefore, represents a direct, modifiable link between nutritional status and CNS effects within the GBA [113,114].

5. Valeric Acid as an Epigenetic Modulator

VA also exerts epigenetic effects, particularly through inhibition of HDACs, leading to chromatin remodelling and regulation of gene expression [115,116]. HDACs are a family of enzymes crucial for regulating gene expression by removing acetyl groups from histone and non-histone proteins. HDACs are broadly categorized into four main classes based on their sequence homology, cellular localization, and cofactor requirements. Class I (HDAC1, 2, 3, and 8) are primarily nuclear, zinc-dependent enzymes. Class II is divided into Class IIa (HDAC4, 5, 7, and 9), which shuttle between the nucleus and cytoplasm, and Class IIb (HDAC6 and 10), with HDAC6 being largely cytoplasmic; both are also zinc-dependent. Class III comprises sirtuins (SIRT1-7), which are NAD+-dependent. Finally, Class IV contains only HDAC11, sharing features of both Class I and Class II, and is also zinc-dependent. Classes I, II, and IV are often referred to as “classical” HDACs, as their activity is inhibited by trichostatin A (TSA), unlike the Class III sirtuins [117]. VA acts as an epigenetic modulator through its selective inhibition of class I histone deacetylases (HDACs), especially HDAC3 (Figure 3) [116]. Accordingly, Yuille et al. demonstrated that VA produced by commensal M. massiliensis promoted a strong HDACi effect in IFN-γ, IL-10, IL-1β, and TNF-α cytokine suppression, contributing to anti-inflammatory effects in ulcerative colitis [116]. Furthermore, Han et al. recently highlighted that VA inhibits the expression of HDAC3 with an IC50 of 16.6 µM, therefore suppressing the proliferation of prostate cancer cells [118]. The most likely mechanism of action for VA’s direct inhibition of HDACs is its binding to the zinc ion (Zn2+) located in the enzyme’s catalytic site [119]. While this molecular mechanism is compelling, much of the current understanding relies on in vitro cell-based assays with limited data. Therefore, further studies are needed to fully elucidate the specific and dose-dependent mechanisms of VA’s epigenetic modulation within the GBA.

6. Valeric Acid: Systemic Effects and Neuroprotection

VA binds to GPR41 and GPR43 in various tissues (liver, skeletal muscle, heart, kidney, pancreas, and brain), modulating key processes such as satiety, insulin sensitivity, and inflammation [101,102,103]. Accordingly, Kumari et al. highlighted that VA inhibited the NF-κB signalling pathway, leading to decreased expression of pro-inflammatory cytokines such as TNF-α and IL-6 [120].
Beyond its local effects, VA also encompasses systemic and neurological implications within the GBA [116,121,122,123]. Direct evidence suggests that VA is a key mediator in the development of Postoperative Cognitive Dysfunction (POCD). The increase of VA in the blood observed after surgery in mice was positively associated with the abundance of Lactobacillus and Anaerotruncus. This systemic elevation is a likely mechanism for the suppression of brain cell genesis and the compromise of synaptic plasticity, dendritic arborization, and cognitive function. The detrimental effect of VA on learning and memory is potentially mediated by the activation of complement C3 signaling. In particular, while peripheral (intraperitoneal) injection of VA impaired cognition in the model, direct central administration (intracerebroventricular) at a low, brain-relevant dose failed to induce cognitive learning and memory impairments. This discrepancy suggests that the adverse effects of high systemic VA in POCD may be primarily mediated by peripheral inflammatory or complement signaling, rather than a direct, toxic effect of VA on CNS neurons at concentrations typically achieved in the brain. Crucially, low-intensity exercise has been shown to stabilize the gut microbiota against surgical insult and mitigate POCD development. These results suggest that reducing blood VA by altering the gut microbiota may represent a primary mechanism for the exercise-induced protection against both the immune/neuroinflammatory responses and the suppression of brain cell genesis. In this context, VA is identified as a potential therapeutic target for brain health in clinical settings involving major surgery [122]. Consistently, a higher level of VA produced by Actinomycetaceae has also been reported in children with autism spectrum disorder (ASD). In particular, a range of VA concentrations among children with ASD was observed, suggesting high inter-individual variability and the possibility of different endophenotypes. Furthermore, intermediate levels of VA in unaffected siblings suggest a contribution from common genetic or environmental factors. This finding suggests that VA in ASD is likely multifactorial, and its physiological functions in regulating the GBA remain under investigation [124].
A key modulatory effect of VA is on the GABAergic system [125]. VA structurally resembles gamma-aminobutyric acid (GABA), the primary inhibitory neurotransmitter in the CNS, and has been shown to bind the GABA_A receptor. This raises the possibility that bacteria involved in VA production and metabolism could be associated with depression, bridging the gap between microbial metabolites and psychiatric symptoms [123]. Anuszkiewicz et al. demonstrated that VA can influence cerebral vascular tone and neuroinflammation through colon-vagus nerve signalling mediated by GPR41/43, unveiling new therapeutic avenues for neurovascular disorders [126]. Foundational pharmacological characterizations of GABA_A receptors by Edvinsson and Krause [127] and Lloyd and Dreksler [128] further affirmed their central role in cerebral vascular regulation and neuronal excitability, mechanisms likely underlying the anxiolytic and neuroprotective effects attributed to VA. Vishwakarma et al. further reported that VA exerts a protective effect against cognitive impairment induced by intracerebroventricular streptozotocin (ICV-STZ) in rats, suggesting a possible GABAergic mechanism underlying its neuroprotective action. In this dementia model, VA improved cognitive performance, likely by enhancing GABAergic transmission and reducing cerebral oxidative stress [109]. Similarly, Nouri and Abbas Abad observed that the aqueous extract of Valeriana officinalis root increases the threshold for picrotoxin (PTZ)-induced clonic seizures in mice, further supporting the involvement of the GABA system as a pharmacological target of VA [129]. These findings suggest that VA acts as a positive modulator of GABA_A receptors, contributing both to the stabilization of neuronal activity and to the potential prevention of neurodegenerative and convulsive disorders. Moreover, Mulyawan et al. reported increased expression of the GABRB3 subunit of the GABA_A receptor following administration of valerian extracts in BALB/c mice, strengthening the evidence for neuroprotective modulation [130].
Taken together, this body of evidence suggests that VA acts not just as a microbial metabolite with local effects but as a bioactive compound with the capacity to systemically modulate neuroprotective effects on host epigenetics. Despite the compelling preclinical findings, its translatability warrants further investigation into strategies that leverage gut-derived interventions to counteract inflammation, cancer, and metabolic disorders [120].

7. From a Pharmacological Drug to a Physiological Mediator: Valproic Acid and Valeric Acid in Epigenetics

Both Valeric Acid (VA) and its pharmacological derivative, Valproic Acid (VPA), have garnered increasing interest due to their shared ability to act as Histone Deacetylase inhibitors (HDACi) [131]. While VA selectively targets Class I HDACs, VPA acts as a well-known, non-selective HDACi, targeting multiple enzyme classes [132,133]. VPA primary targets include Class I HDACs and Class IIa HDACs [117,134,135] with a mechanism that involves the chelation of zinc ions (Zn2+) located within the catalytic site. This disruption leads to an accumulation of acetyl groups on lysine residues within histones, H3K9ac, H3K14ac, and H4K5ac [135]. Beyond directly influencing histone acetylation, VPA can also indirectly alter DNA methylation patterns or interact with chromatin remodeling complexes (SWI/SNF family), significantly broadening its epigenetic impact and strengthening its ability to orchestrate complex changes across the epigenome [136,137]. This fundamental epigenetic mechanism underlies VPA’s neuroprotective and anti-inflammatory effects. VPA activity on the epigenome is particularly relevant in diseases, where epigenetic dysregulations act as both consequences and drivers of pathogenesis (Table 1). Despite preclinical evidence supporting the neuroprotective and anti-inflammatory potential of VA and VPA, translating these findings into clinical practice for neuroinflammatory and neurodegenerative diseases remains complex.
Unlike VPA, which is administered as a pharmaceutical drug acting as a broad-spectrum pharmaceutical derivative, VA acts locally in the gut by modulating the microbiota and then enters systemic circulation to exert its effects, which also include HDAC inhibition at the brain level. This function stems from its intestinal origin. In this sense, VA functions as a more integrated and physiological “epigenetic mediator,” defined here as an endogenously produced metabolite, derived from the host microbiota, which exerts its effects by natural homeostatic mechanisms rather than pharmacological intervention. Indeed, the primary advantage of VA selectively targeting the inhibition of HDAC3 (compared to the non-selective VPA) lies in a better safety profile and greater therapeutic specificity, thereby reducing harmful off-target side effects (Table 2). Moreover, VA’s physiological nature conflicts with the need for a sustained, pharmacologically relevant concentration necessary to maintain an epigenetic effect in the CNS over time. Thus, the potential contribution of VA lies in disease management through nutritional or microbial modulation aimed at optimizing VA levels and maintaining CNS homeostasis within the host physiology and the systemic toxicity of VPA [170,194,195]. Recent studies highlighted the direct epigenetic effects of VA. Jayaraj et al. demonstrated in neurodegeneration models that VA protects dopaminergic neurons by reducing oxidative stress, neuroinflammation, and modulating autophagy pathways—pointing to direct epigenetic mechanisms with potential relevance for Parkinson’s disease (PD) and other neurodegenerative conditions [170]. This evidence supports the therapeutic potential of both VA and VPA in managing diseases where epigenetic dysregulations drive pathogenesis. Despite the compelling preclinical advantages of VA’s selectivity over VPA’s broad action, it is essential to note that the clinical translation of VA’s epigenetic potential is still in its nascent stage.

8. Clinical and Epigenetic Challenges in VA Research

The current stage of VA research is marked by limitations that present unique opportunities for future study. The primary challenge is the absence of dedicated clinical trials on pure VA and its derivatives as epigenetic therapeutics. Most existing data are derived indirectly from studies on its synthetic precursor, VPA, despite significant differences in their pharmacokinetic profiles and HDAC selectivity. Moreover, the absence of validated epigenetic and neuroinflammatory biomarkers in human biofluids remains a major hindrance. However, the major challenge concerns the proper concentration required to engage the epigenetic effect compared to the physiological circulating levels of VA. The circulating level of VA in human plasma is 0.18 µM [118], which is substantially lower than the concentration used in in vitro studies. Furthermore, this discrepancy is further highlighted by the fact that in vitro models do not fully recapitulate the complexities of in vivo pharmacokinetics, compartmentalization, target engagement kinetics, and metabolism in the CNS microenvironment. Therefore, further studies are needed to determine the effective VA concentration necessary to achieve HDAC3 inhibition in vivo at the CNS level. While the literature suggests that VA possesses the capacity to cross the BBB, the critical gap remains the lack of data confirming that VA achieves concentrations sufficient to engage its target in vivo [25,196]. Furthermore, the current literature lacks studies that fully map the specific epigenetic signature and downstream transcriptional changes following VA-induced HDAC3 inhibition in neurons and glia. To bridge the gap between preclinical data and clinical application, researchers should prioritize specific VA-focused studies. This would include utilizing advanced in vitro models, such as those employing induced pluripotent stem cells (iPSCs) derived from patients [197,198,199,200]. These models offer a genetically human context to test the compound and quantify its selective HDAC inhibition on patient-specific cellular mechanisms, providing a crucial intermediate step before large-scale human trials.
Another major practical hurdle is VA’s inherent pharmacological profile. As an endogenously produced SCFA, VA suffers from rapid systemic clearance and substantial first-pass metabolism in the gut and liver [93,99,201]. Achieving therapeutic effectiveness in the CNS would require dedicated pharmacological strategies, such as prodrug development or targeted delivery systems. Furthermore, like other endogenously produced molecules, VA faces regulatory challenges regarding its classification (drug vs. dietary supplement) and requires robust, controlled clinical data to definitively prove its efficacy and safety profile for clinical application.
The critical necessity to overcome these challenges is underlined by the severe limitations of VPA. The drug’s safety profile and significant side effects pose a major barrier to its broader application. These adverse effects at therapeutic dosages include hepatotoxicity (with risks of acute liver failure), teratogenicity (e.g., neural tube defects if taken during pregnancy), common gastrointestinal effects (nausea, vomiting), neurological effects (sedation, tremor, cognitive slowing), hematological effects (thrombocytopenia), and the potential for weight gain and features of metabolic syndrome [170,194,195,202]. These considerable risks substantially limit VPA’s chronic use, especially in vulnerable populations or when clear benefits do not demonstrably outweigh the risks in complex neurodegenerative pathologies.

9. Future Perspectives

In conclusion, although preclinical data suggest valeric acid (VA) could be used as a naturally derived, bioactive compound for neuroprotection, the successful and safe translation of these findings into clinical practice warrants multidisciplinary efforts. This includes the development of innovative pharmacological strategies alongside thorough and methodologically rigorous clinical trials to validate its efficacy and safety profile. Should these efforts yield positive results, VA could emerge as an alternative conceptual approach to valproic acid (VPA) by leveraging selective, host-derived metabolites to address the complex pathophysiology of central nervous system disorders.

Author Contributions

Conceptualization, C.P., M.M. and G.G.; resources: C.P., M.M. and M.D.C.; writing, C.P., M.M. and M.D.C.; writing—review and editing, M.M., C.P., M.D.C. and G.G.; visualization, G.G., A.G., F.S. and F.M.; supervision, G.G., A.G. and F.S.; project administration, G.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
GBAgut–brain axis
CNScentral nervous system
BBBthe blood-brain barrier
SCFAshort-chain fatty acids
VAvaleric acid
HDAChistone deacetylases
HDACiHDAC inhibitors
ROSreactive oxygen species
TNF-αTumor Necrosis Factor-alpha
IL-1β, IL-6Interleukins
COX-2Cyclooxygenase-2
VEGF-AVascular Endothelial Growth Factor-A
MCP-1Monocyte Chemoattractant Protein-1
MMP-9Matrix metalloproteinase-9
NLRP3NOD-like receptor protein 3
ADAlzheimer’s Disease
PDParkinson’s Disease
MSmultiple sclerosis
FTDfrontotemporal dementia
ASDautism spectrum disorder
GABAgamma-aminobutyric acid
CAZymescarbohydrate-active enzymes
MCTsH+-coupled monocarboxylate transporters
SMCTssodium-coupled monocarboxylate transporters
Tregsregulatory T cells
APCsantigen-presenting cells
POCDpostoperative cognitive dysfunction
ICV-STZintracerebroventricular streptozotocin
PTZpicrotoxin

References

  1. Clapp, M.; Aurora, N.; Herrera, L.; Bhatia, M.; Wilen, E.; Wakefield, S. Gut Microbiota’s Effect on Mental Health: The Gut-Brain Axis. Clin. Pract. 2017, 7, 987. [Google Scholar] [CrossRef] [PubMed]
  2. Suganya, K.; Koo, B.S. Gut–brain axis: Role of gut microbiota on neurological disorders and how probiotics/prebiotics beneficially modulate microbial and immune pathways to improve brain functions. Int. J. Mol. Sci. 2020, 21, 7551. [Google Scholar] [CrossRef] [PubMed]
  3. Dong, T.S.; Mayer, E. Advances in Brain–Gut–Microbiome Interactions: A Comprehensive Update on Signaling Mechanisms, Disorders, and Therapeutic Implications. Cell. Mol. Gastroenterol. Hepatol. 2024, 18, 1–13. [Google Scholar] [CrossRef]
  4. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  5. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef]
  6. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Reddy, D.N. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef]
  7. Bonaz, B.; Bazin, T.; Pellissier, S. The vagus nerve at the interface of the microbiota-gut-brain axis. Front. Neurosci. 2018, 12, 49. [Google Scholar] [CrossRef] [PubMed]
  8. Cryan, J.F.; O’riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  9. De Luca, F.; Shoenfeld, Y. The microbiome in autoimmune diseases. Clin. Exp. Immunol. 2019, 195, 74–85. [Google Scholar] [CrossRef]
  10. García-Domínguez, M. Neuroinflammation: Mechanisms, Dual Roles, and Therapeutic Strategies in Neurological Disorders. Curr. Issues Mol. Biol. 2025, 47, 417. [Google Scholar] [CrossRef]
  11. Müller, L.; Di Benedetto, S.; Müller, V. The dual nature of neuroin fl ammation in networked brain. Front. Immunol. 2025, 16, 1659947. [Google Scholar] [CrossRef] [PubMed]
  12. Quigley, E.M.M. Microbiota-Brain-Gut Axis and Neurodegenerative Diseases. Curr. Neurol. Neurosci. Rep. 2017, 17, 94. [Google Scholar] [CrossRef]
  13. Randeni, N.; Bordiga, M.; Xu, B. A Comprehensive Review of the Triangular Relationship among Diet–Gut Microbiota–Inflammation. Int. J. Mol. Sci. 2024, 25, 9366. [Google Scholar] [CrossRef]
  14. Appleton, J. The gut-brain axis: Influence of microbiota on mood and mental health. Integr. Med. 2018, 17, 28–32. [Google Scholar]
  15. Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2015, 7, 17–44. [Google Scholar] [CrossRef] [PubMed]
  16. Boulangé, C.L.; Neves, A.L.; Chilloux, J.; Nicholson, J.K.; Dumas, M.E. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 2016, 8, 42. [Google Scholar] [CrossRef]
  17. Maciel-Fiuza, M.F.; Muller, G.C.; Campos, D.M.S.; do Socorro Silva Costa, P.; Peruzzo, J.; Bonamigo, R.R.; Veit, T.; Vianna, F.S.L. Role of gut microbiota in infectious and inflammatory diseases. Front. Microbiol. 2023, 14, 1098386. [Google Scholar] [CrossRef]
  18. Doifode, T.; Giridharan, V.V.; Generoso, J.S.; Bhatti, G.; Collodel, A.; Schulz, P.E.; Forlenza, O.V.; Barichello, T. The impact of the microbiota-gut-brain axis on Alzheimer’s disease pathophysiology. Pharmacol. Res. 2021, 164, 105314. [Google Scholar] [CrossRef]
  19. Saji, N.; Murotani, K.; Hisada, T.; Tsuduki, T.; Sugimoto, T.; Kimura, A.; Niida, S.; Toba, K.; Sakurai, T. The relationship between the gut microbiome and mild cognitive impairment in patients without dementia: A cross-sectional study conducted in Japan. Sci. Rep. 2019, 9, 19227. [Google Scholar] [CrossRef]
  20. Di Vincenzo, F.; Del Gaudio, A.; Petito, V.; Lopetuso, L.R.; Scaldaferri, F. Gut microbiota, intestinal permeability, and systemic inflammation: A narrative review. Intern. Emerg. Med. 2024, 19, 275–293. [Google Scholar] [CrossRef] [PubMed]
  21. Fasano, A. Leaky gut and autoimmune diseases. Clin. Rev. Allergy Immunol. 2012, 42, 71–78. [Google Scholar] [CrossRef]
  22. Escalante, J.; Artaiz, O.; Diwakarla, S.; McQuade, R.M. Leaky gut in systemic inflammation: Exploring the link between gastrointestinal disorders and age-related diseases. GeroScience 2025, 47, 1–22. [Google Scholar] [CrossRef]
  23. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Guan, N.L.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [PubMed]
  24. Suriano, F.; Nyström, E.E.L.; Sergi, D.; Gustafsson, J.K. Diet, microbiota, and the mucus layer: The guardians of our health. Front. Immunol. 2022, 13, 953196. [Google Scholar] [CrossRef]
  25. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [PubMed]
  26. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef]
  27. Mishra, A.; Bandopadhyay, R.; Singh, P.K.; Mishra, P.S.; Sharma, N.; Khurana, N. Neuroinflammation in neurological disorders: Pharmacotherapeutic targets from bench to bedside. Metab. Brain Dis. 2021, 36, 1591–1626. [Google Scholar] [CrossRef] [PubMed]
  28. Bernier, L.P.; York, E.M.; MacVicar, B.A. Immunometabolism in the Brain: How Metabolism Shapes Microglial Function. Trends Neurosci. 2020, 43, 854–869. [Google Scholar] [CrossRef]
  29. Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of neuroinflammation in neurodegeneration development. Signal Transduct. Target. Ther. 2023, 8, 267. [Google Scholar] [CrossRef]
  30. Saijo, K.; Glass, C.K. Microglial cell origin and phenotypes in health and disease. Nat. Rev. Immunol. 2011, 11, 775–787. [Google Scholar] [CrossRef]
  31. Bachiller, S.; Jiménez-Ferrer, I.; Paulus, A.; Yang, Y.; Swanberg, M.; Deierborg, T.; Boza-Serrano, A. Microglia in neurological diseases: A road map to brain-disease dependent-inflammatory response. Front. Cell. Neurosci. 2018, 12, 488. [Google Scholar] [CrossRef] [PubMed]
  32. Takata, F.; Nakagawa, S.; Matsumoto, J.; Dohgu, S. Blood-Brain Barrier Dysfunction Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction. Front. Cell. Neurosci. 2021, 15, 661838. [Google Scholar] [CrossRef]
  33. Yoon, S.H.; Kim, C.Y.; Lee, E.; Lee, C.; Lee, K.S.; Lee, J.; Park, H.; Choi, B.; Hwang, I.; Kim, J.; et al. Microglial NLRP3-gasdermin D activation impairs blood-brain barrier integrity through interleukin-1β-independent neutrophil chemotaxis upon peripheral inflammation in mice. Nat. Commun. 2025, 16, 699. [Google Scholar] [CrossRef]
  34. Ni, Y.; Teng, T.; Li, R.; Simonyi, A.; Sun, G.Y.; Lee, J.C. TNFα alters occludin and cerebral endothelial permeability: Role of p38MAPK. PLoS ONE 2017, 12, e0170346. [Google Scholar] [CrossRef] [PubMed]
  35. Cheng, C.K.; Yi, M.; Wang, L.; Huang, Y. Role of gasdermin D in inflammatory diseases: From mechanism to therapeutics. Front. Immunol. 2024, 15, 1456244. [Google Scholar] [CrossRef]
  36. Swer, N.M.; Venkidesh, B.S.; Murali, T.S.; Mumbrekar, K.D. Gut microbiota-derived metabolites and their importance in neurological disorders. Mol. Biol. Rep. 2023, 50, 1663–1675. [Google Scholar] [CrossRef] [PubMed]
  37. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed]
  38. Turner-Evans, D.B.; Jensen, K.T.; Ali, S.; Paterson, T.; Sheridan, A.; Ray, R.P.; Wolff, T.; Lauritzen, J.S.; Rubin, G.M.; Bock, D.D.; et al. The Neuroanatomical Ultrastructure and Function of a Biological Ring Attractor. Neuron 2020, 108, 145–163. [Google Scholar] [CrossRef]
  39. Wang, L.; Yu, C.C.; Liu, X.Y.; Deng, X.N.; Tian, Q.; Du, Y.J. Epigenetic Modulation of Microglia Function and Phenotypes in Neurodegenerative Diseases. Neural Plast. 2021, 2021, 9912686. [Google Scholar] [CrossRef]
  40. Ghiglieri, V.; Picconi, B.; Sgobio, C.; Bagetta, V.; Barone, I.; Paillè, V.; Di Filippo, M.; Polli, F.; Gardoni, F.; Altrock, W.; et al. Epilepsy-induced abnormal striatal plasticity in Bassoon mutant mice. Eur. J. Neurosci. 2009, 29, 1979–7993. [Google Scholar] [CrossRef]
  41. Giallongo, S.; Longhitano, L.; Denaro, S.; D’Aprile, S.; Torrisi, F.; La Spina, E.; Giallongo, C.; Mannino, G.; Lo Furno, D.; Zappalà, A.; et al. The Role of Epigenetics in Neuroinflammatory-Driven Diseases. Int. J. Mol. Sci. 2022, 23, 15218. [Google Scholar] [CrossRef] [PubMed]
  42. Kobow, K.; Blümcke, I. The methylation hypothesis: Do epigenetic chromatin modifications play a role in epileptogenesis? Epilepsia 2011, 52, 15–19. [Google Scholar] [CrossRef] [PubMed]
  43. Leus, N.G.J.; Zwinderman, M.R.H.; Dekker, F.J. Histone deacetylase 3 (HDAC 3) as emerging drug target in NF-κB-mediated inflammation. Curr. Opin. Chem. Biol. 2016, 33, 160–168. [Google Scholar] [CrossRef]
  44. Sullivan, K.E.; Reddy, A.B.M.; Dietzmann, K.; Suriano, A.R.; Kocieda, V.P.; Stewart, M.; Bhatia, M. Epigenetic Regulation of Tumor Necrosis Factor Alpha. Mol. Cell. Biol. 2007, 27, 5147–5160. [Google Scholar] [CrossRef] [PubMed]
  45. Pecorelli, A.; Cervellati, C.; Cordone, V.; Hayek, J.; Valacchi, G. Compromised immune/inflammatory responses in Rett syndrome. Free Radic. Biol. Med. 2020, 152, 100–106. [Google Scholar] [CrossRef]
  46. Siniscalco, D.; Cirillo, A.; Bradstreet, J.J.; Antonucci, N. Epigenetic findings in autism: New perspectives for therapy. Int. J. Environ. Res. Public Health 2013, 10, 4261–4273. [Google Scholar] [CrossRef] [PubMed]
  47. Rademakers, R.; Neumann, M.; Mackenzie, I.R. Correction: Advances in understanding the molecular basis of frontotemporal dementia. Nat. Rev. Neurol. 2013, 9, 240. [Google Scholar] [CrossRef]
  48. Boylan, K.; DeJesus-Hernandez, M.; Rush, B.; Desaro, P.; Johnston, A.; Kryston, T.; Rutherford, N.; Baker, M.; Wszolek, Z.; Dickson, D.; et al. Phenotype of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia with ALS (FTD/ALS) associated with the GGGGCC repeat expansion in C9ORF72 (c9FTD/ALS). Neurology 2012, 78, S05.003. [Google Scholar]
  49. Oeckl, P.; Weydt, P.; Steinacker, P.; Anderl-Straub, S.; Nordin, F.; Volk, A.E.; Diehl-Schmid, J.; Andersen, P.M.; Kornhuber, J.; Danek, A.; et al. Different neuroinflammatory profile in amyotrophic lateral sclerosis and frontotemporal dementia is linked to the clinical phase. J. Neurol. Neurosurg. Psychiatry 2019, 90, 4–10. [Google Scholar] [CrossRef]
  50. Woollacott, I.O.C.; Nicholas, J.M.; Heslegrave, A.; Heller, C.; Foiani, M.S.; Dick, K.M.; Russell, L.L.; Paterson, R.W.; Keshavan, A.; Fox, N.C.; et al. Cerebrospinal fluid soluble TREM2 levels in frontotemporal dementia differ by genetic and pathological subgroup. Alzheimers Res. Ther. 2018, 10, 79. [Google Scholar] [CrossRef]
  51. Bright, F.; Werry, E.L.; Dobson-Stone, C.; Piguet, O.; Ittner, L.M.; Halliday, G.M.; Hodges, J.R.; Kiernan, M.C.; Loy, C.T.; Kassiou, M.; et al. Neuroinflammation in frontotemporal dementia. Nat. Rev. Neurol. 2019, 15, 540–555. [Google Scholar] [CrossRef]
  52. Chu, M.; Wen, L.; Jiang, D.; Liu, L.; Nan, H.; Yue, A.; Wang, Y.; Wang, Y.; Qu, M.; Wang, N.; et al. Peripheral inflammation in behavioural variant frontotemporal dementia: Associations with central degeneration and clinical measures. J. Neuroinflamm. 2023, 20, 65. [Google Scholar] [CrossRef]
  53. Veerappan, C.S.; Sleiman, S.; Coppola, G. Epigenetics of Alzheimer’s Disease and Frontotemporal Dementia. Neurotherapeutics 2013, 10, 709–721. [Google Scholar] [CrossRef]
  54. Murthy, M.; Rizzu, P.; Heutink, P.; Mill, J.; Lashley, T.; Bettencourt, C. Epigenetic Age Acceleration in Frontotemporal Lobar Degeneration: A Comprehensive Analysis in the Blood and Brain. Cells 2023, 12, 1922. [Google Scholar] [CrossRef]
  55. Barros, S.P.; Offenbacher, S. Epigenetics: Connecting environment and genotype to phenotype and disease. J. Dent. Res. 2009, 88, 400–408. [Google Scholar] [CrossRef] [PubMed]
  56. Bobetsis, Y.A.; Barros, S.P.; Lin, D.M.; Arce, R.M.; Offenbacher, S. Altered gene expression in murine placentas in an infection-induced intrauterine growth restriction model: A microarray analysis. J. Reprod. Immunol. 2010, 85, 140–148. [Google Scholar] [CrossRef]
  57. Bayarsaihan, D. Epigenetic mechanisms in inflammation. J. Dent. Res. 2011, 90, 9–17. [Google Scholar] [CrossRef]
  58. Doñas, C.; Loyola, A.; Rosemblatt, M. Exploring Epigenetic Drugs in the Regulation of Inflammatory Autoimmune Diseases. In Translational Studies on Inflammation; IntechOpen: London, UK, 2020. [Google Scholar] [CrossRef]
  59. Bonnaud, E.M.; Suberbielle, E.; Malnou, C.E. Histone acetylation in neuronal (dys)function. Biomol. Concepts 2016, 7, 103–116. [Google Scholar] [CrossRef]
  60. Pham, T.M.; Winblad, B.; Granholm, A.C.; Mohammed, A.H. Environmental influences on brain neurotrophins in rats. Pharmacol. Biochem. Behav. 2002, 73, 167–175. [Google Scholar] [CrossRef]
  61. Liyanage, V.R.B.; Rastegar, M. Rett syndrome and MeCP2. NeuroMolecular Med. 2014, 16, 231–264. [Google Scholar] [CrossRef] [PubMed]
  62. Rangasamy, S.; D’Mello, S.R.; Narayanan, V. Epigenetics, Autism Spectrum, and Neurodevelopmental Disorders. Neurotherapeutics 2013, 10, 742–756. [Google Scholar] [CrossRef]
  63. Chiarini, A.; Gui, L.; Viviani, C.; Armato, U.; Dal Prà, I. NLRP3 Inflammasome’s Activation in Acute and Chronic Brain Diseases—An Update on Pathogenetic Mechanisms and Therapeutic Perspectives with Respect to Other Inflammasomes. Biomedicines 2023, 11, 999. [Google Scholar] [CrossRef] [PubMed]
  64. Youm, Y.H.; Nguyen, K.Y.; Grant, R.W.; Goldberg, E.L.; Bodogai, M.; Kim, D.; D’Agostino, D.; Planavsky, N.; Lupfer, C.; Kanneganti, T.D.; et al. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat. Med. 2015, 21, 263–269. [Google Scholar] [CrossRef]
  65. Shimazu, T.; Hirschey, M.D.; Newman, J.; He, W.; Shirakawa, K.; Le Moan, N.; Grueter, C.A.; Lim, H.; Saunders, L.R.; Stevens, R.D.; et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 2013, 339, 211–214. [Google Scholar] [CrossRef]
  66. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef]
  67. Vinolo, M.A.R.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of inflammation by short chain fatty acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef] [PubMed]
  68. Stilling, R.M.; Dinan, T.G.; Cryan, J.F. Microbial genes, brain & behaviour—Epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014, 13, 69–86. [Google Scholar] [CrossRef]
  69. Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T.; Creasy, H.H.; Earl, A.M.; Fitzgerald, M.G.; Fulton, R.S.; et al. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef]
  70. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
  71. Bäckhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef]
  72. Chandrasekaran, P.; Weiskirchen, S.; Weiskirchen, R. Effects of Probiotics on Gut Microbiota: An Overview. Int. J. Mol. Sci. 2024, 25, 6022. [Google Scholar] [CrossRef]
  73. Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef]
  74. Lobionda, S.; Sittipo, P.; Kwon, H.Y.; Lee, Y.K. The role of gut microbiota in intestinal inflammation with respect to diet and extrinsic stressors. Microorganisms 2019, 7, 271. [Google Scholar] [CrossRef] [PubMed]
  75. Candela, M.; Biagi, E.; Turroni, S.; Maccaferri, S.; Figini, P.; Brigidi, P. Dynamic efficiency of the human intestinal microbiota. Crit. Rev. Microbiol. 2015, 41, 165–171. [Google Scholar] [CrossRef]
  76. O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 2006, 7, 688–693. [Google Scholar] [CrossRef] [PubMed]
  77. Di Chiano, M.; Sallustio, F.; Fiocco, D.; Rocchetti, M.T.; Spano, G.; Pontrelli, P.; Moschetta, A.; Gesualdo, L.; Gadaleta, R.M.; Gallone, A. Psychobiotic Properties of Lactiplantibacillus plantarum in Neurodegenerative Diseases. Int. J. Mol. Sci. 2024, 25, 9489. [Google Scholar] [CrossRef]
  78. Petersen, C.; Round, J.L. Defining dysbiosis and its influence on host immunity and disease. Cell. Microbiol. 2014, 16, 1024–1033. [Google Scholar] [CrossRef]
  79. Acevedo-Román, A.; Pagán-Zayas, N.; Velázquez-Rivera, L.I.; Torres-Ventura, A.C.; Godoy-Vitorino, F. Insights into Gut Dysbiosis: Inflammatory Diseases, Obesity, and Restoration Approaches. Int. J. Mol. Sci. 2024, 25, 9715. [Google Scholar] [CrossRef]
  80. Hrncir, T. Gut Microbiota Dysbiosis: Triggers, Consequences, Diagnostic and Therapeutic Options. Microorganisms 2022, 10, 578. [Google Scholar] [CrossRef] [PubMed]
  81. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed]
  82. Fu, J.; Zheng, Y.; Gao, Y.; Xu, W. Dietary Fiber Intake and Gut Microbiota in Human Health. Microorganisms 2022, 10, 2507. [Google Scholar] [CrossRef]
  83. Fassarella, M.; Blaak, E.E.; Penders, J.; Nauta, A.; Smidt, H.; Zoetendal, E.G. Gut microbiome stability and resilience: Elucidating the response to perturbations in order to modulate gut health. Gut 2021, 70, 585–605. [Google Scholar] [CrossRef] [PubMed]
  84. Rocchetti, M.T.; Di Chiano, M.; Elouardi, I.; Fiocco, D. Psychobiotic properties of probiotic lactic acid bacteria and bifidobacteria in paediatric neurological disorders. Glob. Pediatr. 2025, 13, 100266. [Google Scholar] [CrossRef]
  85. Zheng, Y.; Bonfili, L.; Wei, T.; Eleuteri, A.M. Understanding the Gut–Brain Axis and Its Therapeutic Implications for Neurodegenerative Disorders. Nutrients 2023, 15, 4631. [Google Scholar] [CrossRef]
  86. Sonnenburg, E.D.; Sonnenburg, J.L. Starving our microbial self: The deleterious consequences of a diet deficient in microbiota-accessible carbohydrates. Cell Metab. 2014, 20, 779–786. [Google Scholar] [CrossRef] [PubMed]
  87. Van Hul, M.; Cani, P.D.; Petitfils, C.; De Vos, W.M.; Tilg, H.; El-Omar, E.M. What defines a healthy gut microbiome? Gut 2024, 73, 1893–1908. [Google Scholar] [CrossRef] [PubMed]
  88. Vancamelbeke, M.; Vermeire, S. The intestinal barrier: A fundamental role in health and disease. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef]
  89. Wang, K.; Wu, L.Y.; Dou, C.Z.; Guan, X.; Wu, H.G.; Liu, H.R. Research Advance in Intestinal Mucosal Barrier and Pathogenesis of Crohn’s Disease. Gastroenterol. Res. Pract. 2016, 2016, 9686238. [Google Scholar] [CrossRef] [PubMed]
  90. Wells, J.M.; Brummer, R.J.; Derrien, M.; MacDonald, T.T.; Troost, F.; Cani, P.D.; Theodorou, V.; Dekker, J.; Méheust, A.; De Vos, W.M.; et al. Homeostasis of the gut barrier and potential biomarkers. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312, G171–G193. [Google Scholar] [CrossRef]
  91. Knox, E.G.; Aburto, M.R.; Clarke, G.; Cryan, J.F.; O’Driscoll, C.M. The blood-brain barrier in aging and neurodegeneration. Mol. Psychiatry 2022, 27, 2659–2673. [Google Scholar] [CrossRef] [PubMed]
  92. Kurokawa, K.; Itoh, T.; Kuwahara, T.; Oshima, K.; Toh, H.; Toyoda, A.; Takami, H.; Morita, H.; Sharma, V.K.; Srivastava, T.P.; et al. Comparative metagenomics revealed commonly enriched gene sets in human gut microbiomes. DNA Res. 2007, 14, 169–181. [Google Scholar] [CrossRef]
  93. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef]
  94. Knudsen, K.E.B.; Lærke, H.N.; Hedemann, M.S.; Nielsen, T.S.; Ingerslev, A.K.; Nielsen, D.S.G.; Theil, P.K.; Purup, S.; Hald, S.; Schioldan, A.G.; et al. Impact of diet-modulated butyrate production on intestinal barrier function and inflammation. Nutrients 2018, 10, 1499. [Google Scholar] [CrossRef]
  95. Ghanim, H.; Batra, M.; Abuaysheh, S.; Green, K.; Makdissi, A.; Kuhadiya, N.D.; Chaudhuri, A.; Dandona, P. Antiinflammatory and ROS Suppressive Effects of the Addition of Fiber to a High-Fat High-Calorie Meal. J. Clin. Endocrinol. Metab. 2017, 102, 858–869. [Google Scholar] [CrossRef]
  96. Jakobsdottir, G.; Xu, J.; Molin, G.; Ahrné, S.; Nyman, M. High-fat diet reduces the formation of butyrate, but increases succinate, inflammation, liver fat and cholesterol in rats, while dietary fibre counteracts these effects. PLoS ONE 2013, 8, e80476. [Google Scholar] [CrossRef]
  97. Anderson, J.W.; Baird, P.; Davis, R.H.; Ferreri, S.; Knudtson, M.; Koraym, A.; Waters, V.; Williams, C.L. Health benefits of dietary fiber. Nutr. Rev. 2009, 67, 188–205. [Google Scholar] [CrossRef] [PubMed]
  98. Esposito, K.; Giugliano, D. Diet and inflammation: A link to metabolic and cardiovascular diseases. Eur. Heart J. 2006, 27, 15–20. [Google Scholar] [CrossRef] [PubMed]
  99. Cummings, J.H.; Pomare, E.W.; Branch, H.W.J.; Naylor, C.P.E.; MacFarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [PubMed]
  100. Henningsson, Å.M.; Björck, I.M.E.; Nyman, E.M.G.L. Combinations of indigestible carbohydrates affect short-chain fatty acid formation in the hindgut of rats. J. Nutr. 2002, 132, 3098–3104. [Google Scholar] [CrossRef]
  101. Stilling, R.M.; van de Wouw, M.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem. Int. 2016, 99, 110–132. [Google Scholar] [CrossRef]
  102. Richards, L.B.; Li, M.; van Esch, B.C.A.M.; Garssen, J.; Folkerts, G. The effects of short-chain fatty acids on the cardiovascular system. PharmaNutrition 2016, 4, 68–111. [Google Scholar] [CrossRef]
  103. Li, M.; van Esch, B.C.A.M.; Wagenaar, G.T.M.; Garssen, J.; Folkerts, G.; Henricks, P.A.J. Pro- and anti-inflammatory effects of short chain fatty acids on immune and endothelial cells. Eur. J. Pharmacol. 2018, 831, 52–59. [Google Scholar] [CrossRef]
  104. Brown, A.J.; Goldsworthy, S.M.; Barnes, A.A.; Eilert, M.M.; Tcheang, L.; Daniels, D.; Muir, A.I.; Wigglesworth, M.J.; Kinghorn, I.; Fraser, N.J.; et al. The orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J. Biol. Chem. 2003, 278, 11312–11319. [Google Scholar] [CrossRef] [PubMed]
  105. Akimova, T.; Beier, U.H.; Liu, Y.; Wang, L.; Hancock, W.W. Histone/protein deacetylases and T-cell immune responses. Blood 2012, 119, 2443–2451. [Google Scholar] [CrossRef] [PubMed]
  106. Goldberg, I.; Rokem, J.S. Organic and Fatty Acid Production, Microbial. In Encyclopedia of Microbiology, 3rd ed.; Academic Press: New York, NY, USA, 2009. [Google Scholar] [CrossRef]
  107. Khan, A.; Akram, M.; Thiruvengadam, M.; Daniyal, M.; Zakki, S.A.; Munir, N.; Zainab, R.; Heydari, M.; Mosavat, S.H.; Rebezov, M.; et al. Anti-anxiety Properties of Selected Medicinal Plants. Curr. Pharm. Biotechnol. 2021, 23, 1041–1060. [Google Scholar] [CrossRef]
  108. Shinjyo, N.; Waddell, G.; Green, J. Valerian Root in Treating Sleep Problems and Associated Disorders—A Systematic Review and Meta-Analysis. J. Evid.-Based Integr. Med. 2020, 25, 2515690X20967323. [Google Scholar] [CrossRef]
  109. Vishwakarma, S.; Goyal, R.; Gupta, V.; Dhar, K.L. GABAergic effect of valeric acid from Valeriana wallichii in amelioration of ICV STZ induced dementia in rats. Rev. Bras. Farmacogn. 2016, 26, 484–489. [Google Scholar] [CrossRef]
  110. Kim, H.; Jeon, B.S.; Sang, B.I. An Efficient New Process for the Selective Production of Odd-Chain Carboxylic Acids by Simple Carbon Elongation Using Megasphaera hexanoica. Sci. Rep. 2019, 9, 11999. [Google Scholar] [CrossRef]
  111. Dinan, T.G.; Cryan, J.F. The Microbiome-Gut-Brain Axis in Health and Disease. Gastroenterol. Clin. N. Am. 2017, 46, 77–89. [Google Scholar] [CrossRef]
  112. Holscher, H.D. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes 2017, 8, 172–184. [Google Scholar] [CrossRef]
  113. Arboleya, S.; González, S.; Salazar, N. Diet and Microbiome in Health and Aging. Nutrients 2022, 14, 3250. [Google Scholar] [CrossRef]
  114. Moles, L.; Otaegui, D. The impact of diet on microbiota evolution and human health. Is diet an adequate tool for microbiota modulation? Nutrients 2020, 12, 1654. [Google Scholar] [CrossRef] [PubMed]
  115. Seto, E.; Yoshida, M. Erasers of histone acetylation: The histone deacetylase enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef]
  116. Yuille, S.; Reichardt, N.; Panda, S.; Dunbar, H.; Mulder, I.E. Human gut bacteria as potent class I histone deacetylase inhibitors in vitro through production of butyric acid and valeric acid. PLoS ONE 2018, 13, e0201073. [Google Scholar] [CrossRef] [PubMed]
  117. Milazzo, G.; Mercatelli, D.; Di Muzio, G.; Triboli, L.; De Rosa, P.; Perini, G.; Giorgi, F.M. Histone deacetylases (HDACs): Evolution, specificity, role in transcriptional complexes, and pharmacological actionability. Genes 2020, 11, 556. [Google Scholar] [CrossRef]
  118. Han, R.; Yang, H.; Li, Y.; Ling, C.; Lu, L. Valeric acid acts as a novel HDAC3 inhibitor against prostate cancer. Med. Oncol. 2022, 39, 213. [Google Scholar] [CrossRef] [PubMed]
  119. Marks, P.A.; Rifkind, R.A.; Richon, V.M.; Breslow, R.; Miller, T.; Kelly, W.K. Histone deacetylases and cancer: Causes and therapies. Nat. Rev. Cancer 2001, 1, 194–202. [Google Scholar] [CrossRef]
  120. Kumari, B.; Kumari, U.; Singh, D.K.; Husain, G.M.; Patel, D.K.; Shakya, A.; Singh, R.B.; Modi, G.P.; Singh, G.K. Molecular Targets of Valeric Acid: A Bioactive Natural Product for Endocrine, Metabolic, and Immunological Disorders. Endocr. Metab. Immune Disord. Drug Targets 2024, 24, 1506–1517. [Google Scholar] [CrossRef]
  121. Liu, S.; Li, E.; Sun, Z.; Fu, D.; Duan, G.; Jiang, M.; Yu, Y.; Mei, L.; Yang, P.; Tang, Y.; et al. Altered gut microbiota and short chain fatty acids in Chinese children with autism spectrum disorder. Sci. Rep. 2019, 9, 287. [Google Scholar] [CrossRef]
  122. Lai, Z.; Shan, W.; Li, J.; Min, J.; Zeng, X.; Zuo, Z. Appropriate exercise level attenuates gut dysbiosis and valeric acid increase to improve neuroplasticity and cognitive function after surgery in mice. Mol. Psychiatry 2021, 26, 7167–7187. [Google Scholar] [CrossRef]
  123. Naseribafrouei, A.; Hestad, K.; Avershina, E.; Sekelja, M.; Linløkken, A.; Wilson, R.; Rudi, K. Correlation between the human fecal microbiota and depression. Neurogastroenterol. Motil. 2014, 26, 1155–1162. [Google Scholar] [CrossRef]
  124. Wang, L.; Christophersen, C.T.; Sorich, M.J.; Gerber, J.P.; Angley, M.T.; Conlon, M.A. Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig. Dis. Sci. 2012, 57, 2096–2102. [Google Scholar] [CrossRef]
  125. Benke, D.; Barberis, A.; Kopp, S.; Altmann, K.H.; Schubiger, M.; Vogt, K.E.; Rudolph, U.; Möhler, H. GABAA receptors as in vivo substrate for the anxiolytic action of valerenic acid, a major constituent of valerian root extracts. Neuropharmacology 2009, 56, 174–181. [Google Scholar] [CrossRef]
  126. Onyszkiewicz, M.; Gawrys-Kopczynska, M.; Konopelski, P.; Aleksandrowicz, M.; Sawicka, A.; Koźniewska, E.; Samborowska, E.; Ufnal, M. Butyric acid, a gut bacteria metabolite, lowers arterial blood pressure via colon-vagus nerve signaling and GPR41/43 receptors. Pflugers Arch. Eur. J. Physiol. 2019, 471, 1441–1453. [Google Scholar] [CrossRef]
  127. Edvinsson, L.; Krause, D.N. Pharmacological characterization of GABA receptors mediating vasodilation of cerebral arteries in vitro. Brain Res. 1979, 173, 89–97. [Google Scholar] [CrossRef]
  128. Lloyd, K.G.; Dreksler, S. An analysis of [3H]gamma-aminobutyric acid (GABA) binding in the human brain. Brain Res. 1979, 163, 77–87. [Google Scholar] [CrossRef]
  129. Nouri, M.H.K.; Abad, A.N.A. Gabaergic system role in aqueous extract of Valeriana officinalis L. root on PTZ-induced clonic seizure threshold in mice. Afr. J. Pharm. Pharmacol. 2011, 5, 1212–1217. [Google Scholar] [CrossRef]
  130. Mulyawan, E.; Ahmad, M.R.; Islam, A.A.; Massi, M.N.; Hatta, M.; Arif, S.K. Analysis of GABRB3 gene mRNA expression and motor coordination after administration of valerian extracts (Valeriana officinalis) in BALB/c mice. F1000Research 2020, 9, 670. [Google Scholar] [CrossRef]
  131. Göttlicher, M.; Minucci, S.; Zhu, P.; Krämer, O.H.; Schimpf, A.; Giavara, S.; Sleeman, J.P.; Lo Coco, F.; Nervi, C.; Pelicci, P.G.; et al. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J. 2001, 20, 6969–6978. [Google Scholar] [CrossRef] [PubMed]
  132. Al-Khafaji, A.S.K.; Wang, L.M.; Alabdei, H.H.; Liloglou, T. Effect of valproic acid on histone deacetylase expression in oral cancer (Review). Oncol. Lett. 2024, 27, 197. [Google Scholar] [CrossRef] [PubMed]
  133. Romoli, M.; Mazzocchetti, P.; D’Alonzo, R.; Siliquini, S.; Rinaldi, V.E.; Verrotti, A.; Calabresi, P.; Costa, C. Valproic Acid and Epilepsy: From Molecular Mechanisms to Clinical Evidences. Curr. Neuropharmacol. 2018, 17, 926–946. [Google Scholar] [CrossRef] [PubMed]
  134. Curcio, A.; Rocca, R.; Alcaro, S.; Artese, A. The Histone Deacetylase Family: Structural Features and Application of Combined Computational Methods. Pharmaceuticals 2024, 17, 620, Correction in Pharmaceuticals 2024, 17, 1520. https://doi.org/10.3390/ph17111520. [Google Scholar] [CrossRef]
  135. Gurvich, N.; Tsygankova, O.M.; Meinkoth, J.L.; Klein, P.S. Histone Deacetylase Is a Target of Valproic Acid-Mediated Cellular Differentiation. Cancer Res. 2004, 64, 1079–7086. [Google Scholar] [CrossRef]
  136. Milutinovic, S.; Detich, N.; Szyf, M. Valproate induces widespread epigenetic reprogramming which involves demethylation of specific genes. Carcinogenesis 2007, 28, 560–571. [Google Scholar] [CrossRef]
  137. Mello, M.L.S. Sodium Valproate-Induced Chromatin Remodeling. Front. Cell Dev. Biol. 2021, 9, 645518. [Google Scholar] [CrossRef]
  138. Cuddapah, V.A.; Pillai, R.B.; Shekar, K.V.; Lane, J.B.; Motil, K.J.; Skinner, S.A.; Tarquinio, D.C.; Glaze, D.G.; McGwin, G.; Kaufmann, W.E.; et al. Methyl-CpG-binding protein 2 (MECP2) mutation type is associated with disease severity in rett syndrome. J. Med. Genet. 2014, 51, 152–158. [Google Scholar] [CrossRef] [PubMed]
  139. Huppke, P.; Köhler, K.; Brockmann, K.; Stettner, G.M.; Gärtner, J. Treatment of epilepsy in Rett syndrome. Eur. J. Paediatr. Neurol. 2007, 11, 10–16. [Google Scholar] [CrossRef] [PubMed]
  140. Ip, J.P.K.; Mellios, N.; Sur, M. Rett syndrome: Insights into genetic, molecular and circuit mechanisms. Nat. Rev. Neurosci. 2018, 19, 368–382. [Google Scholar] [CrossRef]
  141. Guo, W.; Tsujimura, K.; Otsuka, I.M.; Irie, K.; Igarashi, K.; Nakashima, K.; Zhao, X. VPA alleviates neurological deficits and restores gene expression in a mouse model of rett syndrome. PLoS ONE 2014, 9, e100215. [Google Scholar] [CrossRef]
  142. Landrieu, P.; Baets, J.; De Jonghe, P. Hereditary motor-sensory, motor, and sensory neuropathies in childhood. In Handbook of Clinical Neurology; North-Holland Publishing Company: New York, NY, USA, 2013; Volume 113. [Google Scholar] [CrossRef]
  143. Aicardi, J.; Goutières, F. A Progressive familial encephalopathy in infancy with calcifications of the basal ganglia and chronic cerebrospinal fluid lymphocytosis. Ann. Neurol. 1984, 15, 49–54. [Google Scholar] [CrossRef]
  144. Lebon, P.; Badoual, J.; Ponsot, G.; Goutières, F.; Hémeury-Cukier, F.; Aicardi, J. Intrathecal synthesis of interferon-alpha in infants with progressive familial encephalopathy. J. Neurol. Sci. 1988, 84, 201–208. [Google Scholar] [CrossRef]
  145. Crow, Y.J.; Chase, D.S.; Lowenstein Schmidt, J.; Szynkiewicz, M.; Forte, G.M.A.; Gornall, H.L.; Oojageer, A.; Anderson, B.; Pizzino, A.; Helman, G.; et al. Characterization of human disease phenotypes associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR, and IFIH1. Am. J. Med. Genet. Part A 2015, 167, 296–312. [Google Scholar] [CrossRef] [PubMed]
  146. Livingston, J.H.; Crow, Y.J. Neurologic phenotypes associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR1, and IFIH1: Aicardi-Goutières syndrome and beyond. Neuropediatrics 2016, 47, 355–360. [Google Scholar] [CrossRef]
  147. Uggenti, C.; Lepelley, A.; Depp, M.; Badrock, A.P.; Rodero, M.P.; El-Daher, M.T.; Rice, G.I.; Dhir, S.; Wheeler, A.P.; Dhir, A.; et al. cGAS-mediated induction of type I interferon due to inborn errors of histone pre-mRNA processing. Nat. Genet. 2020, 52, 1364–1372. [Google Scholar] [CrossRef]
  148. Sun, L.; Wu, J.; Du, F.; Chen, X.; Chen, Z.J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013, 339, 786–791. [Google Scholar] [CrossRef]
  149. von Stülpnagel, C.; van Baalen, A.; Borggraefe, I.; Eschermann, K.; Hartlieb, T.; Kiwull, L.; Pringsheim, M.; Wolff, M.; Kudernatsch, M.; Wiegand, G.; et al. Network for Therapy in Rare Epilepsies (NETRE): Lessons from the Past 15 Years. Front. Neurol. 2021, 11, 322510. [Google Scholar] [CrossRef] [PubMed]
  150. Gavazzi, F.; Cross, Z.M.; Woidill, S.; McMann, J.M.; Rand, E.B.; Takanohashi, A.; Ulrick, N.; Shults, J.; Vanderver, A.L.; Adang, L. Hepatic Involvement in Aicardi-Goutières Syndrome. Neuropediatrics 2021, 52, 441–447. [Google Scholar] [CrossRef]
  151. Lim, H.K.; Yoon, J.H.; Song, M. Autism Spectrum Disorder Genes: Disease-Related Networks and Compensatory Strategies. Front. Mol. Neurosci. 2022, 15, 922840. [Google Scholar] [CrossRef] [PubMed]
  152. Aliyev, N. A Double-Blind Placebo-Controlled Trial of Acediprol (Valproate Sodium) for Global Severity in Child Autism Spectrum Disorders. Online J. Neurol. Brain Disord. 2018, 2, 1–5. [Google Scholar] [CrossRef]
  153. Wiśniowiecka-Kowalnik, B.; Nowakowska, B.A. Genetics and epigenetics of autism spectrum disorder—Current evidence in the field. J. Appl. Genet. 2019, 60, 37–47. [Google Scholar] [CrossRef]
  154. Genovese, A.; Butler, M.G. The Autism Spectrum: Behavioral, Psychiatric and Genetic Associations. Genes 2023, 14, 677. [Google Scholar] [CrossRef]
  155. Rylaarsdam, L.; Guemez-Gamboa, A. Genetic Causes and Modifiers of Autism Spectrum Disorder. Front. Cell. Neurosci. 2019, 13, 385. [Google Scholar] [CrossRef] [PubMed]
  156. Fakhoury, M. Autistic spectrum disorders: A review of clinical features, theories and diagnosis. Int. J. Dev. Neurosci. 2015, 43, 70–77. [Google Scholar] [CrossRef]
  157. Li, B.; Xiong, Y.; Li, Y. The Impact of Valproic Acid on Microbiota in a Mouse Model of Autism Spectrum Disorder. Psychiatry Clin. Psychopharmacol. 2025, 35, 6–13. [Google Scholar] [CrossRef] [PubMed]
  158. Csoka, A.B.; El Kouhen, N.; Bennani, S.; Getachew, B.; Aschner, M.; Tizabi, Y. Roles of Epigenetics and Glial Cells in Drug-Induced Autism Spectrum Disorder. Biomolecules 2024, 14, 437. [Google Scholar] [CrossRef]
  159. Fukuchi, M.; Nii, T.; Ishimaru, N.; Minamino, A.; Hara, D.; Takasaki, I.; Tabuchi, A.; Tsuda, M. Valproic acid induces up- or down-regulation of gene expression responsible for the neuronal excitation and inhibition in rat cortical neurons through its epigenetic actions. Neurosci. Res. 2009, 65, 35–43. [Google Scholar] [CrossRef] [PubMed]
  160. Kawanai, T.; Ago, Y.; Watanabe, R.; Inoue, A.; Taruta, A.; Onaka, Y.; Hasebe, S.; Hashimoto, H.; Matsuda, T.; Takuma, K. Prenatal Exposure to Histone Deacetylase Inhibitors Affects Gene Expression of Autism-Related Molecules and Delays Neuronal Maturation. Neurochem. Res. 2016, 41, 2574–2584. [Google Scholar] [CrossRef]
  161. Gazzina, S.; Manes, M.A.; Padovani, A.; Borroni, B. Clinical and biological phenotypes of frontotemporal dementia: Perspectives for disease modifying therapies. Eur. J. Pharmacol. 2017, 817, 76–85. [Google Scholar] [CrossRef]
  162. Bottero, V.; Alrafati, F.; Santiago, J.A.; Potashkin, J.A. Transcriptomic and Network Meta-Analysis of Frontotemporal Dementias. Front. Mol. Neurosci. 2021, 14, 747798. [Google Scholar] [CrossRef]
  163. Blauwendraat, C.; Wilke, C.; Simón-Sánchez, J.; Jansen, I.E.; Reifschneider, A.; Capell, A.; Haass, C.; Castillo-Lizardo, M.; Biskup, S.; Maetzler, W.; et al. The wide genetic landscape of clinical frontotemporal dementia: Systematic combined sequencing of 121 consecutive subjects. Genet. Med. 2018, 20, 240–249. [Google Scholar] [CrossRef]
  164. Ribeiro, A.C.R.; Jahr, F.M.; Hawkins, E.; Kronfol, M.M.; Younis, R.M.; McClay, J.L.; Deshpande, L.S. Epigenetic histone acetylation and Bdnf dysregulation in the hippocampus of rats exposed to repeated, low-dose diisopropylfluorophosphate. Life Sci. 2021, 281, 119765. [Google Scholar] [CrossRef]
  165. Khan, M.S.; Nasiripour, S.; Bopassa, J.C. Parkinson Disease Signaling Pathways, Molecular Mechanisms, and Potential Therapeutic Strategies: A Comprehensive Review. Int. J. Mol. Sci. 2025, 26, 6416. [Google Scholar] [CrossRef]
  166. Larsson, P.; Ulfhammer, E.; Magnusson, M.; Bergh, N.; Lunke, S.; El-Osta, A.; Medcalf, R.L.; Svensson, P.A.; Karlsson, L.; Jern, S. Role of histone acetylation in the stimulatory effect of valproic acid on vascular endothelial tissue-type plasminogen activator expression. PLoS ONE 2012, 7, e31573. [Google Scholar] [CrossRef]
  167. Zhou, Y.; Zhen, Y.; Wang, G.; Liu, B. Deconvoluting the Complexity of Reactive Oxygen Species (ROS) in Neurodegenerative Diseases. Front. Neuroanat. 2022, 16, 910427. [Google Scholar] [CrossRef]
  168. Yang, H.S.; Onos, K.D.; Choi, K.; Keezer, K.J.; Skelly, D.A.; Carter, G.W.; Howell, G.R. Natural genetic variation determines microglia heterogeneity in wild-derived mouse models of Alzheimer’s disease. Cell Rep. 2021, 34, 108739. [Google Scholar] [CrossRef]
  169. Frontiers Production Office. Erratum: Autophagy and neurodegeneration: Unraveling the role of C9ORF72 in the regulation of autophagy and its relationship to ALS-FTD pathology. Front. Cell. Neurosci. 2023, 17, 1225439. [Google Scholar] [CrossRef]
  170. Jayaraj, R.L.; Beiram, R.; Azimullah, S.; Nagoor Meeran, M.F.; Ojha, S.K.; Adem, A.; Jalal, F.Y. Valeric acid protects dopaminergic neurons by suppressing oxidative stress, neuroinflammation and modulating autophagy pathways. Int. J. Mol. Sci. 2020, 21, 7670. [Google Scholar] [CrossRef]
  171. Nijs, M.; Van Damme, P. The genetics of amyotrophic lateral sclerosis. Curr. Opin. Neurol. 2024, 37, 560–569. [Google Scholar] [CrossRef] [PubMed]
  172. Bennett, S.A.; Tanaz, R.; Cobos, S.N.; Torrente, M.P. Epigenetics in amyotrophic lateral sclerosis: A role for histone post-translational modifications in neurodegenerative disease. Transl. Res. 2019, 204, 19–30. [Google Scholar] [CrossRef] [PubMed]
  173. Janssen, C.; Schmalbach, S.; Boeselt, S.; Sarlette, A.; Dengler, R.; Petri, S. Differential histone deacetylase mRNA expression patterns in amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2010, 69, 573–581. [Google Scholar] [CrossRef]
  174. Boutillier, A.L.; Tzeplaeff, L.; Dupuis, L. The dark side of HDAC inhibition in ALS. EBioMedicine 2019, 41, 38–39. [Google Scholar] [CrossRef]
  175. Pigna, E.; Simonazzi, E.; Sanna, K.; Bernadzki, K.M.; Proszynski, T.; Heil, C.; Palacios, D.; Adamo, S.; Moresi, V. Histone deacetylase 4 protects from denervation and skeletal muscle atrophy in a murine model of amyotrophic lateral sclerosis. EBioMedicine 2019, 40, 717–732. [Google Scholar] [CrossRef] [PubMed]
  176. Renzini, A.; Pigna, E.; Rocchi, M.; Cedola, A.; Gigli, G.; Moresi, V.; Coletti, D. Sex and HDAC4 Differently Affect the Pathophysiology of Amyotrophic Lateral Sclerosis in SOD1-G93A Mice. Int. J. Mol. Sci. 2023, 24, 98. [Google Scholar] [CrossRef]
  177. Keinath, M.C.; Prior, D.E.; Prior, T.W. Spinal muscular atrophy: Mutations, testing, and clinical relevance. Appl. Clin. Genet. 2021, 14, 11–25. [Google Scholar] [CrossRef]
  178. Le, T.T.; Pham, L.T.; Butchbach, M.E.R.; Zhang, H.L.; Monani, U.R.; Coovert, D.D.; Gavrilina, T.O.; Xing, L.; Bassell, G.J.; Burghes, A.H.M. SMNΔ7, the major product of the centromeric survival motor neuron (SMN2) gene, extends survival in mice with spinal muscular atrophy and associates with full-length SMN. Hum. Mol. Genet. 2005, 14, 845–857. [Google Scholar] [CrossRef] [PubMed]
  179. Butchbach, M.E.R. Copy number variations in the survival motor neuron genes: Implications for spinal muscular atrophy and other neurodegenerative diseases. Front. Mol. Biosci. 2016, 3, 7. [Google Scholar] [CrossRef]
  180. Monani, U.R.; Sendtner, M.; Coovert, D.D.; Parsons, D.W.; Andreassi, C.; Le, T.T.; Jablonka, S.; Schrank, B.; Rossol, W.; Prior, T.W.; et al. The human centromeric survival motor neuron gene (SMN2) rescues embryonic lethality in Smn(-/-) mice and results in a mouse with spinal muscular atrophy. Hum. Mol. Genet. 2000, 9, 333–339. [Google Scholar] [CrossRef]
  181. Kernochan, L.E.; Russo, M.L.; Woodling, N.S.; Huynh, T.N.; Avila, A.M.; Fischbeck, K.H.; Sumner, C.J. The role of histone acetylation in SMN gene expression. Hum. Mol. Genet. 2005, 14, 1171–1182. [Google Scholar] [CrossRef] [PubMed]
  182. Brichta, L.; Hofmann, Y.; Hahnen, E.; Siebzehnrubi, F.A.; Raschke, H.; Blumcke, I.; Eyupoglu, I.Y.; Wirth, B. Valproic acid increases the SMN2 protein level: A well-known drug as a potential therapy for spinal muscular atrophy. Hum. Mol. Genet. 2003, 12, 2481–2489. [Google Scholar] [CrossRef]
  183. Marasco, L.E.; Dujardin, G.; Sousa-Luís, R.; Liu, Y.H.; Stigliano, J.N.; Nomakuchi, T.; Proudfoot, N.J.; Krainer, A.R.; Kornblihtt, A.R. Counteracting chromatin effects of a splicing-correcting antisense oligonucleotide improves its therapeutic efficacy in spinal muscular atrophy. Cell 2022, 185, 2057–2070. [Google Scholar] [CrossRef]
  184. Swoboda, K.J.; Scott, C.B.; Reyna, S.P.; Prior, T.W.; LaSalle, B.; Sorenson, S.L.; Wood, J.; Acsadi, G.; Crawford, T.O.; Kissel, J.T.; et al. Phase II open label study of valproic acid in spinal muscular atrophy. PLoS ONE 2009, 4, e5268. [Google Scholar] [CrossRef]
  185. Saito, T.; Nurputra, D.K.; Harahap, N.I.F.; Harahap, I.S.K.; Yamamoto, H.; Muneshige, E.; Nishizono, H.; Matsumura, T.; Fujimura, H.; Sakoda, S.; et al. A Study of valproic acid for patients with spinal muscular atrophy. Neurol. Clin. Neurosci. 2015, 3, 49–57. [Google Scholar] [CrossRef]
  186. Merchut, M.P. Multiple Sclerosis; Loyola Univeristy of Chicago: Chicago, IL, USA, 1999; Volume 359, pp. 169–180. [Google Scholar]
  187. Sawcer, S.; Hellenthal, G.; Pirinen, M.; Spencer, C.C.A.; Patsopoulos, N.A.; Moutsianas, L.; Dilthey, A.; Su, Z.; Freeman, C.; Hunt, S.E.; et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 2011, 476, 214–219. [Google Scholar] [CrossRef]
  188. Guan, Y.; Jakimovski, D.; Ramanathan, M.; Weinstock-Guttman, B.; Zivadinov, R. The role of Epstein-Barr virus in multiple sclerosis: From molecular pathophysiology to in vivo imaging. Neural Regen. Res. 2019, 14, 373–386. [Google Scholar] [CrossRef]
  189. Munger, K.L.; Zhang, S.M.; O’Reilly, E.; Hernán, M.A.; Olek, M.J.; Willett, W.C.; Ascherio, A. Vitamin D intake and incidence of multiple sclerosis. Neurology 2004, 62, 60–65. [Google Scholar] [CrossRef] [PubMed]
  190. Zhang, Z.; Zhang, Z.Y.; Wu, Y.; Schluesener, H.J. Valproic acid ameliorates inflammation in experimental autoimmune encephalomyelitis rats. Neuroscience 2012, 221, 140–150. [Google Scholar] [CrossRef]
  191. Chen, P.S.; Wang, C.C.; Bortner, C.D.; Peng, G.S.; Wu, X.; Pang, H.; Lu, R.B.; Gean, P.W.; Chuang, D.M.; Hong, J.S. Valproic acid and other histone deacetylase inhibitors induce microglial apoptosis and attenuate lipopolysaccharide-induced dopaminergic neurotoxicity. Neuroscience 2007, 149, 203–212. [Google Scholar] [CrossRef] [PubMed]
  192. Lv, J.; Du, C.; Wei, W.; Wu, Z.; Zhao, G.; Li, Z.; Xie, X. The antiepileptic drug valproic acid restores T cell homeostasis and ameliorates pathogenesis of experimental autoimmune encephalomyelitis. J. Biol. Chem. 2012, 287, 28656–28665. [Google Scholar] [CrossRef]
  193. Nielsen, N.M.; Svanström, H.; Stenager, E.; Magyari, M.; Koch-Henriksen, N.; Pasternak, B.; Hviid, A. The use of valproic acid and multiple sclerosis. Pharmacoepidemiol. Drug Saf. 2015, 24, 262–268. [Google Scholar] [CrossRef] [PubMed]
  194. Bryant, A.E.; Dreifuss, F.E. Valproic acid hepatic fatalities. III. U.S. experience since 1986. Neurology 1996, 46, 465–469. [Google Scholar] [CrossRef]
  195. Lloyd, K.A. A scientific review: Mechanisms of valproate-mediated teratogenesis. Biosci. Horiz. 2013, 6, hzt003. [Google Scholar] [CrossRef]
  196. Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef] [PubMed]
  197. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed]
  198. Manganelli, M.; Mazzoldi, E.L.; Ferraro, R.M.; Pinelli, M.; Parigi, M.; Aghel, S.A.M.; Bugatti, M.; Collo, G.; Stocco, G.; Vermi, W.; et al. Progesterone receptor is constitutively expressed in induced Pluripotent Stem Cells (iPSCs). Stem Cell Rev. Rep. 2024, 20, 2303–2317. [Google Scholar] [CrossRef]
  199. Lancaster, M.A.; Renner, M.; Martin, C.A.; Wenzel, D.; Bicknell, L.S.; Hurles, M.E.; Homfray, T.; Penninger, J.M.; Jackson, A.P.; Knoblich, J.A. Cerebral organoids model human brain development and microcephaly. Nature 2013, 501, 373–379. [Google Scholar] [CrossRef] [PubMed]
  200. Ferraro, R.M.; Ginestra, P.S.; Seiti, M.; Bugatti, M.; Benini, G.; Ottelli, L.; Vermi, W.; Poliani, P.L.; Ceretti, E.; Giliani, S. Three-Dimensional-Bioprinted Embedded-Based Cerebral Organoids: An Alternative Approach for Mini-Brain In Vitro Modeling Beyond Conventional Generation Methods. Gels 2025, 11, 284. [Google Scholar] [CrossRef] [PubMed]
  201. Xia, X.; Zhou, Y.; Gao, H. Prodrug strategy for enhanced therapy of central nervous system disease. Chem. Commun. 2021, 57, 8842–8855. [Google Scholar] [CrossRef] [PubMed]
  202. Powell-Jackson, P.R.; Tredger, J.M.; Williams, R. Hepatotoxicity to sodium valproate: A review. Gut 1984, 25, 673–681. [Google Scholar] [CrossRef]
Figure 1. The gut–brain axis. The figure illustrates how a healthy gut microbiota (eubiosis) produces beneficial metabolites like short-chain fatty acids (SCFAs), among which valeric acid (VA) to support gut and brain health, while an imbalanced microbiota (dysbiosis) leads to inflammation and neuroinflammation. This figure was created in BioRender.com. (C.P.). (2025) https://www.biorender.com/.
Figure 1. The gut–brain axis. The figure illustrates how a healthy gut microbiota (eubiosis) produces beneficial metabolites like short-chain fatty acids (SCFAs), among which valeric acid (VA) to support gut and brain health, while an imbalanced microbiota (dysbiosis) leads to inflammation and neuroinflammation. This figure was created in BioRender.com. (C.P.). (2025) https://www.biorender.com/.
Cells 14 01823 g001
Figure 2. Schematic representation of Valeric Acid (VA) biosynthesis pathway. Ethanol (C2) and propionate (C3) are condensed to form pentanoic acid (VA, C5) in the gut microbiota. Enzymatic steps: 1: Acetyl-CoA Synthase/Carbon Monoxide Dehydrogenase (ACS/CODH); 2: Acyl-CoA synthetase; 3: Acetyl-CoA C-acetyltransferase; 4: Intermediate oxidoreductases (3-hydroxyacyl-CoA dehydrogenase and enoyl-CoA hydratase); 5: Enoyl-CoA reductase. Et(OH): Ethanol; CoA-SH: Coenzyme A; Ac-CoA: acetyl-CoA; ATP: Adenosine triphosphate.
Figure 2. Schematic representation of Valeric Acid (VA) biosynthesis pathway. Ethanol (C2) and propionate (C3) are condensed to form pentanoic acid (VA, C5) in the gut microbiota. Enzymatic steps: 1: Acetyl-CoA Synthase/Carbon Monoxide Dehydrogenase (ACS/CODH); 2: Acyl-CoA synthetase; 3: Acetyl-CoA C-acetyltransferase; 4: Intermediate oxidoreductases (3-hydroxyacyl-CoA dehydrogenase and enoyl-CoA hydratase); 5: Enoyl-CoA reductase. Et(OH): Ethanol; CoA-SH: Coenzyme A; Ac-CoA: acetyl-CoA; ATP: Adenosine triphosphate.
Cells 14 01823 g002
Figure 3. Valeric acid as an epigenetic modulator. The figure illustrates how valeric acid (VA) acts as a Histone Deacetylase Inhibitor (HDACi), promoting the acetylation of histone proteins. This preservation of histone acetylation leads to chromatin decondensation and consequent modulation of inflammation. This figure was created in BioRender.com. (C.P.). (2025) https://www.biorender.com/.
Figure 3. Valeric acid as an epigenetic modulator. The figure illustrates how valeric acid (VA) acts as a Histone Deacetylase Inhibitor (HDACi), promoting the acetylation of histone proteins. This preservation of histone acetylation leads to chromatin decondensation and consequent modulation of inflammation. This figure was created in BioRender.com. (C.P.). (2025) https://www.biorender.com/.
Cells 14 01823 g003
Table 1. Valproic Acid (VPA) and Valeric Acid (VA) in neurological disorders. Comparative overview of preclinical and clinical evidence for VPA and VA in the epigenetics of neurological disorders.
Table 1. Valproic Acid (VPA) and Valeric Acid (VA) in neurological disorders. Comparative overview of preclinical and clinical evidence for VPA and VA in the epigenetics of neurological disorders.
DisorderGenetic CauseEpigenetic
Mechanism & Rationale
Preclinical/Clinical Evidence (VPA)Valeric Acid (VA)References
Rett SyndromeMECP2 mutationsVPA, as an HDACi, is theoretically relevant to counteract MECP2 dysfunction.Limited clinical evidence. Used for seizure management, not a disease-modifying therapy due to severe adverse effects.N/A[138,139,140,141]
Aicardi-Goutières Syndrome (AGS)Mutations in genes like TREX1, SAMHD1.Chronic activation of the cGAS-STING pathway and Type I interferon production leads to neuroinflammation. VPA might downregulate IFN-stimulated genes (ISGs) and pro-inflammatory cytokines.Primarily preclinical research. Epigenetic modulation remains a theoretical approach.N/A[142,143,144,145,146,147,148,149,150]
Autism Spectrum Disorder (ASD)Multifactorial, including de novo mutations (e.g., ADNP, MECP2, SHANK3) and polygenic risk.Environmental factors and genetic variants shape the epigenome. VPA’s epigenetic action and impact on gut microbiota could modulate ASD-like behaviors.Mixed clinical evidence. Prenatal VPA exposure is a known risk factor for ASD. Therapeutic use in established ASD is limited, though some small trials have shown a reduction in repetitive behaviors.VA as a potential pathological mediator/marker.[121,151,152,153,154,155,156,157,158,159,160]
Genetic FTD, AD, PDMutations in GRN, C9orf72 (FTD), APP, PSEN1 (AD), LRRK2 (PD).Epigenetic dysregulations contribute to pathogenesis, including protein aggregation (TDP-43, alpha-synuclein), mitochondrial dysfunction, and oxidative stress. VPA could shift glial cells from a pro-inflammatory to a neuroprotective phenotype.Variable preclinical results. Use in human studies is restricted by safety concerns, but preclinical models have explored its potential.Protection of dopaminergic neurons by reducing oxidative stress and modulating autophagy.[161,162,163,164,165,166,167,168,169,170]
Amyotrophic Lateral Sclerosis (ALS)Mutations in SOD1, C9orf72, FUS, TDP-43.Epigenetic dysregulation, particularly involving HDACs. HDAC2 levels are increased in some ALS samples. However, the role of specific HDACs like HDAC4 is complex and context-dependent.Complexity in preclinical findings. Studies on HDAC4 inhibition in mouse models have shown detrimental effects, highlighting the nuanced and sometimes contradictory role of individual HDAC isoforms.N/A[171,172,173,174,175,176]
Spinal Muscular Atrophy (SMA)Mutations in SMN1/2 genes.VPA, as an HDACi, can enhance SMN2 expression at RNA and protein levels. This effect is more pronounced in patients with a higher SMN2 copy number.Mixed clinical results as a monotherapy. VPA was shown to increase SMN levels but provided limited benefits for motor deficits. Recent studies suggest VPA may act synergistically with other treatments like nusinersen.N/A[177,178,179,180,181,182,183,184,185]
Multiple Sclerosis (MS)Multifactorial; genetic variants in HLA-DRB1 and other immune genes.Genetic and environmental factors shape the epigenome. VPA modulates immune responses, promotes oligodendrocyte differentiation, and increases myelination gene expression.Promising preclinical evidence in EAE models, showing reduced disease severity, inflammation, and demyelination. Clinical research is ongoing, but therapeutic use is not yet standardized due to safety concerns.N/A[186,187,188,189,190,191,192,193]
Abbreviation: VA: Valeric acid; VPA: Valproic acid; N/A: not available; EAE: Experimental Autoimmune Encephalomyelitis; MECP2: Methyl-CpG-Binding Protein 2; ADNP: Activity-Dependent Neuroprotector Homeobox; SHANK3: SH3 and Multiple Ankyrin Repeat Domains 3; TREX1: 3′-Repair Exonuclease 1; SAMHD1: SAM and HD Domain Containing dNTP Hydrolase 1; GRN: Granulin Precursor Gene; C9orf72: Chromosome 9 Open Reading Frame 72; APP: Amyloid Precursor Protein; PSEN1: Presenilin 1; LRRK2: Leucine-Rich Repeat Kinase 2; SOD1: Superoxide Dismutase 1; FUS: Fused in Sarcoma; TDP-43: TAR DNA-Binding Protein 43; SMN1/2: Survival Motor Neuron 1/2; HLA-DRB1: Human Leukocyte Antigen-DR Beta 1.
Table 2. Similarities and differences between VA and VPA. A comparative analysis of clinical attributes that differentiate the endogenous VA from the pharmaceutical drug VPA.
Table 2. Similarities and differences between VA and VPA. A comparative analysis of clinical attributes that differentiate the endogenous VA from the pharmaceutical drug VPA.
Comparative FeaturesValeric Acid (VA)Valproic Acid (VPA)
OriginGut Microbiota (Endogenous Metabolite)Synthetic Drug (Pharmaceutical)
HDAC TargetsSelective, primarily Class I (HDAC3)Non-selective, Class I & Class IIa
Effective Concentration (EC50)Physiological Circulating levels = 0.18 µM);
HDAC3 IC50 (in vitro) = 16.6 µM
50–125 μg/mL (mM range)
Toxicity/Side EffectsLow/Minimal (Physiological)High/Severe (Hepatotoxicity, Teratogenicity)
Primary RolePhysiological Epigenetic MediatorEstablished Anticonvulsant/Mood Stabilizer
Brain BioavailabilityEffective permeabilityEffective permeability
Clinical StatusPreclinical/Early-stage researchEstablished Drug
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Paciolla, C.; Manganelli, M.; Di Chiano, M.; Montenegro, F.; Gallone, A.; Sallustio, F.; Guida, G. Valeric Acid: A Gut-Derived Metabolite as a Potential Epigenetic Modulator of Neuroinflammation in the Gut–Brain Axis. Cells 2025, 14, 1823. https://doi.org/10.3390/cells14221823

AMA Style

Paciolla C, Manganelli M, Di Chiano M, Montenegro F, Gallone A, Sallustio F, Guida G. Valeric Acid: A Gut-Derived Metabolite as a Potential Epigenetic Modulator of Neuroinflammation in the Gut–Brain Axis. Cells. 2025; 14(22):1823. https://doi.org/10.3390/cells14221823

Chicago/Turabian Style

Paciolla, Chiara, Michele Manganelli, Mariagiovanna Di Chiano, Francesca Montenegro, Anna Gallone, Fabio Sallustio, and Gabriella Guida. 2025. "Valeric Acid: A Gut-Derived Metabolite as a Potential Epigenetic Modulator of Neuroinflammation in the Gut–Brain Axis" Cells 14, no. 22: 1823. https://doi.org/10.3390/cells14221823

APA Style

Paciolla, C., Manganelli, M., Di Chiano, M., Montenegro, F., Gallone, A., Sallustio, F., & Guida, G. (2025). Valeric Acid: A Gut-Derived Metabolite as a Potential Epigenetic Modulator of Neuroinflammation in the Gut–Brain Axis. Cells, 14(22), 1823. https://doi.org/10.3390/cells14221823

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop