1
|
Avagliano C, De Caro C, Cuozzo M, Roberti R, Russo E, La Rana G, Russo R. Sodium Butyrate ameliorates pain and mood disorders in a mouse model of Parkinson disease. Biomed Pharmacother 2025; 184:117903. [PMID: 39938349 DOI: 10.1016/j.biopha.2025.117903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025] Open
Abstract
Pain is one of non-motor features of Parkinson's disease (PD) that significantly impacts on patients' quality of life and increases the risk of developing psychiatric disorders. The mechanisms underlying pain in PD are poorly understood and the classic pharmacological treatments supplying to dopamine depletion have limited therapeutic effects on this symptom. It has been demonstrated that short chain fatty acids (SCFAs) play a key role in several central nervous system diseases including PD; low serum and faecal levels of SCFAs have been described in PD patients. Among SCFAs, the gut microbial metabolite butyrate has a neuroprotective and anti-inflammatory effect, influencing neurological and behavioural processes. Using a 6-hydroxydopamine (6-OHDA) induced-PD mouse model, we evaluated the effects of sodium butyrate (BuNa) treatment on pain and mood-related behaviour, exporing the role of PPARs, opioid and endocannabinoid systems. Our results demonstrated that repeated BuNa treatment (100 mg/kg po) in PD-mice reduced pain hypersensitivity as well as depressive- and anxiety-lke behaviour both on day 7 and day 14 after 6-OHDA injection. Moreover, AM281(CB1R antagonist), GW6471 (PPAR-alpha antagonist), and naloxone (opioid receptor antagonist), reduced BuNa efficacy. Finally, BuNa treatment was associated with a significant reduction of pro-inflammatory cytokines at spinal and supraspinal levels. In conclusion, our results demonstrate that increasing endogenous butyrate concentration reduces PD comorbidities such as pain and psychiatric symptoms, restoring opioidergic and endocannabinergic pathways.
Collapse
Affiliation(s)
- Carmen Avagliano
- CEINGE-Biotechnlogies Advances, via Gaetano Salvatore 486, Naples, Italy; Department of Pharmacy, University of Naples "Federico II", via D. Montesano, 49, Naples 80131, Italy.
| | - Carmen De Caro
- Department of Pharmacy, University of Naples "Federico II", via D. Montesano, 49, Naples 80131, Italy.
| | - Mariarosaria Cuozzo
- CEINGE-Biotechnlogies Advances, via Gaetano Salvatore 486, Naples, Italy; Department of Anatomy and Neuroscience, APC Microbiome, University Collage of Cork, Ireland.
| | - Roberta Roberti
- Department of Health Sciences, School of Medicine, University of Catanzaro "Magna Graecia", Viale Europa, Catanzaro 88100, Italy.
| | - Emilio Russo
- Department of Health Sciences, School of Medicine, University of Catanzaro "Magna Graecia", Viale Europa, Catanzaro 88100, Italy.
| | - Giovanna La Rana
- Department of Pharmacy, University of Naples "Federico II", via D. Montesano, 49, Naples 80131, Italy.
| | - Roberto Russo
- Department of Pharmacy, University of Naples "Federico II", via D. Montesano, 49, Naples 80131, Italy.
| |
Collapse
|
2
|
Urmeneta-Ortíz MF, Tejeda-Martínez AR, González-Reynoso O, Flores-Soto ME. Potential Neuroprotective Effect of the Endocannabinoid System on Parkinson's Disease. PARKINSON'S DISEASE 2024; 2024:5519396. [PMID: 39104613 PMCID: PMC11300097 DOI: 10.1155/2024/5519396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by alterations in motor capacity resulting from a decrease in the neurotransmitter dopamine due to the selective death of dopaminergic neurons of the nigrostriatal pathway. Unfortunately, conventional pharmacological treatments fail to halt disease progression; therefore, new therapeutic strategies are needed, and currently, some are being investigated. The endocannabinoid system (ECS), highly expressed in the basal ganglia (BG) circuit, undergoes alterations in response to dopaminergic depletion, potentially contributing to motor symptoms and the etiopathogenesis of PD. Substantial evidence supports the neuroprotective role of the ECS through various mechanisms, including anti-inflammatory, antioxidative, and antiapoptotic effects. Therefore, the ECS emerges as a promising target for PD treatment. This review provides a comprehensive summary of current clinical and preclinical evidence concerning ECS alterations in PD, along with potential pharmacological targets that may exert the protection of dopaminergic neurons.
Collapse
Affiliation(s)
- María Fernanda Urmeneta-Ortíz
- Chemical Engineering Department, University Center for Exact and Engineering SciencesUniversity of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara C.P. 44430, Jalisco, Mexico
- Cellular and Molecular Neurobiology LaboratoryNeurosciences DivisionWestern Biomedical Research Center (CIBO)Mexican Social Security Institute, Sierra Mojada #800, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| | - Aldo Rafael Tejeda-Martínez
- Cellular and Molecular Neurobiology LaboratoryNeurosciences DivisionWestern Biomedical Research Center (CIBO)Mexican Social Security Institute, Sierra Mojada #800, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| | - Orfil González-Reynoso
- Chemical Engineering Department, University Center for Exact and Engineering SciencesUniversity of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara C.P. 44430, Jalisco, Mexico
| | - Mario Eduardo Flores-Soto
- Cellular and Molecular Neurobiology LaboratoryNeurosciences DivisionWestern Biomedical Research Center (CIBO)Mexican Social Security Institute, Sierra Mojada #800, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
3
|
Branković M, Gmizić T, Dukić M, Zdravković M, Daskalović B, Mrda D, Nikolić N, Brajković M, Gojgić M, Lalatović J, Kralj Đ, Pantić I, Vojnović M, Milovanović T, Đurašević S, Todorović Z. Therapeutic Potential of Palmitoylethanolamide in Gastrointestinal Disorders. Antioxidants (Basel) 2024; 13:600. [PMID: 38790705 PMCID: PMC11117950 DOI: 10.3390/antiox13050600] [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: 02/29/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Palmitoylethanolamide (PEA) is an endocannabinoid-like bioactive lipid mediator belonging to the family of N-acylethanolamines, most abundantly found in peanuts and egg yolk. When the gastrointestinal (GI) effects of PEA are discussed, it must be pointed out that it affects intestinal motility but also modulates gut microbiota. This is due to anti-inflammatory, antioxidant, analgesic, antimicrobial, and immunomodulatory features. Additionally, PEA has shown beneficial effects in several GI diseases, particularly irritable bowel syndrome and inflammatory bowel diseases, as various studies have shown, and it is important to emphasize its relative lack of toxicity, even at high dosages. Unfortunately, there is not enough endogenous PEA to treat disturbed gut homeostasis, even though it is produced in the GI tract in response to inflammatory stimuli, so exogenous intake is mandatory to achieve homeostasis. Intake of PEA could be through animal and/or vegetable food, but bearing in mind that a high dosage is needed to achieve a therapeutic effect, it must be compensated through dietary supplements. There are still open questions pending to be answered, so further studies investigating PEA's effects and mechanisms of action, especially in humans, are crucial to implementing PEA in everyday clinical practice.
Collapse
Affiliation(s)
- Marija Branković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Tijana Gmizić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Marija Dukić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Marija Zdravković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | | | - Davor Mrda
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Novica Nikolić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Milica Brajković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Milan Gojgić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Jovana Lalatović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Đorđe Kralj
- University Hospital Medical Center Zvezdara, 11000 Belgrade, Serbia;
| | - Ivana Pantić
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (I.P.); (M.V.)
| | - Marko Vojnović
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (I.P.); (M.V.)
| | - Tamara Milovanović
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (I.P.); (M.V.)
| | - Siniša Đurašević
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Đaja, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| | - Zoran Todorović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
4
|
Pirozzi C, Coretti L, Opallo N, Bove M, Annunziata C, Comella F, Turco L, Lama A, Trabace L, Meli R, Lembo F, Mattace Raso G. Palmitoylethanolamide counteracts high-fat diet-induced gut dysfunction by reprogramming microbiota composition and affecting tryptophan metabolism. Front Nutr 2023; 10:1143004. [PMID: 37599675 PMCID: PMC10434518 DOI: 10.3389/fnut.2023.1143004] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 07/04/2023] [Indexed: 08/22/2023] Open
Abstract
Obesity is associated with gastrointestinal (GI) tract and central nervous system (CNS) disorders. High-fat diet (HFD) feeding-induced obesity in mice induces dysbiosis, causing a shift toward bacteria-derived metabolites with detrimental effects on metabolism and inflammation: events often contributing to the onset and progression of both GI and CNS disorders. Palmitoylethanolamide (PEA) is an endogenous lipid mediator with beneficial effects in mouse models of GI and CNS disorders. However, the mechanisms underlining its enteroprotective and neuroprotective effects still need to be fully understood. Here, we aimed to study the effects of PEA on intestinal inflammation and microbiota alterations resulting from lipid overnutrition. Ultramicronized PEA (30 mg/kg/die per os) was administered to HFD-fed mice for 7 weeks starting at the 12th week of HFD regimen. At the termination of the study, the effects of PEA on inflammatory factors and cells, gut microbial features and tryptophan (TRP)-kynurenine metabolism were evaluated. PEA regulates the crosstalk between the host immune system and gut microbiota via rebalancing colonic TRP metabolites. PEA treatment reduced intestinal immune cell recruitment, inflammatory response triggered by HFD feeding, and corticotropin-releasing hormone levels. In particular, PEA modulated HFD-altered TRP metabolism in the colon, rebalancing serotonin (5-HT) turnover and reducing kynurenine levels. These effects were associated with a reshaping of gut microbiota composition through increased butyrate-promoting/producing bacteria, such as Bifidobacterium, Oscillospiraceae and Turicibacter sanguinis, with the latter also described as 5-HT sensor. These data indicate that the rebuilding of gut microbiota following PEA supplementation promotes host 5-HT biosynthesis, which is crucial in regulating intestinal function.
Collapse
Affiliation(s)
- Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Lorena Coretti
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Nicola Opallo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Maria Bove
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Chiara Annunziata
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Federica Comella
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Luigia Turco
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Francesca Lembo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
Turco L, Opallo N, Buommino E, De Caro C, Pirozzi C, Mattace Raso G, Lembo F, Coretti L. Zooming into Gut Dysbiosis in Parkinson's Disease: New Insights from Functional Mapping. Int J Mol Sci 2023; 24:ijms24119777. [PMID: 37298727 DOI: 10.3390/ijms24119777] [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: 03/30/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Gut dysbiosis has been involved in the pathogenesis and progression of Parkinson's disease (PD), but the mechanisms through which gut microbiota (GM) exerts its influences deserve further study. Recently, we proposed a two-hit mouse model of PD in which ceftriaxone (CFX)-induced dysbiosis amplifies the neurodegenerative phenotype generated by striatal 6-hydroxydopamine (6-OHDA) injection in mice. Low GM diversity and the depletion of key gut colonizers and butyrate producers were the main signatures of GM alteration in this model. Here, we used the phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt2) to unravel candidate pathways of cell-to-cell communication associated with dual-hit mice and potentially involved in PD progression. We focused our analysis on short-chain fatty acids (SCFAs) metabolism and quorum sensing (QS) signaling. Based on linear discriminant analysis, combined with the effect size results, we found increased functions linked to pyruvate utilization and a depletion of acetate and butyrate production in 6-OHDA+CFX mice. The specific arrangement of QS signaling as a possible result of the disrupted GM structure was also observed. With this exploratory study, we suggested a scenario in which SCFAs metabolism and QS signaling might represent the effectors of gut dysbiosis potentially involved in the designation of the functional outcomes that contribute to the exacerbation of the neurodegenerative phenotype in the dual-hit animal model of PD.
Collapse
Affiliation(s)
- Luigia Turco
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Nicola Opallo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Elisabetta Buommino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Carmen De Caro
- Department of Science of Health, School of Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| | - Francesca Lembo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| | - Lorena Coretti
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| |
Collapse
|
6
|
Shao J, Liu X, Lian M, Mao Y. Citronellol Prevents 6-OHDA-Induced Oxidative Stress, Mitochondrial Dysfunction, and Apoptosis in Parkinson Disease Model of SH-SY5Y Cells via Modulating ROS-NO, MAPK/ERK, and PI3K/Akt Signaling Pathways. Neurotox Res 2022; 40:2221-2237. [PMID: 36097250 DOI: 10.1007/s12640-022-00558-8] [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: 04/26/2022] [Revised: 07/13/2022] [Accepted: 08/09/2022] [Indexed: 12/31/2022]
Abstract
Parkinson disease is a neurodegenerative disorder distinguished by dopaminergic shortage in the striatum and the accumulation of α-synuclein neuronal aggregates in the brains of patients. Since, there is no accurate treatment available for Parkinson disease, researches are designed to alleviate the pathognomonic symptoms such as neuroinflammation, oxidative stress, mitochondrial dysfunction, and apoptosis. Accordingly, a number of compounds have been reported to inhibit these pathognomonic symptoms. In this study, we have assessed the neuroprotective potential of citronellol against 6-OHDA-induced neurotoxicity in SH-SY5Y cells. The results found that citronellol treatment effectively hindered the cell death caused by 6-OHDA and thereby maintaining the cell viability in SH-SY5Y cells at 50 µg/mL concentration. As expected, the citronellol treatment significantly reduced the 6-OHDA-induced secretion of inflammatory factors (IL-1β, IL-6, and TNF-α), which was obtained through ELISA technique. Similarly, citronellol hindered the 6-OHDA-induced oxidative stress by lowering the intracellular ROS and NO level and MDA leakage along with increased expression of SOD level in SH-SY5Y cells. The JC-1 staining showed that 6-OHDA increased the number of green fluorescent dots with ruptured mitochondrial membrane potential, while citronellol increased the amount of red fluorescent, showing the rescue potential against the 6-OHDA-induced mitochondrial dysfunction. Furthermore, citronellol hampered the 6-OHDA-induced apoptosis via the suppression of Bcl-2/Bax pathway. The western blotting results hypothesized that citronellol rescued SH-SY5Y cells from 6-OHDA-induced neurotoxicity via modulating ROS-NO, MAPK/ERK, and PI3K/Akt signaling pathways. However, further clinical trials are required to verify the anti-Parkinson efficacy.
Collapse
Affiliation(s)
- Jiahui Shao
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Xuan Liu
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Mengjia Lian
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Youbing Mao
- Department of Special Inspection Section, The First People's Hospital of Wenling, No. 333, Chuanan South Road, Chengxi StreetZhejiang Province, Wenling, 317500, China.
| |
Collapse
|
7
|
Vellingiri B, Chandrasekhar M, Sri Sabari S, Gopalakrishnan AV, Narayanasamy A, Venkatesan D, Iyer M, Kesari K, Dey A. Neurotoxicity of pesticides - A link to neurodegeneration. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:113972. [PMID: 36029574 DOI: 10.1016/j.ecoenv.2022.113972] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 05/15/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder which mainly targets motor symptoms such as tremor, rigidity, bradykinesia and postural instability. The physiological changes occur due to dopamine depletion in basal ganglia region of the brain. PD aetiology is not yet elucidated clearly but genetic and environmental factors play a prominent role in disease occurrence. Despite of various environmental factors, pesticides exposure has been convicted as major candidate in PD pathogenesis. Among various pesticides 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) has been widely investigated in PD following with paraquat (PQ), maneb (MB), organochlorines (OC) and rotenone. Effect of these pesticides has been suggested to be involved in oxidative stress, alterations in dopamine transporters, mitochondrial dysfunction, α-synuclein (αSyn) fibrillation, and neuroinflammation in PD. The present review discusses the influence of pesticides in neurodegeneration and its related epidemiological studies conducted in PD. Furthermore, we have deliberated the common pesticides involved in PD and its associated genetic alterations and the probable mechanism of them behind PD pathogenesis. Hence, we conclude that pesticides play a prominent role in PD pathogenesis and advance research is needed to investigate the alterations in genetic and mechanistic aspects of PD.
Collapse
Affiliation(s)
- Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India.
| | - Mamatha Chandrasekhar
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - S Sri Sabari
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Livestock Farming and Bioresource Technology, Tamil Nadu, India
| | - Kavindra Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo, 00076, Finland.
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, 700073, West Bengal, India
| |
Collapse
|
8
|
Palese F, Pontis S, Realini N, Torrens A, Ahmed F, Assogna F, Pellicano C, Bossù P, Spalletta G, Green K, Piomelli D. Targeting NAAA counters dopamine neuron loss and symptom progression in mouse models of parkinsonism. Pharmacol Res 2022; 182:106338. [PMID: 35781057 PMCID: PMC9733952 DOI: 10.1016/j.phrs.2022.106338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/19/2022] [Accepted: 06/29/2022] [Indexed: 12/13/2022]
Abstract
The lysosomal cysteine hydrolase N-acylethanolamine acid amidase (NAAA) deactivates palmitoylethanolamide (PEA), a lipid-derived PPAR-α agonist that is critically involved in the control of pain and inflammation. In this study, we asked whether NAAA-regulated PEA signaling might contribute to dopamine neuron degeneration and parkinsonism induced by the mitochondrial neurotoxins, 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In vitro experiments showed that 6-OHDA and MPTP enhanced NAAA expression and lowered PEA content in human SH-SY5Y cells. A similar effect was observed in mouse midbrain dopamine neurons following intra-striatal 6-OHDA injection. Importantly, deletion of the Naaa gene or pharmacological inhibition of NAAA activity substantially attenuated both dopamine neuron death and parkinsonian symptoms in mice treated with 6-OHDA or MPTP. Moreover, NAAA expression was elevated in postmortem brain cortex and premortem blood-derived exosomes from persons with Parkinson's disease compared to age-matched controls. The results identify NAAA-regulated PEA signaling as a molecular control point for dopaminergic neuron survival and a potential target for neuroprotective intervention.
Collapse
Affiliation(s)
- Francesca Palese
- Department of Anatomy and Neurobiology University of California Irvine, 92697-1275 CA, USA
| | - Silvia Pontis
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Natalia Realini
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Alexa Torrens
- Department of Anatomy and Neurobiology University of California Irvine, 92697-1275 CA, USA
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology University of California Irvine, 92697-1275 CA, USA
| | - Francesca Assogna
- Laboratorio di Neuropsichiatria, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Clelia Pellicano
- Laboratorio di Neuropsichiatria, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Paola Bossù
- Laboratorio di Neuropsichiatria, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Gianfranco Spalletta
- Laboratorio di Neuropsichiatria, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Kim Green
- Department of Neurobiology and Behavior, University of California Irvine, 92697-1275 CA, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology University of California Irvine, 92697-1275 CA, USA,Department of Pharmaceutical Sciences, University of California Irvine, 92697-1275 CA, USA,Department of Biological Chemistry, University of California Irvine, 92697-1275 CA, USA
| |
Collapse
|
9
|
Sergeeva OA, Mazur K, Reiner-Link D, Lutsenko K, Haas HL, Alfonso-Prieto M, Stark H. OLHA (N α-oleoylhistamine) modulates activity of mouse brain histaminergic neurons. Neuropharmacology 2022; 215:109167. [PMID: 35750238 DOI: 10.1016/j.neuropharm.2022.109167] [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: 01/28/2022] [Revised: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 11/26/2022]
Abstract
Histaminergic (HA) neurons are located in the tuberomamillary nucleus (TMN) of the posterior hypothalamus, from where they project throughout the whole brain to control wakefulness. We examined the effects of Nα-oleoylhistamine (OLHA), a non-enzymatic condensation product of oleic acid (OLA) and histamine, on activity of mouse HA neurons in brain slices. OLHA bidirectionally modulated the firing of HA neurons. At 10 nM OLHA inhibited or had no action, whereas at 1 μM it evoked excitatory and inhibitory responses. Inhibition was not seen in presence of the histamine receptor H3 (H3R) antagonist clobenpropit and in calcium-free medium. Pre-incubation with a histamine-reuptake blocker prevented the decrease in firing by OLHA. OLHA-evoked increase in firing (EC50 ∼44 nM) was insensitive to blockers of cannabinoid 1 and 2 receptors and of the capsaicin receptor, but was significantly impaired by the peroxisome proliferator-activated receptor-alpha (PPAR-alpha) antagonist MK886, which suppressed also the rise in intracellular calcium level caused by OLHA. The OLHA-evoked excitation was mimicked by synthetic PPAR-alpha agonists (gemfibrozil and GW7647) and was abolished by the PKA inhibitor H-89. The H3R affinity (Ki) for histamine, measured in HEK293 cells with stable expression of human H3R, was higher than for OLHA (Ki: 42 vs 310 nM, respectively). Expression of PPAR-alpha was not different between TMN regions of males and females, responses to OLHA did not differ. Molecular modelling of PPAR-alpha bound to either OLHA or OEA showed similar binding energies. These findings shed light on a novel biotransformation product of histamine which may play a role in health and disease.
Collapse
Affiliation(s)
- Olga A Sergeeva
- Institute of Neural and Sensory Physiology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany; Institute of Clinical Neurosciences and Medical Psychology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.
| | - Karolina Mazur
- Institute of Clinical Neurosciences and Medical Psychology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - David Reiner-Link
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Kiril Lutsenko
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Helmut L Haas
- Institute of Neural and Sensory Physiology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Mercedes Alfonso-Prieto
- Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany; Computational Biomedicine, Institute for Advanced Simulation IAS-5/Institute for Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| |
Collapse
|
10
|
Avagliano C, Coretti L, Lama A, Pirozzi C, De Caro C, De Biase D, Turco L, Mollica MP, Paciello O, Calignano A, Meli R, Lembo F, Mattace Raso G. Dual-Hit Model of Parkinson's Disease: Impact of Dysbiosis on 6-Hydroxydopamine-Insulted Mice-Neuroprotective and Anti-Inflammatory Effects of Butyrate. Int J Mol Sci 2022; 23:ijms23126367. [PMID: 35742813 PMCID: PMC9223521 DOI: 10.3390/ijms23126367] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Recent evidence highlights Parkinson’s disease (PD) initiation in the gut as the prodromal phase of neurodegeneration. Gut impairment due to microbial dysbiosis could affect PD pathogenesis and progression. Here, we propose a two-hit model of PD through ceftriaxone (CFX)-induced dysbiosis and gut inflammation before the 6-hydroxydopamine (6-OHDA) intrastriatal injection to mimic dysfunctional gut-associated mechanisms preceding PD onset. Therefore, we showed that dysbiosis and gut damage amplified PD progression, worsening motor deficits induced by 6-OHDA up to 14 days post intrastriatal injection. This effect was accompanied by a significant increase in neuronal dopaminergic loss (reduced tyrosine hydroxylase expression and increased Bcl-2/Bax ratio). Notably, CFX pretreatment also enhanced systemic and colon inflammation of dual-hit subjected mice. The exacerbated inflammatory response ran in tandem with a worsening of colonic architecture and gut microbiota perturbation. Finally, we demonstrated the beneficial effect of post-biotic sodium butyrate in limiting at once motor deficits, neuroinflammation, and colon damage and re-shaping microbiota composition in this novel dual-hit model of PD. Taken together, the bidirectional communication of the microbiota–gut–brain axis and the recapitulation of PD prodromal/pathogenic features make this new paradigm a useful tool for testing or repurposing new multi-target compounds in the treatment of PD.
Collapse
Affiliation(s)
- Carmen Avagliano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
| | - Lorena Coretti
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
- Correspondence: (A.L.); (G.M.R.); Tel.: +39-081678409 (A.L.); Tel.: +39-081678423 (G.M.R.)
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
| | - Carmen De Caro
- Department of Science of Health, School of Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy;
| | - Davide De Biase
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
| | - Luigia Turco
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Maria Pina Mollica
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte Sant’Angelo, Cupa Nuova Cinthia 21 Edificio, 80126 Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via Delpino, 80137 Naples, Italy;
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
| | - Francesca Lembo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
- Correspondence: (A.L.); (G.M.R.); Tel.: +39-081678409 (A.L.); Tel.: +39-081678423 (G.M.R.)
| |
Collapse
|
11
|
Effects of Palmitoylethanolamide on Neurodegenerative Diseases: A Review from Rodents to Humans. Biomolecules 2022; 12:biom12050667. [PMID: 35625595 PMCID: PMC9138306 DOI: 10.3390/biom12050667] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 02/06/2023] Open
Abstract
Palmitoylethanolamide (PEA) stands out among endogenous lipid mediators for its neuroprotective, anti-inflammatory, and analgesic functions. PEA belonging to the N-acetylanolamine class of phospholipids was first isolated from soy lecithin, egg yolk, and peanut flour. It is currently used for the treatment of different types of neuropathic pain, such as fibromyalgia, osteoarthritis, carpal tunnel syndrome, and many other conditions. The properties of PEA, especially of its micronized or ultra-micronized forms maximizing bioavailability and efficacy, have sparked a series of innovative research to evaluate its possible application as therapeutic agent for neurodegenerative diseases. Neurodegenerative diseases are widespread throughout the world, and although they are numerous and different, they share common patterns of conditions that result from progressive damage to the brain areas involved in mobility, muscle coordination and strength, mood, and cognition. The present review is aimed at illustrating in vitro and in vivo research, as well as human studies, using PEA treatment, alone or in combination with other compounds, in the presence of neurodegeneration. Namely, attention has been paid to the effects of PEA in counteracting neuroinflammatory conditions and in slowing down the progression of diseases, such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, Frontotemporal dementia, Amyotrophic Lateral Sclerosis, and Multiple Sclerosis. Literature research demonstrated the efficacy of PEA in addressing the damage typical of major neurodegenerative diseases.
Collapse
|
12
|
Iranshahy M, Javadi B, Sahebkar A. Protective effects of functional foods against Parkinson's disease: A narrative review on pharmacology, phytochemistry, and molecular mechanisms. Phytother Res 2022; 36:1952-1989. [PMID: 35244296 DOI: 10.1002/ptr.7425] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/25/2022] [Accepted: 02/07/2022] [Indexed: 12/30/2022]
Abstract
In Persian Medicine (PM), PD (brain-based tremor) is a known CNS disorder with several therapeutic and preventive options. In their medical textbooks and pharmacopeias, Persian great scientists such as Rhazes (854-925 AD), Avicenna (980-1037 AD), and Jorjani (1042-1136 AD), have discussed pharmacological and nutritional strategies for the prevention, slowing progression, and treatment of PD. In the present study, we surveyed plant- and animal-based foods recommended by PM for the prevention and treatment of CNS-related tremors. In vivo and in-vitro pharmacological evidence supporting the beneficial effects of PM-recommended foods in prevention and alleviating PD, major active phytochemicals along with the relevant mechanisms of action were studied. Several PM plants possess potent antioxidant, antiinflammatory, and PD preventing properties. Garlic and allicin, cabbage and isothiocyanates, chickpea seed and its O-methylated isoflavones biochanin A and formononetin, cinnamon, and cinnamaldehyde, saffron and its crocin, crocetin, and safranal, black cumin and its thymoquinone, black pepper and piperine, pistachio and genistein and daidzein, and resveratrol are among the most effective dietary itemsagainst PD. They act through attenuating neurotoxin-induced memory loss and behavioral impairment, oxidative stress, and dopaminergic cell death. PM-recommended foods can help alleviate PD progression and also discovering and developing new neuroprotective anti-PD pharmaceuticals.
Collapse
Affiliation(s)
- Milad Iranshahy
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Behjat Javadi
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Angelopoulou E, Paudel YN, Bougea A, Piperi C. Impact of the apelin/APJ axis in the pathogenesis of Parkinson's disease with therapeutic potential. J Neurosci Res 2021; 99:2117-2133. [PMID: 34115895 DOI: 10.1002/jnr.24895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 12/18/2022]
Abstract
The pathogenesis of Parkinson's disease (PD) remains elusive. There is still no available disease-modifying strategy against PD, whose management is mainly symptomatic. A growing amount of preclinical evidence shows that a complex interplay between autophagy dysregulation, mitochondrial impairment, endoplasmic reticulum stress, oxidative stress, and excessive neuroinflammation underlies PD pathogenesis. Identifying key molecules linking these pathological cellular processes may substantially aid in our deeper understanding of PD pathophysiology and the development of novel effective therapeutic approaches. Emerging preclinical evidence indicates that apelin, an endogenous neuropeptide acting as a ligand of the orphan G protein-coupled receptor APJ, may play a key neuroprotective role in PD pathogenesis, via inhibition of apoptosis and dopaminergic neuronal loss, autophagy enhancement, antioxidant effects, endoplasmic reticulum stress suppression, as well as prevention of synaptic dysregulation in the striatum, excessive neuroinflammation, and glutamate-induced excitotoxicity. Underlying signaling pathways involve phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin, extracellular signal-regulated kinase 1/2, and inositol requiring kinase 1α/XBP1/C/EBP homologous protein. Herein, we discuss the role of apelin/APJ axis and associated molecular mechanisms on the pathogenesis of PD in vitro and in vivo and provide evidence for its challenging therapeutic potential.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Department of Neurology, Eginition University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Anastasia Bougea
- Department of Neurology, Eginition University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
14
|
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
|
15
|
Beggiato S, Cassano T, Ferraro L, Tomasini MC. Astrocytic palmitoylethanolamide pre-exposure exerts neuroprotective effects in astrocyte-neuron co-cultures from a triple transgenic mouse model of Alzheimer's disease. Life Sci 2020; 257:118037. [PMID: 32622942 DOI: 10.1016/j.lfs.2020.118037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 12/27/2022]
Abstract
Palmitoylethanolamide (PEA) is an endogenous lipid mediator that, also by blunting astrocyte activation, demonstrated beneficial properties in several in vitro and in vivo models of Alzheimer's disease (AD). In the present study, we used astrocyte-neuron co-cultures from 3xTg-AD mouse (i.e. an animal model of AD) cerebral cortex to further investigate on the role of astrocytes in PEA-induced neuroprotection. To this aim, we evaluated the number of viable cells, apoptotic nuclei, microtubule-associated protein-2 (MAP2) positive cells and morphological parameters in cortical neurons co-cultured with cortical astrocytes pre-exposed, or not, to Aβ42 (0.5 μM; 24 h) or PEA (0.1 μM; 24 h). Pre-exposure of astrocytes to Aβ42 failed to affect the viability, the number of neuronal apoptotic nuclei, MAP2 positive cell number, neuritic aggregations/100 μm, dendritic branches per neuron, the neuron body area, the length of the longest dendrite and number of neurites/neuron in 3xTg-AD mouse astrocyte-neuron co-cultures. Compared to neurons from wild-type (non-Tg) mouse co-cultures, 3xTg-AD mouse neurons co-cultured with astrocytes from this mutant mice displayed higher number of apoptotic nuclei, lower MAP2 immunoreactivity and several morphological changes. These signs of neuronal suffering were significantly counteracted when the 3xTg-AD mouse cortical neurons were co-cultured with 3xTg-AD mouse astrocytes pre-exposed to PEA. The present data suggest that in astrocyte-neuron co-cultures from 3xTg-AD mice, astrocytes contribute to neuronal damage and PEA, by possibly counteracting reactive astrogliosis, improved neuronal survival. These findings further support the role of PEA as a possible new therapeutic opportunity in AD treatment.
Collapse
Affiliation(s)
- Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Borsari, 36-44121 Ferrara, Italy; Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini, 31-66100 Chieti, Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, viale Pinto, 1-71122 Foggia, Italy
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Borsari, 36-44121 Ferrara, Italy; Department of Clinical and Experimental Medicine, University of Foggia, viale Pinto, 1-71122 Foggia, Italy; IRET Foundation, Via Tolara di Sopra 41 - 40064 Ozzano dell'Emilia, Bologna, Italy; Technopole of Ferrara, LTTA Laboratory for the Technologies for Advanced Therapies, Via Fossato di Mortara 70, 44121 Ferrara, Italy.
| | - Maria C Tomasini
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Borsari, 36-44121 Ferrara, Italy
| |
Collapse
|
16
|
Baptista LC, Sun Y, Carter CS, Buford TW. Crosstalk Between the Gut Microbiome and Bioactive Lipids: Therapeutic Targets in Cognitive Frailty. Front Nutr 2020; 7:17. [PMID: 32219095 PMCID: PMC7078157 DOI: 10.3389/fnut.2020.00017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/13/2020] [Indexed: 12/14/2022] Open
Abstract
Cognitive frailty is a geriatric condition defined by the coexistence of cognitive impairment and physical frailty. This "composite" aging phenotype is associated with a higher risk of several adverse health-related outcomes, including dementia. In the last decade, cognitive frailty has gained increased attention from the scientific community that has focused on understanding the clinical impact and the physiological and pathological mechanisms of development and on identifying preventive and/or rehabilitative therapeutic interventions. The emergence of gut microbiome in neural signaling increased the interest in targeting the gut-brain axis as a modulation strategy. Multiple studies on gastroenteric, metabolic, and neurodegenerative diseases support the existence of a wide bidirectional communication network of signaling mediators, e.g., bioactive lipids, that can modulate inflammation, gut permeability, microbiota composition, and the gut-brain axis. This crosstalk between the gut-brain axis, microbiome, and bioactive lipids may emerge as the basis of a promising therapeutic strategy to counteract cognitive frailty. In this review, we summarize the evidence in the literature regarding the link between the gut microbiome, brain, and several families of bioactive lipids. In addition, we also explore the applicability of several bioactive lipid members as a potential routes for therapeutic interventions to combat cognitive frailty.
Collapse
Affiliation(s)
- Liliana C. Baptista
- Division of Gerontology, Geriatrics and Palliative Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yi Sun
- Division of Gerontology, Geriatrics and Palliative Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christy S. Carter
- Division of Gerontology, Geriatrics and Palliative Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: Christy S. Carter
| | - Thomas W. Buford
- Division of Gerontology, Geriatrics and Palliative Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, United States,Thomas W. Buford ; Twitter: @twbuford
| |
Collapse
|
17
|
Lama A, Pirozzi C, Avagliano C, Annunziata C, Mollica MP, Calignano A, Meli R, Mattace Raso G. Nutraceuticals: An integrative approach to starve Parkinson's disease. Brain Behav Immun Health 2020; 2:100037. [PMID: 34589828 PMCID: PMC8474522 DOI: 10.1016/j.bbih.2020.100037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/08/2020] [Accepted: 01/12/2020] [Indexed: 01/15/2023] Open
Abstract
The therapeutic approach of multifactorial complex diseases is always a challenge; Parkinson's disease (PD) is a heterogeneous neurodegenerative disorder triggered by genetic and environmental factors, contributing to its etiology. Indeed, several pathogenic mechanisms lead to selective dopaminergic neuronal injury, including oxidative stress, mitochondrial dysfunction, alteration of endoplasmic reticulum-to-Golgi protein trafficking, excitotoxicity, and neuroinflammation. Current treatment approaches include mainly dopamine replacement therapy or optimizing dopaminergic transmission; however, these strategies that do not counteract the pathogenic mechanisms underlying PD symptoms and often are less effective over time. Recently, there has been growing interest in the therapeutic use of nutraceuticals, that could represent an integrative approach to the pharmacological standard therapy and specifically affect one or more pathogenic pathways. The intake of nutraceuticals or nutritional modifications are generally safe and can be combined with current common drug therapy in most cases to improve the patient's quality of life and/or mitigate PD symptoms. The current review focuses on several key nutritional compounds and dietary modifications that are effective on several pathogenic pathways involved in PD onset and progression, and further highlights the rationale behind their potential use for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Adriano Lama
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Chiara Annunziata
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Maria Pina Mollica
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
- Department of Biology, University of Naples Federico II, Cupa Nuova Cinthia 21-Edificio 7, 80126, Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Rosaria Meli
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
18
|
Udovin LD, Kobiec T, Herrera MI, Toro-Urrego N, Kusnier CF, Kölliker-Frers RA, Ramos-Hryb AB, Luaces JP, Otero-Losada M, Capani F. Partial Reversal of Striatal Damage by Palmitoylethanolamide Administration Following Perinatal Asphyxia. Front Neurosci 2020; 13:1345. [PMID: 31969800 PMCID: PMC6960201 DOI: 10.3389/fnins.2019.01345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 11/29/2019] [Indexed: 01/27/2023] Open
Abstract
Perinatal asphyxia (PA) is a clinical condition brought by a birth temporary oxygen deprivation associated with long-term damage in the corpus striatum, one of the most compromised brain areas. Palmitoylethanolamide (PEA) is a neuromodulator well known for its protective effects in brain injury models, including PA, albeit not deeply studied regarding its particular effects in the corpus striatum following PA. Using Bjelke et al. (1991) PA model, full-term pregnant rats were decapitated, and uterus horns were placed in a water bath at 37°C for 19 min. One hour later, the pups were injected with PEA 10 mg/kg s.c., and placed with surrogate mothers. After 30 days, the animals were perfused, and coronal striatal sections were collected to analyze protein-level expression by Western blot and the reactive area by immunohistochemistry for neuron markers: phosphorylated neurofilament-heavy/medium-chain (pNF-H/M) and microtubule-associated protein-2 (MAP-2), and the astrocyte marker, glial fibrillary acidic protein (GFAP). Results indicated that PA produced neuronal damage and morphological changes. Asphyctic rats showed a decrease in pNF-H/M and MAP-2 reactive areas, GFAP+ cells number, and MAP-2 as well as pNF-H/M protein expression in the striatum. Treatment with PEA largely restored the number of GFAP+ cells. Most important, it ameliorated the decrease in pNF-H/M and MAP-2 reactive areas in asphyctic rats. Noticeably, PEA treatment reversed the decrease in MAP-2 protein expression and largely prevented PA-induced decrease in pNF-H/M protein expression. PA did not affect the GFAP protein level. Treatment with PEA attenuated striatal damage induced by PA, suggesting its therapeutic potential for the prevention of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lucas D Udovin
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Tamara Kobiec
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina.,Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - María I Herrera
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina.,Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Nicolás Toro-Urrego
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Carlos F Kusnier
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Rodolfo A Kölliker-Frers
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Ana B Ramos-Hryb
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Juan P Luaces
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Francisco Capani
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina.,Departamento de Biología, Universidad Argentina John F. Kennedy (UAJK), Buenos Aires, Argentina
| |
Collapse
|
19
|
Kao YC, Wei WY, Tsai KJ, Wang LC. High Fat Diet Suppresses Peroxisome Proliferator-Activated Receptors and Reduces Dopaminergic Neurons in the Substantia Nigra. Int J Mol Sci 2019; 21:ijms21010207. [PMID: 31892244 PMCID: PMC6981702 DOI: 10.3390/ijms21010207] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
Although several epidemiologic and animal studies have revealed correlations between obesity and neurodegenerative disorders, such as Parkinson disease (PD), the underlying pathological mechanisms of obesity-induced PD remain unclear. Our study aimed to assess the effect of diet-induced obesity on the brain dopaminergic pathway. For five months, starting from weaning, we gave C57BL/6 mice a high-fat diet (HFD) to generate an obese mouse model and investigate whether the diet reprogrammed the midbrain dopaminergic system. Tyrosine hydroxylase staining showed that the HFD resulted in fewer dopaminergic neurons in the substantia nigra (SN), but not the striatum. It also induced neuroinflammation, with increased astrogliosis in the SN and striatum. Dendritic spine density in the SN of HFD-exposed mice decreased, which suggested that prolonged HFD altered dopaminergic neuroplasticity. All three peroxisome proliferator-activated receptor (PPAR) subtype (PPAR-α, PPAR-β/δ, PPAR-γ) levels were significantly reduced in the SN and the ventral tegmental area of HFD mice when compared to those in controls. This study showed that a prolonged HFD induced neuroinflammation, suppressed PPAR levels, caused degeneration of midbrain dopaminergic neurons, and resulted in symptoms reminiscent of human PD. To our knowledge, this is the first study documenting the effects of an HFD on PPARs in dopaminergic neurons.
Collapse
Affiliation(s)
- Yu-Chia Kao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-C.K.); (W.-Y.W.)
- Department of Pediatrics, E-DA Hospital, Kaohsiung 82445, Taiwan
| | - Wei-Yen Wei
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-C.K.); (W.-Y.W.)
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-C.K.); (W.-Y.W.)
- Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
- Correspondence: (K.-J.T.); (L.-C.W.); Tel.: +886-6-235-3535-4254 (K.-J.T.); +886-6-235-3535-7212 (L.-C.W.)
| | - Liang-Chao Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-C.K.); (W.-Y.W.)
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
- Correspondence: (K.-J.T.); (L.-C.W.); Tel.: +886-6-235-3535-4254 (K.-J.T.); +886-6-235-3535-7212 (L.-C.W.)
| |
Collapse
|
20
|
Peng T, Liu X, Wang J, Liu Y, Fu Z, Ma X, Li J, Sun G, Ji Y, Lu J, Wan W, Lu H. Fluoxetine-mediated inhibition of endoplasmic reticulum stress is involved in the neuroprotective effects of Parkinson's disease. Aging (Albany NY) 2019; 10:4188-4196. [PMID: 30585175 PMCID: PMC6326670 DOI: 10.18632/aging.101716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/06/2018] [Indexed: 12/25/2022]
Abstract
Background: Accumulating evidence suggests that Fluoxetine (FLX), an anti-depressant drug, has broad neurobiological functions and neuroprotective effects in central nervous system injury, but its roles in Parkinson's disease (PD) remain unclear. In this study, we aimed to evaluate whether fluoxetine attenuates rotenone-induced neurodegeneration in PD. Methods: Male Sprague-Dawley rats were randomly allocated to control, rotenone-treated, rotenone + FLX-treated and FLX-treated groups. Behavioral tests including open field behavioral test and catalepsy measurement were taken to evaluate neurological behavioral measurements. Apoptosis was detected by TUNEL assay. Endoplasmic reticulum (ER)-related gene expressions were detected by qRT-PCR and western blot. Immunohistochemistry was performed to assess dopaminergic neuronal degeneration. Results: We demonstrated that pretreatment with FLX (10.0 mg/kg, i.p.) significantly ameliorated the catalepsy symptom and increased locomotor activity. In addition, FLX markedly reversed the loss of dopaminergic neurons and suppressed the X‑box‑binding protein 1 (XBP1)/caspase-3-activated ER stress. Furthermore, FLX inhibited rotenone-mediated neurodegeneration through caspase-3-mediated neuronal apoptosis. Conclusion: Taken together, our findings indicate that FLX has beneficial neuroprotective effects in PD and FLX might be a potential therapeutic agent for the treatment of PD. In light of its favorable properties, FLX should be evaluated in the treatment of PD as well as related neurologic disorders.
Collapse
Affiliation(s)
- Tao Peng
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Xiaoyan Liu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jingtao Wang
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yu Liu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhenqiang Fu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Xingrong Ma
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Junmin Li
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Guifang Sun
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yangfei Ji
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jingjing Lu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Wencui Wan
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hong Lu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
21
|
Beggiato S, Tomasini MC, Ferraro L. Palmitoylethanolamide (PEA) as a Potential Therapeutic Agent in Alzheimer's Disease. Front Pharmacol 2019; 10:821. [PMID: 31396087 PMCID: PMC6667638 DOI: 10.3389/fphar.2019.00821] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/25/2019] [Indexed: 12/20/2022] Open
Abstract
N-Palmitoylethanolamide (PEA) is a non-endocannabinoid lipid mediator belonging to the class of the N-acylethanolamine phospolipids and was firstly isolated from soy lecithin, egg yolk, and peanut meal. Either preclinical or clinical studies indicate that PEA is potentially useful in a wide range of therapeutic areas, including eczema, pain, and neurodegeneration. PEA-containing products are already licensed for use in humans as a nutraceutical, a food supplement, or a food for medical purposes, depending on the country. PEA is especially used in humans for its analgesic and anti-inflammatory properties and has demonstrated high safety and tolerability. Several preclinical in vitro and in vivo studies have proven that PEA can induce its biological effects by acting on several molecular targets in both central and peripheral nervous systems. These multiple mechanisms of action clearly differentiate PEA from classic anti-inflammatory drugs and are attributed to the compound that has quite unique anti(neuro)inflammatory properties. According to this view, preclinical studies indicate that PEA, especially in micronized or ultramicronized forms (i.e., formulations that maximize PEA bioavailability and efficacy), could be a potential therapeutic agent for the effective treatment of different pathologies characterized by neurodegeneration, (neuro)inflammation, and pain. In particular, the potential neuroprotective effects of PEA have been demonstrated in several experimental models of Alzheimer's disease. Interestingly, a single-photon emission computed tomography (SPECT) case study reported that a mild cognitive impairment (MCI) patient, treated for 9 months with ultramicronized-PEA/luteolin, presented an improvement of cognitive performances. In the present review, we summarized the current preclinical and clinical evidence of PEA as a possible therapeutic agent in Alzheimer's disease. The possible PEA neuroprotective mechanism(s) of action is also described.
Collapse
Affiliation(s)
- Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Technopole of Ferrara, LTTA Laboratory for the Technologies for Advanced Therapies, Ferrara, Italy.,IRET Foundation, Bologna, Italy
| | - Maria Cristina Tomasini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Technopole of Ferrara, LTTA Laboratory for the Technologies for Advanced Therapies, Ferrara, Italy
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Technopole of Ferrara, LTTA Laboratory for the Technologies for Advanced Therapies, Ferrara, Italy.,IRET Foundation, Bologna, Italy
| |
Collapse
|
22
|
Lange KW, Nakamura Y, Chen N, Guo J, Kanaya S, Lange KM, Li S. Diet and medical foods in Parkinson’s disease. FOOD SCIENCE AND HUMAN WELLNESS 2019. [DOI: 10.1016/j.fshw.2019.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Davis MP, Behm B, Mehta Z, Fernandez C. The Potential Benefits of Palmitoylethanolamide in Palliation: A Qualitative Systematic Review. Am J Hosp Palliat Care 2019; 36:1134-1154. [PMID: 31113223 DOI: 10.1177/1049909119850807] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Palmitoylethanolamide (PEA) is a nutraceutical endocannabinoid that was retrospectively discovered in egg yolks. Feeding poor children with known streptococcal infections prevented rheumatic fever. Subsequently, it was found to alter the course of influenza. Unfortunately, there is little known about its pharmacokinetics. Palmitoylethanolamide targets nonclassical cannabinoid receptors rather than CB1 and CB2 receptors. Palmitoylethanolamide will only indirectly activate classical cannabinoid receptors by an entourage effect. There are a significant number of prospective and randomized trials demonstrating the pain-relieving effects of PEA. There is lesser evidence of benefit in patients with nonpain symptoms related to depression, Parkinson disease, strokes, and autism. There are no reported drug-drug interactions and very few reported adverse effects from PEA. Further research is needed to define the palliative benefits to PEA.
Collapse
|
24
|
Zhu J, Dou S, Jiang Y, Chen J, Wang C, Cheng B. Apelin-13 protects dopaminergic neurons in MPTP-induced Parkinson's disease model mice through inhibiting endoplasmic reticulum stress and promoting autophagy. Brain Res 2019; 1715:203-212. [PMID: 30914252 DOI: 10.1016/j.brainres.2019.03.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/11/2019] [Accepted: 03/22/2019] [Indexed: 12/12/2022]
Abstract
The dopaminergic neurodegeneration in the substantia nigrapars compacta (SNpc) and striatum of the midbrain is the important pathological feature of Parkinson's disease (PD). It has been shown that autophagy and endoplasmic reticulum stress (ERS) are involved in the occurrence and development of PD. The neuropeptide Apelin-13 is neuroprotective in the neurological diseases such as PD, Alzheimer's disease and cerebral ischemic stroke. In the present work, we investigated the neuroprotective effects of Apelin-13 on ERS and autophagy in the dopaminergic neurodegeneration of SNpc of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP)-treated mice. The intranigral injection of Apelin-13 alleviated the behavioral dysfunction and dopaminergic neurodegeneration induced by MPTP. After the exposure to MPTP, the expression of tyrosine hydroxylase (TH) was significantly decreased as well as the increased α-synuclein expression, which was significantly reversed by the intranigral injection of Apelin-13. Also, Apelin-13 significantly reversed the decreasing autophagy induced by MPTP which was indicated by the up-regulation of LC3B-II and Beclin1 and down-regulation of p62. And MPTP-induced ERS such as IRE1α, XBP1s, CHOP and GRP78 was significantly inhibited by Apelin-13. Taken together, Apelin-13 protects dopaminergic neurons in MPTP-induced PD model mice in vivo through inhibiting ERS and promoting autophagy, which contributes to the therapy for PD in the future.
Collapse
Affiliation(s)
- Junge Zhu
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Shanshan Dou
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Yunlu Jiang
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Jing Chen
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, 272067 Jining, China.
| | - Baohua Cheng
- Neurobiology Institute, Jining Medical University, 272067 Jining, China.
| |
Collapse
|
25
|
Ferguson LB, Zhang L, Wang S, Bridges C, Harris RA, Ponomarev I. Peroxisome Proliferator Activated Receptor Agonists Modulate Transposable Element Expression in Brain and Liver. Front Mol Neurosci 2018; 11:331. [PMID: 30283300 PMCID: PMC6156381 DOI: 10.3389/fnmol.2018.00331] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator activated receptors (PPARs) are nuclear hormone receptors that act as transcription factors in response to endogenous lipid messengers. The fibrates and thiazolidinediones are synthetic PPAR agonists used clinically to treat dyslipidemia and Type 2 Diabetes Mellitus, respectively, but also improve symptoms of several other diseases. Transposable elements (TEs), repetitive sequences in mammalian genomes, are implicated in many of the same conditions for which PPAR agonists are therapeutic, including neurodegeneration, schizophrenia, and drug addiction. We tested the hypothesis that there is a link between actions of PPAR agonists and TE expression. We developed an innovative application of microarray data by mapping Illumina mouse WG-6 microarray probes to areas of the mouse genome that contain TEs. Using this information, we assessed the effects of systemic administration of three PPAR agonists with different PPAR subtype selectivity: fenofibrate, tesaglitazar, and bezafibrate, on TE probe expression in mouse brain [prefrontal cortex (PFC) and amygdala] and liver. We found that fenofibrate, and bezafibrate to a lesser extent, up-regulated probes mapped to retrotransposons: Short-Interspersed Elements (SINEs) and Long-Interspersed Elements (LINEs), in the PFC. Conversely, all PPAR agonists down-regulated LINEs and tesaglitazar and bezafibrate also down-regulated SINEs in liver. We built gene coexpression networks that partitioned the diverse transcriptional response to PPAR agonists into groups of probes with highly correlated expression patterns (modules). Most of the differentially expressed retrotransposons were within the same module, suggesting coordinated regulation of their expression, possibly by PPAR signaling. One TE module was conserved across tissues and was enriched with genes whose products participate in epigenetic regulation, suggesting that PPAR agonists affect TE expression via epigenetic mechanisms. Other enriched functional categories included phenotypes related to embryonic development and learning and memory, suggesting functional links between these biological processes and TE expression. In summary, these findings suggest mechanistic relationships between retrotransposons and PPAR agonists and provide a basis for future exploration of their functional roles in brain and liver.
Collapse
Affiliation(s)
- Laura B. Ferguson
- Waggoner Center for Alcohol & Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Lingling Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Shi Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Courtney Bridges
- Waggoner Center for Alcohol & Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - R. Adron Harris
- Waggoner Center for Alcohol & Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Igor Ponomarev
- Waggoner Center for Alcohol & Addiction Research, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
26
|
Gallelli CA, Calcagnini S, Romano A, Koczwara JB, de Ceglia M, Dante D, Villani R, Giudetti AM, Cassano T, Gaetani S. Modulation of the Oxidative Stress and Lipid Peroxidation by Endocannabinoids and Their Lipid Analogues. Antioxidants (Basel) 2018; 7:E93. [PMID: 30021985 PMCID: PMC6070960 DOI: 10.3390/antiox7070093] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/10/2018] [Accepted: 07/13/2018] [Indexed: 02/06/2023] Open
Abstract
Growing evidence supports the pivotal role played by oxidative stress in tissue injury development, thus resulting in several pathologies including cardiovascular, renal, neuropsychiatric, and neurodegenerative disorders, all characterized by an altered oxidative status. Reactive oxygen and nitrogen species and lipid peroxidation-derived reactive aldehydes including acrolein, malondialdehyde, and 4-hydroxy-2-nonenal, among others, are the main responsible for cellular and tissue damages occurring in redox-dependent processes. In this scenario, a link between the endocannabinoid system (ECS) and redox homeostasis impairment appears to be crucial. Anandamide and 2-arachidonoylglycerol, the best characterized endocannabinoids, are able to modulate the activity of several antioxidant enzymes through targeting the cannabinoid receptors type 1 and 2 as well as additional receptors such as the transient receptor potential vanilloid 1, the peroxisome proliferator-activated receptor alpha, and the orphan G protein-coupled receptors 18 and 55. Moreover, the endocannabinoids lipid analogues N-acylethanolamines showed to protect cell damage and death from reactive aldehydes-induced oxidative stress by restoring the intracellular oxidants-antioxidants balance. In this review, we will provide a better understanding of the main mechanisms triggered by the cross-talk between the oxidative stress and the ECS, focusing also on the enzymatic and non-enzymatic antioxidants as scavengers of reactive aldehydes and their toxic bioactive adducts.
Collapse
Affiliation(s)
- Cristina Anna Gallelli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Silvio Calcagnini
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Justyna Barbara Koczwara
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Marialuisa de Ceglia
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Donatella Dante
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Rosanna Villani
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Department of Medical and Surgical Sciences, Institute of Internal Medicine, University of Foggia, 71122 Foggia, Italy.
| | - Anna Maria Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, Via Luigi Pinto, c/o Ospedali Riuniti, 71122 Foggia, Italy.
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
27
|
Neuroprotective effect of IDPU (1-(7-imino-3-propyl-2,3-dihydrothiazolo [4,5-d]pyrimidin-6(7H)-yl)urea) in 6-OHDA induced rodent model of hemiparkinson’s disease. Neurosci Lett 2018; 675:74-82. [DOI: 10.1016/j.neulet.2018.03.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 03/02/2018] [Accepted: 03/18/2018] [Indexed: 01/21/2023]
|
28
|
2-Pentadecyl-2-Oxazoline Reduces Neuroinflammatory Environment in the MPTP Model of Parkinson Disease. Mol Neurobiol 2018; 55:9251-9266. [PMID: 29656363 DOI: 10.1007/s12035-018-1064-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
Current pharmacological management of Parkinson disease (PD) does not provide for disease modification, but addresses only symptomatic features. Here, we explore a new approach to neuroprotection based on the use of 2-pentadecyl-2-oxazoline (PEA-OXA), the oxazoline derivative of the fatty acid amide signaling molecule palmitoylethanolamide (PEA), in an experimental model of PD. Daily oral treatment with PEA-OXA (10 mg/kg) significantly reduced behavioral impairments and neuronal cell degeneration of the dopaminergic tract induced by four intraperitoneal injections of the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on 8-week-old male C57 mice. Moreover, PEA-OXA treatment prevented dopamine depletion, increased tyrosine hydroxylase and dopamine transporter activities, and decreased α-synuclein aggregation in neurons. PEA-OXA treatment also diminished nuclear factor-κB traslocation, cyclooxygenase-2, and inducible nitric oxide synthase expression and through upregulation of the nuclear factor E2-related factor 2 pathway, induced activation of Mn-superoxide dismutase and heme oxygenase-1. Further, PEA-OXA modulated microglia and astrocyte activation and preserved microtubule-associated protein-2 alterations. In conclusion, pharmacological activation of nuclear factor E2-related factor 2 pathways with PEA-OXA may be effective in the future therapy of PD.
Collapse
|
29
|
Puglia C, Blasi P, Ostacolo C, Sommella E, Bucolo C, Platania CBM, Romano GL, Geraci F, Drago F, Santonocito D, Albertini B, Campiglia P, Puglisi G, Pignatello R. Innovative Nanoparticles Enhance N-Palmitoylethanolamide Intraocular Delivery. Front Pharmacol 2018; 9:285. [PMID: 29643808 PMCID: PMC5882782 DOI: 10.3389/fphar.2018.00285] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/13/2018] [Indexed: 12/26/2022] Open
Abstract
Nanostructured lipid carriers (NLCs) loaded with palmitoylethanolamide (PEA) were formulated with the aim to enhance ocular bioavailability of PEA, particularly to the back of the eye. Technological characterization (e.g., size, charge) of NLC loaded with PEA formulation (PEA-NLC) was performed, and NLC morphology was characterized by electron microscopy. Ocular pharmacokinetic study, after topical administration of the formulation, was carried out in rabbit eye. Ultra-high performance liquid chromatography tandem mass spectrometry analysis was carried out to detect PEA levels in ocular tissues. Finally, the ocular tolerability of PEA-NLC formulation was assessed in rabbit eye. The novel formulation significantly increased PEA levels in ocular tissues compared to PEA suspension. Vitreous and retinal levels of PEA were significantly higher in the group treated with PEA-NLC formulation versus PEA suspension (PEA-NLC Cmax 5919 ± 541 pmol/g and 315 ± 70 pmol/g in vitreous and retina, respectively). The PEA-NLC formulation was characterized by high stability and robust ocular bioavailability. Therefore, this innovative formulation may be useful in clinical practice to manage retinal diseases.
Collapse
Affiliation(s)
- Carmelo Puglia
- Department of Drug Sciences, University of Catania, Catania, Italy.,NANO-i - Research Centre on Ocular Nanotechnology, University of Catania, Catania, Italy
| | - Paolo Blasi
- School of Pharmacy, University of Camerino, Camerino, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Center for Research in Ocular Pharmacology, University of Catania, Catania, Italy
| | - Chiara B M Platania
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giovanni L Romano
- Bascom Palmer Eye Institute, University of Miami Health System, Miami, FL, United States
| | - Federica Geraci
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Center for Research in Ocular Pharmacology, University of Catania, Catania, Italy
| | | | - Barbara Albertini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Giovanni Puglisi
- Department of Drug Sciences, University of Catania, Catania, Italy.,NANO-i - Research Centre on Ocular Nanotechnology, University of Catania, Catania, Italy
| | - Rosario Pignatello
- Department of Drug Sciences, University of Catania, Catania, Italy.,NANO-i - Research Centre on Ocular Nanotechnology, University of Catania, Catania, Italy
| |
Collapse
|
30
|
Zeng XS, Geng WS, Jia JJ. Neurotoxin-Induced Animal Models of Parkinson Disease: Pathogenic Mechanism and Assessment. ASN Neuro 2018; 10:1759091418777438. [PMID: 29809058 PMCID: PMC5977437 DOI: 10.1177/1759091418777438] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 04/12/2018] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) is the second most common neurodegenerative movement disorder. Pharmacological animal models are invaluable tools to study the pathological mechanisms of PD. Currently, invertebrate and vertebrate animal models have been developed by using several main neurotoxins, such as 6-hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, paraquat, and rotenone. These models achieve to some extent to reproduce the key features of PD, including motor defects, progressive loss of dopaminergic neurons in substantia nigra pars compacta, and the formation of Lewy bodies. In this review, we will highlight the pathogenic mechanisms of those neurotoxins and summarize different neurotoxic animal models with the hope to help researchers choose among them accurately and to promote the development of modeling PD.
Collapse
Affiliation(s)
- Xian-Si Zeng
- College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| | - Wen-Shuo Geng
- College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| | - Jin-Jing Jia
- College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| |
Collapse
|
31
|
Goes AT, Jesse CR, Antunes MS, Lobo Ladd FV, Lobo Ladd AA, Luchese C, Paroul N, Boeira SP. Protective role of chrysin on 6-hydroxydopamine-induced neurodegeneration a mouse model of Parkinson's disease: Involvement of neuroinflammation and neurotrophins. Chem Biol Interact 2018; 279:111-120. [DOI: 10.1016/j.cbi.2017.10.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/15/2017] [Accepted: 10/16/2017] [Indexed: 01/28/2023]
|
32
|
Fu Q, Song R, Yang Z, Shan Q, Chen W. 6-Hydroxydopamine induces brain vascular endothelial inflammation. IUBMB Life 2017; 69:887-895. [PMID: 29048735 DOI: 10.1002/iub.1685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/15/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Qizhi Fu
- Department of Neurology; The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology; Luoyang Henan China
| | - Runluo Song
- Department of Neurology; The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology; Luoyang Henan China
| | - Zhongxi Yang
- Department of Neurosurgery; The First Hospital of Jilin University; Changchun Jilin China
| | - Qi Shan
- Department of Neurology; The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology; Luoyang Henan China
| | - Wenna Chen
- Department of Neurology; The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology; Luoyang Henan China
| |
Collapse
|
33
|
Herrera A, Muñoz P, Steinbusch HWM, Segura-Aguilar J. Are Dopamine Oxidation Metabolites Involved in the Loss of Dopaminergic Neurons in the Nigrostriatal System in Parkinson's Disease? ACS Chem Neurosci 2017; 8:702-711. [PMID: 28233992 DOI: 10.1021/acschemneuro.7b00034] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In 1967, L-dopa was introduced as part of the pharmacological therapy of Parkinson's disease (PD) and, in spite of extensive research, no additional effective drugs have been discovered to treat PD. This brings forward the question: why have no new drugs been developed? We consider that one of the problems preventing the discovery of new drugs is that we still have no information on the pathophysiology of the neurodegeneration of the neuromelanin-containing nigrostriatal dopaminergic neurons. Currently, it is widely accepted that the degeneration of dopaminergic neurons, i.e., in the substantia nigra pars compacta, involves mitochondrial dysfunction, the formation of neurotoxic oligomers of alpha-synuclein, the dysfunction of protein degradation systems, neuroinflammation, and oxidative and endoplasmic reticulum stress. However, the initial trigger of these mechanisms in the nigrostriatal system is still unknown. It has been reported that aminochrome induces the majority of these mechanisms involved in the neurodegeneration process. Aminochrome is formed within the cytoplasm of neuromelanin-containing dopaminergic neurons during the oxidation of dopamine to neuromelanin. The oxidation of dopamine to neuromelanin is a normal and harmless process, because healthy individuals have intact neuromelanin-containing dopaminergic neurons. Interestingly, aminochrome-induced neurotoxicity is prevented by two enzymes: DT-diaphorase and glutathione transferase M2-2, which explains why melanin-containing dopaminergic neurons are intact in healthy human brains.
Collapse
Affiliation(s)
- Andrea Herrera
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
- Department of Neuroscience, Faculty of
Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Patricia Muñoz
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Harry W. M. Steinbusch
- Department of Neuroscience, Faculty of
Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Juan Segura-Aguilar
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
34
|
New preclinical model are required to discover neuroprotective compound in Parkinson's disease. Pharmacol Res 2016; 119:490. [PMID: 27894922 DOI: 10.1016/j.phrs.2016.11.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/09/2016] [Accepted: 11/21/2016] [Indexed: 11/22/2022]
|
35
|
Response to comment by Juan Segura-Aguilar: New preclinical model are required to discover neuroprotective compound in Parkinson's disease. Pharmacol Res 2016; 119:491-492. [PMID: 27890814 DOI: 10.1016/j.phrs.2016.11.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 11/21/2016] [Indexed: 11/20/2022]
|