Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
2.
Brain Behav Immun ; 102: 110-123, 2022 05.
Article in English | MEDLINE | ID: mdl-35176443

ABSTRACT

High-fat diet (HFD) consumption leads to obesity and a chronic state of low-grade inflammation, named metainflammation. Notably, metainflammation contributes to neuroinflammation due to the increased levels of circulating free fatty acids and cytokines. It indicates a strict interplay between peripheral and central counterparts in the pathogenic mechanisms of obesity-related mood disorders. In this context, the impairment of internal hypothalamic circuitry runs in tandem with the alteration of other brain areas associated with emotional processing (i.e., hippocampus and amygdala). Palmitoylethanolamide (PEA), an endogenous lipid mediator belonging to the N-acylethanolamines family, has been extensively studied for its pleiotropic effects both at central and peripheral level. Our study aimed to elucidate PEA capability in limiting obesity-induced anxiety-like behavior and neuroinflammation-related features in an experimental model of HFD-fed obese mice. PEA treatment promoted an improvement in anxiety-like behavior of obese mice and the systemic inflammation, reducing serum pro-inflammatory mediators (i.e., TNF-α, IL-1ß, MCP-1, LPS). In the amygdala, PEA increased dopamine turnover, as well as GABA levels. PEA also counteracted the overactivation of HPA axis, reducing the expression of hypothalamic corticotropin-releasing hormone and its type 1 receptor. Moreover, PEA attenuated the immunoreactivity of Iba-1 and GFAP and reduced pro-inflammatory pathways and cytokine production in both the hypothalamus and hippocampus. This finding, together with the reduced transcription of mast cell markers (chymase 1 and tryptase ß2) in the hippocampus, indicated the weakening of immune cell activation underlying the neuroprotective effect of PEA. Obesity-driven neuroinflammation was also associated with the disruption of blood-brain barrier (BBB) in the hippocampus. PEA limited the albumin extravasation and restored tight junction transcription modified by HFD. To gain mechanistic insight, we designed an in vitro model of metabolic injury using human neuroblastoma SH-SY5Y cells insulted by a mix of glucosamine and glucose. Here, PEA directly counteracted inflammation and mitochondrial dysfunction in a PPAR-α-dependent manner since the pharmacological blockade of the receptor reverted its effects. Our results strengthen the therapeutic potential of PEA in obesity-related neuropsychiatric comorbidities, controlling neuroinflammation, BBB disruption, and neurotransmitter imbalance involved in behavioral dysfunctions.


Subject(s)
Hypothalamo-Hypophyseal System , Neuroinflammatory Diseases , Amides , Animals , Anxiety/drug therapy , Diet, High-Fat , Ethanolamines , Hypothalamo-Hypophyseal System/metabolism , Inflammation , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/metabolism , Palmitic Acids , Pituitary-Adrenal System/metabolism
3.
Animals (Basel) ; 12(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35158670

ABSTRACT

There is growing evidence that perturbation of the gut microbiome, known as "dysbiosis", is associated with the pathogenesis of human and veterinary diseases that are not restricted to the gastrointestinal tract. In this regard, recent studies have demonstrated that dysbiosis is linked to the pathogenesis of central neuroinflammatory disorders, supporting the existence of the so-called microbiome-gut-brain axis. The endocannabinoid system is a recently recognized lipid signaling system and termed endocannabinoidome monitoring a variety of body responses. Accumulating evidence demonstrates that a profound link exists between the gut microbiome and the endocannabinoidome, with mutual interactions controlling intestinal homeostasis, energy metabolism and neuroinflammatory responses during physiological conditions. In the present review, we summarize the latest data on the microbiome-endocannabinoidome mutual link in health and disease, focalizing the attention on gut dysbiosis and/or altered endocannabinoidome tone that may distort the bidirectional crosstalk between these two complex systems, thus leading to gastrointestinal and metabolic diseases (e.g., idiopathic inflammation, chronic enteropathies and obesity) as well as neuroinflammatory disorders (e.g., neuropathic pain and depression). We also briefly discuss the novel possible dietary interventions based not only on probiotics and/or prebiotics, but also, and most importantly, on endocannabinoid-like modulators (e.g., palmitoylethanolamide) for intestinal health and beyond.

4.
Biomolecules ; 13(1)2022 12 24.
Article in English | MEDLINE | ID: mdl-36671418

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a serious inflammatory lung disorder and a complication of SARS-CoV-2 infection. In patients with severe SARS-CoV-2 infection, the transition to ARDS is principally due to the occurrence of a cytokine storm and an exacerbated inflammatory response. The effectiveness of ultra-micronized palmitoylethanolamide (PEA-um) during the earliest stage of COVID-19 has already been suggested. In this study, we evaluated its protective effects as well as the effectiveness of its congener, 2-pentadecyl-2-oxazoline (PEA-OXA), using in vitro models of acute lung injury. In detail, human lung epithelial cells (A549) activated by polyinosinic-polycytidylic acid (poly-(I:C)) or Transforming Growth Factor-beta (TGF-ß) were treated with PEA-OXA or PEA. The release of IL-6 and the appearance of Epithelial-Mesenchymal Transition (EMT) were measured by ELISA and immunofluorescence assays, respectively. A possible mechanism of action for PEA-OXA and PEA was also investigated. Our results showed that both PEA-OXA and PEA were able to counteract poly-(I:C)-induced IL-6 release, as well as to revert TGF-ß-induced EMT. In addition, PEA was able to produce an "entourage" effect on the levels of the two endocannabinoids AEA and 2-AG, while PEA-OXA only increased PEA endogenous levels, in poly-(I:C)-stimulated A549 cells. These results evidence for the first time the superiority of PEA-OXA over PEA in exerting protective effects and point to PEA-OXA as a new promising candidate in the management of acute lung injury.


Subject(s)
Acute Lung Injury , COVID-19 , Humans , Interleukin-6 , SARS-CoV-2 , Transforming Growth Factor beta , Acute Lung Injury/drug therapy
5.
Cells ; 10(2)2021 02 20.
Article in English | MEDLINE | ID: mdl-33672574

ABSTRACT

Palmitoylethanolamide (PEA) is an endogenous anti-inflammatory lipid mediator and a widely used nutraceutical. In this study, we designed, realized, and tested a drug-carrier conjugate between PEA (the active drug) and glucuronic acid (the carrier). The conjugate, named GLUPEA, was characterized for its capability of increasing PEA levels and exerting anti-inflammatory activity both in vitro and in vivo. GLUPEA treatment, compared to the same concentration of PEA, resulted in higher cellular amounts of PEA and the endocannabinoid 2-arachidonoyl glycerol (2-AG), and increased 2-AG-induced transient receptor potential vanilloid type 1 (TRPV1) channel desensitization to capsaicin. GLUPEA inhibited pro-inflammatory monocyte chemoattractant protein 2 (MCP-2) release from stimulated keratinocytes, and it was almost as efficacious as ultra-micronized PEA at reducing colitis in dinitrobenzene sulfonic acid (DNBS)-injected mice when using the same dose. GLUPEA is a novel pro-drug able to efficiently mimic the anti-inflammatory and endocannabinoid enhancing actions of PEA.


Subject(s)
Amides/pharmacology , Drug Delivery Systems , Ethanolamines/pharmacology , Glucuronic Acid/pharmacology , Palmitic Acids/pharmacology , Amides/chemistry , Amides/therapeutic use , Animals , Arachidonic Acids/metabolism , Calcium/metabolism , Chemokine CCL8/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colon/drug effects , Colon/pathology , Dinitrofluorobenzene/analogs & derivatives , Endocannabinoids/metabolism , Ethanolamines/chemistry , Ethanolamines/therapeutic use , Glucuronic Acid/chemistry , Glucuronic Acid/therapeutic use , Glycerides/metabolism , HEK293 Cells , HaCaT Cells , Humans , Ion Channel Gating/drug effects , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Mice, Inbred ICR , Models, Biological , Palmitic Acids/chemistry , Palmitic Acids/therapeutic use , Peroxidase/metabolism , Poly I-C/pharmacology , TRPV Cation Channels/metabolism
6.
Int J Mol Sci ; 21(24)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333772

ABSTRACT

Neuroinflammation is a physiological response aimed at maintaining the homodynamic balance and providing the body with the fundamental resource of adaptation to endogenous and exogenous stimuli. Although the response is initiated with protective purposes, the effect may be detrimental when not regulated. The physiological control of neuroinflammation is mainly achieved via regulatory mechanisms performed by particular cells of the immune system intimately associated with or within the nervous system and named "non-neuronal cells." In particular, mast cells (within the central nervous system and in the periphery) and microglia (at spinal and supraspinal level) are involved in this control, through a close functional relationship between them and neurons (either centrally, spinal, or peripherally located). Accordingly, neuroinflammation becomes a worsening factor in many disorders whenever the non-neuronal cell supervision is inadequate. It has been shown that the regulation of non-neuronal cells-and therefore the control of neuroinflammation-depends on the local "on demand" synthesis of the endogenous lipid amide Palmitoylethanolamide and related endocannabinoids. When the balance between synthesis and degradation of this bioactive lipid mediator is disrupted in favor of reduced synthesis and/or increased degradation, the behavior of non-neuronal cells may not be appropriately regulated and neuroinflammation exceeds the physiological boundaries. In these conditions, it has been demonstrated that the increase of endogenous Palmitoylethanolamide-either by decreasing its degradation or exogenous administration-is able to keep neuroinflammation within its physiological limits. In this review the large number of studies on the benefits derived from oral administration of micronized and highly bioavailable forms of Palmitoylethanolamide is discussed, with special reference to neuroinflammatory disorders.


Subject(s)
Amides/administration & dosage , Amides/metabolism , Ethanolamines/administration & dosage , Ethanolamines/metabolism , Inflammation/diet therapy , Nervous System Diseases/drug therapy , Neurodegenerative Diseases/drug therapy , Palmitic Acids/administration & dosage , Palmitic Acids/metabolism , Alzheimer Disease/diet therapy , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/diet therapy , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Animals , Autism Spectrum Disorder/diet therapy , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Endocannabinoids/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Metabolic Networks and Pathways , Multiple Sclerosis/diet therapy , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Nervous System Diseases/diet therapy , Nervous System Diseases/metabolism , Neurodegenerative Diseases/diet therapy , Neurodegenerative Diseases/metabolism , Pain/diet therapy , Pain/drug therapy , Parkinson Disease/drug therapy , Parkinson Disease/metabolism
7.
Biochem Pharmacol ; 175: 113859, 2020 05.
Article in English | MEDLINE | ID: mdl-32061773

ABSTRACT

The cellular microenvironment plays a critical role in the maintenance of bone marrow-derived mesenchymal stem cells (BM-MSCs) and their subsequent cell lineage differentiation. Recent studies suggested that individuals with adipocyte-related metabolic disorders have altered function and adipogenic potential of adipose stem cell subpopulations, primarily BM-MSCs, increasing the risk of heart attack, stroke or diabetes. In this study, we explored the potential therapeutic effect of some of the most abundant non-euphoric compounds derived from the Cannabis sativa plant (or phytocannabinoids) including tetrahydrocannabivarin (THCV), cannabidiol (CBD), cannabigerol (CBG), cannabidiolic acid (CBDA) and cannabigerolic acid (CBGA), by analysing their pharmacological activity on viability of endogenous BM-MSCs as well as their ability to alter BM-MSC proliferation and differentiation into mature adipocytes. We provide evidence that CBD, CBDA, CBGA and THCV (5 µM) increase the number of viable BM-MSCs; whereas only CBG (5 µM) and CBD (5 µM) alone or in combination promote BM-MSCs maturation into adipocytes via distinct molecular mechanisms. These effects were revealed both in vitro and in vivo. In addition, phytocannabinoids prevented the insulin signalling impairment induced by palmitate in adipocytes differentiated from BM-MSCs. Our study highlights phytocannabinoids as a potential novel pharmacological tool to regain control of functional adipose tissue in unregulated energy homeostasis often occurring in metabolic disorders including type 2 diabetes mellitus (T2DM), aging and lipodystrophy.


Subject(s)
Adipogenesis/drug effects , Cannabinoids/pharmacology , Cannabis/chemistry , Cell Differentiation/drug effects , Mesenchymal Stem Cells/drug effects , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Bone Marrow Cells/cytology , Cannabinoids/isolation & purification , Cell Survival/drug effects , Cells, Cultured , Colony-Forming Units Assay , Endocannabinoids/metabolism , Energy Metabolism/drug effects , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Insulin/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Triglycerides/metabolism
8.
J Neuroinflammation ; 16(1): 274, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31878942

ABSTRACT

BACKGROUND: Palmitoylethanolamide (PEA) is a pleiotropic endogenous lipid mediator currently used as a "dietary food for special medical purposes" against neuropathic pain and neuro-inflammatory conditions. Several mechanisms underlie PEA actions, among which the "entourage" effect, consisting of PEA potentiation of endocannabinoid signaling at either cannabinoid receptors or transient receptor potential vanilloid type-1 (TRPV1) channels. Here, we report novel molecular mechanisms through which PEA controls mast cell degranulation and substance P (SP)-induced histamine release in rat basophilic leukemia (RBL-2H3) cells, a mast cell model. METHODS: RBL-2H3 cells stimulated with SP were treated with PEA in the presence and absence of a cannabinoid type-2 (CB2) receptor antagonist (AM630), or a diacylglycerol lipase (DAGL) enzyme inhibitor (OMDM188) to inhibit the biosynthesis of the endocannabinoid 2-arachidonoylglycerol (2-AG). The release of histamine was measured by ELISA and ß-hexosaminidase release and toluidine blue staining were used as indices of degranulation. 2-AG levels were measured by LC-MS. The mRNA expression of proposed PEA targets (Cnr1, Cnr2, Trpv1, Ppara and Gpr55), and of PEA and endocannabinoid biosynthetic (Napepld, Dagla and Daglb) and catabolic (Faah, Naaa and Mgl) enzymes were also measured. The effects of PEA on the activity of DAGL-α or -ß enzymes were assessed in COS-7 cells overexpressing the human recombinant enzyme or in RBL-2H3 cells, respectively. RESULTS: SP increased the number of degranulated RBL-2H3 cells and triggered the release of histamine. PEA counteracted these effects in a manner antagonized by AM630. PEA concomitantly increased the levels of 2-AG in SP-stimulated RBL-2H3 cells, and this effect was reversed by OMDM188. PEA significantly stimulated DAGL-α and -ß activity and, consequently, 2-AG biosynthesis in cell-free systems. Co-treatment with PEA and 2-AG at per se ineffective concentrations downmodulated SP-induced release of histamine and degranulation, and this effect was reversed by OMDM188. CONCLUSIONS: Activation of CB2 underlies the inhibitory effects on SP-induced RBL-2H3 cell degranulation by PEA alone. We demonstrate for the first time that the effects in RBL-2H3 cells of PEA are due to the stimulation of 2-AG biosynthesis by DAGLs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Degranulation/drug effects , Ethanolamines/pharmacology , Lipoprotein Lipase/metabolism , Mast Cells/drug effects , Palmitic Acids/pharmacology , Amides , Animals , Cell Line, Tumor , In Vitro Techniques , Mast Cells/enzymology , Rats , Substance P/pharmacology
9.
Neurobiol Dis ; 125: 77-91, 2019 05.
Article in English | MEDLINE | ID: mdl-30660740

ABSTRACT

AIM: Recent studies revealed that pharmacological modulation of NAE-hydrolyzing acid amidase (NAAA) can be achieved with PEA oxazoline (PEA-OXA). Hence, the aim of the present work was to thoroughly evaluate the anti-inflammatory and neuroprotective effects of PEA-OXA in an experimental model of vascular dementia (VaD) induced by bilateral carotid arteries occlusion. At 24 h after VaD induction, animals were orally administered with 10 mg/kg of PEA-OXA daily for 15 days. RESULTS: Brain tissues were handled for histological, immunohistochemical, western blot, and immunofluorescence analysis. PEA-OXA treatment evidently reduced the histological alterations and neuronal death induced by VaD and additionally improved behavioral deficits. Further, PEA-OXA decreased GFAP and Iba-1, markers of astrocytes, and microglia activation, as well as increased MAP-2, a marker of neuron development. Moreover, PEA-OXA reduced oxidative stress, modulated Nrf2-mediated antioxidant response, and inhibited the apoptotic process. INNOVATION: Some drugs may demonstrate their healing potential by regulating neuroinflammation, rather than by their habitually attributed actions only. Palmitoylethanolamide (PEA) is a prototype ALIAmide, well-known for its analgesic, anti-inflammatory, and neuroprotective properties. The inhibition of PEA degradation by targeting NAAA, its catabolic enzyme, is a different approach for treating neuroinflammation. This research offers new insight into the mechanism of PEA-OXA-induced neuroprotection. CONCLUSION: Thus, the modulation of intracellular NAAA by PEA-OXA could offer a novel means of controlling neuroinflammatory conditions associated with VaD.


Subject(s)
Carotid Stenosis/complications , Dementia, Vascular/pathology , Neuroprotective Agents/pharmacology , Oxazoles/pharmacology , Oxidative Stress/drug effects , Amidohydrolases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Dementia, Vascular/etiology , Disease Models, Animal , Inflammation/pathology , Male , Mice
10.
Eur J Med Chem ; 161: 239-251, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30359820

ABSTRACT

A set of CB2R ligands, based on the thiophene scaffold, was synthesized and evaluated in in vitro assays. Compounds 8c-i, k, l, bearing the 3-carboxylate and 2-(adamantan-1-yl)carboxamido groups together with apolar alkyl/aryl substituents at 5-position or at 4- and 5-positions of the thiophene ring possess high CB2R affinity at low nanomolar concentration, good receptor selectivity, and agonistic functional activity. The full agonist 8g, showing the best balance between receptor affinity and selectivity, was tested in vitro in an experimental model of allergic contact dermatitis and proved to be able to block the release of MCP-2 in HaCaT cells at 10 µM concentration.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunosuppressive Agents/pharmacology , Inflammation/drug therapy , Receptor, Cannabinoid, CB2/agonists , Skin Diseases/drug therapy , Thiophenes/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/chemistry , Inflammation/pathology , Keratinocytes/drug effects , Molecular Structure , Skin Diseases/pathology , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
11.
Front Pharmacol ; 9: 249, 2018.
Article in English | MEDLINE | ID: mdl-29615912

ABSTRACT

Palmitoylethanolamide (PEA) is a pleiotropic lipid mediator with established anti-inflammatory and anti-hyperalgesic activity. Ultramicronized PEA (PEA-um) has superior oral efficacy compared to naïve (non-micronized) PEA. The aim of the present study was two-fold: (1) to evaluate whether oral PEA-um has greater absorbability compared to naïve PEA, and its ability to reach peripheral and central tissues under healthy and local inflammatory conditions (carrageenan paw edema); (2) to better characterize the molecular pathways involved in PEA-um action, particularly at the spinal level. Rats were dosed with 30 mg/kg of [13C]4-PEA-um or naïve [13C]4-PEA by oral gavage, and [13C]4-PEA levels quantified, as a function of time, by liquid chromatography/atmospheric pressure chemical ionization/mass spectrometry. Overall plasma levels were higher in both healthy and carrageenan-injected rats administered [13C]4-PEA-um as compared to those receiving naïve [13C]4-PEA, indicating the greater absorbability of PEA-um. Furthermore, carrageenan injection markedly favored an increase in levels of [13C]4-PEA in plasma, paw and spinal cord. Oral treatment of carrageenan-injected rats with PEA-um (10 mg/kg) confirmed beneficial peripheral effects on paw inflammation, thermal hyperalgesia and tissue damage. Notably, PEA-um down-regulated distinct spinal inflammatory and oxidative pathways. These last findings instruct on spinal mechanisms involved in the anti-hyperalgesic effect of PEA-um in inflammatory pain.

12.
J Pharmacol Exp Ther ; 365(3): 652-663, 2018 06.
Article in English | MEDLINE | ID: mdl-29632236

ABSTRACT

Phytocannabinoids modulate inflammatory responses by regulating the production of cytokines in several experimental models of inflammation. Cannabinoid type-2 (CB2) receptor activation was shown to reduce the production of the monocyte chemotactic protein-2 (MCP-2) chemokine in polyinosinic-polycytidylic acid [poly-(I:C)]-stimulated human keratinocyte (HaCaT) cells, an in vitro model of allergic contact dermatitis (ACD). We investigated if nonpsychotropic cannabinoids, such as cannabidiol (CBD), produced similar effects in this experimental model of ACD. HaCaT cells were stimulated with poly-(I:C), and the release of chemokines and cytokines was measured in the presence of CBD or other phytocannabinoids (such as cannabidiol acid, cannabidivarin, cannabidivarinic acid, cannabichromene, cannabigerol, cannabigerolic acid, cannabigevarin, tetrahydrocannabivarin, and tetrahydrocannabivarinic acid) and antagonists of CB1, CB2, or transient receptor potential vanilloid type-1 (TRPV1) receptors. HaCaT cell viability following phytocannabinoid treatment was also measured. The cellular levels of endocannabinoids [anandamide (AEA), 2-arachidonoylglycerol] and related molecules (palmitoylethanolamide, oleoylethanolamide) were quantified in poly-(I:C)-stimulated HaCaT cells treated with CBD. We show that in poly-(I:C)-stimulated HaCaT cells, CBD elevates the levels of AEA and dose-dependently inhibits poly-(I:C)-induced release of MCP-2, interleukin-6 (IL-6), IL-8, and tumor necrosis factor-α in a manner reversed by CB2 and TRPV1 antagonists 6-iodopravadoline (AM630) and 5'-iodio-resiniferatoxin (I-RTX), respectively, with no cytotoxic effect. This is the first demonstration of the anti-inflammatory properties of CBD in an experimental model of ACD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cannabidiol/pharmacology , Dermatitis, Allergic Contact/drug therapy , Anti-Inflammatory Agents/therapeutic use , Arachidonic Acids/metabolism , Cannabidiol/therapeutic use , Cell Line , Chemokine CCL8/metabolism , Dermatitis, Allergic Contact/metabolism , Endocannabinoids/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Polyunsaturated Alkamides/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Front Pharmacol ; 8: 308, 2017.
Article in English | MEDLINE | ID: mdl-28611664

ABSTRACT

N-acylethanolamines (NAEs) involve a family of lipid molecules existent in animal and plant, with N-palmitoylethanolamide (PEA) that arouses great attention owing to its anti-inflammatory, analgesic and neuroprotective activities. Because PEA is produced on demand and exerts pleiotropic effects, the modulation of specific amidases for NAEs (and in particular NAE-hydrolyzing acid amidase NAAA, which is more selective for PEA) could be a condition to preserve its levels. Here we investigate the effect of 2-Pentadecyl-2-oxazoline (PEA-OXA) the oxazoline of PEA, on human recombinant NAAA in vitro and in an established model of Carrageenan (CAR)-induced rat paw inflammation. PEA-OXA dose-dependently significantly inhibited recombinant NAAA and, orally administered to rats (10 mg/kg), limiting histological damage, thermal hyperalgesia and the increase of infiltrating inflammatory cells after CAR injection in the rat right hindpaw, compared to ultramicronized PEA given orally at the same dose (10 mg/kg). These effects were accompanied by elevation of paw PEA levels. Moreover, PEA-OXA markedly reduced neutrophil infiltration and pro-inflammatory cytokine release and prevented CAR-induced IκB-α degradation, nuclear translocation of NF-κB p65, the increase of inducible nitric oxide synthase, cyclooxygenase-2, intercellular adhesion molecule-1, and mast cell activation. Experiments in PPAR-α knockout mice showed that the anti-inflammatory effects of PEA-OXA were not dependent on the presence of PPAR-α receptors. In conclusion, NAAA modulators as PEA-OXA could help to maximize the tissue availability of PEA by increasing its levels and anti-inflammatory effects.

14.
Front Neurol ; 8: 233, 2017.
Article in English | MEDLINE | ID: mdl-28634464

ABSTRACT

Vascular dementia (VaD), the second most common cause of cognitive impairment in the population, is a disease that results from reduction in regional cerebral blood flow and involves oxidative stress and inflammation. Co-ultramicronized PEALut (co-ultra PEALut) is a new compound with beneficial effects, which include anti-inflammatory and antioxidant properties. Recently, co-ultraPEALut has been shown to exhibit neuroprotective effects in models of Parkinson's disease, cerebral ischemia and Alzheimer's disease. However, its effects on VaD remain unknown. Therefore, the purpose of the present study was to highlight the potential neuroprotective actions of co-ultraPEALut containing N-palmitoylethanolamine (PEA) and the antioxidant flavonoid luteolin (Lut) (10:1 by mass) in a mouse model of VaD induced by bilateral carotid arteries occlusion. At 24 h after VaD induction, mice were orally treated with 1 mg/kg co-ultraPEALut daily for 15 days. On the 15th day, brain tissues were processed for histological, immunohistochemical, Western blot, and immunofluorescent analysis. Our results clearly demonstrate that co-ultraPEALut improved learning, memory ability, locomotor activity, and the reciprocal social interaction. In addition, the mice subjected to VaD and treated with the co-ultraPEALut showed a reorganization of CA1 and CA3 regions of the hippocampus and restored the number of hippocampal neurons as evidenced by NeuN expression, a specific marker of neurons. Furthermore following carotid arteries ligation, mice treated with co-ultraPEALut showed a modification of proinflammatory, proapoptotic proteins and of oxidative stress as evidenced by the expression of IκB-α, NF-κB p65, Bax, Bcl-2, inducible nitric oxide synthase, and cyclooxygenase-2. In order, co-ultraPEALut treatment restored VaD-induced loss of brain-derived neurotrophic factor and neurotrophins 3 (NT-3) expression in mice. These results confirmed that the neuroprotective effects of co-ultraPEALut were associated with its anti-inflammatory and antioxidant properties.

15.
Br J Pharmacol ; 174(11): 1349-1365, 2017 06.
Article in English | MEDLINE | ID: mdl-27539936

ABSTRACT

Palmitoylethanolamide (PEA) has emerged as a potential nutraceutical, because this compound is naturally produced in many plant and animal food sources, as well as in cells and tissues of mammals, and endowed with important neuroprotective, anti-inflammatory and analgesic actions. Several efforts have been made to identify the molecular mechanism of action of PEA and explain its multiple effects both in the central and the peripheral nervous system. Here, we provide an overview of the pharmacology, efficacy and safety of PEA in neurodegenerative disorders, pain perception and inflammatory diseases. The current knowledge of new formulations of PEA with smaller particle size (i.e. micronized and ultra-micronized) when given alone or in combination with antioxidant flavonoids (i.e. luteolin) and stilbenes (i.e. polydatin) is also reviewed. LINKED ARTICLES: This article is part of a themed section on Principles of Pharmacological Research of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.11/issuetoc.


Subject(s)
Dietary Supplements , Ethanolamines/pharmacology , Palmitic Acids/pharmacology , Amides , Analgesics/administration & dosage , Analgesics/adverse effects , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacology , Chemistry, Pharmaceutical/methods , Ethanolamines/administration & dosage , Ethanolamines/adverse effects , Humans , Inflammation/diet therapy , Neurodegenerative Diseases/diet therapy , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacology , Palmitic Acids/administration & dosage , Palmitic Acids/adverse effects , Particle Size
16.
Eur J Pharmacol ; 791: 669-674, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27720681

ABSTRACT

In mice, 2,4-dinitrofluorobenzene (DNFB) induces contact allergic dermatitis (CAD), which, in a late phase, is characterized by mast cell (MC) infiltration and angiogenesis. Palmitoylethanolamide (PEA), an endogenous anti-inflammatory molecule, acts by down-modulating MCs following activation of the cannabinoid CB2 receptor and peroxisome proliferator-activated receptor-α (PPAR-α). We have previously reported the anti-inflammatory effect of PEA in the early stage of CAD. Here, we examined whether PEA reduces the features of the late stage of CAD including MC activation, angiogenesis and itching. After sensitization to DNFB, female C57BL/6J mice underwent to three DNFB challenges at days 5, 12 and 19 and treatments were given at each challenge and for two more days. CAD was expressed as Δ increase in ear thickness between challenged and un-challenged mice. PEA (5mg/kg/i.p.) reduced: i) the DNFB-induced Δ increase; ii) the number of MCs per tissue area; iii) the expression of VEGF and its receptor Flk-1. These effects were reversed by co-administration of AM630 (1mg/kg/i.p.), a CB2 antagonist, but not GW6471 (1mg/kg/i.p.), a PPAR-α antagonist. Finally, PEA reduced the number of ear scratchings 48h after DNFB challenge and this effect was reversed by both CB2 and PPAR-α antagonists, suggesting the involvement of both receptors. PEA, by reducing the features of late stage CAD in mice, may be beneficial in this pathological condition.


Subject(s)
Dermatitis, Allergic Contact/complications , Dermatitis, Allergic Contact/drug therapy , Ethanolamines/pharmacology , Palmitic Acids/pharmacology , Pruritus/complications , Amides , Animals , Cell Count , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/metabolism , Dinitrofluorobenzene/adverse effects , Disease Models, Animal , Endocannabinoids/metabolism , Ethanolamines/therapeutic use , Female , Gene Expression Regulation/drug effects , Inflammation/complications , Mast Cells/cytology , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Palmitic Acids/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Prog Lipid Res ; 62: 107-28, 2016 04.
Article in English | MEDLINE | ID: mdl-26965148

ABSTRACT

The endocannabinoid system (ECS) is composed of two G protein-coupled receptors (GPCRs), the cannabinoid CB1 and CB2 receptors, and the two main endogenous lipid ligands of such receptors (also known as the "endocannabinoids"), anandamide and 2-arachidonoyl-glycerol. The ECS is a pleiotropic signalling system involved in all aspects of mammalian physiology and pathology, and for this reason it represents a potential target for the design and development of new therapeutic drugs. However, the endocannabinoids as well as some of their congeners also interact with a much wider range of receptors, including members of the Transient Receptor Potential (TRP) channels, Peroxisome Proliferator-Activated Receptors (PPARs), and other GPCRs. Indeed, following the discovery of the endocannabinoids, endocannabinoid-related lipid mediators, which often share the same metabolic pathways of the endocannabinoids, have also been identified or rediscovered. In this review article, we discuss the role of endocannabinoids and related lipids during physiological functions, as well as their involvement in some of the most common neurological disorders.


Subject(s)
Endocannabinoids/biosynthesis , Lipid Metabolism , Receptors, Cannabinoid/metabolism , Animals , Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Glycerides/metabolism , Humans , Metabolic Networks and Pathways , Peroxisome Proliferator-Activated Receptors/metabolism , Polyunsaturated Alkamides/metabolism , Receptors, G-Protein-Coupled/metabolism , Transient Receptor Potential Channels/metabolism
18.
Br J Pharmacol ; 173(7): 1154-62, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25598150

ABSTRACT

BACKGROUND AND PURPOSE: Palmitoylethanolamide (PEA) is an endogenous congener of anandamide and potentiates its actions at cannabinoid CB1 and CB2 receptors, and at transient receptor potential vanilloid type-1 (TRPV1) channels. The other endocannabinoid, 2-arachidonoylglycerol (2-AG), was recently suggested to act as a TRPV1 channel agonist. We investigated if PEA enhanced levels of 2-AG in vitro or in vivo and 2-AG activity at TRPV1 channels. EXPERIMENTAL APPROACH: Endogenous lipid levels were measured by LC-MS in (i) human keratinocytes incubated with PEA (10-20 µM, 40 min, 6 and 24 h, 37°C); (ii) the blood of spontaneously Ascaris suum hypersensitive beagle dogs given a single oral dose of ultramicronized PEA (30 mg·kg(-1), 1, 2, 4 and 8 h from administration); (iii) the blood of healthy volunteers given a single oral dose of micronized PEA (300 mg, 2, 4 and 6 h from administration). Effects of 2-AG at TRPV1 channels were assessed by measuring intracellular Ca(2+) in HEK-293 cells over-expressing human TRPV1 channels. KEY RESULTS: PEA elevated 2-AG levels in keratinocytes (∼3-fold) and in human and canine plasma (∼2 and ∼20-fold respectively). 2-AG dose-dependently raised intracellular Ca(2+) in HEK-293-TRPV1 cells in a TRPV1-dependent manner and desensitized the cells to capsaicin. PEA only slightly enhanced 2-AG activation of TRPV1 channels, but significantly increased 2-AG-induced TRPV1 desensitization to capsaicin (IC50 from 0.75 ± 0.04 to 0.45 ± 0.02 µM, with PEA 2 µM). CONCLUSIONS AND IMPLICATIONS: These observations may explain why several effects of PEA are attenuated by cannabinoid receptor or TRPV1 channel antagonists. LINKED ARTICLES: This article is part of a themed section on Endocannabinoids. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.7/issuetoc.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arachidonic Acids/blood , Endocannabinoids/blood , Ethanolamines/pharmacology , Glycerides/blood , Palmitic Acids/pharmacology , TRPV Cation Channels/metabolism , Adolescent , Adult , Amides , Animals , Calcium/metabolism , Capsaicin/pharmacology , Cell Line , Dogs , Ethanolamines/metabolism , Female , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Middle Aged , Palmitic Acids/metabolism , TRPV Cation Channels/agonists , Young Adult
19.
Biomed Res Int ; 2015: 980983, 2015.
Article in English | MEDLINE | ID: mdl-25866824

ABSTRACT

Allergen exposure may induce changes in the brainstem secondary neurons, with neural sensitization of the nucleus solitary tract (NTS), which in turn can be considered one of the causes of the airway hyperresponsiveness, a characteristic feature of asthma. We evaluated neurofunctional, morphological, and biochemical changes in the NTS of naive or sensitized rats. To evaluate the cell firing activity of NTS, in vivo electrophysiological experiments were performed before and after capsaicin challenge in sensitized or naive rats. Immunohistochemical studies, endocannabinoid, and palmitoylethanolamide quantification in the NTS were also performed. This study provides evidence that allergen sensitization in the NTS induced: (1) increase in the neural firing response to intratracheal capsaicin application, (2) increase of endocannabinoid anandamide and palmitoylethanolamide, a reduction of 2-arachidonoylglycerol levels in the NTS, (3) glial cell activation, and (4) prevention by a Group III metabotropic glutamate receptor activation of neural firing response to intratracheal application of capsaicin in both naïve and sensitized rats. Therefore, normalization of ovalbumin-induced NTS neural sensitization could open up the prospect of new treatments based on the recovery of specific brain nuclei function and for extensive studies on acute or long-term efficacy of selective mGlu ligand, in models of bronchial hyperreactivity.


Subject(s)
Allergens/toxicity , Asthma/metabolism , Capsaicin/administration & dosage , Neuroglia/metabolism , Neurons/metabolism , Amides , Animals , Arachidonic Acids/administration & dosage , Arachidonic Acids/metabolism , Asthma/chemically induced , Asthma/pathology , Brain Stem/drug effects , Brain Stem/pathology , Endocannabinoids/administration & dosage , Endocannabinoids/metabolism , Ethanolamines/administration & dosage , Glycerides/metabolism , Humans , Neuroglia/drug effects , Neuroglia/pathology , Neurons/drug effects , Neurons/pathology , Palmitic Acids/administration & dosage , Polyunsaturated Alkamides/administration & dosage , Propionates/administration & dosage , Rats , Receptors, Glutamate/metabolism , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Solitary Nucleus/drug effects
20.
J Cell Physiol ; 230(12): 2905-14, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25684344

ABSTRACT

The endocannabinoid system, through cannabinoid receptor signaling by endocannabinoids, is involved in a wide range of functions and physiopathological conditions. To date, very little is known concerning the role of the endocannabinoids in the control and regulation of cell proliferation. An anti-proliferative action of CB1 signaling blockade in neurogenesis and angiogenesis argues in favor of proliferation-promoting functions of endocannabinoids through CB1 receptors when pro-growth signals are present. Furthermore, liver regeneration, a useful in vivo model of synchronized cell proliferation, is characterized by a peak of anandamide that elicits through CB1 receptor, the expression of critical mitosis genes. The aim of this study was to focus on the timing of endocannabinoid signaling changes during the different phases of the cell cycle, exploiting the rat liver regeneration model following partial hepatectomy, the most useful to study synchronized cell cycle in vivo. Hepatic regeneration led to increased levels of anandamide and endocannabinoid-like molecules oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) in the G1 phase of the cell cycle, with a concomitant increase in CB1 mRNA levels, whose protein expression peaked later during the S phase. Blocking of CB1 receptor with a low dose of the selective antagonist/inverse agonist SR141716 (0.7 mg/kg/dose) affected cell cycle progression reducing the expression of PCNA, and through the inhibition of pERK and pSTAT3 pathways. These results support the notion that the signaling mediated by anandamide through CB1 receptor may be important for the entry and progression of cells into the cell cycle and hence for their proliferation under mitogenic signals.


Subject(s)
Arachidonic Acids/metabolism , Cell Cycle , Cell Proliferation , Endocannabinoids/metabolism , Liver Regeneration , Liver/metabolism , Polyunsaturated Alkamides/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Cannabinoid Receptor Antagonists/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycerides/metabolism , Hepatectomy , Liver/drug effects , Liver/pathology , Liver Regeneration/drug effects , Male , Proliferating Cell Nuclear Antigen/metabolism , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL