Next Article in Journal
Elevated Expression of TGFB1 in PBMCs Is Associated with Intracranial Aneurysm Formation, but TGFB3 Expression Implicated Rupture
Previous Article in Journal
Malaria Vaccines and Global Equity: A Scoping Review of Current Progress and Future Directions
Previous Article in Special Issue
Cannabidiol-Loaded Retinal Organoid-Derived Extracellular Vesicles Protect Oxidatively Stressed ARPE-19 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

Palmitoylethanolamide: A Multifunctional Molecule for Neuroprotection, Chronic Pain, and Immune Modulation

by
Valeria Di Stefano
1,
Luca Steardo, Jr.
1,*,
Martina D’Angelo
1,
Francesco Monaco
2,3 and
Luca Steardo
4,5
1
Department of Health Sciences, University of Catanzaro Magna Graecia, 88100 Catanzaro, Italy
2
Department of Mental Health, Azienda Sanitaria Locale Salerno, 84132 Salerno, Italy
3
European Biomedical Research Institute of Salerno (EBRIS), 84125 Salerno, Italy
4
Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
5
Department of Clinical Psychology, University Giustino Fortunato, 82100 Benevento, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(6), 1271; https://doi.org/10.3390/biomedicines13061271
Submission received: 10 April 2025 / Revised: 18 May 2025 / Accepted: 20 May 2025 / Published: 22 May 2025
(This article belongs to the Special Issue Therapeutic Potential for Cannabis and Cannabinoids, 3rd Edition)

Abstract

:
Palmitoylethanolamide (PEA) is an endogenous lipid mediator belonging to the N-acyl-ethanolamine family, widely recognized for its multifaceted effects on neuroprotection, chronic pain management, and immune modulation. As a naturally occurring compound, PEA plays a crucial role in maintaining homeostasis under conditions of cellular stress and inflammation. Its pharmacological effects are primarily mediated through peroxisome proliferator-activated receptor-alpha (PPAR-α) activation, alongside indirect modulation of cannabinoid receptors CB1 and CB2, as well as interactions with novel targets such as GPR55 and TRPV1. These molecular mechanisms underpin its broad therapeutic potential, particularly in the management of neuroinflammatory and neurodegenerative disorders, pain syndromes, and immune dysregulation. A major advancement in PEA research has been the development of ultramicronized palmitoylethanolamide (umPEA), which significantly enhances its bioavailability and therapeutic efficacy by facilitating better tissue absorption and interaction with key molecular pathways. Preclinical and clinical studies have demonstrated that umPEA is particularly effective in reducing neuroinflammation, stabilizing mast cells, and enhancing endocannabinoid system activity, making it a promising candidate for integrative approaches in neuropsychiatric and chronic inflammatory diseases. Given its well-established safety profile, umPEA represents an attractive alternative or adjunct to conventional anti-inflammatory and analgesic therapies. This communication provides a comprehensive overview of the mechanisms of action and therapeutic applications of both PEA and umPEA, emphasizing their emerging role in clinical practice and personalized medicine.

1. Introduction

The increasing focus on integrative approaches for the management of neuroinflammatory and immune-related disorders has prompted the investigation of natural compounds with potential modulatory effects. Among these, palmitoylethanolamide (PEA), especially in its ultramicronized palmitoylethanolamide (umPEA), is gaining recognition as a promising therapeutic option for these conditions. Based on growing preclinical and clinical evidence, we argue that umPEA deserves greater consideration within therapeutic protocols, especially in neuropsychiatric care and personalized medicine. PEA, an endogenous bioactive lipid mediator belonging to the N-acyl-ethanolamine (NAE) family, has garnered significant attention due to its diverse pharmacological properties. In response to cellular stress or injury, this compound is synthesized on demand within the lipid bilayer and exerts its effects locally within tissues, including the brain. PEA is upregulated in pathological conditions, indicating its role as a protective and homeostatic agent. Its broad spectrum of biological activities includes anti-inflammatory, analgesic, anticonvulsant, antimicrobial, antipyretic, antiepileptic, immunoregulatory, and neuroprotective effects [1].
PEA is synthesized on demand within the lipid bilayer via a two-step enzymatic process. The initial step involves a calcium- and cAMP-dependent transfer of palmitic acid from phosphatidylcholine to phosphatidylethanolamine, generating N-acylphosphatidylethanolamine (NAPE), which is then hydrolyzed by an NAPE-specific phospholipase D to release PEA. Its degradation is mediated by fatty acid amide hydrolase (FAAH) and a PEA-preferring acid amidase (PAA), both converting PEA into palmitic acid and ethanolamine. Beyond its direct effects, PEA also modulates the endocannabinoid system by inhibiting FAAH, the enzyme responsible for degrading anandamide (AEA). As a result, AEA levels increase, promoting greater activation of cannabinoid receptors CB1 and CB2 and contributing to anti-inflammatory and analgesic actions through the so-called “entourage effect” [2]. PEA has been detected in virtually all mammalian tissues, including the brain. Although the regulation of its endogenous levels is not yet fully understood, several studies indicate that PEA concentrations rise in response to tissue injury and cellular stress. This supports the notion that PEA functions as an intrinsic protective agent, mobilized to restore local homeostasis and counteract inflammation [3]. For instance, in response to cellular damage or tissue injury, macrophages, mast cells, and keratinocytes are known to release PEA as part of the body’s natural response to mitigate inflammation and promote healing [4]. In the brain, PEA has been shown to increase in conditions including neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease, as well as multiple sclerosis and traumatic brain injury [5,6]. PEA also exerts neuroprotective effects, protecting neuronal cells from oxidative damage and neurodegeneration [7,8].
The therapeutic potential of PEA is driven by its interactions with several molecular targets. A primary target is PPAR-α, a nuclear receptor involved in regulating lipid metabolism and inflammation [9]. By promoting fatty acid oxidation and reducing pro-inflammatory cytokines like TNF-α, IL-1β, and IL-6, PEA helps manage conditions such as metabolic syndrome and cardiovascular diseases, highlighting its role in correcting lipid dysregulation and chronic inflammation [10].
The increasing prevalence of chronic degenerative diseases is a major global health concern, driven in part by the nutrition transition, a shift in dietary and lifestyle patterns toward the consumption of highly processed foods, sedentary behaviors, and exposure to environmental stressors. These changes have led to widespread metabolic imbalances, chronic inflammation, and an overall decline in health, predisposing individuals to a range of chronic conditions such as metabolic syndrome, cardiovascular diseases, neurodegenerative disorders, gastrointestinal diseases, and autoimmune conditions [11,12,13,14].
Despite advancements in pharmacological treatments, the toxicity and side effects associated with long-term drug use limit their application in preventive medicine [15]. Therefore, an urgent need is to explore alternative strategies to restore metabolic homeostasis and improve public health outcomes [16]. Nutritional supplementation has emerged as a promising avenue for recreating pre-industrial dietary profiles and mitigating the impact of modern dietary insufficiencies [17,18,19,20]. Ideal supplements should possess anti-inflammatory, antioxidant, immunomodulatory, neuroprotective, and analgesic properties while enhancing cognitive function and overall recovery processes [21].
Additionally, PEA activates TRPV1, a receptor involved in pain perception and neuroinflammation. This activation leads to desensitization of sensory neurons, resulting in reduced pain. Additionally, PEA stabilizes mast cells, reducing allergic and inflammatory responses, underscoring its potential in treating neuropathic pain and chronic pain syndromes [22,23]. PEA also interacts with GPR55 and GPR119, two GPCRs involved in lipid metabolism and pain modulation. While the exact mechanisms are still under investigation, these interactions suggest additional pathways through which PEA may exert therapeutic effects, particularly in metabolic and neurodegenerative diseases [24].
The therapeutic versatility of PEA, particularly in its umPEA, has been demonstrated across a broad spectrum of conditions, including allergic and respiratory disorders, viral infections, chronic pain syndromes, musculoskeletal diseases, psychiatric illnesses, and neurodegenerative disorders [25]. Moreover, umPEA supplementation has been associated with enhanced muscle recovery, improved cognitive performance, mood stabilization, and better sleep regulation [26]. Exogenous administration offers a promising strategy for restoring physiological balance since endogenous PEA levels are often insufficient to counteract the persistent inflammatory stress linked to chronic diseases [4].
Currently, several PEA-containing formulations are available as nutraceuticals, dietary supplements, or medical foods in various countries; however, the ultra-micronized formulation of PEA alone can cross biological barriers, including the blood–brain barrier, thereby ensuring therapeutically active concentrations [27]. These products are typically administered at a recommended daily dosage of 1200 mg [28]. Notably, PEA’s long-established safety profile, supported by both clinical and preclinical research dating back to the 1950s, further reinforces its potential as a valuable adjunctive therapy for managing chronic health conditions [29]. Considering the growing scientific evidence on the role of PEA in modulating inflammation and pain, this communication aims to summarize its key mechanisms of action and explore its potential clinical applications. Specifically, we will discuss the value of PEA, and particularly umPEA, as a safe and effective therapeutic option for chronic inflammatory conditions, neurodegenerative diseases, and immune system disorders.

2. Ultramicronized Palmitoylethanolamide

umPEA is a specifically engineered formulation of PEA, obtained by subjecting standard PEA to a controlled ultramicronization process. Ultramicronization involves mechanically reducing the particle size of PEA crystals to dimensions typically below 10 μm. This significant reduction in particle size increases the surface area of the compound, thereby markedly improving dissolution rates and facilitating rapid and enhanced absorption through the gastrointestinal mucosa [30]. These pharmacological enhancements directly translate into greater clinical effectiveness, allowing lower effective doses and yielding improved therapeutic outcomes across various inflammatory, neuropathic, and neurodegenerative conditions [31,32,33]. Such advantages have been validated by multiple pharmacokinetic studies and clinical trials, reinforcing the therapeutic superiority of umPEA in clinical practice.

Pharmacokinetic and Pharmacodynamic Advantages of umPEA

Standard formulations of PEA are characterized by limited oral bioavailability and suboptimal tissue distribution, restricting their therapeutic effectiveness. umPEA, obtained through an advanced ultramicronization process that reduces particle size to the micrometer scale, significantly enhances PEA’s pharmacokinetic profile [3]. The ultramicronization markedly increases gastrointestinal absorption, leading to higher systemic bioavailability and elevated tissue concentrations. Crucially, this formulation allows umPEA to more effectively cross biological barriers, including the blood–brain barrier, thus achieving therapeutically relevant concentrations within the central nervous system [34]. Consequently, umPEA demonstrates superior pharmacodynamic activity compared to non-micronized PEA, providing enhanced efficacy in reducing neuroinflammation, stabilizing mast cells, modulating glial cell activity, and potentiating endogenous endocannabinoid system function [4]. These pharmacokinetic and pharmacodynamic improvements position umPEA as a clinically preferable alternative, especially for chronic inflammatory, neuropathic, and neuropsychiatric conditions, supported by robust preclinical and emerging clinical evidence [35].

3. PEA and Immunity

3.1. PEA and Immunity

PEA has long been recognized for its immunomodulatory properties, initially highlighted in clinical studies from the 1970s demonstrating its efficacy in reducing the incidence and severity of viral respiratory infections [36]. umPEA enhances nonspecific immune resistance against pathogens while balancing immune responses and mitigating excessive inflammation. Its immunomodulatory action primarily involves modulation of innate immune cells such as mast cells, macrophages, and microglia, preventing excessive activation and consequent neuroinflammation. By reducing cytokine release, stabilizing mast cells, and promoting microglial homeostasis, umPEA acts as an effective neuroprotective and immunomodulatory agent without the adverse effects associated with classical immunosuppressants [37,38]. Preclinical studies underscore its efficacy in conditions characterized by neuroinflammation, including Alzheimer’s, Parkinson’s, and traumatic brain injury, highlighting its potential as a therapeutic strategy for immune-mediated neurodegenerative disorders [37,38,39]. PEA’s extensive immunomodulatory effects, supported by numerous studies, highlight its ability to influence multiple physiological pathways in a coordinated manner to achieve therapeutic benefits. Due to its well-documented safety profile, PEA holds promise as a prophylactic agent for maintaining immune health and supporting overall well-being [40]. Unlike other endocannabinoids, its metabolic breakdown produces non-toxic byproducts, making it suitable for long-term use without significant adverse effects. Incorporating umPEA into health and wellness programs may serve as a viable approach to promoting immune resilience and fostering healthy aging [41].

3.2. Modulation of the Gut–Brain Axis and Gut-Derived Immunity by PEA

PEA contributes significantly to gut homeostasis and gut-derived immune regulation, primarily through its interaction with PPAR-α receptors, enhancing intestinal barrier integrity and mitigating local inflammation [42]. By activating CB2 receptors, PEA reduces gut-derived pro-inflammatory cytokines, limiting systemic inflammation and associated neurological consequences [43]. In experimental colitis models, umPEA reduced intestinal inflammation and immune cell infiltration and promoted epithelial regeneration [42,44]. Additionally, PEA may influence gut–brain interactions via GPR119 receptors, stimulating the secretion of glucagon-like peptide-1 (GLP-1), thus contributing to cognitive and emotional stability [1]. These mechanisms highlight umPEA’s potential therapeutic value in disorders involving gut dysbiosis, inflammation, and neuroimmune dysfunction [45,46,47,48].

4. PEA and Inflammatory Reactions

4.1. PEA’s Antiallergic Effects

Allergic conditions, including allergic rhinitis, dermatitis, and asthma, are primarily characterized by excessive inflammatory responses and the infiltration of immune cells into affected tissues [49]. Upon exposure to allergens, mast cells become activated and release various inflammatory mediators, such as histamine, cytokines, and chemokines, leading to symptom manifestation [50].
The antiallergic potential of PEA was first noted in the mid-20th century when early studies suggested its ability to mitigate allergic reactions. Subsequent research has confirmed PEA’s capacity to modulate immune cell responses, particularly by reducing mast cell activation and degranulation [36]. These protective effects are largely attributed to its ability to interact with specific cellular targets, including PPAR-α and GPR55 receptors, as well as its indirect influence on CB1, CB2, and TRPV1 receptors.
Experimental models have provided compelling evidence of PEA’s efficacy in reducing allergic inflammation [37]. Studies on canine skin mast cells demonstrated that PEA administration significantly inhibited the release of pro-inflammatory mediators such as prostaglandin D2, tumor necrosis factor-alpha (TNF-α), and histamine in a dose-dependent manner [40]. Similarly, in hypersensitive canine models, oral administration of umPEA effectively reduced allergic skin reactions, while in feline models, it alleviated symptoms associated with eosinophilic granuloma. Other research has shown that PEA supplementation helps manage atopic dermatitis and pruritus in dogs by reducing mast cell recruitment and inflammatory responses [38].
In murine models of allergic airway inflammation, reduced bronchial PEA levels have been observed alongside increased CB2 and GPR55 receptor expression [24]. Supplementation with PEA before allergen exposure significantly inhibited leukocyte infiltration, pulmonary inflammation, mast cell recruitment, and leukotriene production, suggesting a regulatory role in airway hypersensitivity reactions. Additionally, experimental models of contact allergic dermatitis have shown that umPEA exerts stronger anti-inflammatory effects compared to standard PEA. In these models, umPEA reduced mast cell numbers and limited angiogenic responses associated with hypersensitivity, indicating its enhanced therapeutic potential [39].
The key difference between PEA and umPEA lies in their bioavailability and efficacy. While both forms of PEA share similar mechanisms of action—modulating mast cell activity, inflammatory mediator release, and immune responses—the ultramicronization process of umPEA significantly improves its absorption and bioavailability. While both forms of PEA share similar mechanisms of action—modulating mast cell activity, inflammatory mediator release, and immune responses—the ultramicronization of PEA markedly enhances its absorption and bioavailability, leading to higher tissue concentrations and improved therapeutic outcomes. This enhanced bioavailability enables umPEA to provide superior efficacy in alleviating symptoms associated with allergic conditions. Preclinical and clinical evidence suggests that umPEA, due to its optimized pharmacokinetic profile, offers a more potent therapeutic solution for the management of chronic inflammatory diseases, including allergic dermatitis and asthma, compared to standard PEA formulations [3,51]. Limited clinical studies in humans have also indicated the potential benefits of PEA in allergic conditions. While these findings are promising, further large-scale clinical trials are needed to fully establish the efficacy of PEA in managing allergic and inflammatory disorders [46,47].

4.2. PEA and Brain Health

Neuroinflammation is increasingly recognized as a fundamental contributor to the development and progression of numerous neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, stroke, traumatic brain injury (TBI), autism spectrum disorder (ASD), epilepsy, and various cognitive, behavioral, and mood disorders. The presence of chronic inflammation in the central nervous system (CNS) can lead to the progressive deterioration of neuronal structures, impairing function and ultimately contributing to disease pathology [5,8,42].
Key players in neuroinflammatory processes include mast cells, microglia, and astrocytes, which, when persistently activated, release inflammatory mediators that exacerbate neuronal damage [43]. Although the detailed molecular mechanisms of neuroinflammation are complex, it is widely accepted that chronic activation of these pathways disrupts neuronal homeostasis and accelerates neurodegenerative processes. Consequently, therapeutic strategies aimed at mitigating neuroinflammation hold promise for preserving brain health and function [48].
Exogenous administration of umPEA has been explored in preclinical and clinical settings, where it has demonstrated beneficial effects in conditions such as stroke, neurotrauma, and neurodegenerative diseases. By enhancing endogenous neuroprotective mechanisms and dampening harmful inflammatory cascades, umPEA may support long-term brain health and function [25,44].
Moreover, recent meta-analytical evidence further supports the cognitive benefits of PEA supplementation. Specifically, Colizzi et al. (2022), in their systematic review and preliminary meta-analysis, demonstrated that PEA significantly improves cognitive function, as evidenced by enhanced Mini-Mental State Examination (MMSE) scores of 3.80 points (95% CI: −0.16 to 7.75), indicating a trend toward cognitive enhancement, and positively affects executive function, working memory, language deficits, and daily living activities. These findings strongly reinforce the therapeutic potential of PEA in neurocognitive disorders, suggesting its role not merely in symptom management but potentially in slowing cognitive decline progression [52].
Additionally, PEA has shown efficacy in mitigating negative symptoms commonly associated with psychotic and manic disorders, including schizophrenia and bipolar disorder, supporting its tolerability and possible use as a safe and effective therapeutic intervention in the psychiatric setting [53].
Given the growing body of research supporting umPEA’s role in neuroinflammation and neuroprotection, its application in the management of neurodegenerative diseases and neurological disorders is an area of increasing interest [54,55]. Further studies are needed to fully elucidate its mechanisms of action, optimal dosing strategies, and long-term efficacy in maintaining cognitive function and preventing neurodegeneration.

4.3. PEA’s Effects on Mood, Cognition, and Behavior

PEA’s role extends beyond inflammation but also has a direct impact on mood, anxiety, stress, and cognitive function through its interactions with the endocannabinoid system and other neuromodulatory pathways. The significance of endocannabinoids and N-acyl-ethanolamines in regulating cognitive function, mood, and behavior is well-established [56,57]. Endogenous PEA levels have been shown to increase in response to acute psychosocial stress, acting as an adaptive mechanism to counteract inflammatory and excitotoxic cascades within the central nervous system, as happens in PTSD and depression. Interestingly, individuals with PTSD often self-medicate with cannabis, possibly due to its effects on the endocannabinoid system [58]. Higher PEA concentrations have been observed in affected individuals, correlating with symptom severity [59]. This suggests that while PEA is upregulated as part of an adaptive response, endogenous levels may be insufficient or depleted to counteract chronic stress, proposing that supplementation could help restore neurochemical balance [60,61].
At the same time, antidepressants have been found to increase brain PEA levels, further supporting its relevance in mood regulation [62]. Furthermore, in patients with major depressive disorder, the addition of umPEA to standard antidepressant treatment led to significantly greater improvements in depressive symptoms compared to placebo. In animal models of Alzheimer’s disease and traumatic brain injury, PEA supplementation has been shown to improve memory function while reducing anxiety, aggression, and depressive symptoms [25,63]. Reports from case studies on individuals across the autism spectrum, both children and adults, suggest that PEA supplementation improves social behaviors, cognitive function, and overall adaptability [64,65].
Instead, umPEA is more effective on the CNS due to its ability to cross the blood–brain barrier.
The therapeutic potential of umPEA in these neuropsychiatric conditions is increasingly being explored in preclinical and clinical studies that have shown promising beneficial effects (personal data, paper in preparation).
Studies have demonstrated that umPEA, in its micronized form, exhibits enhanced pharmacokinetic properties, including improved bioavailability and more efficient distribution within the CNS compared to conventional PEA [66]. This enhanced profile makes umPEA particularly advantageous for conditions characterized by neuroinflammation, neurodegeneration, or chronic nociceptive and neuropathic pain. The ability of umPEA to effectively cross the blood–brain barrier and achieve central distribution allows it to directly interact with the brain and spinal cord, enhancing its therapeutic potential for disorders such as neuroinflammatory and neurodegenerative conditions [67].
PEA’s ability to support neurogenesis and synaptic remodeling likely plays a fundamental role in its effects on cognition and behavior. Moreover, as chronic pain and mood disorders negatively impact memory and executive function, PEA’s concurrent antidepressant, anxiolytic, and analgesic properties may contribute to its cognitive-enhancing potential. Given its ability to target neuroinflammation, modulate pain perception, alleviate depressive symptoms, and promote neuroplasticity, umPEA emerges as a promising therapeutic candidate for neuropsychiatric and neurocognitive disorders [68]. While current findings are encouraging, further clinical trials are necessary to establish optimal dosing strategies and confirm their long-term benefits in both clinical populations and healthy individuals aiming to support brain health.

4.4. Inflammation and Pain

Inflammation plays a crucial role in pain, serving both as a protective and a pathological mechanism. While acute inflammation is essential for tissue healing and defense against injury, chronic inflammation contributes to the persistence of pain and the development of neuropathic conditions [69]. Non-neuronal cells, such as mast cells, microglia, and astrocytes, play a key role in amplifying and sustaining inflammatory responses, sensitizing pain pathways. This interaction leads to central and peripheral sensitization, where normally non-painful stimuli are perceived as painful [70].

4.4.1. umPEA Mechanism of Action

Inflammatory pain refers to pain resulting from tissue injury and the associated immune response, typically characterized by the activation of peripheral nociceptors due to the release of pro-inflammatory mediators such as cytokines and prostaglandins. Persistent pain, often overlapping with chronic pain, is defined as pain that lasts beyond normal tissue healing time, generally more than three months, and may involve continued inflammation, sensitization of neural pathways, or maladaptive neuroimmune mechanisms [71].
Palmitoylethanolamide (umPEA), an endogenous lipid molecule, plays a key role in pain relief by modulating this neuroimmune response. It activates PPAR-α and GPR55 receptors and exerts indirect effects on CB1, CB2, and TRPV1 receptors. By inhibiting mast cell activation, umPEA suppresses the release of key pro-inflammatory mediators such as nerve growth factor (NGF), cyclooxygenase-2 (COX-2), tumor necrosis factor-alpha (TNF-α), and inducible nitric oxide synthase (iNOS) [10]. Additionally, it reduces microglial and astrocyte activation, preserving peripheral nerve morphology, reducing endoneurial edema, and limiting macrophage infiltration in inflamed tissues.

4.4.2. Differences in umPEA Action Across Pain Types

Pain syndromes can be broadly classified into nociceptive, neuropathic, and nociplastic pain, each characterized by distinct pathophysiological mechanisms. Nociceptive pain arises from tissue injury and inflammation, triggering peripheral nociceptors. umPEA addresses this pain type primarily through its anti-inflammatory properties, stabilizing mast cells, and inhibiting cytokine release. Neuropathic pain, resulting from damage or dysfunction within the nervous system itself, involves neuroinflammation and sensitization of neural pathways [72]. umPEA exerts its therapeutic effects in neuropathic conditions by modulating microglial activation, reducing pro-inflammatory cytokines, and normalizing sensory neuron excitability via interactions with PPAR-α, CB2, and TRPV1 receptors [10,73]. Finally, nociplastic pain represents pain without clear tissue or nerve injury, largely driven by abnormal central processing of pain signals. Here, umPEA’s ability to cross the blood–brain barrier, modulate central glial cells, and enhance endogenous analgesic mechanisms through the endocannabinoid system is particularly beneficial [23,32,74]. Thus, umPEA offers targeted, multimodal relief tailored to the specific underlying mechanisms of each distinct pain type.

4.4.3. Preclinical and Clinical Evidence

Preclinical and clinical studies have documented the efficacy of umPEA in the treatment of inflammatory and chronic pain, with applications in conditions such as fibromyalgia, osteoarthritis, postoperative pain, and endometriosis [75,76,77]. In cases of chronic pain, endogenous PEA levels may be reduced, but supplementing with exogenous umPEA can restore its protective and anti-inflammatory effects [70].

4.4.4. Comparison with Pharmaceutical Analgesics

Although conventional analgesic medications, such as NSAIDs, opioids, and corticosteroids, are commonly used for chronic pain, their long-term use carries significant risks of gastrointestinal, hepatic, cardiovascular, and renal complications [78]. In contrast, umPEA stands out for its favorable safety profile, offering a promising, natural, and well-tolerated alternative for individuals suffering from chronic pain and inflammation. Further clinical research is necessary to optimize therapeutic strategies and dosing for long-term pain management [79]. A comparative overview of the pharmacological and clinical advantages of standard PEA versus umPEA is summarized in Table 1.

4.5. PEA’s Effects on Primary Headaches

Headaches are one of the most prevalent neurological disorders, affecting nearly all individuals at some point in their lives. Among these, primary headaches, including migraines and tension-type headaches (TTH), constitute a significant global health burden. These conditions can occur as isolated episodic events or develop into chronic disorders, severely impacting quality of life and daily functioning [82,83].
Chronic TTH is believed to involve central sensitization and a deficiency in endogenous pain control mechanisms, with nociceptive input originating from myofascial tissues around the cranium. In migraines, early stages are thought to be triggered by the activation of trigeminal nociceptors due to neurovascular inflammation in the meninges and cranial blood vessels [84]. Once sensitization occurs in the perivascular trigeminal nerve terminals, non-painful stimuli can provoke pain responses. Moreover, dural mast cells and associated glial cells contribute to migraine pathology by releasing pronociceptive and pro-inflammatory mediators, which further drive sensitization [85,86].
umPEA has emerged as a potential therapeutic agent for primary headaches due to its ability to modulate inflammation and pain signaling. By regulating the release of inflammatory mediators and stabilizing mast cells, umPEA may help reduce the excessive nociceptive input that contributes to headache pathophysiology [87]. Clinical studies have explored the efficacy of umPEA in treating migraines [88,89]. Research has demonstrated that sublingual administration of PEA leads to a reduction in headache frequency, duration, and intensity, as well as a decrease in the use of analgesic medications among individuals with migraine [88]. Additionally, PEA supplementation has shown benefits in patients with migraines with aura who were concurrently using non-steroidal anti-inflammatory drugs (NSAIDs), leading to significant reductions in pain severity, attack frequency, and duration. Similar findings have been observed in pediatric migraine populations, where PEA supplementation resulted in a marked decrease in attack frequency and severity over a three-month period [89].
These findings suggest that PEA holds promise not only as a migraine prophylactic but also as a potential therapeutic option for tension-type headaches. Its ability to regulate neuroinflammation, stabilize nociceptive signaling pathways, and support endogenous pain control mechanisms makes it an attractive candidate for future headache management strategies [9]. Further clinical trials are necessary to establish optimal dosing and long-term efficacy in broader populations.

4.6. PEA and Sleep

Chronic pain is particularly known to interfere with sleep quality, with conditions such as neuropathic pain, osteoarthritis, diabetic neuropathy, and carpal tunnel syndrome being strongly associated with sleep disruptions. Circadian rhythms regulate numerous essential physiological processes, including sleep–wake cycles, cognitive performance, and overall homeostasis [90]. Disruptions to these rhythms can negatively impact both physical and mental well-being, leading to increased sympathetic nervous system activity, stress susceptibility, mood disorders, and cognitive decline [91]. Persistent disturbances in sleep patterns have been linked to impairments in synaptic plasticity, deficits in attention and working memory, and increased susceptibility to depression and neurodegenerative conditions [92].
Various external and internal factors, including environmental stressors, psychosocial pressures, and underlying medical conditions, can contribute to sleep disturbances [93]. While prescription sedatives and tranquilizers are commonly used to manage sleep disorders, they come with risks of dependence and adverse side effects [94,95]. Cannabinoid-based treatments offer an alternative, but prolonged use may result in withdrawal-induced sleep deprivation, underscoring the need for natural, well-tolerated sleep aids [96,97].

PEA’s Role in Sleep Regulation

Given the close interplay between pain, stress, and sleep disturbances, umPEA’s analgesic and anxiolytic properties make it a promising candidate for improving sleep quality [69]. By modulating TRPV1 channels and enhancing anandamide signaling via the entourage effect, PEA may help alleviate sleep disturbances associated with pain and anxiety. Additionally, its ability to modulate the hypothalamic–pituitary–adrenal (HPA) axis may mitigate stress-induced sleep disorders.
Emerging clinical research supports umPEA’s role in improving sleep patterns. In individuals experiencing chronic pain, PEA supplementation has been shown to reduce stress, anxiety, and associated sleep disruptions [70]. A study involving patients with osteoarthritis demonstrated that PEA not only alleviated joint pain but also improved overall sleep quality [98]. Furthermore, in patients recovering from carpal tunnel syndrome surgery, PEA supplementation significantly enhanced sleep–wake rhythms and overall restfulness [99].
Declining levels of anandamide have been associated with sleep disturbances in aging populations [100]. Through its role in supporting endocannabinoid signaling, umPEA may help maintain circadian balance and cognitive function in both healthy individuals and those at risk for sleep disorders. While further research is needed to fully elucidate its long-term benefits, PEA presents a compelling option for promoting restorative sleep and overall well-being without the adverse effects associated with conventional sleep aids.

5. Limitations

Despite growing interest in PEA, especially umPEA, several limitations persist. The current evidence is primarily based on heterogeneous preclinical studies and small-scale clinical trials, often lacking robust designs such as adequate randomization and control groups, which compromises validity and generalizability [25]. Additionally, significant variability in formulations, dosing regimens, and study outcomes hampers the comparability and reproducibility of results, limiting definitive conclusions and robust meta-analyses [29]. To establish umPEA’s therapeutic role clearly, future studies must include larger, randomized controlled trials and standardized protocols addressing pharmacokinetic variability and clinical efficacy across diverse populations [99].

6. Conclusions and Future Perspectives: The Anti-Inflammatory Potential of PEA in Clinical Practice

PEA, and particularly its umPEA, has demonstrated significant potential as a safe and effective anti-inflammatory agent with broad applications in the management of neurodegenerative diseases, chronic pain syndromes, and immune-related disorders [101]. Its multimodal mechanism of action via PPAR-α activation, mast cell stabilization, and indirect modulation of the endocannabinoid system confers a unique therapeutic versatility that distinguishes it from conventional anti-inflammatory and analgesic drugs [102].
Unlike NSAIDs or corticosteroids, umPEA does not carry the risk of gastrointestinal bleeding, cardiovascular complications, or renal impairment, which often limit the long-term use of traditional agents in chronic conditions [103]. Moreover, its favorable tolerability profile, supported by decades of clinical and preclinical data, renders it a compelling candidate for long-term, integrative treatment strategies in both preventive and therapeutic contexts.
In our opinion, umPEA is not merely a complementary option—it represents a paradigm shift in how we approach chronic inflammation, particularly in neuropsychiatric and pain-related disorders. We believe its incorporation into clinical protocols should be actively pursued, especially in patients with poor tolerability to standard treatments or in those with complex, multisystemic symptoms.
Nonetheless, the integration of umPEA into mainstream medical practice requires a concerted effort to overcome regulatory inertia, establish standardized guidelines, and generate high-quality evidence through rigorous clinical trials. In this regard, we strongly advocate for a translational research agenda that prioritizes head-to-head comparisons, personalized dosing strategies, and mechanistic studies aimed at identifying predictive biomarkers of response.
In summary, we believe that umPEA, by virtue of its safety, pleiotropic effects, and neuroimmune targeting capacity, should be considered not only as an adjunct but as a central pillar in the future of anti-inflammatory therapy, particularly within the framework of precision medicine.
Given the accumulating preclinical and clinical evidence, and in light of its excellent safety profile, we urge clinicians and researchers to reconsider the role of umPEA not as a marginal nutraceutical but as a scientifically grounded therapeutic agent deserving full clinical dignity. We advocate for its inclusion in clinical guidelines, particularly for complex syndromes where inflammation, neuroglial dysfunction, and immune dysregulation converge. In an era that increasingly demands personalized, multimodal, and well-tolerated therapies, umPEA represents an underutilized asset. The time has come to move beyond skepticism and integrate this molecule into the therapeutic arsenal with the scientific rigor and clinical openness it deserves.

7. Practical Clinical Recommendations for umPEA Use

As a well-tolerated, pleiotropic molecule acting across neuroimmune, glial, and metabolic pathways, umPEA should be actively considered in the clinical management of complex chronic conditions. Its use is particularly valuable when conventional therapies are limited by adverse effects, polypharmacy, or suboptimal response.

7.1. Chronic Pain and Inflammatory Disorders

  • Neuropathic Pain and Fibromyalgia. umPEA should be used as an adjunct to gabapentinoids or antidepressants to enhance analgesia while minimizing dose-dependent side effects. A starting dose of 600 mg twice daily is recommended, with individual titration based on clinical response [77,80,104,105].
  • Autoimmune and Systemic Inflammation. In diseases such as rheumatoid arthritis or inflammatory bowel disease, umPEA should be considered as a supportive agent to downregulate immune hyperactivity and systemic inflammation, particularly in patients with contraindications to long-term steroid or NSAID use [77,106].

7.2. Neuroinflammation and Neurodegenerative Conditions

  • Alzheimer’s and Parkinson’s Disease. Given its neuroprotective and anti-inflammatory actions, umPEA (600–1200 mg/day) is a rational adjunct in early-phase disease management aimed at slowing cognitive and functional decline [5,25].
  • Stroke and Traumatic Brain Injury (TBI). Post-acute supplementation with umPEA (600 mg twice daily) may enhance recovery, reduce neuroinflammation, and promote neuroplasticity, as supported by preclinical and emerging clinical evidence [107,108].

7.3. Gut–Brain Axis and Immune Regulation

  • Leaky Gut and Microbiota Dysregulation. umPEA (300–600 mg/day) in combination with targeted probiotics can restore gut barrier function, mitigate dysbiosis, and reduce systemic endotoxemia, especially in IBS, Crohn’s disease, or colitis [73,109].
  • Immune Support and Viral Infections. Historical and experimental data support the use of umPEA for enhancing non-specific immune defense. 600 mg/day is recommended for immune support, with 1200 mg/day during acute respiratory infections or high-risk exposure periods [110].
The clinical indications are summarized in Table 2.
Table 3 details the observed clinical outcomes associated with umPEA administration in different conditions, highlighting model types and key mechanisms involved.

8. Final Considerations

In our view, umPEA should not be relegated to the margins of integrative medicine but rather promoted as a core tool in personalized anti-inflammatory strategies. Its exceptional tolerability, combined with a multimodal mechanism of action, makes it uniquely suited for long-term management of inflammation-driven disorders. While additional large-scale RCTs are warranted to refine dosage and indications, the current evidence justifies its immediate, judicious incorporation into clinical algorithms, particularly in patients for whom conventional therapies have proven insufficient or intolerable. Although pharmacokinetic and preclinical studies have demonstrated superior bioavailability and tissue distribution of micronized and ultra-micronized PEA compared to naïve formulations, direct head-to-head clinical trials comparing umPEA with other PEA formulations remain limited, highlighting the need for further randomized controlled studies [22,112,113]
Table 1 Legend: Comparison of pharmacological and clinical differences between standard PEA and ultramicronized PEA (umPEA). The ultramicronization process significantly reduces particle size (<10 µm), greatly enhancing oral bioavailability, systemic absorption, and blood–brain barrier penetration. Consequently, umPEA achieves higher tissue concentrations, faster therapeutic onset, and greater clinical efficacy at lower doses compared to standard PEA formulations. These pharmacokinetic and pharmacodynamic advantages position umPEA as a preferable therapeutic option for inflammatory, neuropathic, and neurodegenerative conditions.

Author Contributions

Conceptualization, L.S.J.; methodology, L.S.J.; formal analysis, L.S.J.; investigation, F.M., M.D., V.D.S. and L.S.; data curation, L.S.J. and F.M.; writing—original draft preparation, M.D., V.D.S. and L.S.J.; writing—review and editing, M.D., V.D.S., L.S., F.M. and L.S.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Clayton, P.; Hill, M.; Bogoda, N.; Subah, S.; Venkatesh, R. Palmitoylethanolamide: A Natural Compound for Health Management. Int. J. Mol. Sci. 2021, 22, 5305. [Google Scholar] [CrossRef] [PubMed]
  2. Rankin, L.; Fowler, C.J. The Basal Pharmacology of Palmitoylethanolamide. Int. J. Mol. Sci. 2020, 21, 7942. [Google Scholar] [CrossRef]
  3. Petrosino, S.; Di Marzo, V. The Pharmacology of Palmitoylethanolamide and First Data on the Therapeutic Efficacy of Some of Its New Formulations. Br. J. Pharmacol. 2017, 174, 1349–1365. [Google Scholar] [CrossRef] [PubMed]
  4. Skaper, S.D.; Facci, L. Mast Cell-Glia Axis in Neuroinflammation and Therapeutic Potential of the Anandamide Congener I confirm Palmitoylethanolamide. Philos. Trans. R. Soc. B Biol. Sci. 2012, 367, 3312–3325. [Google Scholar] [CrossRef]
  5. Beggiato, S.; Tomasini, M.C.; Ferraro, L. Palmitoylethanolamide (PEA) as a Potential Therapeutic Agent in Alzheimer’s Disease. Front. Pharmacol. 2019, 10, 821. [Google Scholar] [CrossRef]
  6. Talarico, G.; Trebbastoni, A.; Bruno, G.; de Lena, C. Modulation of the Cannabinoid System: A New Perspective for the Treatment of the Alzheimer’s Disease. Curr. Neuropharmacol. 2019, 17, 176–183. [Google Scholar] [CrossRef] [PubMed]
  7. Esposito, E.; Impellizzeri, D.; Mazzon, E.; Paterniti, I.; Cuzzocrea, S. Neuroprotective Activities of Palmitoylethanolamide in an Animal Model of Parkinson’s Disease. PLoS ONE 2012, 7, e41880. [Google Scholar] [CrossRef]
  8. Bronzuoli, M.R.; Facchinetti, R.; Steardo, L.; Romano, A.; Stecca, C.; Passarella, S.; Steardo, L.; Cassano, T.; Scuderi, C. Palmitoylethanolamide Dampens Reactive Astrogliosis and Improves Neuronal Trophic Support in a Triple Transgenic Model of Alzheimer’s Disease: In Vitro and In Vivo Evidence. Oxidative Med. Cell. Longev. 2018, 2018, 4720532. [Google Scholar] [CrossRef]
  9. Keppel Hesselink, J. Evolution in Pharmacologic Thinking around the Natural Analgesic Palmitoylethanolamide: From Nonspecific Resistance to PPAR-α Agonist and Effective Nutraceutical. J. Pain Res. 2013, 625, 625–634. [Google Scholar] [CrossRef]
  10. Costa, B.; Comelli, F.; Bettoni, I.; Colleoni, M.; Giagnoni, G. The Endogenous Fatty Acid Amide, Palmitoylethanolamide, Has Anti-Allodynic and Anti-Hyperalgesic Effects in a Murine Model of Neuropathic Pain: Involvement of CB1, TRPV1 and PPARγ Receptors and Neurotrophic Factors. Pain 2008, 139, 541–550. [Google Scholar] [CrossRef]
  11. Popkin, B.M. Global Nutrition Dynamics: The World Is Shifting Rapidly toward a Diet Linked with Noncommunicable Diseases. Am. J. Clin. Nutr. 2006, 84, 289–298. [Google Scholar] [CrossRef] [PubMed]
  12. Corbett, S.; Courtiol, A.; Lummaa, V.; Moorad, J.; Stearns, S. The Transition to Modernity and Chronic Disease: Mismatch and Natural Selection. Nat. Rev. Genet. 2018, 19, 419–430. [Google Scholar] [CrossRef] [PubMed]
  13. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic Inflammation in the Etiology of Disease across the Life Span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
  14. Carrera-Bastos, P.; Fontes; O’Keefe, J.; Lindeberg, S.; Cordain, L. The Western Diet and Lifestyle and Diseases of Civilization. Res. Rep. Clin. Cardiol. 2011, 2, 15–35. [Google Scholar] [CrossRef]
  15. Kim, H.S.; Cho, J.H.; Yoon, K.H. New Directions in Chronic Disease Management. Endocrinol. Metab. 2015, 30, 159–166. [Google Scholar] [CrossRef]
  16. Clemente-Suárez, V.J.; Martín-Rodríguez, A.; Redondo-Flórez, L.; López-Mora, C.; Yáñez-Sepúlveda, R.; Tornero-Aguilera, J.F. New Insights and Potential Therapeutic Interventions in Metabolic Diseases. Int. J. Mol. Sci. 2023, 24, 10672. [Google Scholar] [CrossRef]
  17. Rautiainen, S.; Manson, J.E.; Lichtenstein, A.H.; Sesso, H.D. Dietary Supplements and Disease Prevention—A Global Overview. Nat. Rev. Endocrinol. 2016, 12, 407–420. [Google Scholar] [CrossRef] [PubMed]
  18. Blumberg, J.B.; Bailey, R.L.; Sesso, H.D.; Ulrich, C.M. The Evolving Role of Multivitamin/Multimineral Supplement Use among Adults in the Age of Personalized Nutrition. Nutrients 2018, 10, 248. [Google Scholar] [CrossRef]
  19. Wienecke, E.; Gruenwald, J. Nutritional Supplementation: Is It Necessary for Everybody? Adv. Ther. 2007, 24, 1126–1135. [Google Scholar] [CrossRef]
  20. Ward, E. Addressing Nutritional Gaps with Multivitamin and Mineral Supplements. Nutr. J. 2014, 13, 72. [Google Scholar] [CrossRef]
  21. Nau, R.; Ribes, S.; Djukic, M.; Eiffert, H. Strategies to Increase the Activity of Microglia as Efficient Protectors of the Brain against Infections. Front. Cell. Neurosci. 2014, 8, 138. [Google Scholar] [CrossRef]
  22. Petrosino, S.; Schiano Moriello, A.; Cerrato, S.; Fusco, M.; Puigdemont, A.; De Petrocellis, L.; Di Marzo, V. The Anti-Inflammatory Mediator Palmitoylethanolamide Enhances the Levels of 2-Arachidonoyl-Glycerol and Potentiates Its Actions at TRPV1 Cation Channels. Br. J. Pharmacol. 2016, 173, 1154–1162. [Google Scholar] [CrossRef]
  23. Bettoni, I.; Comelli, F.; Colombo, A.; Bonfanti, P.; Costa, B. Non-Neuronal Cell Modulation Relieves Neuropathic Pain: Efficacy of the Endogenous Lipid Palmitoylethanolamide. CNS Neurol. Disord. Drug Targets 2013, 12, 34–44. [Google Scholar] [CrossRef]
  24. Ryberg, E.; Larsson, N.; Sjögren, S.; Hjorth, S.; Hermansson, N.-O.; Leonova, J.; Elebring, T.; Nilsson, K.; Drmota, T.; Greasley, P.J. The Orphan Receptor GPR55 Is a Novel Cannabinoid Receptor. Br. J. Pharmacol. 2007, 152, 1092–1101. [Google Scholar] [CrossRef]
  25. Scuderi, C.; Bronzuoli, M.R.; Facchinetti, R.; Pace, L.; Ferraro, L.; Broad, K.D.; Serviddio, G.; Bellanti, F.; Palombelli, G.; Carpinelli, G.; et al. Ultramicronized Palmitoylethanolamide Rescues Learning and Memory Impairments in a Triple Transgenic Mouse Model of Alzheimer’s Disease by Exerting Anti-Inflammatory and Neuroprotective Effects. Transl. Psychiatry 2018, 8, 32. [Google Scholar] [CrossRef]
  26. Kim, N.; Parolin, B.; Renshaw, D.; Deb, S.K.; Zariwala, M.G. Formulated Palmitoylethanolamide Supplementation Improves Parameters of Cognitive Function and BDNF Levels in Young, Healthy Adults: A Randomised Cross-Over Trial. Nutrients 2024, 16, 489. [Google Scholar] [CrossRef] [PubMed]
  27. Paterniti, I.; Impellizzeri, D.; Di Paola, R.; Navarra, M.; Cuzzocrea, S.; Esposito, E. A New Co-Ultramicronized Composite Including Palmitoylethanolamide and Luteolin to Prevent Neuroinflammation in Spinal Cord Injury. J. Neuroinflammation 2013, 10, 91. [Google Scholar] [CrossRef]
  28. Galla, R.; Mulè, S.; Ferrari, S.; Grigolon, C.; Molinari, C.; Uberti, F. Palmitoylethanolamide as a Supplement: The Importance of Dose-Dependent Effects for Improving Nervous Tissue Health in an In Vitro Model. Int. J. Mol. Sci. 2024, 25, 9079. [Google Scholar] [CrossRef] [PubMed]
  29. Varrassi, G.; Rekatsina, M.; Leoni, M.L.G.; Cascella, M.; Finco, G.; Sardo, S.; Corno, C.; Tiso, D.; Schweiger, V.; Fornasari, D.M.M.; et al. A Decades-Long Journey of Palmitoylethanolamide (PEA) for Chronic Neuropathic Pain Management: A Comprehensive Narrative Review. Pain Ther. 2025, 14, 81–101. [Google Scholar] [CrossRef]
  30. Nobili, S.; Micheli, L.; Lucarini, E.; Toti, A.; Ghelardini, C.; Di Cesare Mannelli, L. Ultramicronized N-Palmitoylethanolamine Associated with Analgesics: Effects against Persistent Pain. Pharmacol. Ther. 2024, 258, 108649. [Google Scholar] [CrossRef]
  31. Basavarajappa, B.S.; Shivakumar, M.; Joshi, V.; Subbanna, S. Endocannabinoid System in Neurodegenerative Disorders. J. Neurochem. 2017, 142, 624–648. [Google Scholar] [CrossRef] [PubMed]
  32. Palazzo, E.; Luongo, L.; Guida, F.; de Novellis, V.; Boccella, S.; Cristiano, C.; Marabese, I.; Maione, S. Role of N-Acylethanolamines in the Neuroinflammation: Ultramicronized Palmitoylethanolamide in the Relief of Chronic Pain and Neurodegenerative DiseasesRole of N-Acylethanolamines in the Neuroinflammation: Ultramicronized Palmitoylethanolamide in the Relief of Chronic Pain and Neurodegenerative Diseases. Neuropsychiatry 2019, 9, 1–12. [Google Scholar] [CrossRef]
  33. Tadijan, A.; Vlašić, I.; Vlainić, J.; Đikić, D.; Oršolić, N.; Jazvinšćak Jembrek, M. Intracellular Molecular Targets and Signaling Pathways Involved in Antioxidative and Neuroprotective Effects of Cannabinoids in Neurodegenerative Conditions. Antioxidants 2022, 11, 2049. [Google Scholar] [CrossRef] [PubMed]
  34. Petrosino, S.; Cordaro, M.; Verde, R.; Schiano Moriello, A.; Marcolongo, G.; Schievano, C.; Siracusa, R.; Piscitelli, F.; Peritore, A.F.; Crupi, R.; et al. Oral Ultramicronized Palmitoylethanolamide: Plasma and Tissue Levels and Spinal Anti-Hyperalgesic Effect. Front. Pharmacol. 2018, 9, 249. [Google Scholar] [CrossRef]
  35. Impellizzeri, D.; Bruschetta, G.; Cordaro, M.; Crupi, R.; Siracusa, R.; Esposito, E.; Cuzzocrea, S. Micronized/Ultramicronized Palmitoylethanolamide Displays Superior Oral Efficacy Compared to Nonmicronized Palmitoylethanolamide in a Rat Model of Inflammatory Pain. J. Neuroinflamm. 2014, 11, 136. [Google Scholar] [CrossRef]
  36. Maisto, M.; Piccolo, V.; Marzocchi, A.; Maresca, D.C.; Romano, B.; Summa, V.; Tenore, G.C.; Ercolano, G.; Ianaro, A. Nutraceutical Formulation Based on a Synergic Combination of Melatonin and Palmitoylethanolamide for the Management of Allergic Events. Front. Nutr. 2024, 11, 1417747. [Google Scholar] [CrossRef]
  37. Brown, J.M.; Wilson, T.M.; Metcalfe, D.D. The Mast Cell and Allergic Diseases: Role in Pathogenesis and Implications for Therapy. Clin. Exp. Allergy 2008, 38, 4–18. [Google Scholar] [CrossRef]
  38. Cerrato, S.; Brazis, P.; della Valle, M.F.; Miolo, A.; Puigdemont, A. Effects of Palmitoylethanolamide on Immunologically Induced Histamine, PGD2 and TNFα Release from Canine Skin Mast Cells. Vet. Immunol. Immunopathol. 2010, 133, 9–15. [Google Scholar] [CrossRef]
  39. Vaia, M.; Petrosino, S.; De Filippis, D.; Negro, L.; Guarino, A.; Carnuccio, R.; Di Marzo, V.; Iuvone, T. Palmitoylethanolamide Reduces Inflammation and Itch in a Mouse Model of Contact Allergic Dermatitis. Eur. J. Pharmacol. 2016, 791, 669–674. [Google Scholar] [CrossRef]
  40. Nelissen, S.; Lemmens, E.; Geurts, N.; Kramer, P.; Maurer, M.; Hendriks, J.; Hendrix, S. The Role of Mast Cells in Neuroinflammation. Acta Neuropathol. 2013, 125, 637–650. [Google Scholar] [CrossRef]
  41. Belizário, J.E.; Faintuch, J.; Garay-Malpartida, M. Gut Microbiome Dysbiosis and Immunometabolism: New Frontiers for Treatment of Metabolic Diseases. Mediat. Inflamm. 2018, 2018, 1–12. [Google Scholar] [CrossRef] [PubMed]
  42. Scuderi, C.; Stecca, C.; Valenza, M.; Ratano, P.; Bronzuoli, M.R.; Bartoli, S.; Steardo, L.; Pompili, E.; Fumagalli, L.; Campolongo, P.; et al. Palmitoylethanolamide Controls Reactive Gliosis and Exerts Neuroprotective Functions in a Rat Model of Alzheimer’s Disease. Cell Death Dis. 2014, 5, e1419. [Google Scholar] [CrossRef] [PubMed]
  43. Skaper, S.D.; Facci, L.; Giusti, P. Mast Cells, Glia and Neuroinflammation: Partners in Crime? Immunology 2014, 141, 314–327. [Google Scholar] [CrossRef]
  44. Bellanti, F.; Bukke, V.N.; Moola, A.; Villani, R.; Scuderi, C.; Steardo, L.; Palombelli, G.; Canese, R.; Beggiato, S.; Altamura, M.; et al. Effects of Ultramicronized Palmitoylethanolamide on Mitochondrial Bioenergetics, Cerebral Metabolism, and Glutamatergic Transmission: An Integrated Approach in a Triple Transgenic Mouse Model of Alzheimer’s Disease. Front. Aging Neurosci. 2022, 14, 890855. [Google Scholar] [CrossRef] [PubMed]
  45. Fakhri, S.; Yarmohammadi, A.; Yarmohammadi, M.; Farzaei, M.H.; Echeverria, J. Marine Natural Products: Promising Candidates in the Modulation of Gut-Brain Axis towards Neuroprotection. Mar. Drugs 2021, 19, 165. [Google Scholar] [CrossRef]
  46. Bertolino, B.; Crupi, R.; Impellizzeri, D.; Bruschetta, G.; Cordaro, M.; Siracusa, R.; Esposito, E.; Cuzzocrea, S. Beneficial Effects of Co-Ultramicronized Palmitoylethanolamide/Luteolin in a Mouse Model of Autism and in a Case Report of Autism. CNS Neurosci. Ther. 2017, 23, 87–98. [Google Scholar] [CrossRef]
  47. Bortoletto, R.; Comacchio, C.; Garzitto, M.; Piscitelli, F.; Balestrieri, M.; Colizzi, M. Palmitoylethanolamide Supplementation for Human Health: A State-of-the-Art Systematic Review of Randomized Controlled Trials in Patient Populations. Brain Behav. Immun. Health 2025, 43, 100927. [Google Scholar] [CrossRef]
  48. Adamu, A.; Li, S.; Gao, F.; Xue, G. The Role of Neuroinflammation in Neurodegenerative Diseases: Current Understanding and Future Therapeutic Targets. Front. Aging Neurosci. 2024, 16, 1347987. [Google Scholar] [CrossRef]
  49. Petrosino, S.; Cristino, L.; Karsak, M.; Gaffal, E.; Ueda, N.; Tüting, T.; Bisogno, T.; De Filippis, D.; D’Amico, A.; Saturnino, C.; et al. Protective Role of Palmitoylethanolamide in Contact Allergic Dermatitis. Allergy 2010, 65, 698–711. [Google Scholar] [CrossRef]
  50. Amin, K. The Role of Mast Cells in Allergic Inflammation. Respir. Med. 2012, 106, 9–14. [Google Scholar] [CrossRef]
  51. Eberlein, B.; Eicke, C.; Reinhardt, H.-W.; Ring, J. Adjuvant Treatment of Atopic Eczema: Assessment of an Emollient Containing N-Palmitoylethanolamine (ATOPA Study). J. Eur. Acad. Dermatol. Venereol. 2008, 22, 73–82. [Google Scholar] [CrossRef]
  52. Colizzi, M.; Bortoletto, R.; Colli, C.; Bonomo, E.; Pagliaro, D.; Maso, E.; Di Gennaro, G.; Balestrieri, M. Therapeutic Effect of Palmitoylethanolamide in Cognitive Decline: A Systematic Review and Preliminary Meta-Analysis of Preclinical and Clinical Evidence. Front. Psychiatry 2022, 13, 1038122. [Google Scholar] [CrossRef] [PubMed]
  53. Bortoletto, R.; Piscitelli, F.; Candolo, A.; Bhattacharyya, S.; Balestrieri, M.; Colizzi, M. Questioning the Role of Palmitoylethanolamide in Psychosis: A Systematic Review of Clinical and Preclinical Evidence. Front. Psychiatry 2023, 14, 1231710. [Google Scholar] [CrossRef]
  54. Citraro, R.; Russo, E.; Scicchitano, F.; Van Rijn, C.M.; Cosco, D.; Avagliano, C.; Russo, R.; D’Agostino, G.; Petrosino, S.; Guida, F.; et al. Antiepileptic Action of N-Palmitoylethanolamine through CB1 and PPAR-α Receptor Activation in a Genetic Model of Absence Epilepsy. Neuropharmacology 2013, 69, 115–126. [Google Scholar] [CrossRef]
  55. D’Agostino, G.; Russo, R.; Avagliano, C.; Cristiano, C.; Meli, R.; Calignano, A. Palmitoylethanolamide Protects against the Amyloid-Β25-35-Induced Learning and Memory Impairment in Mice, an Experimental Model of Alzheimer Disease. Neuropsychopharmacology 2012, 37, 1784–1792. [Google Scholar] [CrossRef] [PubMed]
  56. Hauer, D.; Schelling, G.; Gola, H.; Campolongo, P.; Morath, J.; Roozendaal, B.; Hamuni, G.; Karabatsiakis, A.; Atsak, P.; Vogeser, M.; et al. Plasma Concentrations of Endocannabinoids and Related Primary Fatty Acid Amides in Patients with Post-Traumatic Stress Disorder. PLoS ONE 2013, 8, e62741. [Google Scholar] [CrossRef] [PubMed]
  57. Hill, M.N.; Miller, G.E.; Carrier, E.J.; Gorzalka, B.B.; Hillard, C.J. Circulating Endocannabinoids and N-Acyl Ethanolamines Are Differentially Regulated in Major Depression and Following Exposure to Social Stress. Psychoneuroendocrinology 2009, 34, 1257–1262. [Google Scholar] [CrossRef]
  58. Bassir Nia, A.; Bender, R.; Harpaz-Rotem, I. Endocannabinoid System Alterations in Posttraumatic Stress Disorder: A Review of Developmental and Accumulative Effects of Trauma. Chronic Stress 2019, 3. [Google Scholar] [CrossRef]
  59. Berardi, A.; Schelling, G.; Campolongo, P. The Endocannabinoid System and Post Traumatic Stress Disorder (PTSD): From Preclinical Findings to Innovative Therapeutic Approaches in Clinical Settings. Pharmacol. Res. 2016, 111, 668–678. [Google Scholar] [CrossRef]
  60. Morena, M.; Patel, S.; Bains, J.S.; Hill, M.N. Neurobiological Interactions Between Stress and the Endocannabinoid System. Neuropsychopharmacology 2016, 41, 80–102. [Google Scholar] [CrossRef]
  61. Hill, M.N.; Bierer, L.M.; Makotkine, I.; Golier, J.A.; Galea, S.; McEwen, B.S.; Hillard, C.J.; Yehuda, R. Reductions in Circulating Endocannabinoid Levels in Individuals with Post-Traumatic Stress Disorder Following Exposure to the World Trade Center Attacks. Psychoneuroendocrinology 2013, 38, 2952–2961. [Google Scholar] [CrossRef] [PubMed]
  62. De Gregorio, D.; Manchia, M.; Carpiniello, B.; Valtorta, F.; Nobile, M.; Gobbi, G.; Comai, S. Role of Palmitoylethanolamide (PEA) in Depression: Translational Evidence. J. Affect. Disord. 2019, 255, 195–200. [Google Scholar] [CrossRef] [PubMed]
  63. Guida, F.; Luongo, L.; Boccella, S.; Giordano, M.E.; Romano, R.; Bellini, G.; Manzo, I.; Furiano, A.; Rizzo, A.; Imperatore, R.; et al. Palmitoylethanolamide Induces Microglia Changes Associated with Increased Migration and Phagocytic Activity: Involvement of the CB2 Receptor. Sci. Rep. 2017, 7, 375. [Google Scholar] [CrossRef]
  64. Cristiano, C.; Pirozzi, C.; Coretti, L.; Cavaliere, G.; Lama, A.; Russo, R.; Lembo, F.; Mollica, M.P.; Meli, R.; Calignano, A.; et al. Palmitoylethanolamide Counteracts Autistic-like Behaviours in BTBR T+tf/J Mice: Contribution of Central and Peripheral Mechanisms. Brain Behav. Immun. 2018, 74, 166–175. [Google Scholar] [CrossRef]
  65. Bortoletto, R.; Piscitelli, F.; Basaldella, M.; Scipioni, C.; Comacchio, C.; Fiorino, R.; Fornasaro, S.; Barbieri, P.; Pagliaro, D.; Sepulcri, O.; et al. Assessing the Biobehavioral Effects of Ultramicronized-Palmitoylethanolamide Monotherapy in Autistic Adults with Different Severity Levels: A Report of Two Cases. Front. Psychiatry 2024, 15, 1463849. [Google Scholar] [CrossRef]
  66. Ardizzone, A.; Fusco, R.; Casili, G.; Lanza, M.; Impellizzeri, D.; Esposito, E.; Cuzzocrea, S. Effect of Ultra-Micronized-Palmitoylethanolamide and Acetyl-l-Carnitine on Experimental Model of Inflammatory Pain. Int. J. Mol. Sci. 2021, 22, 1967. [Google Scholar] [CrossRef] [PubMed]
  67. de Sá, M.C.I.; Castor, M.G.M. Therapeutic Use of Palmitoylethanolamide as an Anti-Inflammatory and Immunomodulator. Future Pharmacol. 2023, 3, 951–977. [Google Scholar] [CrossRef]
  68. Skaper, S.D.; Facci, L.; Barbierato, M.; Zusso, M.; Bruschetta, G.; Impellizzeri, D.; Cuzzocrea, S.; Giusti, P. N-Palmitoylethanolamine and Neuroinflammation: A Novel Therapeutic Strategy of Resolution. Mol. Neurobiol. 2015, 52, 1034–1042. [Google Scholar] [CrossRef]
  69. D’Angelo, M.; Steardo, L. Cannabinoids and Sleep: Exploring Biological Mechanisms and Therapeutic Potentials. Int. J. Mol. Sci. 2024, 25, 3603. [Google Scholar] [CrossRef]
  70. Di Cesare Mannelli, L.; D’Agostino, G.; Pacini, A.; Russo, R.; Zanardelli, M.; Ghelardini, C.; Calignano, A. Palmitoylethanolamide Is a Disease-Modifying Agent in Peripheral Neuropathy: Pain Relief and Neuroprotection Share a PPAR-Alpha-Mediated Mechanism. Mediat. Inflamm. 2013, 2013, 1–12. [Google Scholar] [CrossRef]
  71. Fang, X.-X.; Zhai, M.-N.; Zhu, M.; He, C.; Wang, H.; Wang, J.; Zhang, Z.-J. Inflammation in Pathogenesis of Chronic Pain: Foe and Friend. Mol. Pain 2023, 19, 541–550. [Google Scholar] [CrossRef] [PubMed]
  72. Skaper, S.D.; Facci, L.; Giusti, P. Glia and Mast Cells as Targets for Palmitoylethanolamide, an Anti-Inflammatory and Neuroprotective Lipid Mediator. Mol. Neurobiol. 2013, 48, 340–352. [Google Scholar] [CrossRef]
  73. Esposito, G.; Capoccia, E.; Turco, F.; Palumbo, I.; Lu, J.; Steardo, A.; Cuomo, R.; Sarnelli, G.; Steardo, L. Palmitoylethanolamide Improves Colon Inflammation through an Enteric Glia/Toll like Receptor 4-Dependent PPAR-α Activation. Gut 2014, 63, 1300–1312. [Google Scholar] [CrossRef]
  74. Manzanares, J.; Julian, M.D.; Carrascosa, A. Role of the Cannabinoid System in Pain Control and Therapeutic Implications for the Management of Acute and Chronic Pain Episodes. Curr. Neuropharmacol. 2006, 4, 239–257. [Google Scholar] [CrossRef] [PubMed]
  75. Giugliano, E.; Cagnazzo, E.; Soave, I.; Lo Monte, G.; Wenger, J.M.; Marci, R. The Adjuvant Use of N-Palmitoylethanolamine and Transpolydatin in the Treatment of Endometriotic Pain. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 168, 209–213. [Google Scholar] [CrossRef] [PubMed]
  76. Luongo, L.; Guida, F.; Boccella, S.; Bellini, G.; Gatta, L.; Rossi, F.; de Novellis, V.; Maione, S. Palmitoylethanolamide Reduces Formalin-Induced Neuropathic-Like Behaviour Through Spinal Glial/Microglial Phenotypical Changes in Mice. CNS Neurol. Disord. Drug Targets 2013, 12, 45–54. [Google Scholar] [CrossRef]
  77. Steels, E.; Venkatesh, R.; Steels, E.; Vitetta, G.; Vitetta, L. A Double-Blind Randomized Placebo Controlled Study Assessing Safety, Tolerability and Efficacy of Palmitoylethanolamide for Symptoms of Knee Osteoarthritis. Inflammopharmacology 2019, 27, 475–485. [Google Scholar] [CrossRef]
  78. van de Laar, M.; Pergolizzi, J.V.; Mellinghoff, H.-U.; Merchante, I.M.; Nalamachu, S.; O’Brien, J.; Perrot, S.; Raffa, R.B. Pain Treatment in Arthritis-Related Pain: Beyond NSAIDs. Open Rheumatol. J. 2012, 6, 320–330. [Google Scholar] [CrossRef]
  79. Sohail, R.; Mathew, M.; Patel, K.K.; Reddy, S.A.; Haider, Z.; Naria, M.; Habib, A.; Abdin, Z.U.; Razzaq Chaudhry, W.; Akbar, A. Effects of Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) and Gastroprotective NSAIDs on the Gastrointestinal Tract: A Narrative Review. Cureus 2023, 15, e37080. [Google Scholar] [CrossRef]
  80. Paladini, A.; Fusco, M.; Cenacchi, T.; Schievano, C.; Piroli, A.; Varrassi, G. Palmitoylethanolamide, a Special Food for Medical Purposes, in the Treatment of Chronic Pain: A Pooled Data Meta-Analysis. Pain. Physician 2016, 19, 11–24. [Google Scholar]
  81. Schweiger, V.; Martini, A.; Bellamoli, P.; Donadello, K.; Schievano, C.; Balzo, G.D.; Sarzi-Puttini, P.; Parolini, M.; Polati, E. Ultramicronized Palmitoylethanolamide (Um-PEA) as Add-on Treatment in Fibromyalgia Syndrome (FMS): Retrospective Observational Study on 407 Patients. CNS Neurol. Disord. Drug Targets 2019, 18, 326–333. [Google Scholar] [CrossRef] [PubMed]
  82. Straube, A.; Andreou, A. Primary Headaches during Lifespan. J. Headache Pain 2019, 20, 35. [Google Scholar] [CrossRef] [PubMed]
  83. Rizzoli, P.; Mullally, W.J. Headache. Am. J. Med. 2018, 131, 17–24. [Google Scholar] [CrossRef]
  84. Bendtsen, L. Central Sensitization in Tension-Type Headache—Possible Pathophysiological Mechanisms. Cephalalgia 2000, 20, 486–508. [Google Scholar] [CrossRef] [PubMed]
  85. Stovner, L.J.; Nichols, E.; Steiner, T.J.; Abd-Allah, F.; Abdelalim, A.; Al-Raddadi, R.M.; Ansha, M.G.; Barac, A.; Bensenor, I.M.; Doan, L.P.; et al. Global, Regional, and National Burden of Migraine and Tension-Type Headache, 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018, 17, 954–976. [Google Scholar] [CrossRef]
  86. Andreou, A.P.; Edvinsson, L. Mechanisms of Migraine as a Chronic Evolutive Condition. J. Headache Pain 2019, 20, 117. [Google Scholar] [CrossRef]
  87. Gatti, A.; Lazzari, M.; Gianfelice, V.; Di Paolo, A.; Sabato, E.; Sabato, A.F. Palmitoylethanolamide in the Treatment of Chronic Pain Caused by Different Etiopathogenesis. Pain Med. 2012, 13, 1121–1130. [Google Scholar] [CrossRef]
  88. Chirchiglia, D.; Cione, E.; Caroleo, M.C.; Wang, M.; Di Mizio, G.; Faedda, N.; Giacolini, T.; Siviglia, S.; Guidetti, V.; Gallelli, L. Effects of Add-On Ultramicronized N-Palmitol Ethanol Amide in Patients Suffering of Migraine With Aura: A Pilot Study. Front. Neurol. 2018, 9, 674. [Google Scholar] [CrossRef]
  89. Papetti, L.; Sforza, G.; Tullo, G.; Alaimo di Loro, P.; Moavero, R.; Ursitti, F.; Ferilli, M.A.N.; Tarantino, S.; Vigevano, F.; Valeriani, M. Tolerability of Palmitoylethanolamide in a Pediatric Population Suffering from Migraine: A Pilot Study. Pain Res. Manag. 2020, 2020, 1–7. [Google Scholar] [CrossRef]
  90. Killgore, W.D.S. Effects of Sleep Deprivation on Cognition. Prog. Brain Res. 2010, 185, 105–129. [Google Scholar]
  91. Hermesdorf, M.; Berger, K.; Baune, B.T.; Wellmann, J.; Ruscheweyh, R.; Wersching, H. Pain Sensitivity in Patients With Major Depression: Differential Effect of Pain Sensitivity Measures, Somatic Cofactors, and Disease Characteristics. J. Pain. 2016, 17, 606–616. [Google Scholar] [CrossRef] [PubMed]
  92. Medic, G.; Wille, M.; Hemels, M.E. Short- and Long-Term Health Consequences of Sleep Disruption. Nat. Sci. Sleep 2017, 9, 151–161. [Google Scholar] [CrossRef] [PubMed]
  93. Kalmbach, D.A.; Anderson, J.R.; Drake, C.L. The Impact of Stress on Sleep: Pathogenic Sleep Reactivity as a Vulnerability to Insomnia and Circadian Disorders. J. Sleep Res. 2018, 27, e12710. [Google Scholar] [CrossRef] [PubMed]
  94. Pogorzelski, R.; Kułakowska, A.; Halicka, D.; Drozdowski, W. Neurological and Emotional Profile of Carpal Tunnel Syndrome Patients. Przegl Lek. 2011, 68, 269–273. [Google Scholar]
  95. Jerosch-Herold, C.; Houghton, J.; Blake, J.; Shaikh, A.; Wilson, E.C.; Shepstone, L. Association of Psychological Distress, Quality of Life and Costs with Carpal Tunnel Syndrome Severity: A Cross-Sectional Analysis of the PALMS Cohort. BMJ Open 2017, 7, e017732. [Google Scholar] [CrossRef]
  96. Kaul, M.; Zee, P.C.; Sahni, A.S. Effects of Cannabinoids on Sleep and Their Therapeutic Potential for Sleep Disorders. Neurotherapeutics 2021, 18, 217–227. [Google Scholar] [CrossRef]
  97. Leinen, Z.J.; Mohan, R.; Premadasa, L.S.; Acharya, A.; Mohan, M.; Byrareddy, S.N. Therapeutic Potential of Cannabis: A Comprehensive Review of Current and Future Applications. Biomedicines 2023, 11, 2630. [Google Scholar] [CrossRef]
  98. Rao, A.; Ebelt, P.; Mallard, A.; Briskey, D. Palmitoylethanolamide for Sleep Disturbance. A Double-Blind, Randomised, Placebo-Controlled Interventional Study. Sleep. Sci. Pract. 2021, 5, 12. [Google Scholar] [CrossRef]
  99. Evangelista, M.; Cilli; De Vitis, R.; Militerno, A.; Fanfani, F. Ultra-Micronized Palmitoylethanolamide Effects on Sleep-Wake Rhythm and Neuropathic Pain Phenotypes in Patients with Carpal Tunnel Syndrome: An Open-Label, Randomized Controlled Study. CNS Neurol. Disord. Drug Targets 2018, 17, 291–298. [Google Scholar] [CrossRef]
  100. Murillo-Rodríguez, E.; Budde, H.; Veras, A.B.; Rocha, N.B.; Telles-Correia, D.; Monteiro, D.; Cid, L.; Yamamoto, T.; Machado, S.; Torterolo, P. The Endocannabinoid System May Modulate Sleep Disorders in Aging. Curr. Neuropharmacol. 2020, 18, 97–108. [Google Scholar] [CrossRef]
  101. Cocito, D.; Peci, E.; Torrieri, M.C.; Clerico, M. Ultramicronized Palmitoylethanolamide in the Management of Neuropathic Pain Related to Chronic Inflammatory Demyelinating Polyneuropathy: A Proof-of-Concept Study. J. Clin. Med. 2024, 13, 2787. [Google Scholar] [CrossRef]
  102. Iannotti, F.A.; Vitale, R.M. The Endocannabinoid System and PPARs: Focus on Their Signalling Crosstalk, Action and Transcriptional Regulation. Cells 2021, 10, 586. [Google Scholar] [CrossRef] [PubMed]
  103. Tai, F.W.D.; McAlindon, M.E. Non-Steroidal Anti-Inflammatory Drugs and the Gastrointestinal Tract. Clin. Med. 2021, 21, 131–134. [Google Scholar] [CrossRef]
  104. Gabrielsson, L.; Mattsson, S.; Fowler, C.J. Palmitoylethanolamide for the Treatment of Pain: Pharmacokinetics, Safety and Efficacy. Br. J. Clin. Pharmacol. 2016, 82, 932–942. [Google Scholar] [CrossRef] [PubMed]
  105. Lang-Illievich, K.; Klivinyi, C.; Lasser, C.; Brenna, C.T.A.; Szilagyi, I.S.; Bornemann-Cimenti, H. Palmitoylethanolamide in the Treatment of Chronic Pain: A Systematic Review and Meta-Analysis of Double-Blind Randomized Controlled Trials. Nutrients 2023, 15, 1350. [Google Scholar] [CrossRef]
  106. Blake, D.R.; Robson, P.; Ho, M.; Jubb, R.W.; McCabe, C.S. Preliminary Assessment of the Efficacy, Tolerability and Safety of a Cannabis-Based Medicine (Sativex) in the Treatment of Pain Caused by Rheumatoid Arthritis. Rheumatology 2006, 45, 50–52. [Google Scholar] [CrossRef]
  107. Cordaro, M.; Impellizzeri, D.; Paterniti, I.; Bruschetta, G.; Siracusa, R.; De Stefano, D.; Cuzzocrea, S.; Esposito, E. Neuroprotective Effects of Co-UltraPEALut on Secondary Inflammatory Process and Autophagy Involved in Traumatic Brain Injury. J. Neurotrauma 2016, 33, 132–146. [Google Scholar] [CrossRef] [PubMed]
  108. Ahmad, A.; Crupi, R.; Impellizzeri, D.; Campolo, M.; Marino, A.; Esposito, E.; Cuzzocrea, S. Administration of Palmitoylethanolamide (PEA) Protects the Neurovascular Unit and Reduces Secondary Injury after Traumatic Brain Injury in Mice. Brain Behav. Immun. 2012, 26, 1310–1321. [Google Scholar] [CrossRef]
  109. Branković, M.; Gmizić, T.; Dukić, M.; Zdravković, M.; Daskalović, B.; Mrda, D.; Nikolić, N.; Brajković, M.; Gojgić, M.; Lalatović, J.; et al. Therapeutic Potential of Palmitoylethanolamide in Gastrointestinal Disorders. Antioxidants 2024, 13, 600. [Google Scholar] [CrossRef]
  110. Del Re, A.; Corpetti, C.; Pesce, M.; Seguella, L.; Steardo, L.; Palenca, I.; Rurgo, S.; De Conno, B.; Sarnelli, G.; Esposito, G. Ultramicronized Palmitoylethanolamide Inhibits NLRP3 Inflammasome Expression and Pro-Inflammatory Response Activated by SARS-CoV-2 Spike Protein in Cultured Murine Alveolar Macrophages. Metabolites 2021, 11, 592. [Google Scholar] [CrossRef]
  111. D’Amico, R.; Impellizzeri, D.; Cuzzocrea, S.; Di Paola, R. ALIAmides Update: Palmitoylethanolamide and Its Formulations on Management of Peripheral Neuropathic Pain. Int. J. Mol. Sci. 2020, 21, 5330. [Google Scholar] [CrossRef] [PubMed]
  112. Schweiger, V.; Schievano, C.; Martini, A.; Polati, L.; Del Balzo, G.; Simari, S.; Milan, B.; Finco, G.; Varrassi, G.; Polati, E. Extended Treatment with Micron-Size Oral Palmitoylethanolamide (PEA) in Chronic Pain: A Systematic Review and Meta-Analysis. Nutrients 2024, 16, 1653. [Google Scholar] [CrossRef] [PubMed]
  113. Impellizzeri, D.; Esposito, E.; Di Paola, R.; Ahmad, A.; Campolo, M.; Peli, A.; Morittu, V.M.; Britti, D.; Cuzzocrea, S. Palmitoylethanolamide and Luteolin Ameliorate Development of Arthritis Caused by Injection of Collagen Type II in Mice. Arthritis Res. Ther. 2013, 15, 4055. [Google Scholar] [CrossRef] [PubMed]
Table 1. Comparison of standard PEA and umPEA: key pharmacological and clinical differences.
Table 1. Comparison of standard PEA and umPEA: key pharmacological and clinical differences.
AspectStandard PEAUltramicronized PEA (umPEA)
Particle SizeLarge particles (>100 µm)Reduced particles (<10 µm)
BioavailabilityLow oral bioavailability, slow absorptionEnhanced oral bioavailability, rapid absorption
Blood–brain barrier penetrationLimited CNS penetrationImproved CNS penetration
PharmacokineticsLower peak plasma and tissue concentrationsHigher peak plasma and tissue concentrations
Onset of ActionSlower onsetFaster onset
Therapeutic EfficacyModerate efficacySuperior efficacy
Dosage RequirementsHigher required dosagesLower effective dosages
Clinical OutcomesModerate reduction in inflammation and painGreater reduction in inflammation, pain severity, and improved neurological outcomes
Clinical EvidenceSupported by preclinical studies and limited clinical trials [3,80]Strongly supported by robust clinical and meta-analytic evidence [25,29,81]
PEA: palmitoylethanolamide; umPEA: ultramicronized palmitoylethanolamide; CNS: central nervous system; BBB: blood–brain barrier; BID: bis in die (twice daily).
Table 2. Clinical indications.
Table 2. Clinical indications.
Clinical ConditionRecommended Dose (mg/Day)Therapeutic RationaleReferences
Neuropathic Pain and Fibromyalgia600 mg BIDEnhances analgesia, reduces central sensitizationPaladini et al., 2016 [80]; Gabrielsson et al., 2016 [104]; Schweiger et al., 2019 [81]
Autoimmune and Inflammatory Diseases600–1200 mgRegulates immune overactivation, supports toleranceSteels et al., 2019 [77]; Blake et al., 2006 [106]
Alzheimer’s and Parkinson’s Disease600–1200 mgNeuroprotection, slows progression, cognitive supportScuderi et al., 2018 [25]; Beggiato et al., 2019 [5]
Stroke and Traumatic Brain Injury600 mg BIDReduces neuroinflammation, promotes recoveryCordaro et al., 2016 [107]; Ahmad et al., 2012 [108]
Leaky Gut and Microbiome Support300–600 mgRestores gut barrier, modulates microbiotaEsposito et al., 2014; [73] Branković et al., 2024 [109]
Viral Infections and Immune Support600–1200 mgEnhances innate immunity, reduces inflammationRe et al., 2021 [110]
Table 3. Clinical doses, therapeutic indications, and observed outcomes with umPEA in clinical practice.
Table 3. Clinical doses, therapeutic indications, and observed outcomes with umPEA in clinical practice.
Clinical ConditionRecommended DosageTherapeutic ContextMain OutcomesModel TypeMechanisms InvestigatedReferences
Neuropathic Pain600–1200 mg/day (divided into 2 daily doses)Peripheral neuropathy, diabetic neuropathy, sciatic painReduction in pain severity, improved quality of life, decreased analgesic usageClinical (human)Anti-inflammatory, analgesic, mast cell stabilizationPaladini et al., 2016 [80]; Gabrielsson et al., 2016 [104]
Fibromyalgia600–1200 mg/dayChronic widespread pain, fatigue, muscle tendernessReduced pain intensity, improved sleep quality, reduced fatiguePreclinical (mouse model of Alzheimer’s)Anti-inflammatory, neuroprotective, glial modulationSchweiger et al., 2019 [81]
Sciatica and Low Back Pain600 mg twice dailyRadicular pain, chronic low back painSignificant reduction in pain intensity, improved functional recoveryPreclinical (rat model of Parkinson’s)Neuroprotection, antioxidant activityPaladini et al., 2016 [80]; D’Amico et al., 2020 [111]
Pelvic Pain (Endometriosis)600–1200 mg/dayChronic pelvic pain, dysmenorrhea, endometriosisPain relief, reduced inflammation, improved daily activitiesPreclinical (Alzheimer’s triple transgenic mouse)Neuroinflammation modulation, astroglial regulationGiugliano et al., 2013 [75]
Osteoarthritis600 mg twice dailyKnee osteoarthritis, joint inflammation, and painReduction in pain and joint stiffness, improved joint functionClinical (human, endometriosis)Pain modulation, anti-inflammatory actionSteels et al., 2019 [77]
Migraine and Primary Headache1200 mg/dayMigraine, tension-type headacheDecreased attack frequency, reduced duration and intensity of headaches, reduced analgesic consumptionClinical (human)Analgesia, mast cell stabilizationLang-Illievich et al., 2023 [105]; Gabrielsson et al., 2016 [104]
Neurodegenerative Disorders (Alzheimer’s, Parkinson’s)600–1200 mg/dayEarly cognitive decline, motor impairmentNeuroinflammation reduction, cognitive stabilization, improved motor functionClinical (human)Anti-inflammatory, pain reliefScuderi et al., 2018 [25]; Beggiato et al., 2019 [5]
Postoperative Pain600 mg twice dailyPost-surgical inflammation, pain managementReduced post-operative pain severity, reduced reliance on opioid analgesicsClinical (human)Neuropathic pain reliefPaladini et al., 2016 [80]
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

Di Stefano, V.; Steardo, L., Jr.; D’Angelo, M.; Monaco, F.; Steardo, L. Palmitoylethanolamide: A Multifunctional Molecule for Neuroprotection, Chronic Pain, and Immune Modulation. Biomedicines 2025, 13, 1271. https://doi.org/10.3390/biomedicines13061271

AMA Style

Di Stefano V, Steardo L Jr., D’Angelo M, Monaco F, Steardo L. Palmitoylethanolamide: A Multifunctional Molecule for Neuroprotection, Chronic Pain, and Immune Modulation. Biomedicines. 2025; 13(6):1271. https://doi.org/10.3390/biomedicines13061271

Chicago/Turabian Style

Di Stefano, Valeria, Luca Steardo, Jr., Martina D’Angelo, Francesco Monaco, and Luca Steardo. 2025. "Palmitoylethanolamide: A Multifunctional Molecule for Neuroprotection, Chronic Pain, and Immune Modulation" Biomedicines 13, no. 6: 1271. https://doi.org/10.3390/biomedicines13061271

APA Style

Di Stefano, V., Steardo, L., Jr., D’Angelo, M., Monaco, F., & Steardo, L. (2025). Palmitoylethanolamide: A Multifunctional Molecule for Neuroprotection, Chronic Pain, and Immune Modulation. Biomedicines, 13(6), 1271. https://doi.org/10.3390/biomedicines13061271

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