1
|
Batacan R, Briskey D, Bajagai YS, Smith C, Stanley D, Rao A. Effect of Palmitoylethanolamide Compared to a Placebo on the Gut Microbiome and Biochemistry in an Overweight Adult Population: A Randomised, Placebo Controlled, Double-Blind Study. Biomedicines 2024; 12:1620. [PMID: 39062193 PMCID: PMC11274356 DOI: 10.3390/biomedicines12071620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
This study investigates the effects of palmitoylethanolamide (PEA) on the gut microbiome of overweight adults. Fifty-eight participants (twenty males, thirty-eight females) aged 18-65 years with a BMI range of 30-40 kg/m2 were recruited. Participants were randomised to receive PEA (n = 36) or a placebo (n = 22) for 12 weeks. Microbiota composition, richness, diversity, and metabolic functions, faecal short chain fatty acids and calprotectin, pathology markers, and health-related questionnaires were analysed throughout the 12 weeks of supplementation. PEA supplementation significantly reduced triglyceride levels and IL-2 concentrations. No significant differences were found in the overall microbiota composition between the groups, and microbiota richness and diversity remained consistent for both groups. Functional analysis demonstrated no differences in functional richness and diversity, but specific pathways were modified. PEA supplementation resulted in a decrease in the abundance of pathways related to aromatic compound degradation, NAD interconversion, and L-glutamate degradation, while pathways associated with molybdopterin biosynthesis and O-antigen building blocks exhibited increased abundance. Increased production of O-antigen results in smooth LPS associated with reduced pathogenic stealth and persistence. PEA supplementation may influence specific microbial species, metabolic pathways, and reduce serum triglyceride and IL-2 concentration, shedding light on the intricate relationship between PEA, the microbiome, and host health.
Collapse
Affiliation(s)
- Romeo Batacan
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia (Y.S.B.); (D.S.)
| | - David Briskey
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD 4006, Australia;
- RDC Clinical, Brisbane, QLD 4006, Australia;
| | - Yadav Sharma Bajagai
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia (Y.S.B.); (D.S.)
| | - Chelsie Smith
- RDC Clinical, Brisbane, QLD 4006, Australia;
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Dana Stanley
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia (Y.S.B.); (D.S.)
| | - Amanda Rao
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD 4006, Australia;
- RDC Clinical, Brisbane, QLD 4006, Australia;
| |
Collapse
|
2
|
Munzen ME, Reguera Gomez M, Hamed MF, Enriquez V, Charles-Niño CL, Dores MR, Alviña K, Martinez LR. Palmitoylethanolamide shows limited efficacy in controlling cerebral cryptococcosis in vivo. Antimicrob Agents Chemother 2023; 67:e0045923. [PMID: 37750714 PMCID: PMC10583666 DOI: 10.1128/aac.00459-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/13/2023] [Indexed: 09/27/2023] Open
Abstract
Cryptococcus neoformans (Cn) is an encapsulated neurotropic fungal pathogen and the causative agent of cryptococcal meningoencephalitis (CME) in humans. Recommended treatment for CME is Amphotericin B (AmpB) and 5-fluorocytosine (5-FC). Though effective, AmpB has displayed numerous adverse side effects due to its potency and nephrotoxicity, prompting investigation into alternative treatments. Palmitoylethanolamide (PEA) is an immunomodulatory compound capable of promoting neuroprotection and reducing inflammation. To investigate the efficacy of PEA as a therapeutic alternative for CME, we intracerebrally infected mice with Cn and treated them with PEA or AmpB alone or in combination. Our results demonstrate that PEA alone does not significantly prolong survival nor reduce fungal burden, but when combined with AmpB, PEA exerts an additive effect and promotes both survivability and fungal clearance. However, we compared this combination to traditional AmpB and 5-FC treatment in a survivability study and observed lower efficacy. Overall, our study revealed that PEA alone is not effective as an antifungal agent in the treatment of CME. Importantly, we describe the therapeutic capability of PEA in the context of Cn infection and show that its immunomodulatory properties may confer limited protection when combined with an effective fungicidal agent.
Collapse
Affiliation(s)
- Melissa E. Munzen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Marta Reguera Gomez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Mohamed F. Hamed
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Vanessa Enriquez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Claudia L. Charles-Niño
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Michael R. Dores
- Department of Biology, Hofstra University, Hempstead, New York, USA
| | - Karina Alviña
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
- Center for Translational Research in Neurodegenerative Disease, Gainesville, Florida, USA
| | - Luis R. Martinez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
- Center for Translational Research in Neurodegenerative Disease, Gainesville, Florida, USA
- Center for Immunology and Transplantation, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
3
|
Voicu V, Brehar FM, Toader C, Covache-Busuioc RA, Corlatescu AD, Bordeianu A, Costin HP, Bratu BG, Glavan LA, Ciurea AV. Cannabinoids in Medicine: A Multifaceted Exploration of Types, Therapeutic Applications, and Emerging Opportunities in Neurodegenerative Diseases and Cancer Therapy. Biomolecules 2023; 13:1388. [PMID: 37759788 PMCID: PMC10526757 DOI: 10.3390/biom13091388] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
In this review article, we embark on a thorough exploration of cannabinoids, compounds that have garnered considerable attention for their potential therapeutic applications. Initially, this article delves into the fundamental background of cannabinoids, emphasizing the role of endogenous cannabinoids in the human body and outlining their significance in studying neurodegenerative diseases and cancer. Building on this foundation, this article categorizes cannabinoids into three main types: phytocannabinoids (plant-derived cannabinoids), endocannabinoids (naturally occurring in the body), and synthetic cannabinoids (laboratory-produced cannabinoids). The intricate mechanisms through which these compounds interact with cannabinoid receptors and signaling pathways are elucidated. A comprehensive overview of cannabinoid pharmacology follows, highlighting their absorption, distribution, metabolism, and excretion, as well as their pharmacokinetic and pharmacodynamic properties. Special emphasis is placed on the role of cannabinoids in neurodegenerative diseases, showcasing their potential benefits in conditions such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. The potential antitumor properties of cannabinoids are also investigated, exploring their potential therapeutic applications in cancer treatment and the mechanisms underlying their anticancer effects. Clinical aspects are thoroughly discussed, from the viability of cannabinoids as therapeutic agents to current clinical trials, safety considerations, and the adverse effects observed. This review culminates in a discussion of promising future research avenues and the broader implications for cannabinoid-based therapies, concluding with a reflection on the immense potential of cannabinoids in modern medicine.
Collapse
Affiliation(s)
- Victor Voicu
- Pharmacology, Toxicology and Clinical Psychopharmacology, “Carol Davila” University of Medicine and Pharmacy in Bucharest, 020021 Bucharest, Romania;
- Medical Section within the Romanian Academy, 010071 Bucharest, Romania
| | - Felix-Mircea Brehar
- Neurosurgery Department, Emergency Clinical Hospital Bagdasar-Arseni, 041915 Bucharest, Romania
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Antonio Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Andrei Bordeianu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
4
|
Xiao J, Zhou Y, Sun L, Wang H. Role of integrating cannabinoids and the endocannabinoid system in neonatal hypoxic-ischaemic encephalopathy. Front Mol Neurosci 2023; 16:1152167. [PMID: 37122621 PMCID: PMC10130673 DOI: 10.3389/fnmol.2023.1152167] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/16/2023] [Indexed: 05/02/2023] Open
Abstract
Neonatal hypoxic-ischaemic events, which can result in long-term neurological impairments or even cell death, are among the most significant causes of brain injury during neurodevelopment. The complexity of neonatal hypoxic-ischaemic pathophysiology and cellular pathways make it difficult to treat brain damage; hence, the development of new neuroprotective medicines is of great interest. Recently, numerous neuroprotective medicines have been developed to treat brain injuries and improve long-term outcomes based on comprehensive knowledge of the mechanisms that underlie neuronal plasticity following hypoxic-ischaemic brain injury. In this context, understanding of the medicinal potential of cannabinoids and the endocannabinoid system has recently increased. The endocannabinoid system plays a vital neuromodulatory role in numerous brain regions, ensuring appropriate control of neuronal activity. Its natural neuroprotection against adult brain injury or acute brain injury also clearly demonstrate the role of endocannabinoid signalling in modulating neuronal activity in the adult brain. The goal of this review is to examine how cannabinoid-derived compounds can be used to treat neonatal hypoxic-ischaemic brain injury and to assess the critical function of the endocannabinoid system and its potential for use as a new neuroprotective treatment for neonatal hypoxic-ischaemic brain injury.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Pathology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Yue Zhou
- Department of Pharmacy, Xindu District People’s Hospital of Chengdu, Chengdu, China
| | - Luqiang Sun
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haichuan Wang
- Department of Paediatrics, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Haichuan Wang,
| |
Collapse
|
5
|
Palmisano M, Gargano A, Olabiyi BF, Lutz B, Bilkei-Gorzo A. Hippocampal Deletion of CB1 Receptor Impairs Social Memory and Leads to Age-Related Changes in the Hippocampus of Adult Mice. Int J Mol Sci 2022; 24:ijms24010026. [PMID: 36613469 PMCID: PMC9819823 DOI: 10.3390/ijms24010026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Endocannabinoid system activity declines with age in the hippocampus, along with the density of the cannabinoid receptor type-1 (CB1). This process might contribute to brain ageing, as previous studies showed that the constitutive deletion of the CB1 receptor in mice leads to early onset of memory deficits and histological signs of ageing in the hippocampus including enhanced pro-inflammatory glial activity and reduced neurogenesis. Here we asked whether the CB1 receptor exerts its activity locally, directly influencing hippocampal ageing or indirectly, accelerating systemic ageing. Thus, we deleted the CB1 receptor site-specifically in the hippocampus of 2-month-old CB1flox/flox mice using stereotaxic injections of rAAV-Cre-Venus viruses and assessed their social recognition memory four months later. Mice with hippocampus-specific deletion of the CB1 receptor displayed a memory impairment, similarly as observed in constitutive knockouts at the same age. We next analysed neuroinflammatory changes in the hippocampus, neuronal density and cell proliferation. Site-specific mutant mice had enhanced glial cell activity, up-regulated levels of TNFα in the hippocampus and decreased cell proliferation, specifically in the subgranular zone of the dentate gyrus. Our data indicate that a local activity of the CB1 receptor in the hippocampus is required to maintain neurogenesis and to prevent neuroinflammation and cognitive decline.
Collapse
Affiliation(s)
- Michela Palmisano
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53125 Bonn, Germany
| | - Alessandra Gargano
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53125 Bonn, Germany
| | | | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53125 Bonn, Germany
- Correspondence: ; Tel.: +49-0228-6885-317
| |
Collapse
|
6
|
Wu W, Teng Y, Tian M, Huang B, Deng Y, Li H, Yuan H, Chen J, Li X, Zhou C. Tissue-specific metabolomic profiling after cardiopulmonary bypass in fetal sheep. Front Cardiovasc Med 2022; 9:1009165. [PMID: 36578834 PMCID: PMC9791045 DOI: 10.3389/fcvm.2022.1009165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
Objective Fetal cardiopulmonary bypass (CPB) is essential to fetal heart surgery, while its development is limited by vital organ dysfunction after CPB. Studying organ metabolism may help to solve this problem. The objective of this study was to describe the tissue-specific metabolic fingerprints of fetal sheep under CPB and to associate them with organ functions. Methods Ten pregnant ewes at 90-120 days of gestation were randomly divided into two groups. The bypass group underwent a 1-h fetal CPB, whereas the control group underwent only a fetal sternotomy. During bypass, echocardiography, blood gases, and blood biochemistry were measured. After bypass, lambs were sacrificed, and tissues of the heart, liver, brain, kidney, and placenta were harvested. The metabolites extracted from these tissues were analyzed using non-targeted metabolomics based on liquid chromatography-mass spectrometry techniques. Results All tissues except the placenta displayed significant metabolic changes, and the fetal heart displayed obvious functional changes. Fetal sheep that underwent CPB had common and tissue-specific metabolic signatures. These changes can be attributed to dysregulated lipid metabolism, altered amino acid metabolism, and the accumulation of plasticizer metabolism. Conclusion Fetal CPB causes tissue-specific metabolic changes in fetal sheep. Studying these metabolic changes, especially cardiac metabolism, is of great significance for the study of fetal CPB.
Collapse
Affiliation(s)
- Wentao Wu
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yun Teng
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Miao Tian
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Bingxin Huang
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuhang Deng
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huili Li
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Haiyun Yuan
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jimei Chen
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaohong Li
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,*Correspondence: Xiaohong Li
| | - Chengbin Zhou
- Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,Chengbin Zhou
| |
Collapse
|
7
|
Abedini T, Hosseyni R, Ghannadi F, Moghaddam HS, Ardakani MRK, Talaei A, Akhondzadeh S. Efficacy and safety of palmitoylethanolamide as an adjunctive treatment for acute mania: A randomized, double-blind, placebo-controlled trial. Psychiatry Clin Neurosci 2022; 76:505-511. [PMID: 35737597 DOI: 10.1111/pcn.13441] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/07/2022] [Accepted: 06/19/2022] [Indexed: 11/30/2022]
Abstract
AIM Palmitoylethanolamide is an endogenous fatty acid amide with neuroprotective and anti-inflammatory actions. We performed a randomized, double-blind, placebo-controlled clinical trial to investigate the efficacy and safety of palmitoylethanolamide combination therapy in acute mania. METHODS Patients in the acute phase of mania were assigned into two parallel groups given either lithium (blood level of 0.8-1.1 mEq/L) and risperidone 3 mg plus palmitoylethanolamide 600 mg or placebo twice per day for 6 weeks. All participants were assessed with the Young Mania Rating Scale (YMRS), Hamilton Depression Rating Scale (HDRS), and Extrapyramidal Symptom Rating Scale (ESRS) at baseline and at weeks 1, 2, 4, and 6. RESULTS A total of 63 patients (32 in palmitoylethanolamide and 31 in placebo groups) completed the trial. We found a significant effect for time×treatment interaction on the YMRS score (F = 5.22, d.f. = 2.34, P= 0.004) from baseline to study end point. Results from independent t test showed a significantly greater decrease in YMRS scores in the palmitoylethanolamide group, compared with the placebo group, from baseline to weeks 4 and 6 (P= 0.018 and P= 0.002, respectively). There was no significant difference between palmitoylethanolamide and placebo groups based on ESRS scores or ESRS changes in scores (P>0.05). CONCLUSIONS Our findings provide preliminary evidence that palmitoylethanolamide is an effective adjunctive medication that improves manic symptoms and overall clinical status in acute episodes of mania. However, larger sample sizes and more extended follow-up therapy are needed in future studies to confirm our findings.
Collapse
Affiliation(s)
- Talieh Abedini
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Hosseyni
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Ghannadi
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Ali Talaei
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Pickering E, Steels EL, Steadman KJ, Rao A, Vitetta L. A randomized controlled trial assessing the safety and efficacy of palmitoylethanolamide for treating diabetic-related peripheral neuropathic pain. Inflammopharmacology 2022; 30:2063-2077. [PMID: 36057884 PMCID: PMC9700575 DOI: 10.1007/s10787-022-01033-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/04/2022]
Abstract
Background Peripheral neuropathy is a common complication of diabetes. The management of the associated neuropathic pain remains difficult to treat. Objective This study explored the safety, tolerability and efficacy of a palmitoylethanolamide (PEA) formulation in treating diabetic-related peripheral neuropathic pain (PNP). Secondary outcomes included systemic inflammation, sleep and mood changes in patients diagnosed with type 1 and type 2 diabetes and PNP. Design This study was a single-centre, quadruple-blinded, placebo-controlled trial with 70 participants receiving 600 mg of PEA or placebo daily, for 8 weeks, with a 94% rate of study participation completion. Primary outcomes were neuropathic pain and specific pain types (the BPI-DPN and NPSI). The secondary outcomes were sleep quality (MOS sleep scale), mood (DASS-21), glucose metabolism and inflammation. Results There was a significant reduction (P ≤ 0.001) in BPI-DPN total pain and pain interference, NPSI total score and sub-scores, except for evoked pain (P = 0.09) in the PEA group compared with the placebo group. The MOS sleep problem index and sub-scores significantly improved (P ≤ 0.001). DASS-21 depression scores significantly reduced (P = 0.03), but not anxiety or stress scores. Interleukin-6 and elevated C-reactive protein levels significantly reduced in the PEA group (P = 0.05), with no differences in fibrinogen between groups (P = 0.78) at treatment completion. There were no changes in safety pathology parameters, and the treatment was well tolerated. Conclusions The study demonstrated that the PEA formulation reduced diabetic peripheral neuropathic pain and inflammation along with improving mood and sleep. Further studies on the mechanistic effectiveness of PEA as an adjunct medicine and as a monotherapy pain analgesic are warranted. Clinical Trial Registration Registry name: Australian New Zealand Clinical Trials Registry (ANZCTR), Registration number: ACTRN12620001302943, Registration link: https://anzctr.org.au/Trial/Registration/TrialReview.aspx?id=380826, Actual study start date: 20 November 2020.
Collapse
Affiliation(s)
- Emily Pickering
- School of Pharmacy, University of Queensland, PACE Precinct, 20 Cornwall Street, Wooloongabba, Brisbane, QLD, 4102, Australia.,Evidence Sciences Pty. Ltd., Brisbane, QLD, Australia
| | - Elizabeth L Steels
- School of Pharmacy, University of Queensland, PACE Precinct, 20 Cornwall Street, Wooloongabba, Brisbane, QLD, 4102, Australia. .,Evidence Sciences Pty. Ltd., Brisbane, QLD, Australia.
| | - Kathryn J Steadman
- School of Pharmacy, University of Queensland, PACE Precinct, 20 Cornwall Street, Wooloongabba, Brisbane, QLD, 4102, Australia
| | - Amanda Rao
- School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, QLD, 4102, Australia
| | - Luis Vitetta
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Vicente-Acosta A, Ceprian M, Sobrino P, Pazos MR, Loría F. Cannabinoids as Glial Cell Modulators in Ischemic Stroke: Implications for Neuroprotection. Front Pharmacol 2022; 13:888222. [PMID: 35721207 PMCID: PMC9199389 DOI: 10.3389/fphar.2022.888222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is the second leading cause of death worldwide following coronary heart disease. Despite significant efforts to find effective treatments to reduce neurological damage, many patients suffer from sequelae that impair their quality of life. For this reason, the search for new therapeutic options for the treatment of these patients is a priority. Glial cells, including microglia, astrocytes and oligodendrocytes, participate in crucial processes that allow the correct functioning of the neural tissue, being actively involved in the pathophysiological mechanisms of ischemic stroke. Although the exact mechanisms by which glial cells contribute in the pathophysiological context of stroke are not yet completely understood, they have emerged as potentially therapeutic targets to improve brain recovery. The endocannabinoid system has interesting immunomodulatory and protective effects in glial cells, and the pharmacological modulation of this signaling pathway has revealed potential neuroprotective effects in different neurological diseases. Therefore, here we recapitulate current findings on the potential promising contribution of the endocannabinoid system pharmacological manipulation in glial cells for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Andrés Vicente-Acosta
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maria Ceprian
- ERC Team, PGNM, INSERM U1315, CNRS UMR5261, University of Lyon 1, University of Lyon, Lyon, France
| | - Pilar Sobrino
- Departamento de Neurología, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Maria Ruth Pazos
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Frida Loría
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| |
Collapse
|
10
|
van den Hoogen NJ, Harding EK, Davidson CED, Trang T. Cannabinoids in Chronic Pain: Therapeutic Potential Through Microglia Modulation. Front Neural Circuits 2022; 15:816747. [PMID: 35069129 PMCID: PMC8777271 DOI: 10.3389/fncir.2021.816747] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/17/2021] [Indexed: 12/16/2022] Open
Abstract
Chronic pain is a complex sensory, cognitive, and emotional experience that imposes a great personal, psychological, and socioeconomic burden on patients. An estimated 1.5 billion people worldwide are afflicted with chronic pain, which is often difficult to treat and may be resistant to the potent pain-relieving effects of opioid analgesics. Attention has therefore focused on advancing new pain therapies directed at the cannabinoid system because of its key role in pain modulation. Endocannabinoids and exogenous cannabinoids exert their actions primarily through Gi/o-protein coupled cannabinoid CB1 and CB2 receptors expressed throughout the nervous system. CB1 receptors are found at key nodes along the pain pathway and their activity gates both the sensory and affective components of pain. CB2 receptors are typically expressed at low levels on microglia, astrocytes, and peripheral immune cells. In chronic pain states, there is a marked increase in CB2 expression which modulates the activity of these central and peripheral immune cells with important consequences for the surrounding pain circuitry. Growing evidence indicate that interventions targeting CB1 or CB2 receptors improve pain outcomes in a variety of preclinical pain models. In this mini-review, we will highlight recent advances in understanding how cannabinoids modulate microglia function and its implications for cannabinoid-mediated analgesia, focusing on microglia-neuron interactions within the spinal nociceptive circuitry.
Collapse
Affiliation(s)
- Nynke J. van den Hoogen
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Erika K. Harding
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Chloé E. D. Davidson
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Tuan Trang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- *Correspondence: Tuan Trang
| |
Collapse
|
11
|
Hashiesh HM, Jha NK, Sharma C, Gupta PK, Jha SK, Patil CR, Goyal SN, Ojha SK. Pharmacological potential of JWH133, a cannabinoid type 2 receptor agonist in neurodegenerative, neurodevelopmental and neuropsychiatric diseases. Eur J Pharmacol 2021; 909:174398. [PMID: 34332924 DOI: 10.1016/j.ejphar.2021.174398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/15/2021] [Accepted: 07/28/2021] [Indexed: 12/09/2022]
Abstract
The pharmacological activation of cannabinoid type 2 receptors (CB2R) gained attention due to its ability to mitigate neuroinflammatory events without eliciting psychotropic actions, a limiting factor for the drugs targeting cannabinoid type 1 receptors (CB1R). Therefore, ligands activating CB2R are receiving enormous importance for therapeutic targeting in numerous neurological diseases including neurodegenerative, neuropsychiatric and neurodevelopmental disorders as well as traumatic injuries and neuropathic pain where neuroinflammation is a common accompaniment. Since the characterization of CB2R, many CB2R selective synthetic ligands have been developed with high selectivity and functional activity. Among numerous ligands, JWH133 has been found one of the compounds with high selectivity for CB2R. JWH133 has been reported to exhibit numerous pharmacological activities including antioxidant, anti-inflammatory, anticancer, cardioprotective, hepatoprotective, gastroprotective, nephroprotective, and immunomodulatory. Recent studies have shown that JWH133 possesses potent neuroprotective properties in several neurological disorders, including neuropathic pain, anxiety, epilepsy, depression, alcoholism, psychosis, stroke, and neurodegeneration. Additionally, JWH133 showed to protect neurons from oxidative damage and inflammation, promote neuronal survival and neurogenesis, and serve as an immunomodulatory agent. The present review comprehensively examined neuropharmacological activities of JWH133 in neurological disorders including neurodegenerative, neurodevelopmental and neuropsychiatric using synoptic tables and elucidated pharmacological mechanisms based on reported observations. Considering the cumulative data, JWH133 appears to be a promising CB2R agonist molecule for further evaluation and it can be a prototype agent in drug discovery and development for a unique class of agents in neurotherapeutics. Further, regulatory toxicology and pharmacokinetic studies are required to determine safety and proceed for clinical evaluation.
Collapse
Affiliation(s)
- Hebaallah Mamdouh Hashiesh
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, PO Box - 17666, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, PO Box - 17666, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Piyush Kumar Gupta
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, 201310, Uttar Pradesh, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Chandragouda R Patil
- Department of Pharmacology, Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, New Delhi, 110017, India
| | - Sameer N Goyal
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Shreesh K Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, PO Box - 17666, United Arab Emirates University, Al Ain, United Arab Emirates.
| |
Collapse
|
12
|
Parisi S, Ditto MC, Borrelli R, Fusaro E. Efficacy of a fixed combination of palmitoylethanolamide and acetyl-l-carnitine (PEA+ALC FC) in the treatment of neuropathies secondary to rheumatic diseases. Minerva Med 2021; 112:492-499. [PMID: 34056884 DOI: 10.23736/s0026-4806.21.07486-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The neurologic complications of rheumatic diseases (RDs) are highly variable, and their manifestations are linked to the pathogenesis and clinical phenotype of the specific RDs. In rheumatoid arthritis, for example, the peripheral nervous system is most commonly involved and mononeuritis multiplex, nerve entrapment and vasculitic sensorimotor neuropathies are not uncommon. Often the therapy for these disorders is not easy and is characterized by the use of different drugs. Palmitoylethanolamide (PEA) has been tested in a wide variety of animal models and has been evaluated in several clinical studies for nerve compression syndromes, demonstrating that PEA acts as an effective and safe analgesic compound. Acetyl-L-Carnitine (ALC) has also been shown to be an effective and safe treatment in painful peripheral neuropathy. In the last years the synergistic effect between PEA and ALC has been demonstrated. The aim of our study was to evaluate the efficacy of supplementation of standard therapy (STh) with Kalanit® (Chiesi Italia Spa; Parma, Italy) in patients with peripheral neuropathy secondary to RDs. METHODS Patients at the time of enrollment were affected by RDs with neuropathy from <12 months, documented by electromyography. The analyzed patients were treated with the STh chosen according to their rheumatic disease (RA or SpA) and for their neuropathy (e.g. analgesic, NSAIDs, pregabalin or gabapentin) as per clinical practice. The sample was divided into 2 groups: group 1, patients treated with STh, to which a fixed combination of PEA (600 mg) + ALC (500 mg) (Kalanit®) was added twice a day for 2 weeks and then once a day for 6 months; group 2, patients treated only with STh. Each patient underwent clinical evaluations and questionnaires were administered in order to evaluate their neuropathy and the efficacy of the therapy. RESULTS In group 1, 18 patients suffering from sciatic pain, 16 patients from carpal tunnel syndrome and 8 patients with peripheral neuropathy of the lower limbs were included and PEA + ALC FC was added to STh. These patients were compared with patients from group 2, who had the same pathology and demographic characteristics: 20 patients with sciatic pain, 15 with carpal tunnel syndrome and 5 with peripheral neuropathy of the lower limbs, respectively; this group was treated with STh only. Patients treated with PEA + ALC FC had a significant improvement in pain VAS compared to patients treated with group 2 in all the diseases analyzed (P value: sciatic pain 0.032, carpal tunnel syndrome 0.025 and lower limbs neuropathy 0.041). Patients in group 1 showed a significant improvement compared to patients treated in group 2 also from a specific score. Specifically, LBP-IQ showed significant improvement in group one (P value: 0.031), as did CHFD (P=0.011) and NPQ (P=0.025). CONCLUSIONS The synergistic effect of PEA and ALC seems to have a further advantage in the treatment of this type of pathology, including the anti-inflammatory effect but also in terms of therapy optimization and therefore of better adherence to treatments. Our study shows that it is important to identify the type of pain to follow an accurate diagnostic algorithm, considering the clinical characteristics of the patient and carefully evaluate the indication, preferring a multimodal approach.
Collapse
Affiliation(s)
- Simone Parisi
- Unit of Rheumatology, Città della Salute e della Scienza, Turin, Italy -
| | - Maria C Ditto
- Unit of Rheumatology, Città della Salute e della Scienza, Turin, Italy
| | - Richard Borrelli
- Unit of Rheumatology, Città della Salute e della Scienza, Turin, Italy
| | - Enrico Fusaro
- Unit of Rheumatology, Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
13
|
Armeli F, Bonucci A, Maggi E, Pinto A, Businaro R. Mediterranean Diet and Neurodegenerative Diseases: The Neglected Role of Nutrition in the Modulation of the Endocannabinoid System. Biomolecules 2021; 11:biom11060790. [PMID: 34073983 PMCID: PMC8225112 DOI: 10.3390/biom11060790] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/17/2022] Open
Abstract
Neurodegenerative disorders are a widespread cause of morbidity and mortality worldwide, characterized by neuroinflammation, oxidative stress and neuronal depletion. The broad-spectrum neuroprotective activity of the Mediterranean diet is widely documented, but it is not yet known whether its nutritional and caloric balance can induce a modulation of the endocannabinoid system. In recent decades, many studies have shown how endocannabinoid tone enhancement may be a promising new therapeutic strategy to counteract the main hallmarks of neurodegeneration. From a phylogenetic point of view, the human co-evolution between the endocannabinoid system and dietary habits could play a key role in the pro-homeostatic activity of the Mediterranean lifestyle: this adaptive balance among our ancestors has been compromised by the modern Western diet, resulting in a “clinical endocannabinoid deficiency syndrome”. This review aims to evaluate the evidence accumulated in the literature on the neuroprotective, immunomodulatory and antioxidant properties of the Mediterranean diet related to the modulation of the endocannabinoid system, suggesting new prospects for research and clinical interventions against neurodegenerative diseases in light of a nutraceutical paradigm.
Collapse
Affiliation(s)
- Federica Armeli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica, 79, 04100 Latina, Italy; (F.A.); (A.B.); (E.M.)
| | - Alessio Bonucci
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica, 79, 04100 Latina, Italy; (F.A.); (A.B.); (E.M.)
| | - Elisa Maggi
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica, 79, 04100 Latina, Italy; (F.A.); (A.B.); (E.M.)
| | - Alessandro Pinto
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica, 79, 04100 Latina, Italy; (F.A.); (A.B.); (E.M.)
- Correspondence:
| |
Collapse
|
14
|
Kasatkina LA, Rittchen S, Sturm EM. Neuroprotective and Immunomodulatory Action of the Endocannabinoid System under Neuroinflammation. Int J Mol Sci 2021; 22:ijms22115431. [PMID: 34063947 PMCID: PMC8196612 DOI: 10.3390/ijms22115431] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/17/2022] Open
Abstract
Endocannabinoids (eCBs) are lipid-based retrograde messengers with a relatively short half-life that are produced endogenously and, upon binding to the primary cannabinoid receptors CB1/2, mediate multiple mechanisms of intercellular communication within the body. Endocannabinoid signaling is implicated in brain development, memory formation, learning, mood, anxiety, depression, feeding behavior, analgesia, and drug addiction. It is now recognized that the endocannabinoid system mediates not only neuronal communications but also governs the crosstalk between neurons, glia, and immune cells, and thus represents an important player within the neuroimmune interface. Generation of primary endocannabinoids is accompanied by the production of their congeners, the N-acylethanolamines (NAEs), which together with N-acylneurotransmitters, lipoamino acids and primary fatty acid amides comprise expanded endocannabinoid/endovanilloid signaling systems. Most of these compounds do not bind CB1/2, but signal via several other pathways involving the transient receptor potential cation channel subfamily V member 1 (TRPV1), peroxisome proliferator-activated receptor (PPAR)-α and non-cannabinoid G-protein coupled receptors (GPRs) to mediate anti-inflammatory, immunomodulatory and neuroprotective activities. In vivo generation of the cannabinoid compounds is triggered by physiological and pathological stimuli and, specifically in the brain, mediates fine regulation of synaptic strength, neuroprotection, and resolution of neuroinflammation. Here, we review the role of the endocannabinoid system in intrinsic neuroprotective mechanisms and its therapeutic potential for the treatment of neuroinflammation and associated synaptopathy.
Collapse
Affiliation(s)
- Ludmila A. Kasatkina
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sonja Rittchen
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
| | - Eva M. Sturm
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
- Correspondence:
| |
Collapse
|
15
|
Petrosino S, Schiano Moriello A. Palmitoylethanolamide: A Nutritional Approach to Keep Neuroinflammation within Physiological Boundaries-A Systematic Review. Int J Mol Sci 2020; 21:E9526. [PMID: 33333772 PMCID: PMC7765232 DOI: 10.3390/ijms21249526] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/30/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
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.
Collapse
Affiliation(s)
- Stefania Petrosino
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Napoli, Italy;
- Epitech Group SpA, Via Einaudi 13, 35030 Padova, Italy
| | - Aniello Schiano Moriello
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Napoli, Italy;
- Epitech Group SpA, Via Einaudi 13, 35030 Padova, Italy
| |
Collapse
|
16
|
Palmitoylethanolamide and Related ALIAmides: Prohomeostatic Lipid Compounds for Animal Health and Wellbeing. Vet Sci 2020; 7:vetsci7020078. [PMID: 32560159 PMCID: PMC7355440 DOI: 10.3390/vetsci7020078] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023] Open
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.
Collapse
|
17
|
Tolerability of Palmitoylethanolamide in a Pediatric Population Suffering from Migraine: A Pilot Study. Pain Res Manag 2020; 2020:3938640. [PMID: 32377286 PMCID: PMC7196162 DOI: 10.1155/2020/3938640] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/28/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022]
Abstract
Background Palmitoylethanolamide (PEA) is emerging as a new therapeutic approach in pain and inflammatory conditions, and it has been evaluated in studies on various painful diseases. The aim of this open-label study was to evaluate the efficacy of ultramicronized PEA (umPEA) in the prophylactic treatment of migraine. Methods The study included 70 patients with mean age of 10.3 ± 2.7 (24.5% M and 75.5% F). All patients had a diagnosis of migraine without aura (ICHD 3 criteria) and received umPEA (600 mg/day orally) for three months. We compared the attack frequency (AF) and attack intensity at baseline and after three months. Patients were asked to classify the intensity of the attack with a value ranging from 1 to 3, where 1 means mild attack, 2 moderate, and 3 severe attack. Results Nine patients discontinued treatment before the target time of 12 weeks. After 3 months of treatment with umPEA, the headache frequency was reduced by >50% per month in 63.9% patients. The number of monthly attacks at T1 decreased significantly compared with the baseline assessment (from 13.9 ± 7.5 SD of T0 to 6.5 ± 5.9 SD of T1; p < 0.001). The mean intensity of the attacks dropped from 1.67 ± 0.6 (T0) to 1.16 ± 0.5 (T1) (p < 0.001), and the percentage of patients with severe attacks decreased after treatment (from 8.2% to 1.6%; p < 0.05). The monthly assumptions of drugs for the attack reduced from 9.5 ± 4.4 to 4.9 ± 2.5 (p < 0.001). Only one patient developed mild side effects (nausea and floating). Conclusions Our preliminary data show that umPEA administered for three month reduces pain intensity and the number of attacks per month in pediatric patients with migraine. Although the small number of patients and the lack of control group do not allow us to consider these initial results as definitely reliable, they encourage us to expand the sample.
Collapse
|
18
|
Xin M, Feng J, Hao Y, You J, Wang X, Yin X, Shang P, Ma D. Cyclic adenosine monophosphate in acute ischemic stroke: some to update, more to explore. J Neurol Sci 2020; 413:116775. [PMID: 32197118 DOI: 10.1016/j.jns.2020.116775] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/14/2022]
Abstract
The development of effective treatment for ischemic stroke, which is a common cause of morbidity and mortality worldwide, remains an unmet goal because the current first-line treatment management interventional therapy has a strict time window and serious complications. In recent years, a growing body of evidence has shown that the elevation of intracellular and extracellular cyclic adenosine monophosphate (cAMP) alleviates brain damage after ischemic stroke by attenuating neuroinflammation in the central nervous system and peripheral immune system. In the central nervous system, upregulated intracellular cAMP signaling can alleviate immune-mediated damage by restoring neuronal morphology and function, inhibiting microglia migration and activation, stabilizing the membrane potential of astrocytes and improving the cellular functions of endothelial cells and oligodendrocytes. Enhancement of the extracellular cAMP signaling pathway can improve neurological function by activating the cAMP-adenosine pathway to reduce immune-mediated damage. In the peripheral immune system, cAMP can act on various immune cells to suppress peripheral immune function, which can alleviate the inflammatory response in the central nervous system and improve the prognosis of acute cerebral ischemic injury. Therefore, cAMP may play key roles in reducing post-stroke neuroinflammatory damage. The protective roles of the cAMP indicate that the cAMP enhancing drugs such as cAMP supplements, phosphodiesterase inhibitors, adenylate cyclase agonists, which are currently used in the treatment of heart and lung diseases. They are potentially able to be applied as a new therapeutic strategy in ischemic stroke. This review focuses on the immune-regulating roles and the clinical implication of cAMP in acute ischemic stroke.
Collapse
Affiliation(s)
- Meiying Xin
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Jiachun Feng
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China.
| | - Yulei Hao
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Jiulin You
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Xinyu Wang
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Xiang Yin
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Pei Shang
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Di Ma
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China.
| |
Collapse
|
19
|
Design and synthesis of fluorescent ligands for the detection of cannabinoid type 2 receptor (CB2R). Eur J Med Chem 2020; 188:112037. [PMID: 31954990 DOI: 10.1016/j.ejmech.2020.112037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/17/2019] [Accepted: 01/03/2020] [Indexed: 12/28/2022]
Abstract
The Cannabinoid 2 receptor, CB2R, belonging to the endocannabinoid system, ECS, is involved in the first steps of neurodegeneration and cancer evolution and progression and thus its modulation may be exploited in the therapeutic and diagnostic fields. However, CB2Rs distribution and signaling pathways in physiological and pathological conditions are still controversial mainly because of the lack of reliable diagnostic tools. With the aim to produce green and safe systems to detect CB2R, we designed a series of fluorescent ligands with three different green fluorescent moieties (4-dimethylaminophthalimide, 4-DMAP, 7-nitro-4-yl-aminobenzoxadiazole, NBD, and Fluorescein-thiourea, FTU) linked to the N1-position of the CB2R pharmacophore N-adamantyl-4-oxo-1,4-dihydroquinoline-3-carboxamide through polymethylene chains. Compound 28 emerged for its compromise between good pharmacodynamic properties (CB2R Ki = 130 nM and no affinity vs the other subtype CB1R) and optimal fluorescent spectroscopic properties. Therefore, compound 28 was studied through FACS (saturation and competitive binding studies) and fluorescence microscopy (visualization and competitive binding) in engineered cells (CB2R-HEK293 cells) and in diverse tumour cells. The fluoligand binding assays were successfully set up, and affinity values for the two reference compounds GW405833 and WIN55,212-2, comparable to the values obtained by radioligand binding assays, were obtained. Fluoligand 28 also allowed the detection of the presence and quantification of the CB2R in the same cell lines. The interactions of compound 28 within the CB2R binding site were also investigated by molecular docking simulations, and indications for the improvement of the CB2R affinity of this class of compounds were provided. Overall, the results obtained through these studies propose compound 28 as a safe and green alternative to the commonly used radioligands for in vitro investigations.
Collapse
|
20
|
Cannabinoids and the expanded endocannabinoid system in neurological disorders. Nat Rev Neurol 2019; 16:9-29. [PMID: 31831863 DOI: 10.1038/s41582-019-0284-z] [Citation(s) in RCA: 542] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
|
21
|
Hohmann U, Pelzer M, Kleine J, Hohmann T, Ghadban C, Dehghani F. Opposite Effects of Neuroprotective Cannabinoids, Palmitoylethanolamide, and 2-Arachidonoylglycerol on Function and Morphology of Microglia. Front Neurosci 2019; 13:1180. [PMID: 31787870 PMCID: PMC6853843 DOI: 10.3389/fnins.2019.01180] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/18/2019] [Indexed: 12/19/2022] Open
Abstract
Various studies performed in cultured cells and in in vivo models of neuronal damage showed that cannabinoids exert a neuroprotective effect. The increase in cannabinoids and cannabinoid like substances after stroke has been postulated to limit the content of neuronal injury. As well-accepted, inflammation, and neuronal damage are coupled processes and microglial cells as the main intrinsic immunological effector within the brain play a central role in their regulation. Treatment with the endocannabinoid, 2-arachidonoylglycerol (2-AG) or the endocannabinoid-like substance, palmitoylethanolamide (PEA) affected microglial cells and led to a decrease in the number of damaged neurons after excitotoxical lesion in organotypic hippocampal slice cultures (OHSC). 2-AG activated abnormal cannabidiol (abn-CBD) receptor, PEA was shown to mediate neuroprotection via peroxisome proliferator-activated receptor (PPAR)α. Despite the known neuroprotective and anti-inflammatory properties, the potential synergistic effect, namely possible entourage effect after treatment with the combination of these two protective cannabinoids has not been examined yet. After excitotoxical lesion OHSC were treated with PEA, 2-AG or a combination of both and the number of damaged neurons was evaluated. To investigate the role of microglial cells in PEA and 2-AG mediated protection, primary microglial cell cultures were treated with lipopolysaccharide (LPS) and 2-AG, PEA or a combination of those. Thereafter, we measured NO production, ramification index, proliferation and PPARα distribution in microglial cells. While PEA or 2-AG alone were neuroprotective, their co-application vanished the protective effect. This behavior was independent of microglial cells. Furthermore, PEA and 2-AG had contrary effects on ramification index and on NO production. No significant changes were observed in the proliferation rate of microglial cells after treatment. The expression of PPARα was not changed upon stimulation with PEA or 2-AG, but the distribution was significantly altered. 2-AG and PEA mediated neuroprotection was abolished when co-applied. Both cannabinoids exert contrary effects on morphology and function of microglial cells. Co-application of both cannabinoids with different targets did not lead to a positive additive effect as expected, presumably due to the contrary polarization of microglial cells.
Collapse
Affiliation(s)
- Urszula Hohmann
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Markus Pelzer
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Joshua Kleine
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Tim Hohmann
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chalid Ghadban
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
22
|
Martin GG, Seeger DR, McIntosh AL, Milligan S, Chung S, Landrock D, Dangott LJ, Golovko MY, Murphy EJ, Kier AB, Schroeder F. Sterol Carrier Protein-2/Sterol Carrier Protein-x/Fatty Acid Binding Protein-1 Ablation Impacts Response of Brain Endocannabinoid to High-Fat Diet. Lipids 2019; 54:583-601. [PMID: 31487051 DOI: 10.1002/lipd.12192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Abstract
Brain endocannabinoids (EC) such as arachidonoylethanolamine (AEA) and 2-arachidonoylglycerol (2-AG) primarily originate from serum arachidonic acid (ARA), whose level is regulated in part by a cytosolic ARA-binding protein, that is, liver fatty acid binding protein-1 (FABP1), not expressed in the brain. Ablation of the Fabp1 gene (LKO) increases brain AEA and 2-AG by decreasing hepatic uptake of ARA to increase serum ARA, thereby increasing ARA availability for uptake by the brain. The brain also expresses sterol carrier protein-2 (SCP-2), which is also a cytosolic ARA-binding protein. To further resolve the role of SCP-2 independent of FABP1, mice ablated in the Scp-2/Scp-x gene (DKO) were crossed with mice ablated in the Fabp1 gene (LKO) mice to generate triple knock out (TKO) mice. TKO impaired the ability of LKO to increase brain AEA and 2-AG. While a high-fat diet (HFD) alone increased brain AEA, TKO impaired this effect. Overall, these TKO-induced blocks were not attributable to altered expression of brain proteins in ARA uptake, AEA/2-AG synthesis, or AEA/2-AG degrading enzymes. Instead, TKO reduced serum levels of free ARA and/or total ARA and thereby decreased ARA availability for uptake to the brain and downstream synthesis of AEA and 2-AG therein. In summary, Scp-2/Scp-x gene ablation in Fabp1 null (LKO) mice antagonized the impact of LKO and HFD on brain ARA and, subsequently, EC levels. Thus, both FABP1 and SCP-2 participate in regulating the EC system in the brain.
Collapse
Affiliation(s)
- Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Drew R Seeger
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Avery L McIntosh
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Sherrelle Milligan
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Sarah Chung
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Lawrence J Dangott
- Protein Chemistry Laboratory, Texas A&M University, College Station, TX, 77843-2128, USA
| | - Mikhail Y Golovko
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Eric J Murphy
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| |
Collapse
|
23
|
Eyo UB, Wu LJ. Microglia: Lifelong patrolling immune cells of the brain. Prog Neurobiol 2019; 179:101614. [PMID: 31075285 PMCID: PMC6599472 DOI: 10.1016/j.pneurobio.2019.04.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/11/2019] [Accepted: 04/19/2019] [Indexed: 02/02/2023]
Abstract
Microglial cells are the predominant parenchymal immune cell of the brain. Recent evidence suggests that like peripheral immune cells, microglia patrol the brain in health and disease. Reviewing these data, we first examine the evidence that microglia invade the brain mesenchyme early in embryonic development, establish residence therein, proliferate and subsequently maintain their numbers throughout life. We, then, summarize established and novel evidence for microglial process surveillance in the healthy and injured brain. Finally, we discuss emerging evidence for microglial cell body dynamics that challenge existing assumptions of their sessile nature. We conclude that microglia are long-lived immune cells that patrol the brain through both cell body and process movements. This recognition has significant implications for neuroimmune interactions throughout the animal lifespan.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
24
|
Rivera P, Silva-Peña D, Blanco E, Vargas A, Arrabal S, Serrano A, Pavón FJ, Bindila L, Lutz B, Rodríguez de Fonseca F, Suárez J. Oleoylethanolamide restores alcohol-induced inhibition of neuronal proliferation and microglial activity in striatum. Neuropharmacology 2019; 146:184-197. [DOI: 10.1016/j.neuropharm.2018.11.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/08/2018] [Accepted: 11/25/2018] [Indexed: 01/19/2023]
|
25
|
Martin GG, Landrock D, Dangott LJ, McIntosh AL, Kier AB, Schroeder F. Human Liver Fatty Acid Binding Protein-1 T94A Variant, Nonalcohol Fatty Liver Disease, and Hepatic Endocannabinoid System. Lipids 2019; 53:27-40. [PMID: 29488637 DOI: 10.1002/lipd.12008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022]
Abstract
Hepatic endocannabinoids (EC) and their major binding/"chaperone" protein (i.e., liver fatty acid binding protein-1 [FABP1]) are associated with development of nonalcoholic fatty liver (NAFLD) in animal models and humans. Since expression of the highly prevalent human FABP1 T94A variant induces serum lipid accumulation, it is important to determine its impact on hepatic lipid accumulation and the EC system. This issue was addressed in livers from human subjects expressing only wild-type (WT) FABP1 T94T (TT genotype) or T94A variant (TC or CC genotype). WT FABP1 males had lower total lipids (both neutral cholesteryl esters, triacylglycerols) and phospholipids than females. WT FABP1 males' lower lipids correlated with lower levels of the N-acylethanolamide DHEA and 2-monoacylglycerols (2-MAG) (2-OG, 2-PG). T94A expression in males increased the hepatic total lipids (triacylglycerol, cholesteryl ester), which is consistent with their higher level of CB1-potentiating 2-OG and lower antagonistic EPEA. In contrast, in females, T94A expression did not alter the total lipids, neutral lipids, or phospholipids, which is attributable to the higher cannabinoid receptor-1 (CB1) agonist arachidonoylethanolamide (AEA) and its CB1-potentiator OEA being largely offset by reduced potentiating 2-OG and increased antagonistic EPEA. Taken together, these findings indicate that T94A-induced alterations in the hepatic EC system contribute at least in part to the hepatic accumulation of lipids associated with NAFLD, especially in males.
Collapse
Affiliation(s)
- Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Lawrence J Dangott
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843-2128, USA
| | - Avery L McIntosh
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4467, USA
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843-4466, USA
| |
Collapse
|
26
|
Cristiano C, Pirozzi C, Coretti L, Cavaliere G, Lama A, Russo R, Lembo F, Mollica MP, Meli R, Calignano A, Mattace Raso G. Palmitoylethanolamide counteracts autistic-like behaviours in BTBR T+tf/J mice: Contribution of central and peripheral mechanisms. Brain Behav Immun 2018; 74:166-175. [PMID: 30193877 DOI: 10.1016/j.bbi.2018.09.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/27/2018] [Accepted: 09/03/2018] [Indexed: 12/23/2022] Open
Abstract
Autism spectrum disorders (ASD) are a group of heterogeneous neurodevelopmental conditions characterized by impaired social interaction, and repetitive stereotyped behaviours. Interestingly, functional and inflammatory gastrointestinal diseases are often reported as a comorbidity in ASDs, indicating gut-brain axis as a novel emerging approach. Recently, a central role for peroxisome-proliferator activated receptor (PPAR)-α has been addressed in neurological functions, associated with the behaviour. Among endogenous lipids, palmitoylethanolamide (PEA), a PPAR-α agonist, has been extensively studied for its anti-inflammatory effects both at central and peripheral level. Based on this background, the aim of this study was to investigate the pharmacological effects of PEA on autistic-like behaviour of BTBR T+tf/J mice and to shed light on the contributing mechanisms. Our results showed that PEA reverted the altered behavioural phenotype of BTBR mice, and this effect was contingent to PPAR-α activation. Moreover, PEA was able to restore hippocampal BDNF signalling pathway, and improve mitochondrial dysfunction, both pathological aspects, known to be consistently associated with ASDs. Furthermore, PEA reduced the overall inflammatory state of BTBR mice, reducing the expression of pro-inflammatory cytokines at hippocampal, serum, and colonic level. The analysis of gut permeability and the expression of colonic tight junctions showed a reduction of leaky gut in PEA-treated BTBR mice. This finding together with PEA effect on gut microbiota composition suggests an involvement of microbiota-gut-brain axis. In conclusion, our results demonstrated a therapeutic potential of PEA in limiting ASD symptoms, through its pleiotropic mechanism of action, supporting neuroprotection, anti-inflammatory effects, and the modulation of gut-brain axis.
Collapse
Affiliation(s)
- Claudia Cristiano
- Department of Pharmacy, University of Naples "Federico II", 80131 Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples "Federico II", 80131 Naples, Italy
| | - Lorena Coretti
- Task Force on Microbiome Studies, University of Naples "Federico II", 80131 Naples, Italy; Institute for Experimental Endocrinology and Oncology, IEOS, Consiglio Nazionale delle Ricerche CNR, Via S. Pansini, 5, 80131, Naples, Italy
| | - Gina Cavaliere
- Department of Biology, University of Naples "Federico II", 80131 Naples, Italy
| | - Adriano Lama
- Department of Pharmacy, University of Naples "Federico II", 80131 Naples, Italy; Task Force on Microbiome Studies, University of Naples "Federico II", 80131 Naples, Italy
| | - Roberto Russo
- Department of Pharmacy, University of Naples "Federico II", 80131 Naples, Italy
| | - Francesca Lembo
- Department of Pharmacy, University of Naples "Federico II", 80131 Naples, Italy; Task Force on Microbiome Studies, University of Naples "Federico II", 80131 Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples "Federico II", 80131 Naples, Italy
| | - Rosaria Meli
- Department of Pharmacy, University of Naples "Federico II", 80131 Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples "Federico II", 80131 Naples, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, University of Naples "Federico II", 80131 Naples, Italy; Task Force on Microbiome Studies, University of Naples "Federico II", 80131 Naples, Italy.
| |
Collapse
|
27
|
Davidson JO, Dhillon SK, Wassink G, Zhou KQ, Bennet L, Gunn AJ. Endogenous neuroprotection after perinatal hypoxia-ischaemia: the resilient developing brain. J R Soc N Z 2018. [DOI: 10.1080/03036758.2018.1529685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Joanne O. Davidson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Simerdeep K. Dhillon
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Guido Wassink
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Kelly Q. Zhou
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Alistair J. Gunn
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
28
|
Holubiec MI, Romero JI, Suárez J, Portavella M, Fernández-Espejo E, Blanco E, Galeano P, de Fonseca FR. Palmitoylethanolamide prevents neuroinflammation, reduces astrogliosis and preserves recognition and spatial memory following induction of neonatal anoxia-ischemia. Psychopharmacology (Berl) 2018; 235:2929-2945. [PMID: 30058012 DOI: 10.1007/s00213-018-4982-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/23/2018] [Indexed: 11/27/2022]
Abstract
RATIONAL Neonatal anoxia-ischemia (AI) particularly affects the central nervous system. Despite the many treatments that have been tested, none of them has proven to be completely successful. Palmitoylethanolamide (PEA) and oleoylethanolamide (OEA) are acylethanolamides that do not bind to CB1 or CB2 receptors and thus they do not present cannabinoid activity. These molecules are agonist compounds of peroxisome proliferator-activator receptor alpha (PPARα), which modulates the expression of different genes that are related to glucose and lipid metabolism, inflammation, differentiation and proliferation. OBJECTIVE In the present study, we analyzed the effects that the administration of PEA or OEA, after a neonatal AI event, has over different areas of the hippocampus. METHODS To this end, 7-day-old rats were subjected to AI and then treated with vehicle, OEA (2 or 10 mg/kg) or PEA (2 or 10 mg/kg). At 30 days of age, animals were subjected to behavioral tests followed by immunohistochemical studies. RESULTS Results showed that neonatal AI was associated with decreased locomotion, as well as recognition and spatial memory impairments. Furthermore, these deficits were accompanied with enhanced neuroinflammation and astrogliosis, as well as a decreased PPARα expression. PEA treatment was able to prevent neuroinflammation, reduce astrogliosis and preserve cognitive functions. CONCLUSIONS These results indicate that the acylethanolamide PEA may play an important role in the mechanisms underlying neonatal AI, and it could be a good candidate for further studies regarding neonatal AI treatments.
Collapse
Affiliation(s)
- Mariana I Holubiec
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Patricias Argentinas 435, C1405BWE, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I Romero
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Patricias Argentinas 435, C1405BWE, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan Suárez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain
| | - Manuel Portavella
- Laboratorio de Conducta Animal y Neurociencia, Departamento de Psicología Experimental, Facultad de Psicología, Universidad de Sevilla, C/Camilo José Cela s/n, 41018, Sevilla, Spain
| | - Emilio Fernández-Espejo
- Laboratorio de Neurofisiología y Neurología Molecular, Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009, Sevilla, Spain
| | - Eduardo Blanco
- Lleida Institute for Biomedical Research, Dr. Pifarré Foundation (IRBLleida), University of Lleida, Av. Alcalde Rovira Roure 80, 25198, Lleida, Spain
| | - Pablo Galeano
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain.
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Patricias Argentinas 435, C1405BWE, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain.
| |
Collapse
|
29
|
Portavella M, Rodriguez-Espinosa N, Galeano P, Blanco E, Romero JI, Holubiec MI, Rodriguez de Fonseca F, Fernández-Espejo E. Oleoylethanolamide and Palmitoylethanolamide Protect Cultured Cortical Neurons Against Hypoxia. Cannabis Cannabinoid Res 2018; 3:171-178. [PMID: 30255158 PMCID: PMC6148719 DOI: 10.1089/can.2018.0013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Introduction: Perinatal hypoxic-ischemic (HI) encephalopathy is defined as a neurological syndrome where the newborn suffers from acute ischemia and hypoxia during the perinatal period. New therapies are needed. The acylethanolamides, oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), possess neuroprotective properties, and they could be effective against perinatal HI. These lipid mediators act through peroxisome proliferator-activated receptors subtype α (PPARα), or transient receptor potential vanilloid (TRPV), such as TRPV subtype 1 and 4. Materials and Methods: The objectives of this study were to discern: (1) the neuroprotective role of OEA and PEA in parietotemporal cortical neurons of newborn rats and mice subjected to hypoxia, and (2) the role of the receptors, PPARα, TRPV1, and TRPV4, in neuroprotective effects. Cell culture of cortical neurons and the lactate dehydrogenase assay was carried out. The role of receptors was discerned by using selective antagonist and agonist ligands, as well as knockout (KO) PPARα mice. Results: The findings indicate that OEA and PEA exert neuroprotective effects on cultured cortical neurons subjected to a hypoxic episode. These protective effects are not mediated by the receptors, PPARα, TRPV1, or TRPV4, because neither PPARα KO mice nor receptor ligands significantly modify OEA and PEA-induced effects. Blocking TRPV4 with RN1734 is neuroprotective per se, and cotreatment with OEA and PEA is able to enhance neuroprotective effects of the acylethanolamides. Since stimulating TRPV4 was devoid of effects on OEA and PEA-induced protective effects, effects of RN1734 cotreatment seem to be a consequence of additive actions. Conclusion: The lipid mediators, OEA and PEA, exert neuroprotective effects on cultured cortical neurons subjected to hypoxia. Coadministration of OEA or PEA, and the TRPV4 antagonist RN1734 is able to enhance neuroprotective effects. These in vitro results could be of utility for developing new therapeutic tools against perinatal HI.
Collapse
Affiliation(s)
- Manuel Portavella
- Laboratory of Animal Behavior and Neuroscience, Department of Experimental Psychology, Faculty of Psychology, Universidad de Sevilla, Seville, Spain
| | - Nieves Rodriguez-Espinosa
- Neurophysiology and Molecular Neurology Lab, Department of Medical Physiology and Biophysics, Faculty of Medicine, Universidad de Sevilla, Seville, Spain
| | - Pablo Galeano
- Biochemical Research Institute of Buenos Aires (IIBBA-CONICET), Buenos Aires, Argentina
| | - Eduardo Blanco
- University of Lleida, Medical Research Institute, Dr. Pifarré Foundation (IRBLleida), Lleida, Spain
| | - Juan I Romero
- Biochemical Research Institute of Buenos Aires (IIBBA-CONICET), Buenos Aires, Argentina
| | - Mariana I Holubiec
- Biochemical Research Institute of Buenos Aires (IIBBA-CONICET), Buenos Aires, Argentina
| | | | - Emilio Fernández-Espejo
- Neurophysiology and Molecular Neurology Lab, Department of Medical Physiology and Biophysics, Faculty of Medicine, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
30
|
Martin GG, Seeger DR, McIntosh AL, Chung S, Milligan S, Landrock D, Dangott LJ, Golovko MY, Murphy EJ, Kier AB, Schroeder F. Scp-2/Scp-x ablation in Fabp1 null mice differentially impacts hepatic endocannabinoid level depending on dietary fat. Arch Biochem Biophys 2018; 650:93-102. [PMID: 29763591 PMCID: PMC6033332 DOI: 10.1016/j.abb.2018.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/07/2018] [Accepted: 05/11/2018] [Indexed: 12/21/2022]
Abstract
Dysregulation of the hepatic endocannabinoid (EC) system and high fat diet (HFD) are associated with non-alcoholic fatty liver disease. Liver cytosol contains high levels of two novel endocannabinoid binding proteins-liver fatty acid binding protein (FABP1) and sterol carrier protein-2 (SCP-2). While Fabp1 gene ablation significantly increases hepatic levels of arachidonic acid (ARA)-containing EC and sex-dependent response to pair-fed high fat diet (HFD), the presence of SCP-2 complicates interpretation. These issues were addressed by ablating Scp-2/Scp-x in Fabp1 null mice (TKO). In control-fed mice, TKO increased hepatic levels of arachidonoylethanolamide (AEA) in both sexes. HFD impacted hepatic EC levels by decreasing AEA in TKO females and decreasing 2-arachidonoyl glycerol (2-AG) in WT of both sexes. Only TKO males on HFD had increased hepatic 2-AG levels. Hepatic ARA levels were decreased in control-fed TKO of both sexes. Changes in hepatic AEA/2-AG levels were not associated with altered amounts of hepatic proteins involved in AEA/2-AG synthesis or degradation. These findings suggested that ablation of the Scp-2/Scp-x gene in Fabp1 null mice exacerbated hepatic EC accumulation and antagonized the impact of HFD on hepatic EC levels-suggesting both proteins play important roles in regulating the hepatic EC system.
Collapse
Affiliation(s)
- Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA.
| | - Drew R Seeger
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037 USA
| | - Avery L McIntosh
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA
| | - Sarah Chung
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, USA
| | - Sherrelle Milligan
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, USA
| | - Lawrence J Dangott
- Protein Chemistry Laboratory, Texas A&M University, College Station, TX 77843-2128, USA
| | - Mikhail Y Golovko
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037 USA
| | - Eric J Murphy
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037 USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, USA
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA.
| |
Collapse
|
31
|
Rahmani MR, Shamsizadeh A, Moghadam-Ahmadi A, Bazmandegan G, Allahtavakoli M. JZL184, as a monoacylglycerol lipase inhibitor, down-regulates inflammation in a cannabinoid pathway dependent manner. Biomed Pharmacother 2018; 103:1720-1726. [DOI: 10.1016/j.biopha.2018.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023] Open
|
32
|
Varrassi G, Fusco M, Skaper SD, Battelli D, Zis P, Coaccioli S, Pace MC, Paladini A. A Pharmacological Rationale to Reduce the Incidence of Opioid Induced Tolerance and Hyperalgesia: A Review. Pain Ther 2018; 7:59-75. [PMID: 29594972 PMCID: PMC5993687 DOI: 10.1007/s40122-018-0094-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Indexed: 02/06/2023] Open
Abstract
Chronic pain is an important health and social problem. Misuse and abuse of opioids in chronic non-cancer pain management seem to be a huge problem, in some countries. This could probably affect the normal use of such analgesics in patients in need of them. Basic and clinical researches should find the solution to mitigate the potential damage. Dysregulation of mast cell and microglia activation plays an important role in the pathogenesis and management of chronic pain. Persistent mast cell activation sensitizes nociceptors and initiates central nervous system inflammatory processes, involving microglial cell activation and sensitization of spinal somatosensory neurons. Exposure of mast cells and microglia to opioids is well known to provoke activation of these non-neuronal immune cell populations, thereby contributing to an exacerbation of pro-inflammatory and pro-nociceptive processes and promoting, over the long-term, opioid-induced hyperalgesia and tolerance. This review is intended to provide the reader with an overview of the role for these non-neuronal cells in opioid-induced chronic pain and tolerance as a consequence of prolonged exposure to these drugs. In addition, we will examine a potential strategy with the aim to modulate opioid-induced over-activation of glia and mast cells, based on endogenous defense mechanisms and fatty acid amide signaling molecules.
Collapse
Affiliation(s)
- Giustino Varrassi
- Department of Anesthesia and Pain Medicine, University of L'Aquila, L'Aquila, Italy.
| | - Mariella Fusco
- Center for Medical Documentation and Information, Epitech, Padua, Italy
| | | | - Daniele Battelli
- Department of Anesthesia and Pain Medicine, San Marino Hospital, San Marino, San Marino
| | - Panagiotis Zis
- Academic Department of Neurosciences, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Stefano Coaccioli
- Department of Internal Medicine, University of Perugia, Terni, Italy
| | - Maria Caterina Pace
- Department of Anesthesia and Pain Medicine, University of Napoli, Naples, Italy
| | - Antonella Paladini
- Department of Anesthesia and Pain Medicine, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
33
|
Ghazizadeh-Hashemi M, Ghajar A, Shalbafan MR, Ghazizadeh-Hashemi F, Afarideh M, Malekpour F, Ghaleiha A, Ardebili ME, Akhondzadeh S. Palmitoylethanolamide as adjunctive therapy in major depressive disorder: A double-blind, randomized and placebo-controlled trial. J Affect Disord 2018; 232:127-133. [PMID: 29486338 DOI: 10.1016/j.jad.2018.02.057] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/03/2018] [Accepted: 02/19/2018] [Indexed: 12/15/2022]
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.
Collapse
Affiliation(s)
| | - Alireza Ghajar
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Mohsen Afarideh
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Malekpour
- Mental Health Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Ghaleiha
- Research Center for Behavioral Disorders and Substance Abuse, Hamadan University of Medical Sciences. Hamadan, Iran
| | | | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Di Zazzo A, Roberti G, Mashaghi A, Abud TB, Pavese D, Bonini S. Use of Topical Cannabinomimetic Palmitoylethanolamide in Ocular Surface Disease Associated with Antiglaucoma Medications. J Ocul Pharmacol Ther 2018; 33:670-677. [PMID: 29045169 DOI: 10.1089/jop.2016.0117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Chronic use of topical hypotensive therapies in glaucoma patients leads to chronic inflammation of the ocular surface, which decreases the success rate of long-term glaucoma management. The aim of this study is to evaluate the effect of topical palmitoylethanolamide (PEA) (Defluxa©), a well-known anti-inflammatory and analgesic agent, in suppressing the ocular surface inflammation associated with the use of hypotensive eye drops. METHODS In a pilot clinical trial, we enrolled 15 glaucomatous patients who received topical PEA (Defluxa) in addition to the current antiglaucoma drugs, while 15 glaucomatous patients did not receive any additional treatment. At 3 different time points (day 0, 15, and 30), signs of ocular surface involvement, adverse events, visual acuity, and intraocular pressure were assessed. RESULTS Topical PEA (Defluxa) was effective in increasing the Schirmer test (P < 0.05) and the tear film breakup time (T-BUT) (P < 0.0001), and improving the conjunctival hyperemia (P < 0.0001) by day 30, compared to baseline. Compared to control, by day 15, the conjunctival hyperemia score was significantly decreased in the PEA (Defluxa) group (P < 0.01), while the T-BUT and the Schirmer Test achieved a significant improvement by day 30 (P < 0.05; P < 0.01). DISCUSSION Our data suggests that topical PEA (Defluxa) is a safe, effective, and generally well-tolerated treatment to prevent or suppress ocular surface inflammation attributable to chronic glaucoma treatment.
Collapse
Affiliation(s)
| | | | - Alireza Mashaghi
- 2 Faculty of Mathematics and Natural Sciences, Leiden Academic Centre for Drug Research, Leiden University , Leiden, The Netherlands .,3 Schepens Eye Research Institute , Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts.,4 Basir Eye Health Research Center , Tehran, Iran
| | - Tulio Batista Abud
- 5 Department of Ophthalmology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Daniela Pavese
- 6 Department of Ophthalmology, Campus Bio-Medico University, Rome, Italy
| | - Stefano Bonini
- 6 Department of Ophthalmology, Campus Bio-Medico University, Rome, Italy
| |
Collapse
|
35
|
McIntosh AL, Martin GG, Huang H, Landrock D, Kier AB, Schroeder F. Δ 9-Tetrahydrocannabinol induces endocannabinoid accumulation in mouse hepatocytes: antagonism by Fabp1 gene ablation. J Lipid Res 2018; 59:646-657. [PMID: 29414765 PMCID: PMC5880504 DOI: 10.1194/jlr.m082644] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/03/2018] [Indexed: 01/06/2023] Open
Abstract
Phytocannabinoids, such as Δ9-tetrahydrocannabinol (THC), bind and activate cannabinoid (CB) receptors, thereby "piggy-backing" on the same pathway's endogenous endocannabinoids (ECs). The recent discovery that liver fatty acid binding protein-1 (FABP1) is the major cytosolic "chaperone" protein with high affinity for both Δ9-THC and ECs suggests that Δ9-THC may alter hepatic EC levels. Therefore, the impact of Δ9-THC or EC treatment on the levels of endogenous ECs, such as N-arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG), was examined in cultured primary mouse hepatocytes from WT and Fabp1 gene-ablated (LKO) mice. Δ9-THC alone or 2-AG alone significantly increased AEA and especially 2-AG levels in WT hepatocytes. LKO alone markedly increased AEA and 2-AG levels. However, LKO blocked/diminished the ability of Δ9-THC to further increase both AEA and 2-AG. In contrast, LKO potentiated the ability of exogenous 2-AG to increase the hepatocyte level of AEA and 2-AG. These and other data suggest that Δ9-THC increases hepatocyte EC levels, at least in part, by upregulating endogenous AEA and 2-AG levels. This may arise from Δ9-THC competing with AEA and 2-AG binding to FABP1, thereby decreasing targeting of bound AEA and 2-AG to the degradative enzymes, fatty acid amide hydrolase and monoacylglyceride lipase, to decrease hydrolysis within hepatocytes.
Collapse
Affiliation(s)
- Avery L McIntosh
- Departments of Physiology and Pharmacology Texas A&M University, College Station, TX 77843
| | - Gregory G Martin
- Departments of Physiology and Pharmacology Texas A&M University, College Station, TX 77843
| | - Huan Huang
- Departments of Physiology and Pharmacology Texas A&M University, College Station, TX 77843
| | - Danilo Landrock
- Departments of Pathobiology, Texas A&M University, College Station, TX 77843
| | - Ann B Kier
- Departments of Pathobiology, Texas A&M University, College Station, TX 77843
| | - Friedhelm Schroeder
- Departments of Physiology and Pharmacology Texas A&M University, College Station, TX 77843.
| |
Collapse
|
36
|
Skaper SD, Facci L, Zusso M, Giusti P. An Inflammation-Centric View of Neurological Disease: Beyond the Neuron. Front Cell Neurosci 2018; 12:72. [PMID: 29618972 PMCID: PMC5871676 DOI: 10.3389/fncel.2018.00072] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/27/2018] [Indexed: 12/13/2022] Open
Abstract
Inflammation is a complex biological response fundamental to how the body deals with injury and infection to eliminate the initial cause of cell injury and effect repair. Unlike a normally beneficial acute inflammatory response, chronic inflammation can lead to tissue damage and ultimately its destruction, and often results from an inappropriate immune response. Inflammation in the nervous system (“neuroinflammation”), especially when prolonged, can be particularly injurious. While inflammation per se may not cause disease, it contributes importantly to disease pathogenesis across both the peripheral (neuropathic pain, fibromyalgia) and central [e.g., Alzheimer disease, Parkinson disease, multiple sclerosis, motor neuron disease, ischemia and traumatic brain injury, depression, and autism spectrum disorder] nervous systems. The existence of extensive lines of communication between the nervous system and immune system represents a fundamental principle underlying neuroinflammation. Immune cell-derived inflammatory molecules are critical for regulation of host responses to inflammation. Although these mediators can originate from various non-neuronal cells, important sources in the above neuropathologies appear to be microglia and mast cells, together with astrocytes and possibly also oligodendrocytes. Understanding neuroinflammation also requires an appreciation that non-neuronal cell—cell interactions, between both glia and mast cells and glia themselves, are an integral part of the inflammation process. Within this context the mast cell occupies a key niche in orchestrating the inflammatory process, from initiation to prolongation. This review will describe the current state of knowledge concerning the biology of neuroinflammation, emphasizing mast cell-glia and glia-glia interactions, then conclude with a consideration of how a cell's endogenous mechanisms might be leveraged to provide a therapeutic strategy to target neuroinflammation.
Collapse
Affiliation(s)
- Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
37
|
Petrosino S, Cordaro M, Verde R, Schiano Moriello A, Marcolongo G, Schievano C, Siracusa R, Piscitelli F, Peritore AF, Crupi R, Impellizzeri D, Esposito E, Cuzzocrea S, Di Marzo V. Oral Ultramicronized Palmitoylethanolamide: Plasma and Tissue Levels and Spinal Anti-hyperalgesic Effect. Front Pharmacol 2018; 9:249. [PMID: 29615912 PMCID: PMC5870042 DOI: 10.3389/fphar.2018.00249] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/06/2018] [Indexed: 12/22/2022] Open
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.
Collapse
Affiliation(s)
- Stefania Petrosino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Napoli, Italy
- Epitech Group SpA, Padova, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Science University of Messina, Messina, Italy
| | - Roberta Verde
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Napoli, Italy
| | - Aniello Schiano Moriello
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Napoli, Italy
- Epitech Group SpA, Padova, Italy
| | | | | | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Science University of Messina, Messina, Italy
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Napoli, Italy
| | - Alessio F. Peritore
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Napoli, Italy
| | - Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Science University of Messina, Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Science University of Messina, Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Science University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Science University of Messina, Messina, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Napoli, Italy
| |
Collapse
|
38
|
Cilia R. Molecular Imaging of the Cannabinoid System in Idiopathic Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 141:305-345. [DOI: 10.1016/bs.irn.2018.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
|
39
|
Fowler CJ, Doherty P, Alexander SPH. Endocannabinoid Turnover. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 80:31-66. [PMID: 28826539 DOI: 10.1016/bs.apha.2017.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this review, we consider the biosynthetic, hydrolytic, and oxidative metabolism of the endocannabinoids anandamide and 2-arachidonoylglycerol. We describe the enzymes associated with these events and their characterization. We identify the inhibitor profile for these enzymes and the status of therapeutic exploitation, which to date has been limited to clinical trials for fatty acid amide hydrolase inhibitors. To bring the review to a close, we consider whether point block of a single enzyme is likely to be the most successful approach for therapeutic exploitation of the endocannabinoid system.
Collapse
Affiliation(s)
| | - Patrick Doherty
- Wolfson Centre for Age-Related Disease, King's College London, London, United Kingdom
| | | |
Collapse
|
40
|
Anti-Inflammatory, Antioxidant and Crystallographic Studies of N-Palmitoyl-ethanol Amine (PEA) Derivatives. Molecules 2017; 22:molecules22040616. [PMID: 28398240 PMCID: PMC6154659 DOI: 10.3390/molecules22040616] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 12/12/2022] Open
Abstract
N-Palmitoyl-ethanolamine (PEA) is an anti-inflammatory component of egg yolk that is usually employed for the prevention of respiratory apparatus virus infection and then frequently used for its efficient anti-inflammatory and analgesic effects in experimental models of visceral, neuropathic, and inflammatory diseases. Nevertheless, data of its use in animal or human therapy are still scarce and further studies are needed. Herein, we report the biological evaluation of a small library of N-palmitoyl-ethanolamine analogues or derivatives, characterized by a protected acid function (either as palmitoyl amides or hexadecyl esters), useful to decrease their hydrolysis rate in vitro and prolong their biological activity. Two of these compounds—namely phenyl-carbamic acid hexadecyl ester (4) and 2-methyl-pentadecanoic acid (4-nitro-phenyl)-amide (5)—have shown good anti-inflammatory and antioxidant properties, without affecting the viability of J774A.1 macrophages. Finally, crystals suitable for X-ray analysis of compound 4 have been obtained, and its solved crystal structure is here reported. Our outcomes may be helpful for a rational drug design based on new PEA analogues/derivatives with improved biological properties.
Collapse
|
41
|
Guida F, Luongo L, Boccella S, Giordano ME, Romano R, Bellini G, Manzo I, Furiano A, Rizzo A, Imperatore R, Iannotti FA, D'Aniello E, Piscitelli F, Sca Rossi F, Cristino L, Di Marzo V, de Novellis V, Maione S. Palmitoylethanolamide induces microglia changes associated with increased migration and phagocytic activity: involvement of the CB2 receptor. Sci Rep 2017; 7:375. [PMID: 28336953 PMCID: PMC5428303 DOI: 10.1038/s41598-017-00342-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 02/22/2017] [Indexed: 12/22/2022] Open
Abstract
The endogenous fatty acid amide palmitoylethanolamide (PEA) has been shown to exert anti-inflammatory actions mainly through inhibition of the release of pro-inflammatory molecules from mast cells, monocytes and macrophages. Indirect activation of the endocannabinoid (eCB) system is among the several mechanisms of action that have been proposed to underlie the different effects of PEA in vivo. In this study, we used cultured rat microglia and human macrophages to evaluate whether PEA affects eCB signaling. PEA was found to increase CB2 mRNA and protein expression through peroxisome proliferator-activated receptor-α (PPAR-α) activation. This novel gene regulation mechanism was demonstrated through: (i) pharmacological PPAR-α manipulation, (ii) PPAR-α mRNA silencing, (iii) chromatin immunoprecipitation. Moreover, exposure to PEA induced morphological changes associated with a reactive microglial phenotype, including increased phagocytosis and migratory activity. Our findings suggest indirect regulation of microglial CB2R expression as a new possible mechanism underlying the effects of PEA. PEA can be explored as a useful tool for preventing/treating the symptoms associated with neuroinflammation in CNS disorders.
Collapse
Affiliation(s)
- F Guida
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - L Luongo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - S Boccella
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - M E Giordano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - R Romano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - G Bellini
- Department of Women, Child and General and Specialistic Surgery, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - I Manzo
- Department of Women, Child and General and Specialistic Surgery, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - A Furiano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - A Rizzo
- Department of Experimental Medicine, Section of Microbiology and Clinical Microbiology, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - R Imperatore
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Department of Science and Technology, University of Sannio, Benevento, Italy
| | - F A Iannotti
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - E D'Aniello
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - F Piscitelli
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - F Sca Rossi
- Department of Women, Child and General and Specialistic Surgery, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - L Cristino
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - V Di Marzo
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - V de Novellis
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - S Maione
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy. .,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy.
| |
Collapse
|
42
|
Cordaro M, Impellizzeri D, Gugliandolo E, Siracusa R, Crupi R, Esposito E, Cuzzocrea S. Adelmidrol, a Palmitoylethanolamide Analogue, as a New Pharmacological Treatment for the Management of Inflammatory Bowel Disease. Mol Pharmacol 2016; 90:549-561. [DOI: 10.1124/mol.116.105668] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
43
|
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 2016; 174:1349-1365. [PMID: 27539936 DOI: 10.1111/bph.13580] [Citation(s) in RCA: 225] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/01/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
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.
Collapse
Affiliation(s)
- Stefania Petrosino
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Pozzuoli (NA), Italy.,Epitech Group S.p.A., Saccolongo (PD), Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Pozzuoli (NA), Italy
| |
Collapse
|
44
|
Martin GG, Chung S, Landrock D, Landrock KK, Dangott LJ, Peng X, Kaczocha M, Murphy EJ, Kier AB, Schroeder F. Female Mice are Resistant to Fabp1 Gene Ablation-Induced Alterations in Brain Endocannabinoid Levels. Lipids 2016; 51:1007-20. [PMID: 27450559 PMCID: PMC5418128 DOI: 10.1007/s11745-016-4175-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/14/2016] [Indexed: 10/21/2022]
Abstract
Although liver fatty acid binding protein (FABP1, L-FABP) is not detectable in the brain, Fabp1 gene ablation (LKO) markedly increases endocannabinoids (EC) in brains of male mice. Since the brain EC system of females differs significantly from that of males, it was important to determine if LKO differently impacted the brain EC system. LKO did not alter brain levels of arachidonic acid (ARA)-containing EC, i.e. arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG), but decreased non-ARA-containing N-acylethanolamides (OEA, PEA) and 2-oleoylglycerol (2-OG) that potentiate the actions of AEA and 2-AG. These changes in brain potentiating EC levels were not associated with: (1) a net decrease in levels of brain membrane proteins associated with fatty acid uptake and EC synthesis; (2) a net increase in brain protein levels of cytosolic EC chaperones and enzymes in EC degradation; or (3) increased brain protein levels of EC receptors (CB1, TRVP1). Instead, the reduced or opposite responsiveness of female brain EC levels to loss of FABP1 (LKO) correlated with intrinsically lower FABP1 level in livers of WT females than males. These data show that female mouse brain endocannabinoid levels were unchanged (AEA, 2-AG) or decreased (OEA, PEA, 2-OG) by complete loss of FABP1 (LKO).
Collapse
Affiliation(s)
- Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Sarah Chung
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Kerstin K Landrock
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Lawrence J Dangott
- Protein Chemistry Laboratory, Texas A&M University, College Station, TX, 77843-2128, USA
| | - Xiaoxue Peng
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Eric J Murphy
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA.
| |
Collapse
|
45
|
Bisogno T, Oddi S, Piccoli A, Fazio D, Maccarrone M. Type-2 cannabinoid receptors in neurodegeneration. Pharmacol Res 2016; 111:721-730. [DOI: 10.1016/j.phrs.2016.07.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/14/2016] [Accepted: 07/19/2016] [Indexed: 01/01/2023]
|
46
|
Martin GG, Chung S, Landrock D, Landrock KK, Huang H, Dangott LJ, Peng X, Kaczocha M, Seeger DR, Murphy EJ, Golovko MY, Kier AB, Schroeder F. FABP-1 gene ablation impacts brain endocannabinoid system in male mice. J Neurochem 2016; 138:407-22. [PMID: 27167970 PMCID: PMC4961623 DOI: 10.1111/jnc.13664] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/14/2016] [Accepted: 04/26/2016] [Indexed: 12/15/2022]
Abstract
Liver fatty acid-binding protein (FABP1, L-FABP) has high affinity for and enhances uptake of arachidonic acid (ARA, C20:4, n-6) which, when esterified to phospholipids, is the requisite precursor for synthesis of endocannabinoids (EC) such as arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG). The brain derives most of its ARA from plasma, taking up ARA and transporting it intracellularly via cytosolic fatty acid-binding proteins (FABPs 3,5, and 7) localized within the brain. In contrast, the much more prevalent cytosolic FABP1 is not detectable in the brain but is instead highly expressed in the liver. Therefore, the possibility that FABP1 outside the central nervous system may regulate brain AEA and 2-AG was examined in wild-type (WT) and FABP1 null (LKO) male mice. LKO increased brain levels of AA-containing EC (AEA, 2-AG), correlating with increased free and total ARA in brain and serum. LKO also increased brain levels of non-ARA that contain potentiating endocannabinoids (EC*) such as oleoyl ethanolamide (OEA), PEA, 2-OG, and 2-PG. Concomitantly, LKO decreased serum total ARA-containing EC, but not non-ARA endocannabinoids. LKO did not elicit these changes in the brain EC and EC* as a result of compensatory up-regulation of brain protein levels of enzymes in EC synthesis (NAPEPLD, DAGLα) or cytosolic EC chaperone proteins (FABPs 3, 5, 7, SCP-2, HSP70), or cannabinoid receptors (CB1, TRVP1). These data show for the first time that the non-CNS fatty acid-binding protein FABP1 markedly affected brain levels of both ARA-containing endocannabinoids (AEA, 2-AG) as well as their non-ARA potentiating endocannabinoids. Fatty acid-binding protein-1 (FABP-1) is not detectable in brain but instead is highly expressed in liver. The possibility that FABP1 outside the central nervous system may regulate brain endocannabinoids arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG) was examined in wild-type (WT) and FABP-1 null (LKO) male mice. LKO increased brain levels of arachidonic acid-containing endocannabinoids (AEA, 2-AG), correlating with increased free and total arachidonic acid in brain and serum. Read the Editorial Highlight for this article on page 371.
Collapse
Affiliation(s)
- Gregory G. Martin
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Sarah Chung
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467
| | - Kerstin K. Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467
| | - Huan Huang
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Lawrence J. Dangott
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843-2128
| | - Xiaoxue Peng
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794
| | - Drew R. Seeger
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037 USA
| | - Eric J. Murphy
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037 USA
| | - Mikhail Y. Golovko
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037 USA
| | - Ann B. Kier
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466
| |
Collapse
|
47
|
Reichenbach ZW, Li H, Ward SJ, Tuma RF. The CB1 antagonist, SR141716A, is protective in permanent photothrombotic cerebral ischemia. Neurosci Lett 2016; 630:9-15. [PMID: 27453059 DOI: 10.1016/j.neulet.2016.07.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 01/24/2023]
Abstract
Modulation of the endocannabinoid system has been shown to have a significant impact on outcomes in animal models of stroke. We have previously reported a protective effect of the CB1 antagonist, SR141716A, in a transient reperfusion mouse model of cerebral ischemia. This protective effect was in part mediated by activation of the 5HT1A receptor. Here we have examined its effect in a mouse model of permanent ischemia induced by photoinjury. The CB1 antagonist was found to be protective in this model. As was the case following transient ischemia reperfusion, SR141716A (5mg/kg) resulted in smaller infarct fractions and stroke volumes when utilized both as a pretreatment and as a post-treatment. In contrast to the effect in a transient ischemia model, the pretreatment effect did not depend on the 5HT1A receptor. Neurological function correlated favorably to the reduction in stroke size when SR141716A was given as a pretreatment. With the incidence of stroke predicted to rise in parallel with an ever aging population, understanding mechanisms underlying ischemia and therapeutics remains a paramount goal of research.
Collapse
Affiliation(s)
- Zachary Wilmer Reichenbach
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Medical Education and Research Building, 8th floor Philadelphia, PA, 19140, United States.
| | - Hongbo Li
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Medical Education and Research Building, 8th floor Philadelphia, PA, 19140, United States.
| | - Sara Jane Ward
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Medical Education and Research Building, 8th floor Philadelphia, PA, 19140, United States.
| | - Ronald F Tuma
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Medical Education and Research Building, 8th floor Philadelphia, PA, 19140, United States.
| |
Collapse
|
48
|
Guo S, Liu Y, Ma R, Li J, Su B. Neuroprotective effect of endogenous cannabinoids on ischemic brain injury induced by the excess microglia-mediated inflammation. Am J Transl Res 2016; 8:2631-2640. [PMID: 27398146 PMCID: PMC4931157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/29/2016] [Indexed: 06/06/2023]
Abstract
Increasing evidence has demonstrated the role of endogenous cannabinoids system (ECS) on protecting brain injury caused by ischemia (IMI). Papers reported that microglia-mediated inflammation has become one of the most pivotal mechanisms for IMI. This study was aimed to investigate the potential roles of ECS on neuron protection under microglia-mediated inflammation. Inflammatory cytokines level both in vitro (BV-2 cells) and in vivo (brain tissue from constructed IMI model and brain-isolated microglia) was detected. ECS levels were detected, and its effects on inflammations was also analyzed. Influence of microglia-mediated inflammation on neuron injury was analyzed. Moreover, the effects of ECS on protecting neuron injury were also analyzed. Our results showed that the levels of inflammatory cytokines including TNFα and IL-1β were higher while IKBα was lower in IMI model brain tissue, brain-isolated microglia and BV-2 cells compared to the control. Inflammation was activated in microglia, as well as the activation of ECS characterized by the increasing level of AEA and 2-AG. Furthermore, the activated microglia-mediated self-inflammation performed harmful influence on neurons via suppressing cell viability and inducing apoptosis. Moreover, ECS functioned as a protector on neuron injury though promoting cell proliferation and suppressing cell apoptosis which were caused by the activated BV-2 cells (LPS induced for 3 h). Our data suggested that ECS may play certain neuroprotective effects on microglia-mediated inflammations-induced IMI through anti-inflammatory function.
Collapse
Affiliation(s)
- Shuyun Guo
- Department of Pharmaceutics, Tangdu Hospital, The Fourth Military Medical UniversityXi’an, Shaanxi 710038, China
| | - Yanwu Liu
- Institute of Orthopedics, Xijing Hospital, The Fourth Military Medical UniversityXi’an, Shaanxi 710032, China
| | - Rui Ma
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical UniversityXi’an, Shaanxi 710032, China
| | - Jun Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical UniversityXi’an, Shaanxi 710032, China
| | - Binxiao Su
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical UniversityXi’an, Shaanxi 710032, China
| |
Collapse
|
49
|
Schroeder F, McIntosh AL, Martin GG, Huang H, Landrock D, Chung S, Landrock KK, Dangott LJ, Li S, Kaczocha M, Murphy EJ, Atshaves BP, Kier AB. Fatty Acid Binding Protein-1 (FABP1) and the Human FABP1 T94A Variant: Roles in the Endocannabinoid System and Dyslipidemias. Lipids 2016; 51:655-76. [PMID: 27117865 PMCID: PMC5408584 DOI: 10.1007/s11745-016-4155-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/11/2016] [Indexed: 01/01/2023]
Abstract
The first discovered member of the mammalian FABP family, liver fatty acid binding protein (FABP1, L-FABP), occurs at high cytosolic concentration in liver, intestine, and in the case of humans also in kidney. While the rat FABP1 is well studied, the extent these findings translate to human FABP1 is not clear-especially in view of recent studies showing that endocannabinoids and cannabinoids represent novel rat FABP1 ligands and FABP1 gene ablation impacts the hepatic endocannabinoid system, known to be involved in non-alcoholic fatty liver (NAFLD) development. Although not detectable in brain, FABP1 ablation nevertheless also impacts brain endocannabinoids. Despite overall tertiary structure similarity, human FABP1 differs significantly from rat FABP1 in secondary structure, much larger ligand binding cavity, and affinities/specificities for some ligands. Moreover, while both mouse and human FABP1 mediate ligand induction of peroxisome proliferator activated receptor-α (PPARα), they differ markedly in pattern of genes induced. This is critically important because a highly prevalent human single nucleotide polymorphism (SNP) (26-38 % minor allele frequency and 8.3 ± 1.9 % homozygous) results in a FABP1 T94A substitution that further accentuates these species differences. The human FABP1 T94A variant is associated with altered body mass index (BMI), clinical dyslipidemias (elevated plasma triglycerides and LDL cholesterol), atherothrombotic cerebral infarction, and non-alcoholic fatty liver disease (NAFLD). Resolving human FABP1 and the T94A variant's impact on the endocannabinoid and cannabinoid system is an exciting challenge due to the importance of this system in hepatic lipid accumulation as well as behavior, pain, inflammation, and satiety.
Collapse
Affiliation(s)
- Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA.
| | - Avery L McIntosh
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Huan Huang
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Sarah Chung
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Kerstin K Landrock
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Lawrence J Dangott
- Department of Biochemistry and Biophysics, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Shengrong Li
- Avanti Polar Lipids, 700 Industrial Park Dr., Alabaster, AL, 35007-9105, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Eric J Murphy
- Department of Pharmacology, Physiology, and Therapeutics and Chemistry, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Barbara P Atshaves
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| |
Collapse
|
50
|
Herrera MI, Kölliker-Frers R, Barreto G, Blanco E, Capani F. Glial Modulation by N-acylethanolamides in Brain Injury and Neurodegeneration. Front Aging Neurosci 2016; 8:81. [PMID: 27199733 PMCID: PMC4844606 DOI: 10.3389/fnagi.2016.00081] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/30/2016] [Indexed: 12/14/2022] Open
Abstract
Neuroinflammation involves the activation of glial cells and represents a key element in normal aging and pathophysiology of brain damage. N-acylethanolamides (NAEs), naturally occurring amides, are known for their pro-homeostatic effects. An increase in NAEs has been reported in vivo and in vitro in the aging brain and in brain injury. Treatment with NAEs may promote neuroprotection and exert anti-inflammatory actions via PPARα activation and/or by counteracting gliosis. This review aims to provide an overview of endogenous and exogenous properties of NAEs in neuroinflammation and to discuss their interaction with glial cells.
Collapse
Affiliation(s)
- María I Herrera
- Instituto de Investigaciones Cardiológicas, Facultad de Medicina, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina; Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología, Universidad Católica ArgentinaBuenos Aires, Argentina
| | - Rodolfo Kölliker-Frers
- Instituto de Investigaciones Cardiológicas, Facultad de Medicina, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas Buenos Aires, Argentina
| | - George Barreto
- Department of Nutrition and Biochemistry, Faculty of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Eduardo Blanco
- Departament de Pedagogia i Psicologia, Facultat d'Educació, Psicologia i Treball Social, Universitat de Lleida Lleida, Spain
| | - Francisco Capani
- Instituto de Investigaciones Cardiológicas, Facultad de Medicina, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina; Facultad de Psicología, Universidad Católica ArgentinaBuenos Aires, Argentina; Departamento de Biología, Universidad Argentina John F. KennedyBuenos Aires, Argentina; Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de ChileSantiago, Chile
| |
Collapse
|