Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(14)2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39063011

ABSTRACT

Sepsis-induced multiple organ dysfunction arises from the highly complex pathophysiology encompassing the interplay of inflammation, oxidative stress, endothelial dysfunction, mitochondrial damage, cellular energy failure, and dysbiosis. Over the past decades, numerous studies have been dedicated to elucidating the underlying molecular mechanisms of sepsis in order to develop effective treatments. Current research underscores liver and cardiac dysfunction, along with acute lung and kidney injuries, as predominant causes of mortality in sepsis patients. This understanding of sepsis-induced organ failure unveils potential therapeutic targets for sepsis treatment. Various novel therapeutics, including melatonin, metformin, palmitoylethanolamide (PEA), certain herbal extracts, and gut microbiota modulators, have demonstrated efficacy in different sepsis models. In recent years, the research focus has shifted from anti-inflammatory and antioxidative agents to exploring the modulation of energy metabolism and gut microbiota in sepsis. These approaches have shown a significant impact in preventing multiple organ damage and mortality in various animal sepsis models but require further clinical investigation. The accumulation of this knowledge enriches our understanding of sepsis and is anticipated to facilitate the development of effective therapeutic strategies in the future.


Subject(s)
Multiple Organ Failure , Sepsis , Humans , Sepsis/complications , Sepsis/metabolism , Sepsis/drug therapy , Sepsis/microbiology , Multiple Organ Failure/etiology , Multiple Organ Failure/metabolism , Animals , Gastrointestinal Microbiome , Oxidative Stress , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology
2.
Molecules ; 29(7)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38611871

ABSTRACT

Oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) are endogenous lipids that act as agonists of the peroxisome proliferator-activated receptor α (PPARα). Recently, an interest in the role of these lipids in malignant tumors has emerged. Nevertheless, the effects of OEA and PEA on human neuroblastoma cells are still not documented. Type I interferons (IFNs) are immunomodulatory cytokines endowed with antiviral and anti-proliferative actions and are used in the treatment of various pathologies such as different cancer forms (i.e., non-Hodgkin's lymphoma, melanoma, leukemia), hepatitis B, hepatitis C, multiple sclerosis, and many others. In this study, we investigated the effect of OEA and PEA on human neuroblastoma SH-SY5Y cells treated with IFNß. We focused on evaluating cell viability, cell proliferation, and cell signaling. Co-exposure to either OEA or PEA along with IFNß leads to increased apoptotic cell death marked by the cleavage of caspase 3 and poly-(ADP ribose) polymerase (PARP) alongside a decrease in survivin and IKBα levels. Moreover, we found that OEA and PEA did not affect IFNß signaling through the JAK-STAT pathway and the STAT1-inducible protein kinase R (PKR). OEA and PEA also increased the phosphorylation of p38 MAP kinase and programmed death-ligand 1 (PD-L1) expression both in full cell lysate and surface membranes. Furthermore, GW6471, a PPARα inhibitor, and the genetic silencing of the receptor were shown to lower PD-L1 and cleaved PARP levels. These results reveal the presence of a novel mechanism, independent of the IFNß-prompted pathway, by which OEA and PEA can directly impair cell survival, proliferation, and clonogenicity through modulating and potentiating the intrinsic apoptotic pathway in human SH-SY5Y cells.


Subject(s)
Amides , Endocannabinoids , Ethanolamines , Neuroblastoma , Oleic Acids , Humans , Neuroblastoma/drug therapy , B7-H1 Antigen , Janus Kinases , PPAR alpha , Poly(ADP-ribose) Polymerase Inhibitors , STAT Transcription Factors , Signal Transduction , Apoptosis , Palmitic Acids/pharmacology
3.
Nat Prod Res ; : 1-20, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38269580

ABSTRACT

Intraocular pressure (IOP) positively correlates with both normal and high-tension glaucoma. To date, IOP targeting remains the validated pharmacological approach in counteracting glaucoma progression as well as in halting vision loss. Among the different adjuvant compounds, evidence highlighted the potential effectiveness of Palmitoylethanolamide (PEA), an endogenous fatty acid amide. Thus, a systematic review of the literature was conducted, thoroughly evaluating PEA treatment regimen in decreasing IOP in patients with eye disorders. We checked for articles across the scientific databases Pubmed (MEDLINE), Embase (OVID), and Web of Science from the inception to 30 August 2023, and a total of 828 articles were recovered. Six of these studies (199 patients) were included in the systematic review after the study selection process, and three studies for meta-analysia. Overall, PEA showed significant efficacy in reducing IOP in patients, this encourages its clinical use in glaucoma as well as across different forms of eye disorders.

4.
Biomedicines ; 11(8)2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37626685

ABSTRACT

Post-Covid Olfactory Dysfunction (PCOD) is characterized by olfactory abnormalities, hyposmia, and anosmia, which are among the most often enduring symptoms in individuals who have recovered from SARS-CoV-2 infection. This disorder has been reported to persist in subsets of patients well after 12 months following infection, significantly affecting their quality of life. Despite the high prevalence of PCOD among patients who suffered from SARS-CoV-2 infection, specific therapeutic strategies are still limited. Among these, emerging evidence seems to indicate the administration of CoUltraPEALut, a combination of micronized Palmitoylethanolamide (PEA), an endogenous fatty acid amide, and Luteolin, a natural antioxidant flavonoid, as a viable therapy, especially when given as an adjuvant to olfactory training. Based on the above, a systematic review and a meta-analysis of the literature were conducted, with the aim of evaluating the efficacy of CoUltraPEALut as an addition to olfactory training (OT), in treating PCOD symptoms. Pubmed (MEDLINE), Embase (OVID), and Web of Science scientific databases were screened from the inception until 31 May 2023, and a total of 407 articles were recovered; only five of these studies (441 total patients between treated and control groups) were included in the systematic review. CoUltraPEALut demonstrated significant efficacy in the overall recovery of the olfactory function, compared to the conventional therapy, suggesting that it could represent a possible future adjuvant treatment for PCOD.

5.
Pharmacol Res ; 185: 106491, 2022 11.
Article in English | MEDLINE | ID: mdl-36244543

ABSTRACT

Psoriasis is an incurable autoimmune disease that affects 2-3% of the world's population. Limited understanding of its pathogenesis hinders the development of therapies for the disease. Herein, we reported that N-acylethanolamine acid amidase (NAAA), a cysteine enzyme that catalyzes the hydrolysis of fatty acid ethanolamides (FAEs), was upregulated in psoriasis patients and imiquimod (IMQ)-induced mouse model of psoriasis. The upregulated NAAA contributes to the progression of psoriasis via enhancing dendritic cell (DCs) maturation. Transgenic expression of NAAA in mice accelerated the development of psoriasis, whereas genetic ablation of NAAA or local administration of NAAA inhibitor F96 ameliorated psoriasis. NAAA expressed in dendritic cells (DCs), but not in macrophages, T cells, or keratinocytes plays a critical role in psoriasis development. In addition, the results showed that NAAA degrades palmitoylethanolamide (PEA) and reduces PEA-PPARα-mediated dissociation of NF-κB p65 from Sirtuin 1 (SIRT1), subsequently, repressing the acetylation of p65 and down-regulating IL10 production. The decreased IL10 then leads to the maturation of DCs, thus promoting the development of psoriasis. These results provide new insights into the pathophysiological mechanism of psoriasis and identify NAAA as a novel target for the treatment of psoriasis.


Subject(s)
Interleukin-10 , Psoriasis , Mice , Animals , Enzyme Inhibitors/pharmacology , Amidohydrolases , Inflammation , Psoriasis/drug therapy , Dendritic Cells/metabolism
6.
Eur J Pharmacol ; 912: 174561, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34655598

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of death worldwide, for which there is currently no comprehensive treatment available. Preventing blood-brain barrier (BBB) disruption is crucial for TBI treatment. N-acylethanolamine acid amidase (NAAA)-regulated palmitoylethanolamide (PEA) signaling play an important role in the control of inflammation. However, the role of NAAA in BBB dysfunction following TBI remains unclear. In the present study, we found that TBI induces the increase of PEA levels in the injured cortex, which prevent the disruption of BBB after TBI. TBI also induces the infiltration of NAAA-contained neutrophils, increasing the contribution of NAAA to the PEA degradation. Neutrophil-derived NAAA weakens PEA/PPARα-mediated BBB protective effects after TBI, facilitates the accumulation of immune cells, leading to secondary expansion of tissue injury. Inactivation of NAAA increased PEA levels in injured site, prevents early BBB damage and improves secondary injury, thereby eliciting long-term functional improvements after TBI. This study identified a new role of NAAA in TBI, suggesting that NAAA is a new important target for BBB dysfunction related CNS diseases.


Subject(s)
Amidohydrolases/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic/metabolism , Enzyme Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Oxazolidinones/pharmacology , Amides/metabolism , Amidohydrolases/antagonists & inhibitors , Animals , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/pathology , Cell Line , Disease Models, Animal , Endothelial Cells/drug effects , Enzyme Inhibitors/therapeutic use , Ethanolamines/metabolism , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Neutrophils/metabolism , Oxazolidinones/therapeutic use , PPAR alpha/deficiency , PPAR alpha/genetics , Palmitic Acids/metabolism
7.
Inflammopharmacology ; 29(5): 1475-1486, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34468900

ABSTRACT

Novel treatment strategies are urgently required for osteoarthritis (OA). Palmitoylethanolamide (PEA) is a naturally occurring fatty acid amide with analgesic and anti-inflammatory effects. We aimed to examine its effect on OA and elucidate the molecular mechanism of actions in monosodium iodoacetate (MIA)-induced OA Sprague-Dawley rats. The experimental animals were divided into normal control group (injected with saline + treated with phosphate-buffered saline (PBS), NOR), control group (injected with MIA + treated with PBS, CON), 50 or 100 mg/kg body weight (BW)/day PEA-treated group (injected with MIA + treated with 50 or 100 mg of PEA/kg BW/day, PEA50 or PEA100), and positive control group (injected with MIA + treated with 6 mg of diclofenac/kg BW/day, DiC). The changes in blood parameters, body parameters, gene expression of inflammatory mediators and cytokines, knee thickness, and joint tissue were observed. Oral administration of PEA had no adverse effects on the BW, liver, or kidneys. PEA reduced knee joint swelling and cartilage degradation in MIA-induced OA rats. The serum levels of leukotriene B4, nitric oxide, tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and prostaglandin E2 considerably reduced in the PEA100 group compared with those in the CON group. In the synovia of knee joints, the mRNA expression of iNOS, 5-Lox, Cox-2, Il-1ß, Tnf-α, and Mmp-2, -3, -9, and -13 apparently increased with MIA administration. Meanwhile, Timp-1 mRNA expression apparently decreased in the CON group but increased to the normal level with PEA treatment. Thus, PEA can be an effective therapeutic agent for OA.


Subject(s)
Amides/pharmacology , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Ethanolamines/pharmacology , Osteoarthritis/drug therapy , Palmitic Acids/pharmacology , Administration, Oral , Amides/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Dose-Response Relationship, Drug , Ethanolamines/administration & dosage , Iodoacetic Acid , Knee Joint/drug effects , Knee Joint/pathology , Male , Palmitic Acids/administration & dosage , Rats , Rats, Sprague-Dawley
8.
Biochem Biophys Res Commun ; 566: 164-169, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34126347

ABSTRACT

Palmitoylethanolamide (PEA) offers a strong protection against BBB disruption and neurological deficits after cerebral ischaemic/reperfusion (I/R) injury. To date, these BBB protective effects of PEA are mainly attributed to PPARα-mediated actions. However, whether PEA protects against BBB disruption through direct regulation of cytoskeletal microfilaments remains unknown. Here, we identified PEA as a Rho-associated protein kinase (ROCK2) inhibitor (IC50 = 38.4 ± 4.8 µM). In vitro data suggested that PEA reduced the activation of ROCK/MLC signaling and stress fiber formation within microvascular endothelial cells (ECs) after oxygen-glucose deprivation (OGD), and consequently attenuated early (0-4 h) EC barrier disruption. These actions of PEA could not be blocked by the PPARα antagonist GW6471. In summary, the present study described a previously unexplored role of PEA as a ROCK2 inhibitor, and propose a PPARα-independent mechanism for pharmacological effects of PEA.


Subject(s)
Amides/therapeutic use , Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Ethanolamines/therapeutic use , Myosin Light Chains/metabolism , Palmitic Acids/therapeutic use , Reperfusion Injury/drug therapy , rho-Associated Kinases/metabolism , Amides/pharmacology , Animals , Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Cell Line , Ethanolamines/pharmacology , Humans , Mice , Palmitic Acids/pharmacology , Reperfusion Injury/metabolism , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors
9.
Int J Mol Sci ; 22(4)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671213

ABSTRACT

Palmitoylethanolamide (PEA), a fatty acid amide, has been widely investigated for its analgesic and anti-inflammatory properties. The ultra-micronized formulation of PEA (um-PEA), that has an enhanced rate of dissolution, is extensively used. Acetyl-l-carnitine (LAC), employed for the treatment of neuropathic pain in humans, is able to cause analgesia by up-regulating type-2 metabotropic glutamate (mGlu2) receptors. In the present study, we tested different associations of um-PEA, LAC and non-micronized PEA (non-m-PEA) in a rat model of carrageenan (CAR)-induced paw edema. Intraplantar injection of CAR into the hind paw of animals caused edema, thermal hyperalgesia, accumulation of infiltrating inflammatory cells and augmented myeloperoxidase (MPO) activity. All these parameters were decreased in a significantly manner by oral administration of a compound constituted by a mixture of um-PEA and LAC in relation 1:1 (5 mg/kg), but not with the association of single compounds administered one after the other. These findings showed the superior anti-inflammatory and anti-nociceptive action displayed by oral administration of um-PEA and LAC versus LAC plus, separate but consecutive, um-PEA in the rat paw CAR model of inflammatory pain.


Subject(s)
Acetylcarnitine/therapeutic use , Amides/therapeutic use , Ethanolamines/therapeutic use , Inflammation/drug therapy , Pain/drug therapy , Palmitic Acids/therapeutic use , Acetylcarnitine/pharmacology , Amides/pharmacology , Animals , Carrageenan , Cell Count , Cyclooxygenase 2/metabolism , Disease Models, Animal , Edema/complications , Edema/drug therapy , Edema/pathology , Ethanolamines/pharmacology , Hyperalgesia/complications , Hyperalgesia/drug therapy , Inflammation/complications , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/metabolism , Male , Mast Cells/drug effects , Nitric Oxide Synthase Type II/metabolism , Pain/complications , Pain/pathology , Palmitic Acids/pharmacology , Peroxidase/metabolism , Rats, Sprague-Dawley , Time Factors , Tumor Necrosis Factor-alpha/metabolism
10.
Prostaglandins Other Lipid Mediat ; 154: 106540, 2021 06.
Article in English | MEDLINE | ID: mdl-33636368

ABSTRACT

Coronavirus Disease 2019 (COVID-19) is upsetting the world and innovative therapeutic solutions are needed in an attempt to counter this new pandemic. Great hope lies in vaccines, but drugs to cure the infected patient are just as necessary. In the most severe forms of the disease, a cytokine storm with neuroinflammation occurs, putting the patient's life at serious risk, with sometimes long-lasting sequelae. Palmitoylethanolamide (PEA) is known to possess anti-inflammatory and neuroprotective properties, which make it an ideal candidate to be assumed in the earliest stage of the disease. Here, we provide a mini-review on the topic, pointing out phospholipids consumption in COVID-19, the possible development of an antiphospholipid syndrome secondary to SARS-CoV-2 infection, and reporting our preliminary single-case experience concerning to a 45-year-old COVID-19 female patient recently treated with success by micronized / ultramicronized PEA.


Subject(s)
Amides/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Antiphospholipid Syndrome/drug therapy , COVID-19 Drug Treatment , Ethanolamines/administration & dosage , Neuroprotective Agents/administration & dosage , Palmitic Acids/administration & dosage , SARS-CoV-2/metabolism , Antiphospholipid Syndrome/etiology , Antiphospholipid Syndrome/metabolism , Antiphospholipid Syndrome/pathology , COVID-19/complications , COVID-19/metabolism , COVID-19/pathology , Female , Humans , Middle Aged
11.
Biochem Pharmacol ; 184: 114398, 2021 02.
Article in English | MEDLINE | ID: mdl-33385371

ABSTRACT

Postoperative adhesions and scarring are the particular complication after strabismus surgery, for which there is currently no comprehensive treatment available. Preventing inflammation and fibrosis in the extraocular muscle are crucial for treatment of postoperative adhesions. In the present study, we found that administration of palmitoylethanolamide (PEA) attenuated postoperative inflammation and fibroproliferation through activating peroxisome proliferator-activated receptor α (PPARα), thus prevented scar formation. Inhibition of PEA degradation by N-Acylethanolamine acid amidase (NAAA) inhibitor F96 led to the same pharmacological results. PPARα activation suppressed both canonical and non-canonical TGFß signaling. Mechanistically, we found that PPARα directly bound to TGFß-activated kinase 1 (TAK1), thus preventing its hyperphosphorylation and the activation of downstream p38 and JNK1/2 signaling. Taken together, current study suggested that PEA could be a novel therapeutic approach for postoperative adhesions after strabismus surgery.


Subject(s)
Amides/pharmacology , Ethanolamines/pharmacology , Palmitic Acids/pharmacology , Postoperative Complications/drug therapy , Strabismus/surgery , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Fibrosis , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Mice , NIH 3T3 Cells , Oxazolidinones/pharmacology , PPAR alpha/metabolism , Postoperative Complications/etiology , Rabbits , Tissue Adhesions/drug therapy
12.
Front Pharmacol ; 12: 817603, 2021.
Article in English | MEDLINE | ID: mdl-35069223

ABSTRACT

N-Acylethanolamine acid amidase (NAAA) is a lysosomal enzyme responsible for the hydrolysis of fatty acid ethanolamides (FAEs). However, the role of NAAA in FAEs metabolism and regulation of pain and inflammation remains mostly unknown. Here, we generated NAAA-deficient (NAAA-/-) mice using CRISPR-Cas9 technique, and found that deletion of NAAA increased PEA and AEA levels in bone marrow (BM) and macrophages, and elevated AEA levels in lungs. Unexpectedly, genetic blockade of NAAA caused moderately effective anti-inflammatory effects in lipopolysaccharides (LPS)-induced acute lung injury (ALI), and poor analgesic effects in carrageenan-induced hyperalgesia and sciatic nerve injury (SNI)-induced mechanical allodynia. These data contrasted with acute (single dose) or chronic NAAA inhibition by F96, which produced marked anti-inflammation and analgesia in these models. BM chimera experiments indicated that these phenotypes were associated with the absence of NAAA in non-BM cells, whereas deletion of NAAA in BM or BM-derived cells in rodent models resulted in potent analgesic and anti-inflammatory phenotypes. When combined, current study suggested that genetic blockade of NAAA regulated FAEs metabolism and inflammatory responses in a cell-specifical manner.

13.
Pharmacol Res ; 160: 105064, 2020 10.
Article in English | MEDLINE | ID: mdl-32634582

ABSTRACT

N-Acylethanolamine acid amidase (NAAA) deactivates the endogenous peroxisome proliferator-activated receptor-α (PPAR-α) agonist palmitoylethanolamide (PEA). NAAA-regulated PEA signaling participates in the control of peripheral inflammation, but evidence suggests also a role in the modulation of neuroinflammatory pathologies such as multiple sclerosis (MS). Here we show that disease progression in the mouse experimental autoimmune encephalomyelitis (EAE) model of MS is accompanied by induction of NAAA expression in spinal cord, which in presymptomatic animals is confined to motor neurons and oligodendrocytes but, as EAE progresses, extends to microglia/macrophages and other cell types. As previously reported for NAAA inhibition, genetic NAAA deletion delayed disease onset and attenuated symptom intensity in female EAE mice, suggesting that accrued NAAA expression may contribute to pathology. To further delineate the role of NAAA in EAE, we generated a mouse line that selectively overexpresses the enzyme in macrophages, microglia and other monocyte-derived cells. Non-stimulated alveolar macrophages from these NaaaCD11b+ mice contain higher-than-normal levels of inducible nitric oxide synthase and display an activated morphology. Furthermore, intranasal lipopolysaccharide injections cause greater alveolar leukocyte accumulation in NaaaCD11b+ than in control mice. NaaaCD11b+ mice also display a more aggressive clinical response to EAE induction, compared to their wild-type littermates. The results identify NAAA as a critical control step in EAE pathogenesis, and point to this enzyme as a possible target for the treatment of MS.


Subject(s)
Amidohydrolases/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/pathology , Multiple Sclerosis/enzymology , Multiple Sclerosis/pathology , Amidohydrolases/genetics , Animals , Disease Progression , Female , Lipopolysaccharides , Macrophages/enzymology , Male , Mice , Mice, Inbred C57BL , Microglia/enzymology , Motor Neurons/enzymology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Oligodendroglia/metabolism , Spinal Cord/enzymology
14.
Vet Sci ; 7(2)2020 Jun 16.
Article in English | MEDLINE | ID: mdl-32560159

ABSTRACT

Virtually every cellular process is affected by diet and this represents the foundation of dietary management to a variety of small animal disorders. Special attention is currently being paid to a family of naturally occurring lipid amides acting through the so-called autacoid local injury antagonism, i.e., the ALIA mechanism. The parent molecule of ALIAmides, palmitoyl ethanolamide (PEA), has being known since the 1950s as a nutritional factor with protective properties. Since then, PEA has been isolated from a variety of plant and animal food sources and its proresolving function in the mammalian body has been increasingly investigated. The discovery of the close interconnection between ALIAmides and the endocannabinoid system has greatly stimulated research efforts in this field. The multitarget and highly redundant mechanisms through which PEA exerts prohomeostatic functions fully breaks with the classical pharmacology view of "one drug, one target, one disease", opening a new era in the management of animals' health, i.e., an according-to-nature biomodulation of body responses to different stimuli and injury. The present review focuses on the direct and indirect endocannabinoid receptor agonism by PEA and its analogues and also targets the main findings from experimental and clinical studies on ALIAmides in animal health and wellbeing.

15.
Antioxidants (Basel) ; 9(4)2020 Apr 02.
Article in English | MEDLINE | ID: mdl-32252390

ABSTRACT

TBI (traumatic brain injury) is a major cause of death among youth in industrialized societies. Brain damage following traumatic injury is a result of direct and indirect mechanisms; indirect or secondary injury involves the initiation of an acute inflammatory response, including the breakdown of the blood-brain barrier (BBB), brain edema, infiltration of peripheral blood cells, and activation of resident immunocompetent cells, as well as the release of numerous immune mediators such as interleukins and chemotactic factors. TBI can cause changes in molecular signaling and cellular functions and structures, in addition to tissue damage, such as hemorrhage, diffuse axonal damages, and contusions. TBI typically disturbs brain functions such as executive actions, cognitive grade, attention, memory data processing, and language abilities. Animal models have been developed to reproduce the different features of human TBI, better understand its pathophysiology, and discover potential new treatments. For many years, the first approach to manage TBI has been treatment of the injured tissue with interventions designed to reduce the complex secondary-injury cascade. Several studies in the literature have stressed the importance of more closely examining injuries, including endothelial, microglia, astroglia, oligodendroglia, and precursor cells. Significant effort has been invested in developing neuroprotective agents. The aim of this work is to review TBI pathophysiology and existing and potential new therapeutic strategies in the management of inflammatory events and behavioral deficits associated with TBI.

16.
Bioorg Med Chem ; 28(1): 115195, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31761726

ABSTRACT

N-acylethanolamine acid amidase (NAAA) inhibition represents an exciting novel approach to treat inflammation and pain. NAAA is a cysteine amidase which preferentially hydrolyzes the endogenous biolipids palmitoylethanolamide (PEA) and oleoylethanolamide (OEA). PEA is an endogenous agonist of the nuclear peroxisome proliferator-activated receptor-α (PPAR-α), which is a key regulator of inflammation and pain. Thus, blocking the degradation of PEA with NAAA inhibitors results in augmentation of the PEA/PPAR-α signaling pathway and regulation of inflammatory and pain processes. We have prepared a new series of NAAA inhibitors exploring the azetidine-nitrile (cyanamide) pharmacophore that led to the discovery of highly potent and selective compounds. Key analogs demonstrated single-digit nanomolar potency for hNAAA and showed >100-fold selectivity against serine hydrolases FAAH, MGL and ABHD6, and cysteine protease cathepsin K. Additionally, we have identified potent and selective dual NAAA-FAAH inhibitors to investigate a potential synergism between two distinct anti-inflammatory molecular pathways, the PEA/PPAR-α anti-inflammatory signaling pathway,1-4 and the cannabinoid receptors CB1 and CB2 pathways which are known for their antiinflammatory and antinociceptive properties.5-8 Our ligand design strategy followed a traditional structure-activity relationship (SAR) approach and was supported by molecular modeling studies of reported X-ray structures of hNAAA. Several inhibitors were evaluated in stability assays and demonstrated very good plasma stability (t1/2 > 2 h; human and rodents). The disclosed cyanamides represent promising new pharmacological tools to investigate the potential role of NAAA inhibitors and dual NAAA-FAAH inhibitors as therapeutic agents for the treatment of inflammation and pain.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Cyanamide/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Amidohydrolases/metabolism , Animals , Cyanamide/chemical synthesis , Cyanamide/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Mice , Models, Molecular , Molecular Structure , Rats , Structure-Activity Relationship
17.
Pharmacol Res ; 145: 104264, 2019 07.
Article in English | MEDLINE | ID: mdl-31063807

ABSTRACT

Osteoarthritis (OA), characterized by cartilage damage, synovitis inflammation and chronic pain, is a common degenerative joint disease that may lead to physical disability. In the present study, we first explored the association between N-Acylethanolamine acid amidase (NAAA) and OA progression, and then examined the capability of the NAAA inhibitor F215 to attenuate osteoarthritis. Increased NAAA expressions and decreased PEA levels in synovial membrane and lumbar spinal cord were observed in MIA induced osteoarthritic rats. F215 (i.a., and i.p.) significantly protected against cartilage damage and synovial inflammation by directly increasing PEA levels in joints, or normalization of PEA levels and resolution of inflammation in spinal cord. Moreover, F215 also markedly alleviated osteoarthritic pain in rats, and the therapeutic effects of F215 were blocked by the PPAR-α antagonist MK886. The results revealed that NAAA may has been implicated in OA progression, and treatment with NAAA inhibitor F215 alleviated OA development by preventing cartilage damage, reducing inflammation, and alleviating pain. Our study suggested that NAAA inhibitor might be a novel therapeutic agent for OA treatment.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Inflammatory Agents/therapeutic use , Osteoarthritis/drug therapy , Oxazolidinones/therapeutic use , Pain/drug therapy , Pyrrolidines/therapeutic use , Amides , Amidohydrolases/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cartilage/drug effects , Cartilage/pathology , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Ethanolamines/pharmacology , Injections, Intra-Articular , Injections, Intraperitoneal , Interleukin-6/genetics , Knee Joint/drug effects , Knee Joint/pathology , Male , Matrix Metalloproteinase 13/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Oxazolidinones/pharmacology , PPAR alpha/metabolism , Pain/metabolism , Pain/pathology , Palmitic Acids/pharmacology , Pyrrolidines/pharmacology , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/pathology
18.
Psychopharmacology (Berl) ; 236(9): 2623-2633, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30993360

ABSTRACT

RATIONALE: Oleoyl glycine (OlGly), a recently discovered fatty acid amide that is structurally similar to N- acylethanolamines, which include the endocannabinoid, anandamide (AEA), as well as endogenous peroxisome proliferator-activated receptor alpha (PPARα) agonists oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), has been shown to interfere with nicotine reward and dependence in mice. OBJECTIVES AND METHODS: Behavioral and molecular techniques were used to investigate the ability of OlGly to interfere with the affective properties of morphine and morphine withdrawal (MWD) in male Sprague-Dawley rats. RESULTS: Synthetic OlGly (1-30 mg/kg, intraperitoneal [ip]) produced neither a place preference nor aversion on its own; however, at doses of 1 and 5 mg/kg, ip, it blocked the aversive effects of MWD in a place aversion paradigm. This effect was reversed by the cannabinoid 1 (CB1) receptor antagonist, AM251 (1 mg/kg, ip), but not the PPARα antagonist, MK886 (1 mg/kg, ip). OlGly (5 or 30 mg/kg, ip) did not interfere with a morphine-induced place preference or reinstatement of a previously extinguished morphine-induced place preference. Ex vivo analysis of tissue (nucleus accumbens, amygdala, prefrontal cortex, and interoceptive insular cortex) collected from rats experiencing naloxone-precipitated MWD revealed that OlGly was selectively elevated in the nucleus accumbens. MWD did not modify levels of the endocannabinoids 2-AG and AEA, nor those of the PPARα ligands, OEA and PEA, in any region evaluated. CONCLUSION: Here, we show that OlGly interferes with the aversive properties of acute naloxone-precipitated morphine withdrawal in rats. These results suggest that OlGly may reduce the impact of MWD and may possess efficacy in treating opiate withdrawal.


Subject(s)
Analgesics, Opioid/adverse effects , Glycine/analogs & derivatives , Morphine/adverse effects , Naloxone/toxicity , Oleic Acids/administration & dosage , Reward , Substance Withdrawal Syndrome/drug therapy , Amygdala/drug effects , Amygdala/metabolism , Animals , Dose-Response Relationship, Drug , Glycine/administration & dosage , Glycine/metabolism , Male , Mice , Narcotic Antagonists/toxicity , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Oleic Acids/metabolism , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
19.
Pharmacol Res ; 132: 7-14, 2018 06.
Article in English | MEDLINE | ID: mdl-29572189

ABSTRACT

N-Acylethanolamine acid amidase (NAAA) is a cysteine enzyme that catalyzes the hydrolysis of palmitoylethanolamide (PEA). Pharmacological blockage of NAAA elevates PEA levels and exerts powerful anti-inflammatory activities. We have recently identified a highly potent NAAA inhibitor F215. Here, we demonstrated that F215 was an unusual inflammation-restricted NAAA inhibitor. In lipopolysaccharides (LPS) induced acute lung injury (ALI) model, F215 markedly accelerated inflammation resolution, promoted clearance of neutrophils infiltration and alveolar repair in the lungs. F215 efficiently inhibited NAAA and protected endogenous PEA from degradation in ALI model, but it cannot readily suppress the NAAA activity in naïve mice. The inflammation-restricted effect of F215 was further confirmed in the alveolar macrophage, F215 only increased PEA levels and exerted anti-inflammatory effects in activated macrophages, but not in unstimulated macrophages. Moreover, we also showed that the pharmacological effects of F215 were restricted to the local inflamed skin elicited by 12-o-tetradecanoylphorbol-13-acetate (TPA), but not the normal tissues. We believe that F215 could be a useful probe to investigate the function of NAAA, as well as a potent anti-inflammatory agent, and its inflammation-restricted feature might offer a new approach to prevent potential side effects of systemic enzyme inhibition.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Inflammatory Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Oxazoles/therapeutic use , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Edema/chemically induced , Edema/drug therapy , Enzyme Inhibitors/pharmacology , Lipopolysaccharides , Macrophages, Alveolar/drug effects , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Oxazoles/pharmacology , Tetradecanoylphorbol Acetate/analogs & derivatives
20.
J Affect Disord ; 232: 127-133, 2018 05.
Article in English | MEDLINE | ID: mdl-29486338

ABSTRACT

BACKGROUND: Experimental studies provide evidence for antidepressant effects of Palmitoylethanolamide (PEA) in animal models of depression. We aimed to evaluate the efficacy and tolerability of PEA add-on therapy in treatment of patients with major depressive disorder (MDD). METHODS: In a randomized double-blind, and placebo-controlled study, 58 patients with MDD (DSM-5) and Hamilton Depression Rating Scale (HAM-D) score ≥ 19 were randomized to receive either 600 mg twice daily Palmitoylethanolamide or placebo in addition to citalopram for six weeks. Patients were assessed using the HAM-D scale at baseline and weeks 2, 4, and 6. RESULTS: Fifty-four individuals completed the trial. At week 2, patients in the PEA group demonstrated significantly greater reduction in HAM-D scores compared to the placebo group (8.30 ± 2.41 vs. 5.81 ± 3.57, P = .004). The PEA group also demonstrated significantly greater improvement in depressive symptoms [F (3, 156) = 3.35, P = .021] compared to the placebo group throughout the trial period. The patients in the PEA group experienced more response rate (≥ 50% reduction in the HAM-D score) than the placebo group (100% vs. 74% respectively, P = .01) at the end of the trial. Baseline parameters and frequency of side effects were not significantly different between the two groups. LIMITATIONS: The population size in this study was small and the follow-up period was relatively short. CONCLUSIONS: Palmitoylethanolamide adjunctive therapy to citalopram can effectively improve symptoms of patients (predominantly male gender) with major depressive disorder. PEA showed rapid-onset antidepressant effects which need further investigation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antidepressive Agents/therapeutic use , Citalopram/therapeutic use , Depressive Disorder, Major/drug therapy , Ethanolamines/therapeutic use , Palmitic Acids/therapeutic use , Adult , Amides , Chemotherapy, Adjuvant , Depressive Disorder, Major/physiopathology , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL