Therapeutic effect of palmitoylethanolamide in cognitive decline: A systematic review and preliminary meta-analysis of preclinical and clinical evidence

Marco Colizzi, Riccardo Bortoletto, Chiara Colli, Enrico Bonomo, Daniele Pagliaro, Elisa Maso, Gianfranco Di Gennaro, Matteo Balestrieri, Marco Colizzi, Riccardo Bortoletto, Chiara Colli, Enrico Bonomo, Daniele Pagliaro, Elisa Maso, Gianfranco Di Gennaro, Matteo Balestrieri

Abstract

Cognitive decline is believed to be associated with neurodegenerative processes involving excitotoxicity, oxidative damage, inflammation, and microvascular and blood-brain barrier dysfunction. Interestingly, research evidence suggests upregulated synthesis of lipid signaling molecules as an endogenous attempt to contrast such neurodegeneration-related pathophysiological mechanisms, restore homeostatic balance, and prevent further damage. Among these naturally occurring molecules, palmitoylethanolamide (PEA) has been independently associated with neuroprotective and anti-inflammatory properties, raising interest into the possibility that its supplementation might represent a novel therapeutic approach in supporting the body-own regulation of many pathophysiological processes potentially contributing to neurocognitive disorders. Here, we systematically reviewed all human and animal studies examining PEA and its biobehavioral correlates in neurocognitive disorders, finding 33 eligible outputs. Studies conducted in animal models of neurodegeneration indicate that PEA improves neurobehavioral functions, including memory and learning, by reducing oxidative stress and pro-inflammatory and astrocyte marker expression as well as rebalancing glutamatergic transmission. PEA was found to promote neurogenesis, especially in the hippocampus, neuronal viability and survival, and microtubule-associated protein 2 and brain-derived neurotrophic factor expression, while inhibiting mast cell infiltration/degranulation and astrocyte activation. It also demonstrated to mitigate β-amyloid-induced astrogliosis, by modulating lipid peroxidation, protein nytrosylation, inducible nitric oxide synthase induction, reactive oxygen species production, caspase3 activation, amyloidogenesis, and tau protein hyperphosphorylation. Such effects were related to PEA ability to indirectly activate cannabinoid receptors and modulate proliferator-activated receptor-α (PPAR-α) activity. Importantly, preclinical evidence suggests that PEA may act as a disease-modifying-drug in the early stage of a neurocognitive disorder, while its protective effect in the frank disorder may be less relevant. Limited human research suggests that PEA supplementation reduces fatigue and cognitive impairment, the latter being also meta-analytically confirmed in 3 eligible studies. PEA improved global executive function, working memory, language deficits, daily living activities, possibly by modulating cortical oscillatory activity and GABAergic transmission. There is currently no established cure for neurocognitive disorders but only treatments to temporarily reduce symptom severity. In the search for compounds able to protect against the pathophysiological mechanisms leading to neurocognitive disorders, PEA may represent a valid therapeutic option to prevent neurodegeneration and support endogenous repair processes against disease progression.

Keywords: Alzheimer's disease; Parkinson's disease; acylethanolamines; cannabinoids; dementia; immune response; neurocognitive disorder.

Conflict of interest statement

Author MC has been a consultant/advisor to GW Pharma Limited, GW Pharma Italy SRL and F. Hoffmann-La Roche Limited, outside of this work. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Colizzi, Bortoletto, Colli, Bonomo, Pagliaro, Maso, Di Gennaro and Balestrieri.

Figures

Figure 1
Figure 1
PRISMA flowchart of search strategy for systematic review.
Figure 2
Figure 2
Forest plot showing the pooled Mini-Mental State Examination (MMSE) change from baseline. Homogeneity (I-squared): 98.40%, p < 0.001; Estimation by DerSimonian and Laird random-effects model.

References

    1. Shinosaki K, Nishikawa T, Takeda M. Neurobiological basis of behavioral and psychological symptoms in dementia of the Alzheimer type. Psychiatry Clin Neurosci. (2000) 54:611–20. 10.1046/j.1440-1819.2000.00773.x
    1. Halliday GM, Leverenz JB, Schneider JS, Adler CH. The neurobiological basis of cognitive impairment in Parkinson's disease. Mov Disord. (2014) 29:634–50. 10.1002/mds.25857
    1. Young JJ, Lavakumar M, Tampi D, Balachandran S, Tampi RR. Frontotemporal dementia: latest evidence and clinical implications. Ther Adv Psychopharmacol. (2018) 8:33–48. 10.1177/2045125317739818
    1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Washington, DC: American Psychiatric Association; (2013).
    1. Fereshtehnejad SM, Vosoughi K, Heydarpour P, Sepanlou SG, Farzadfar F, Tehrani-Banihashemi A, et al. . Burden of neurodegenerative diseases in the Eastern Mediterranean Region, 1990-2016: findings from the Global Burden of Disease Study 2016. Eur J Neurol. (2019) 26:1252–65. 10.1111/ene.13972
    1. Collaborators GD. Global, regional, and national burden of Alzheimer's disease and other dementias, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. (2019) 18:88–106. 10.1016/S1474-4422(18)30403-4
    1. Collaborators GDF. Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: an analysis for the Global Burden of Disease Study 2019. Lancet Public Health. (2022) 7:e105–e25. 10.1016/S2468-2667(21)00249-8
    1. Kempuraj D, Thangavel R, Natteru PA, Selvakumar GP, Saeed D, Zahoor H, et al. . Neuroinflammation Induces Neurodegeneration. J Neurol Neurosurg Spine. (2016) 1:1003–17.
    1. Kwon HS, Koh SH. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl Neurodegener. (2020) 9:42. 10.1186/s40035-020-00221-2
    1. Moss DE. Improving Anti-Neurodegenerative Benefits of Acetylcholinesterase Inhibitors in Alzheimer's Disease: Are Irreversible Inhibitors the Future? Int J Mol Sci. (2020) 21:3438–55. 10.3390/ijms21103438
    1. Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG, Ramos-Escobar N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front Pharmacol. (2019) 10:1008. 10.3389/fphar.2019.01008
    1. Kasatkina LA, Rittchen S, Sturm EM. Neuroprotective and immunomodulatory action of the endocannabinoid system under neuroinflammation. Int J Mol Sci. (2021) 22:5431. 10.3390/ijms22115431
    1. Di Marzo V. 'Endocannabinoids' and other fatty acid derivatives with cannabimimetic properties: biochemistry and possible physiopathological relevance. Biochim Biophys Acta. (1998) 1392:153–75. 10.1016/S0005-2760(98)00042-3
    1. Rankin L, Fowler CJ. The Basal Pharmacology of Palmitoylethanolamide. Int J Mol Sci. (2020) 21:7942–62. 10.3390/ijms21217942
    1. Bortoletto R, Balestrieri M, Bhattacharyya S, Colizzi M. Is It Time to Test the Antiseizure Potential of Palmitoylethanolamide in Human Studies? A systematic review of preclinical evidence. Brain Sci. (2022) 12:101. 10.3390/brainsci12010101
    1. Lo Verme J, Fu J, Astarita G, La Rana G, Russo R, Calignano A, et al. . The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol Pharmacol. (2005) 67:15–9. 10.1124/mol.104.006353
    1. Solorzano C, Zhu C, Battista N, Astarita G, Lodola A, Rivara S, et al. . Selective N-acylethanolamine-hydrolyzing acid amidase inhibition reveals a key role for endogenous palmitoylethanolamide in inflammation. Proc Natl Acad Sci U S A. (2009) 106:20966–71. 10.1073/pnas.0907417106
    1. Jaggar SI, Hasnie FS, Sellaturay S, Rice AS. The anti-hyperalgesic actions of the cannabinoid anandamide and the putative CB2 receptor agonist palmitoylethanolamide in visceral and somatic inflammatory pain. Pain. (1998) 76:189–99. 10.1016/S0304-3959(98)00041-4
    1. Pesce M, Seguella L, Cassarano S, Aurino L, Sanseverino W, Lu J, et al. . Phytotherapics in COVID19: Why palmitoylethanolamide? Phytother Res. (2021) 35:2514–22. 10.1002/ptr.6978
    1. Scuderi C, Bronzuoli M, Facchinetti R, Pace L, Ferraro L, Broad K, 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. 10.1038/s41398-017-0076-4
    1. 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) 174:1349–65. 10.1111/bph.13580
    1. Paterniti I, Cordaro M, Campolo M, Siracusa R, Cornelius C, Navarra M, et al. . Neuroprotection by association of palmitoylethanolamide with luteolin in experimental Alzheimer's disease models: the control of neuroinflammation. CNS Neurol Disord Drug Targets. (2014) 13:1530–41. 10.2174/1871527313666140806124322
    1. Altamura C, Ventriglia M, Martini MG, Montesano D, Errante Y, Piscitelli F, et al. . Elevation of Plasma 2-Arachidonoylglycerol Levels in Alzheimer's Disease Patients as a Potential Protective Mechanism against Neurodegenerative Decline. J Alzheimers Dis. (2015) 46:497–506. 10.3233/JAD-142349
    1. Caltagirone C, Cisari C, Schievano C, Di Paola R, Cordaro M, Bruschetta G, et al. . Co-ultramicronized Palmitoylethanolamide/Luteolin in the Treatment of Cerebral Ischemia: from Rodent to Man. Transl Stroke Res. (2016) 7:54–69. 10.1007/s12975-015-0440-8
    1. Cipriano M, Esposito G, Negro L, Capoccia E, Sarnelli G, Scuderi C, et al. . Palmitoylethanolamide Regulates Production of Pro-Angiogenic Mediators in a Model of beta Amyloid-Induced Astrogliosis In vitro. CNS Neurol Disord Drug Targets. (2015) 14:828–37. 10.2174/1871527314666150317224155
    1. Brotini S, Schievano C, Guidi L. Ultra-micronized palmitoylethanolamide: an efficacious adjuvant therapy for Parkinson's Disease. CNS Neurol Disord Drug Targets. (2017) 16:705–13. 10.2174/1871527316666170321124949
    1. Assogna M, Casula EP, Borghi I, Bonnì S, Samà D, Motta C, et al. . Effects of palmitoylethanolamide combined with luteoline on frontal lobe functions, high frequency oscillations, and GABAergic transmission in patients with frontotemporal dementia. J Alzheimers Dis. (2020) 76:1297–308. 10.3233/JAD-200426
    1. Campolo M, Crupi R, Cordaro M, Cardali SM, Ardizzone A, Casili G, et al. . Co-Ultra PEALut enhances endogenous repair response following moderate traumatic brain injury. Int J Mol Sci. (2021) 22:8717. 10.3390/ijms22168717
    1. Scuderi C, Esposito G, Blasio A, Valenza M, Arietti P, Steardo L, et al. . Palmitoylethanolamide counteracts reactive astrogliosis induced by β-amyloid peptide. J Cell Mol Med. (2011) 15:2664–74. 10.1111/j.1582-4934.2011.01267.x
    1. Benito C, Tolón RM, Castillo AI, Ruiz-Valdepeñas L, Martínez-Orgado JA, Fernández-Sánchez FJ, et al. . β-Amyloid exacerbates inflammation in astrocytes lacking fatty acid amide hydrolase through a mechanism involving PPAR-α, PPAR-γ and TRPV1, but not CB1 or CB2 receptors. Br J Pharmacol. (2012) 166:1474–89. 10.1111/j.1476-5381.2012.01889.x
    1. 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) 37:1784–92. 10.1038/npp.2012.25
    1. Scuderi C, Valenza M, Stecca C, Esposito G, Carratù MR, Steardo L. Palmitoylethanolamide exerts neuroprotective effects in mixed neuroglial cultures and organotypic hippocampal slices via peroxisome proliferator-activated receptor-α. J Neuroinflammation. (2012) 9:49. 10.1186/1742-2094-9-49
    1. Scuderi C, Steardo L. Neuroglial roots of neurodegenerative diseases: therapeutic potential of palmitoylethanolamide in models of Alzheimer's disease. CNS Neurol Disord Drug Targets. (2013) 12:62–9. 10.2174/1871527311312010011
    1. Scuderi C, Stecca C, Valenza M, Ratano P, Bronzuoli MR, Bartoli S, et al. . Palmitoylethanolamide controls reactive gliosis and exerts neuroprotective functions in a rat model of Alzheimer's disease. Cell Death Dis. (2014) 5:e1419. 10.1038/cddis.2014.376
    1. Tomasini MC, Borelli AC, Beggiato S, Ferraro L, Cassano T, Tanganelli S, et al. . Differential Effects of Palmitoylethanolamide against Amyloid-β Induced Toxicity in Cortical Neuronal and Astrocytic Primary Cultures from Wild-Type and 3xTg-AD Mice. J Alzheimers Dis. (2015) 46:407–21. 10.3233/JAD-143039
    1. Siracusa R, Impellizzeri D, Cordaro M, Crupi R, Esposito E, Petrosino S, et al. . Anti-Inflammatory and Neuroprotective Effects of Co-UltraPEALut in a Mouse Model of Vascular Dementia. Front Neurol. (2017) 8:233. 10.3389/fneur.2017.00233
    1. Beggiato S, Borelli AC, Ferraro L, Tanganelli S, Antonelli T, Tomasini MC. Palmitoylethanolamide blunts amyloid-β42-induced astrocyte activation and improves neuronal survival in primary mouse cortical astrocyte-neuron co-cultures. J Alzheimers Dis. (2018) 61:389–99. 10.3233/JAD-170699
    1. Bronzuoli M, Facchinetti R, Steardo L, Romano A, Stecca C, Passarella S, et al. . Palmitoylethanolamide dampens reactive astrogliosis and improves neuronal trophic support in a triple transgenic model of Alzheimer's Disease: in vitro and in vivo evidence. Oxid Med Cell Longev. (2018) 2018:14. 10.1155/2018/4720532
    1. Crupi R, Impellizzeri D, Cordaro M, Siracusa R, Casili G, Evangelista M, et al. . N-palmitoylethanolamide Prevents Parkinsonian Phenotypes in Aged Mice. Mol Neurobiol. (2018) 55:8455–72. 10.1007/s12035-018-0959-2
    1. Boccella S, Marabese I, Iannotta M, Belardo C, Neugebauer V, Mazzitelli M, et al. . Metabotropic glutamate receptor 5 and 8 modulate the ameliorative effect of ultramicronized palmitoylethanolamide on cognitive decline associated with neuropathic pain. Int J Mol Sci. (2019) 20:1757–71. 10.3390/ijms20071757
    1. Boccella S, Cristiano C, Romano R, Iannotta M, Belardo C, Farina A, et al. . Ultra-micronized palmitoylethanolamide rescues the cognitive decline-associated loss of neural plasticity in the neuropathic mouse entorhinal cortex-dentate gyrus pathway. Neurobiol Dis. (2019) 121:106–19. 10.1016/j.nbd.2018.09.023
    1. Impellizzeri D, Siracusa R, Cordaro M, Crupi R, Peritore AF, Gugliandolo E, et al. . N-Palmitoylethanolamine-oxazoline (PEA-OXA): A new therapeutic strategy to reduce neuroinflammation, oxidative stress associated to vascular dementia in an experimental model of repeated bilateral common carotid arteries occlusion. Neurobiol Dis. (2019) 125:77–91. 10.1016/j.nbd.2019.01.007
    1. Piscitelli F, Coccurello R, Totaro A, Leuti A, Giacovazzo G, Verde R, et al. . Targeted lipidomics investigation of N-acylethanolamines in a transgenic mouse model of AD: A longitudinal study. Eur J Lipid Sci Technol. (2019) 121:7. 10.1002/ejlt.201900015
    1. Zimmermann T, Bartsch JC, Beer A, Lomazzo E, Guggenhuber S, Lange MD, et al. . Impaired anandamide/palmitoylethanolamide signaling in hippocampal glutamatergic neurons alters synaptic plasticity, learning, and emotional responses. Neuropsychopharmacology. (2019) 44:1377–88. 10.1038/s41386-018-0274-7
    1. Beggiato S, Cassano T, Ferraro L, Tomasini MC. Astrocytic palmitoylethanolamide pre-exposure exerts neuroprotective effects in astrocyte-neuron co-cultures from a triple transgenic mouse model of Alzheimer's disease. Life Sci. (2020) 257:118037. 10.1016/j.lfs.2020.118037
    1. Beggiato S, Tomasini MC, Cassano T, Ferraro L. Chronic oral palmitoylethanolamide administration rescues cognitive deficit and reduces neuroinflammation, oxidative stress, and glutamate levels in A transgenic murine model of Alzheimer's Disease. J Clin Med. (2020) 9:428–46. 10.3390/jcm9020428
    1. Facchinetti R, Valenza M, Bronzuoli M, Menegoni G, Ratano P, Steardo L, et al. . Looking for a treatment for the early stage of alzheimer's disease: preclinical evidence with co-ultramicronized palmitoylethanolamide and luteolin. Int J Mol Sci. (2020) 21:3802–18. 10.3390/ijms21113802
    1. Lama A, Pirozzi C, Annunziata C, Morgese MG, Senzacqua M, Severi I, et al. . Palmitoylethanolamide counteracts brain fog improving depressive-like behaviour in obese mice: Possible role of synaptic plasticity and neurogenesis. Br J Pharmacol. (2021) 178:845–59. 10.1111/bph.15071
    1. Boccella S, Guida F, Iannotta M, Iannotti FA, Infantino R, Ricciardi F, et al. . 2-Pentadecyl-2-oxazoline ameliorates memory impairment and depression-like behaviour in neuropathic mice: possible role of adrenergic alpha2- and H3 histamine autoreceptors. Mol Brain. (2021) 14:28. 10.1186/s13041-020-00724-z
    1. D'Antongiovanni V, Pellegrini C, Antonioli L, Benvenuti L, Di Salvo C, Flori L, et al. . Palmitoylethanolamide counteracts enteric inflammation and bowel motor dysfunctions in a mouse model of Alzheimer's Disease. Front Pharmacol. (2021) 12:748021. 10.3389/fphar.2021.748021
    1. Gaspar JC, Healy C, Ferdousi MI, Roche M, Finn DP. Pharmacological blockade of PPARα exacerbates inflammatory pain-related impairment of spatial memory in rats. Biomedicines. (2021) 9:610. 10.3390/biomedicines9060610
    1. Gatta N, Parente A, Guida F, Maione S, Gentile V. Neuronutraceuticals Modulate Lipopolysaccharide- or Amyloid-beta 1-42 Peptide-Induced Transglutaminase 2 Overexpression as a Marker of Neuroinflammation in Mouse Microglial Cells. Appl Sci. (2021) 11:5718–28. 10.3390/app11125718
    1. Colizzi M, Bortoletto R, Costa R, Zoccante L. Palmitoylethanolamide and Its biobehavioral correlates in autism spectrum disorder: a systematic review of human and animal evidence. Nutrients. (2021) 13:1346. 10.3390/nu13041346
    1. West S, King V, Carey TS, Lohr KN, McKoy N, Sutton SF, et al. . Systems to rate the strength of scientific evidence. Evid Rep Technol Assess. (2002) 1–11.
    1. DerSimonian R, Laird N. Meta-analysis in clinical trials revisited. Contemp Clin Trials. (2015) 45(Pt A):139–45. 10.1016/j.cct.2015.09.002
    1. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. (1975) 12:189–98. 10.1016/0022-3956(75)90026-6
    1. Landolfo E, Cutuli D, Petrosini L, Caltagirone C. Effects of palmitoylethanolamide on neurodegenerative diseases: a review from rodents to humans. Biomolecules. (2022) 12:667. 10.3390/biom12050667
    1. Hampel H, Mesulam MM, Cuello AC, Farlow MR, Giacobini E, Grossberg GT, et al. . The cholinergic system in the pathophysiology and treatment of Alzheimer's disease. Brain. (2018) 141:1917–33. 10.1093/brain/awy132
    1. Yoshiyama Y, Kojima A, Ishikawa C, Arai K. Anti-inflammatory action of donepezil ameliorates tau pathology, synaptic loss, and neurodegeneration in a tauopathy mouse model. J Alzheimers Dis. (2010) 22:295–306. 10.3233/JAD-2010-100681
    1. Citraro R, Russo E, Scicchitano F, van Rijn CM, Cosco D, Avagliano C, et al. . Antiepileptic action of N-palmitoylethanolamine through CB1 and PPAR-α receptor activation in a genetic model of absence epilepsy. Neuropharmacology. (2013) 69:115–26. 10.1016/j.neuropharm.2012.11.017
    1. Abedini T, Hosseyni R, Ghannadi F, Sanjari Moghaddam H, Khodaei Ardakani MR, Talaei A, et al. . Efficacy and safety of palmitoylethanolamide as an adjunctive treatment for acute mania: a randomized, double-blind and placebo-controlled trial. Psychiatry Clin Neurosci. (2022) 76:505–11. 10.1111/pcn.13441
    1. Salehi A, Namaei P, TaghaviZanjani F, Bagheri S, Moradi K, Khodaei Ardakani MR, et al. . Adjuvant palmitoylethanolamide therapy with risperidone improves negative symptoms in patients with schizophrenia: a randomized, double-blinded, placebo-controlled trial. Psychiatry Res. (2022) 316:114737. 10.1016/j.psychres.2022.114737

Source: PubMed

3
Předplatit