1
|
Zhu Y, Chen S, Lurong Q, Qi Z. Recent Advances in Antivirals for Japanese Encephalitis Virus. Viruses 2023; 15:v15051033. [PMID: 37243122 DOI: 10.3390/v15051033] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Culex mosquitoes are the primary vectors of the Japanese encephalitis virus (JEV). Since its discovery in 1935, Japanese encephalitis (JE), caused by JEV, has posed a significant threat to human health. Despite the widespread implementation of several JEV vaccines, the transmission chain of JEV in the natural ecosystem has not changed, and the vector of transmission cannot be eradicated. Therefore, JEV is still the focus of attention for flaviviruses. At present, there is no clinically specific drug for JE treatment. JEV infection is a complex interaction between the virus and the host cell, which is the focus of drug design and development. An overview of antivirals that target JEV elements and host factors is presented in this review. In addition, drugs that balance antiviral effects and host protection by regulating innate immunity, inflammation, apoptosis, or necrosis are reviewed to treat JE effectively.
Collapse
Affiliation(s)
- Yongzhe Zhu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Shenglin Chen
- Department of Clinic Laboratory Diagnostics, General Hospital of Tibet Military Area Command of PLA, Lhasa 850007, China
| | - Qilin Lurong
- Department of Geriatrics, General Hospital of Tibet Military Area Command of PLA, Lhasa 850007, China
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
2
|
Shou JW, Li XX, Tang YS, Lim-Ho Kong B, Wu HY, Xiao MJ, Cheung CK, Shaw PC. Novel mechanistic insight on the neuroprotective effect of berberine: The role of PPARδ for antioxidant action. Free Radic Biol Med 2022; 181:62-71. [PMID: 35093536 DOI: 10.1016/j.freeradbiomed.2022.01.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/07/2022] [Accepted: 01/23/2022] [Indexed: 02/06/2023]
Abstract
Cerebral ischemic stroke ranks the second leading cause of death and the third leading cause of disability in lifetime all around the world, urgently necessitating effective therapeutic interventions. Reactive oxygen species (ROS) have been implicated in stroke pathogenesis and peroxisome proliferator-activated receptors (PPARs) are prominent targets for ROS management. Although recent research has shown antioxidant effect of berberine (BBR), little is known regarding its effect upon ROS-PPARs signaling in stroke. The aim of this study is to explore whether BBR could target on ROS-PPARs pathway to ameliorate middle cerebral artery occlusion (MCAO)-induced stroke. Herein, we report that BBR is able to scavenge ROS in oxidation-damaged C17.2 neural stem cells and stroked mice. PPARδ, rather than PPARα or PPARγ, is involved in the anti-ROS effect of BBR, as evidenced by the siRNA transfection and specific antagonist treatment data. Further, we have found BBR could upregulate NF-E2 related factor-1/2 (NRF1/2) and NAD(P)H:quinone oxidoreductase 1 (NQO1) following a PPARδ-dependent manner. Mechanistic study has revealed that BBR acts as a potent ligand (Kd = 290 ± 92 nM) to activate PPARδ and initiates the transcriptional regulation functions, thus promoting the expression of PPARδ, NRF1, NRF2 and NQO1. Collectively, our results indicate that BBR confers neuroprotective effects by activating PPARδ to scavenge ROS, providing a novel mechanistic insight for the antioxidant action of BBR.
Collapse
Affiliation(s)
- Jia-Wen Shou
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao-Xiao Li
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yun-Sang Tang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Bobby Lim-Ho Kong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hoi-Yan Wu
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Meng-Jie Xiao
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chun-Kai Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Pang-Chui Shaw
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China; Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants and Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Akhtar MJ, Ahamed M, Alhadlaq H. Anti-Inflammatory CeO 2 Nanoparticles Prevented Cytotoxicity Due to Exogenous Nitric Oxide Donors via Induction Rather Than Inhibition of Superoxide/Nitric Oxide in HUVE Cells. Molecules 2021; 26:5416. [PMID: 34500851 PMCID: PMC8434366 DOI: 10.3390/molecules26175416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 11/16/2022] Open
Abstract
The mechanism behind the cytoprotective potential of cerium oxide nanoparticles (CeO2 NPs) against cytotoxic nitric oxide (NO) donors and H2O2 is still not clear. Synthesized and characterized CeO2 NPs significantly ameliorated the lipopolysaccharide (LPS)-induced cytokines IL-1β and TNF-α. The main goal of this study was to determine the capacities of NPs regarding signaling effects that could have occurred due to reactive oxygen species (ROS) and/or NO, since NP-induced ROS/NO did not lead to toxicity in HUVE cells. Concentrations that induced 50% cell death (i.e., IC50s) of two NO donors (DETA-NO; 1250 ± 110 µM and sodium nitroprusside (SNP); 950 ± 89 µM) along with the IC50 of H2O2 (120 ± 7 µM) were utilized to evaluate cytoprotective potential and its underlying mechanism. We determined total ROS (as a collective marker of hydrogen peroxide, superoxide radical (O2•-), hydroxyl radical, etc.) by DCFH-DA and used a O2•- specific probe DHE to decipher prominent ROS. The findings revealed that signaling effects mediated mainly by O2•- and/or NO are responsible for the amelioration of toxicity by CeO2 NPs at 100 µg/mL. The unaltered effect on mitochondrial membrane potential (MMP) due to NP exposure and, again, CeO2 NPs-mediated recovery in the loss of MMP due to exogenous NO donors and H2O2 suggested that NP-mediated O2•- production might be extra-mitochondrial. Data on activated glutathione reductase (GR) and unaffected glutathione peroxidase (GPx) activities partially explain the mechanism behind the NP-induced gain in GSH and persistent cytoplasmic ROS. The promoted antioxidant capacity due to non-cytotoxic ROS and/or NO production, rather than inhibition, by CeO2 NP treatment may allow cells to develop the capacity to tolerate exogenously induced toxicity.
Collapse
Affiliation(s)
- Mohd Javed Akhtar
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Maqusood Ahamed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Hisham Alhadlaq
- Department of Physics and Astronomy, College of Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
| |
Collapse
|
4
|
CAILLAUD M, PATEL NH, WHITE A, WOOD M, Contreras KM, TOMA W, Alkhlaif Y, ROBERTS JL, Tran TH, JACKSON AB, POKLIS J, GEWIRTZ DA, DAMAJ MI. Targeting Peroxisome Proliferator-Activated Receptor-α (PPAR- α) to reduce paclitaxel-induced peripheral neuropathy. Brain Behav Immun 2021; 93:172-185. [PMID: 33434562 PMCID: PMC8226373 DOI: 10.1016/j.bbi.2021.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND AND PURPOSE Paclitaxel, a widely used anti-cancer drug, is frequently associated with prolonged and severe peripheral neuropathies (PIPN), associated with neuroinflammation. Currently, PIPN effective treatments are lacking. Peroxisome Proliferator-Activated Receptor-α (PPAR-⍺) can modulate inflammatory responses. Thus, the use of PPAR-⍺ agonists, such as fibrates (fenofibrate and choline-fenofibrate), currently used in dyslipidemia treatment, could represent an interesting therapeutic approach in PIPN. EXPERIMENTAL APPROACH Our studies tested the efficacy of fenofibrate (150 mg/kg, daily, i.p.) and choline fenofibrate (60 mg/kg daily, p.o.) in reversing and preventing the development of PIPN (paclitaxel: 8 mg/kg, i.p., every other day for 4 days) in male and female C57BL/6J mice. Mechanical and cold hypersensitivity, conditioned place preference, sensory nerve action potential (SNAP), as well as the expression of PPAR-⍺, TNF-⍺, IL-1β and IL-6 mRNA were evaluated. KEY RESULTS While fenofibrate treatment partially reversed and prevented the development of mechanical hypersensitivity, this was completely reversed and prevented by choline-fenofibrate. Both fibrates were able to completely reverse and prevent cold hypersensitivity induced by paclitaxel. The reduction of SNAP amplitude induced by paclitaxel was also reversed by both fenofibrate and choline-fenofibrate. Our results indicate that suppression of paclitaxel-induced hypersensitivity by fibrates involves the regulation of PPAR-⍺ expression and decrease neuroinflammation in DRG. Finally, the co-treatment of Paclitaxel and fenofibric acid (fibrates active metabolite) was tested on different cancer cell lines, no decrease in the antitumoral effect of paclitaxel was observed. CONCLUSIONS AND IMPLICATIONS Taken together, our results show for the first time the therapeutic potential (prevention and reversal) of fibrates in PIPN and opens to a potential pharmacological repurposing of these drugs.
Collapse
Affiliation(s)
- Martial CAILLAUD
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA.,Corresponding authors:,
| | - Nipa H. PATEL
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Alyssa WHITE
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Mackinsey WOOD
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Katherine M. Contreras
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Wisam TOMA
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Yasmin Alkhlaif
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Jane L. ROBERTS
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Tammy H. Tran
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Asti B. JACKSON
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Justin POKLIS
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - David A. GEWIRTZ
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - M. Imad DAMAJ
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA.,Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA.,Corresponding authors:,
| |
Collapse
|
5
|
Shehata AHF, Ahmed ASF, Abdelrehim AB, Heeba GH. The impact of single and combined PPAR-α and PPAR-γ activation on the neurological outcomes following cerebral ischemia reperfusion. Life Sci 2020; 252:117679. [PMID: 32325134 DOI: 10.1016/j.lfs.2020.117679] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/20/2022]
Abstract
AIM The neuronal damage and accompanied functional deficits induced by cerebral ischemia are among the most common causes of disabilities in adults. Activation of subtypes of peroxisome proliferator-activated receptors (PPARs); PPAR-α and PPAR-γ have shown neuroprotective effects in different neurodegenerative diseases including stroke. Thus, this study aimed to compare the effects of two different agonists: PPAR-α (fenofibrate) and PPAR-γ (pioglitazone) as well as the effect of their combination in ameliorating post-ischemia behavioral deficits. METHODS Male Wistar rats were either pretreated with vehicle, fenofibrate (100 mg/kg/day p.o), pioglitazone (10 mg/kg/day p.o) or their combination for 14 days prior to bilateral common carotid artery occlusion followed by reperfusion for 24 hoursh. The sensory motor functions of rats were assessed, then rats were sacrificed to determine infarct volume and histopathological changes as well as oxidative stress, inflammatory and apoptotic markers in the brain tissue. KEY FINDINGS Pre-treatment with fenofibrate and pioglitazone in addition to their combination improved neurobehavioral dysfunction, reduced cerebral infarct volume, attenuated inflammatory and apoptotic markers and ameliorated histopathological changes in I/R injured rats. The effect of pioglitazone in cerebral cortex was higher than its corresponding effect in fenofibrate while the combined administration of both drugs had additive neuroprotective effect and normalized inflammatory and apoptotic mediators in ischemic rats. SIGNIFICANCE The study compared the neuroprotective effects of PPAR-α and PPAR-γ agonists, and tested the impact of their combination. We concluded that no additional benefits on the functional outcomes might be gained upon their combination.
Collapse
Affiliation(s)
- Alaa H F Shehata
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Egypt
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Egypt.
| | - Amany B Abdelrehim
- Department of Biochemistry and Toxicology, Faculty of Pharmacy, Minia University, Egypt
| | - Gehan H Heeba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Egypt
| |
Collapse
|
6
|
Oyagbemi AA, Adebiyi OE, Adigun KO, Ogunpolu BS, Falayi OO, Hassan FO, Folarin OR, Adebayo AK, Adejumobi OA, Asenuga ER, Ola-Davies OE, Omobowale TO, Olopade JO, Saba AB, Adedapo AA, Nkadimeng SM, McGaw LJ, Oguntibeju OO, Yakubu MA. Clofibrate, a PPAR-α agonist, abrogates sodium fluoride-induced neuroinflammation, oxidative stress, and motor incoordination via modulation of GFAP/Iba-1/anti-calbindin signaling pathways. ENVIRONMENTAL TOXICOLOGY 2020; 35:242-253. [PMID: 31710167 DOI: 10.1002/tox.22861] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/20/2019] [Accepted: 10/10/2019] [Indexed: 06/10/2023]
Abstract
Fluoride is an environmental contaminant that is ubiquitously present in air, water, and soil. It is commonly added in minute quantity to drinking water, toothpaste, and mouth rinses to prevent tooth decay. Epidemiological findings have demonstrated that exposure to fluoride induced neurodevelopmental toxicity, developmental neurotoxicity, and motor disorders. The neuroprotective effect of clofibrate, a peroxisome proliferator-activated receptor alpha agonist, was investigated in the present study. Forty male Wistar rats were used for this study and randomly grouped into 10 rats per group as control, sodium fluoride (NaF) alone (300 ppm), NaF plus clofibrate (250 mg/kg), and NaF plus lisinopril (10 mg/kg), respectively, for 7 days. NaF was administered in drinking water while clofibrate and lisinopril were administered by oral gavage. Markers of neuronal inflammation and oxidative stress, acetylcholinesterase activity, and neurobehavioral (hanging wire and open field) tests were performed. Immunohistochemistry was performed on brain tissues, and they were probed with glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, and cerebellar Ca2+ -binding protein calbindin-D28k. The results showed that NaF significantly increased of oxidative stress and neuroinflammation and inhibited AChE activity. Immunostaining showed reactive astrocytes, microgliosis, loss of dendritic spines, and arborization in Purkinje cells in rats administered only NaF. Neurobehavioral results showed that cotreatment of NaF with clofibrate improved muscular strength and locomotion, reduced anxiety, and significantly reduced astrocytic count. Overall, cotreatment of NaF with either clofibrate or lisinopril showed neuroprotective effects by mitigating neuronal inflammation and oxidative and motor incoordination. Hence, clofibrate could be seen as a novel drug candidate against neurodegeneration and motor disorders.
Collapse
Affiliation(s)
- Ademola A Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olamide E Adebiyi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Kabirat O Adigun
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Blessing S Ogunpolu
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olufunke O Falayi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Fasilat O Hassan
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Oluwabusayo R Folarin
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Benin, Nigeria
- Department of Medical Laboratory Science, College of Health Sciences, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Adedeji K Adebayo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olumuyiwa A Adejumobi
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Ebunoluwa R Asenuga
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Benin, Nigeria
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olufunke E Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Temidayo O Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - James O Olopade
- Department of Medical Laboratory Science, College of Health Sciences, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Adebowale B Saba
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Adeolu A Adedapo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Sanah M Nkadimeng
- Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Faculty of Veterinary Science, Onderstepoort, South Africa
| | - Lyndy J McGaw
- Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Faculty of Veterinary Science, Onderstepoort, South Africa
| | - Oluwafemi O Oguntibeju
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Momoh A Yakubu
- Department of Environmental & Interdisciplinary Sciences, College of Science, Engineering & Technology, Texas Southern University, Houston, Texas
| |
Collapse
|
7
|
Fransen M, Lismont C. Redox Signaling from and to Peroxisomes: Progress, Challenges, and Prospects. Antioxid Redox Signal 2019; 30:95-112. [PMID: 29433327 DOI: 10.1089/ars.2018.7515] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Peroxisomes are organelles that are best known for their role in cellular lipid and hydrogen peroxide (H2O2) metabolism. Emerging evidence suggests that these organelles serve as guardians and modulators of cellular redox balance, and that alterations in their redox metabolism may contribute to aging and the development of chronic diseases such as neurodegeneration, diabetes, and cancer. Recent Advances: H2O2 is an important signaling messenger that controls many cellular processes by modulating protein activity through cysteine oxidation. Somewhat surprisingly, the potential involvement of peroxisomes in H2O2-mediated signaling processes has been overlooked for a long time. However, recent advances in the development of live-cell approaches to monitor and modulate spatiotemporal fluxes in redox species at the subcellular level have opened up new avenues for research in redox biology and boosted interest in the concept of peroxisomes as redox signaling platforms. CRITICAL ISSUES This review first introduces the reader to what is known about the role of peroxisomes in cellular H2O2 production and clearance, with a focus on mammalian cells. Next, it briefly describes the benefits and drawbacks of current strategies used to investigate the complex interplay between peroxisome metabolism and cellular redox state. Furthermore, it integrates and critically evaluates literature dealing with the interrelationship between peroxisomal redox metabolism, cell signaling, and human disease. FUTURE DIRECTIONS As the precise molecular mechanisms underlying many of these associations are still poorly understood, a key focus for future research should be the identification of primary targets for peroxisome-derived H2O2.
Collapse
Affiliation(s)
- Marc Fransen
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| | - Celien Lismont
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| |
Collapse
|
8
|
Low Phytanic Acid-Concentrated DHA Prevents Cognitive Deficit and Regulates Alzheimer Disease Mediators in an ApoE -/- Mice Experimental Model. Nutrients 2018; 11:nu11010011. [PMID: 30577526 PMCID: PMC6356727 DOI: 10.3390/nu11010011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is the main cause of dementia and cognitive impairment. It has been associated with a significant diminution of omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) levels in the brain. Clinical trials with DHA as a treatment in neurological diseases have shown inconsistent results. Previously, we reported that the presence of phytanic acid (PhA) in standard DHA compositions could be blunting DHA’s beneficial effects. Therefore, we aimed to analyze the effects of a low PhA-concentrated DHA and a standard PhA-concentrated DHA in Apolipoprotein E knockout (ApoE−/−) mice. Behavioral tests and protein expression of pro-inflammatory, pro-oxidant, antioxidant factors, and AD-related mediators were evaluated. Low PhA-concentrated DHA decreased Aβ, ß-amyloid precursor protein (APP), p-tau, Ca2+/calmodulin-dependent protein kinase II (CAMKII), caspase 3, and catalase, and increased brain derived neurotrophic factor (BDNF) when compared to standard PhA-concentrated DHA. Low PhA-concentrated DHA decreased interleukin (IL)-6 and tumor necrosis factor alpha (TNF-α) protein expression in ApoE−/− mice when compared to standard PhA-concentrated DHA. No significant differences were found in p22phox, inducible nitric oxide synthase (iNOS), glutathione peroxidase (GPx), superoxide dismutase 1 (SOD-1), and tau protein expression. The positive actions of a low PhA-concentrated DHA were functionally reflected by improving the cognitive deficit in the AD experimental model. Therefore, reduction of PhA content in DHA compositions could highlight a novel pathway for the neurodegeneration processes related to AD.
Collapse
|
9
|
Oleoylethanolamide treatment reduces neurobehavioral deficits and brain pathology in a mouse model of Gulf War Illness. Sci Rep 2018; 8:12921. [PMID: 30150699 PMCID: PMC6110778 DOI: 10.1038/s41598-018-31242-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 08/15/2018] [Indexed: 12/12/2022] Open
Abstract
There are nearly 250,000 Gulf War (GW) veterans who suffer from Gulf War Illness (GWI), a multi-symptom condition that remains untreatable. The main objective was to determine if targeting peroxisomal function could be of therapeutic value in GWI. We performed a pilot study that showed accumulation of very long chain fatty acids (VLCFA), which are metabolized in peroxisomes, in plasma from veterans with GWI. We then examined if targeting peroxisomal β-oxidation with oleoylethanolamide (OEA) restores these lipids to the normal levels and mitigates neuroinflammation and neurobehavioral deficits in a well-established mouse model of GWI. In GWI mice, treatment with OEA corresponded with cognitive benefits and reduced fatigue and disinhibition-like behavior in GWI mice. Biochemical and molecular analysis of the brain tissue showed reduced astroglia and microglia staining, decreased levels of chemokines and cytokines, and decreased NFκB phosphorylation. Treatment with OEA reduced accumulation of peroxisome specific VLCFA in the brains of GWI mice. These studies further support the translational value of targeting peroxisomes. We expect that OEA may be a potential therapy for treating neurobehavioral symptoms and the underlying lipid dysfunction and neuroinflammation associated with GWI. Oleoylethanolamide is available as a dietary supplement, making it appealing for human translational studies.
Collapse
|
10
|
Lin C, Chen P, Chan H, Huang Y, Chang NW. Peroxisome proliferator‐activated receptor alpha accelerates neuronal differentiation and this might involve the mitogen‐activated protein kinase pathway. Int J Dev Neurosci 2018; 71:46-51. [DOI: 10.1016/j.ijdevneu.2018.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023] Open
Affiliation(s)
- Chingju Lin
- Department of PhysiologyCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Pei‐Yi Chen
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Hsu‐Chin Chan
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Yi‐Ping Huang
- Department of PhysiologyCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Nai Wen Chang
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| |
Collapse
|
11
|
Metzger J, Nolte A, Uhde AK, Hewicker-Trautwein M, Distl O. Whole genome sequencing identifies missense mutation in MTBP in Shar-Pei affected with Autoinflammatory Disease (SPAID). BMC Genomics 2017; 18:348. [PMID: 28472921 PMCID: PMC5418765 DOI: 10.1186/s12864-017-3737-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/27/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Autoinflammatory diseases in dogs are characterized by complex disease processes with varying clinical signs. In Shar-Pei, signs of inflammation including fever and arthritis are known to be related with a breed-specific predisposition for Shar-Pei Autoinflammatory Disease (SPAID). RESULTS Clinical and histopathological examinations of two severely SPAID-affected Shar-Pei revealed signs of inflammation including fever, arthritis, and perivascular and diffuse dermatitis in both dogs. A multifocal accumulation of amyloid in different organs was found in one SPAID-affected case. Whole genome sequencing resulted in 37 variants, which were homozygous mutant private mutations in SPAID-affected Shar-Pei. Nine SNVs with predicted damaging effects and three INDELs were further investigated in 102 Shar-Pei affected with SPAID, 62 unaffected Shar-Pei and 162 controls from 11 different dog breeds. The results showed the missense variant MTBP:g.19383758G > A in MTBP to be highly associated with SPAID in Shar-Pei. In the region of this gene a large ROH (runs of homozygosity) region could be detected exclusively in the two investigated SPAID-affected Shar-Pei compared to control dog breeds. No further SPAID-associated variant with predicted high or moderate effects could be found in genes identified in ROH regions. This MTBP variant was predicted to affect the MDN2-binding protein domain and consequently promote proinflammatory reactions. In the investigated group of Shar-Pei older than six years all dogs with the mutant genotype A/A were SPAID-affected whereas SPAID-unaffected dogs harbored the homozygous wildtype (G/G). Shar-Pei with a heterozygous genotype (G/A) were shown to have a 2.13-fold higher risk for disease development, which gave evidence for an incomplete dominant mode of inheritance. CONCLUSIONS The results of this study give strong evidence for a variant in MTBP related with proinflammatory processes via MTBP-MDM2 pathway. Thus, these results enable a reliable detection of SPAID in Shar-Pei dogs.
Collapse
Affiliation(s)
- Julia Metzger
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17p, 30559 Hannover, Germany
| | - Anna Nolte
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany
| | - Ann-Kathrin Uhde
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany
| | - Marion Hewicker-Trautwein
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany
| | - Ottmar Distl
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17p, 30559 Hannover, Germany
| |
Collapse
|
12
|
WY-14643, a selective agonist of peroxisome proliferator-activated receptor-α, ameliorates lipopolysaccharide-induced depressive-like behaviors by preventing neuroinflammation and oxido-nitrosative stress in mice. Pharmacol Biochem Behav 2017; 153:97-104. [PMID: 28017641 DOI: 10.1016/j.pbb.2016.12.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 11/27/2016] [Accepted: 12/19/2016] [Indexed: 11/22/2022]
|
13
|
Krishnan ML, Wang Z, Silver M, Boardman JP, Ball G, Counsell SJ, Walley AJ, Montana G, Edwards AD. Possible relationship between common genetic variation and white matter development in a pilot study of preterm infants. Brain Behav 2016; 6:e00434. [PMID: 27110435 PMCID: PMC4821839 DOI: 10.1002/brb3.434] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/16/2015] [Accepted: 12/19/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The consequences of preterm birth are a major public health concern with high rates of ensuing multisystem morbidity, and uncertain biological mechanisms. Common genetic variation may mediate vulnerability to the insult of prematurity and provide opportunities to predict and modify risk. OBJECTIVE To gain novel biological and therapeutic insights from the integrated analysis of magnetic resonance imaging and genetic data, informed by prior knowledge. METHODS We apply our previously validated pathway-based statistical method and a novel network-based method to discover sources of common genetic variation associated with imaging features indicative of structural brain damage. RESULTS Lipid pathways were highly ranked by Pathways Sparse Reduced Rank Regression in a model examining the effect of prematurity, and PPAR (peroxisome proliferator-activated receptor) signaling was the highest ranked pathway once degree of prematurity was accounted for. Within the PPAR pathway, five genes were found by Graph Guided Group Lasso to be highly associated with the phenotype: aquaporin 7 (AQP7), malic enzyme 1, NADP(+)-dependent, cytosolic (ME1), perilipin 1 (PLIN1), solute carrier family 27 (fatty acid transporter), member 1 (SLC27A1), and acetyl-CoA acyltransferase 1 (ACAA1). Expression of four of these (ACAA1, AQP7, ME1, and SLC27A1) is controlled by a common transcription factor, early growth response 4 (EGR-4). CONCLUSIONS This suggests an important role for lipid pathways in influencing development of white matter in preterm infants, and in particular a significant role for interindividual genetic variation in PPAR signaling.
Collapse
Affiliation(s)
- Michelle L Krishnan
- Centre for the Developing Brain King's College London St Thomas' Hospital London SE1 7EH UK
| | - Zi Wang
- Department of Biomedical Engineering King's College London St Thomas' Hospital London SE1 7EH UK
| | - Matt Silver
- Department of Population Health London School of Hygiene and Tropical Medicine London WC1E 7HT UK
| | - James P Boardman
- MRC Centre for Reproductive Health University of Edinburgh Edinburgh EH16 4TJ UK
| | - Gareth Ball
- Centre for the Developing Brain King's College London St Thomas' Hospital London SE1 7EH UK
| | - Serena J Counsell
- Centre for the Developing Brain King's College London St Thomas' Hospital London SE1 7EH UK
| | - Andrew J Walley
- School of Public Health Faculty of Medicine Imperial College London Norfolk Place London W2 1PG UK
| | - Giovanni Montana
- Department of Biomedical Engineering King's College London St Thomas' Hospital London SE1 7EH UK
| | - Anthony David Edwards
- Centre for the Developing Brain King's College London St Thomas' Hospital London SE1 7EH UK
| |
Collapse
|
14
|
Yang C, Liu J, Li L, Hu M, Long Y, Liu X, Zhu T, Huang X, Zhao S, Liu S, Rong R. Proteome Analysis of Renoprotection Mediated by a Novel Cyclic Helix B Peptide in Acute Kidney Injury. Sci Rep 2015; 5:18045. [PMID: 26655840 PMCID: PMC4674932 DOI: 10.1038/srep18045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 11/11/2015] [Indexed: 01/18/2023] Open
Abstract
We developed a novel, erythropoietin-derived, non-erythropoiesis, cyclic helix B peptide (CHBP) that displays potent renoprotection against acute kidney injury (AKI). To determine the mechanism of CHBP-mediated protection, we investigated the proteomic profile of mice treated with CHBP in a kidney ischemia-reperfusion (IR) injury model. The isobaric tags for relative and absolute quantitation (iTRAQ)-labeled samples were analyzed using a QSTAR XL LC/MS system. In total, 38 differentially expressed proteins (DEPs) were shared by all experimental groups, while 3 DEPs were detected specifically in the IR + CHBP group. Eight significant pathways were identified, and oxidative phosphorylation was shown to be the most important pathway in CHBP-mediated renoprotection. The significant DEPs in the oxidative phosphorylation pathway elicited by CHBP are NADH-ubiquinone oxidoreductase Fe-S protein 6 (NDUFS6), alpha-aminoadipic semialdehyde synthase (AASS) and ATP-binding cassette sub-family D member 3 (ABCD3). The DEPs mentioned above were verified by RT-qPCR and immunostaining in mouse kidneys. We tested 6 DEPs in human biopsy samples from kidney transplant recipients. The trend of differential expression was consistent with that in the murine model. In conclusion, this study helps to elucidate the pharmacological mechanisms of CHBP before clinical translation.
Collapse
Affiliation(s)
- Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China.,Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Junjun Liu
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Long Li
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Meiyu Hu
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China.,Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yaqiu Long
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Material Medical, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Xiaohui Liu
- Department of Chemistry /Institutes of Biomedical Science, Fudan University, Shanghai, 200433, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Xiao Huang
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Shouliang Zhao
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shangfeng Liu
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China.,Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
15
|
Secretions from placenta, after hypoxia/reoxygenation, can damage developing neurones of brain under experimental conditions. Exp Neurol 2014; 261:386-95. [DOI: 10.1016/j.expneurol.2014.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/25/2014] [Accepted: 05/01/2014] [Indexed: 01/10/2023]
|
16
|
Cheng YH, Lai SW, Chen PY, Chang JH, Chang NW. PPARα activation attenuates amyloid-β-dependent neurodegeneration by modulating Endo G and AIF translocation. Neurotox Res 2014; 27:55-68. [PMID: 25048111 DOI: 10.1007/s12640-014-9485-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 06/19/2014] [Accepted: 07/04/2014] [Indexed: 12/31/2022]
Abstract
The accumulation of a large amount of amyloid-β (Aβ42) in brain neurons is one of the debilitating characteristics of Alzheimer's disease. In this study, we determined the effects of peroxisome proliferator-activated receptor alpha (PPARα) activation on neuronal degeneration using a model of Aβ42-induced cytotoxicity. We found that 0.5 μM Aβ42 induced DNA damage and apoptosis in NT2N cells after 6 h of treatment. Co-treatment of Aβ42-treated cells with Wy14643, a PPARα ligand, significantly increased cell viability after 24 h compared with cells treated with Aβ42 alone. There were no differences in the protein levels of caspase-3, Bcl-2/Bax or p53 between cells treated with Aβ42 alone and those treated with both Aβ42 and Wy14643. However, the addition of Wy14643 significantly suppressed the Aβ42-induced upregulation of Endo G and AIF protein levels. Immunohistochemical analyses further demonstrated that Wy14643 reduced the expression of Endo G and AIF translocated from the cytoplasm into the nucleus, which occurred concomitantly with the decrease in DNA damage in Aβ42-treated cells. Our data clearly show that PPARα activation prevents DNA damage and neuronal cell apoptosis by decreasing the expression and translocation of AIF/Endo G to the nucleus in a caspase-3- and p53-independent pathway in the NT2N cell model. This role of PPARα in promoting neuron survival suggests a possible clinical application in treating Aβ42-associated neurotoxicity in Alzheimer's disease.
Collapse
Affiliation(s)
- Ya-Hsin Cheng
- Department of Physiology, School of Medicine, China Medical University, Taichung, 40402, Taiwan, ROC
| | | | | | | | | |
Collapse
|
17
|
Zhang Y, Wang Z, Chen H, Chen Z, Tian Y. Antioxidants: potential antiviral agents for Japanese encephalitis virus infection. Int J Infect Dis 2014; 24:30-6. [PMID: 24780919 DOI: 10.1016/j.ijid.2014.02.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/10/2014] [Accepted: 02/11/2014] [Indexed: 12/26/2022] Open
Abstract
Japanese encephalitis (JE) is prevalent throughout eastern and southern Asia and the Pacific Rim. It is caused by the JE virus (JEV), which belongs to the family Flaviviridae. Despite the importance of JE, little is known about its pathogenesis. The role of oxidative stress in the pathogenesis of viral infections has led to increased interest in its role in JEV infections. This review focuses mainly on the role of oxidative stress in the pathogenesis of JEV infection and the antiviral effect of antioxidant agents in inhibiting JEV production. First, this review summarizes the pathogenesis of JE. The pathological changes include neuronal death, astrocyte activation, and microglial proliferation. Second, the relationship between oxidative stress and JEV infection is explored. JEV infection induces the generation of oxidants and exhausts the supply of antioxidants, which activates specific signaling pathways. Finally, the therapeutic efficacy of a variety of antioxidants as antiviral agents, including minocycline, arctigenin, fenofibrate, and curcumin, was studied. In conclusion, antioxidants are likely to be developed into antiviral agents for the treatment of JE.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Histology and Embryology, School of Basic Medicine, Third Military Medical University, Chongqing 400038, PR China; Squadron 13 of Cadet Brigade, College of Medical Laboratory Technology, Third Military Medical University, Chongqing, PR China
| | - Zehua Wang
- Department of Histology and Embryology, School of Basic Medicine, Third Military Medical University, Chongqing 400038, PR China; Squadron 17, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Huan Chen
- Department of Histology and Embryology, School of Basic Medicine, Third Military Medical University, Chongqing 400038, PR China; Squadron 13 of Cadet Brigade, College of Medical Laboratory Technology, Third Military Medical University, Chongqing, PR China
| | - Zongtao Chen
- Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China.
| | - Yanping Tian
- Department of Histology and Embryology, School of Basic Medicine, Third Military Medical University, Chongqing 400038, PR China.
| |
Collapse
|
18
|
Revisiting the neuropathogenesis of Zellweger syndrome. Neurochem Int 2014; 69:1-8. [DOI: 10.1016/j.neuint.2014.02.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/11/2014] [Accepted: 02/24/2014] [Indexed: 01/27/2023]
|
19
|
Yakunin E, Kisos H, Kulik W, Grigoletto J, Wanders RJA, Sharon R. The regulation of catalase activity by PPAR γ is affected by α-synuclein. Ann Clin Transl Neurol 2014; 1:145-59. [PMID: 25356396 PMCID: PMC4184544 DOI: 10.1002/acn3.38] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 12/27/2013] [Accepted: 01/07/2014] [Indexed: 01/25/2023] Open
Abstract
Objective While evidence for oxidative injury is frequently detected in brains of humans affected by Parkinson's disease (PD) and in relevant animal models, there is uncertainty regarding its cause. We tested the potential role of catalase in the oxidative injury that characterizes PD. Methods Utilizing brains of A53T α-Syn and ntg mice, and cultured cells, we analyzed catalase activity and expression, and performed biochemical analyses of peroxisomal metabolites. Results Lower catalase expression and lower activity levels were detected in A53T α-Syn brains and α-Syn-expressing cells. The effect on catalase activity was independent of disease progression, represented by mouse age and α-Syn mutation, suggesting a potential physiological function for α-Syn. Notably, catalase activity and expression were unaffected in brains of mice modeling Alzheimer's disease. Moreover, we found that α-Syn expression downregulate the peroxisome proliferator-activated receptor (PPAR)γ, which controls catalase transcription. Importantly, activation of either PPARγ2, PPARα or retinoic X receptor eliminated the inhibiting effect of α-Syn on catalase activity. In addition, activation of these nuclear receptors enhanced the accumulation of soluble α-Syn oligomers, resulting in a positive association between the degree of soluble α-Syn oligomers and catalase activity. Of note, a comprehensive biochemical analysis of specific peroxisomal metabolites indicated no signs of dysfunction in specific peroxisomal activities in brains of A53T α-Syn mice. Interpretation Our results suggest that α-Syn expression may interfere with the complex and overlapping network of nuclear receptors transcription activation. In result, catalase activity is affected through mechanisms involved in the regulation of soluble α-Syn oligomers.
Collapse
Affiliation(s)
- Eugenia Yakunin
- Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University Jerusalem, 91120, Israel
| | - Haya Kisos
- Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University Jerusalem, 91120, Israel
| | - Willem Kulik
- Genetic Metabolic Diseases Lab, Academic Medical Center, University of Amsterdam Amsterdam, 1105 AZ, The Netherlands
| | - Jessica Grigoletto
- Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University Jerusalem, 91120, Israel
| | - Ronald J A Wanders
- Genetic Metabolic Diseases Lab, Academic Medical Center, University of Amsterdam Amsterdam, 1105 AZ, The Netherlands
| | - Ronit Sharon
- Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University Jerusalem, 91120, Israel
| |
Collapse
|
20
|
Aleshin S, Reiser G. Role of the peroxisome proliferator-activated receptors (PPAR)-α, β/δ and γ triad in regulation of reactive oxygen species signaling in brain. Biol Chem 2013; 394:1553-70. [DOI: 10.1515/hsz-2013-0215] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/04/2013] [Indexed: 01/25/2023]
Abstract
Abstract
Overwhelming evidence shows that oxidative stress is a major cause in development of brain disorders. Low activity of the reactive oxygen species (ROS)-degrading system as well as high levels of oxidative damage markers have been observed in brain tissue of patients with neurodegenerative and other brain diseases to a larger extent than in healthy individuals. Many studies aimed to develop effective and safe antioxidant strategies for the therapy or prevention of brain diseases. Nevertheless, it became clear that rigorous suppression of ROS is deleterious for normal cell functioning. Thus, approaches that can regulate the ROS levels over a wide range, from inhibition to induction, will be a powerful tool for neuroprotection. A most prominent target for such ROS management is the family of peroxisome proliferator-activated receptors (PPARs). All three members (PPAR-α, -β/δ and -γ) of this nuclear receptor subfamily form a tightly connected triad. For individual PPAR isoforms, neuroprotective properties have been well proven. Their involvement in regulation of ROS production and degradation underlies the therapeutic effects. Nevertheless, the current paradigms of the involvement of PPAR in neuroprotective therapy ignore such interconnections of PPARs and aim at antioxidant effects of individual PPAR isoforms, but do not take into account the necessity of careful regulation of ROS levels. The present review (i) summarizes the data, which support the concept of the PPAR triad in brain, (ii) demonstrates that usage of the PPAR triad allows the regulation of PPAR-dependent genes over a wide range, from inhibition to upregulation, and (iii) summarizes the known data concerning the PPAR triad involvement in regulation of ROS. Our report opens new directions in the field of PPAR/ROS-related neuroscience research.
Collapse
|
21
|
Ouk T, Potey C, Laprais M, Gautier S, Hanf R, Darteil R, Staels B, Duriez P, Bordet R. PPARα is involved in the multitargeted effects of a pretreatment with atorvastatin in experimental stroke. Fundam Clin Pharmacol 2013; 28:294-302. [PMID: 23701370 DOI: 10.1111/fcp.12035] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 03/25/2013] [Accepted: 04/15/2013] [Indexed: 01/04/2023]
Abstract
There is now substantial data in the literature showing that statins can protect against cerebral ischemia. This neuroprotective potency is related to their pleiotropic effects that modulate various pathways implicated in the pathophysiology of stroke. It has been demonstrated that statins exert anti-inflammatory and vasculoprotective effects, thus contributing to a reduction in infarct size. The underlying mechanisms are still incompletely known. As a cross-talk between statins and the nuclear receptor PPARα has been described, we hypothesized that this cross-talk is necessary to neuroprotection in stroke. We studied the effects of a 14-day preventive atorvastatin treatment (10 mg/kg/day) on C57Bl6 wild-type and PPARα-KO mice submitted to experimental stroke. PPARα was involved in the atorvastatin-induced neuroprotective effect, as confirmed by the measurement of infarct volumes. We also evidenced that the anti-inflammatory action of atorvastatin is mediated, at least partly, by PPARα. The decrease in IL-6 plasmatic levels was PPARα dependent. The cerebral expression of the adhesion molecules ICAM-1 and vascular cell adhesion molecule was reduced by the atorvastatin treatment, and this effect was PPARα dependent in the cortex but not in the striatum of treated animals. Atorvastatin also diminished the cerebral expression of iNOS in the cortex, but had no effect in the striatum of treated mice, whatever the PPARα status. At the vascular level, we found that the atorvastatin-related endothelial nitric oxide synthase upregulation was regulated by PPARα in the aorta, while there was no effect in the brain. We demonstrate here that PPARα is a key mediator of the multitargeted neuroprotective effects of statins in stroke.
Collapse
Affiliation(s)
- Thavarak Ouk
- Department of Medical Pharmacology, EA 1046, University of Lille Nord de France, UDSL, Faculty of Medicine, Lille University Hospital, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Nitric oxide donors as neuroprotective agents after an ischemic stroke-related inflammatory reaction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:297357. [PMID: 23691263 PMCID: PMC3649699 DOI: 10.1155/2013/297357] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 12/17/2022]
Abstract
Cerebral ischemia initiates a cascade of detrimental events including glutamate-associated excitotoxicity, intracellular calcium accumulation, formation of Reactive oxygen species (ROS), membrane lipid degradation, and DNA damage, which lead to the disruption of cellular homeostasis and structural damage of ischemic brain tissue. Cerebral ischemia also triggers acute inflammation, which exacerbates primary brain damage. Therefore, reducing oxidative stress (OS) and downregulating the inflammatory response are options that merit consideration as potential therapeutic targets for ischemic stroke. Consequently, agents capable of modulating both elements will constitute promising therapeutic solutions because clinically effective neuroprotectants have not yet been discovered and no specific therapy for stroke is available to date. Because of their ability to modulate both oxidative stress and the inflammatory response, much attention has been focused on the role of nitric oxide donors (NOD) as neuroprotective agents in the pathophysiology of cerebral ischemia-reperfusion injury. Given their short therapeutic window, NOD appears to be appropriate for use during neurosurgical procedures involving transient arterial occlusions, or in very early treatment of acute ischemic stroke, and also possibly as complementary treatment for neurodegenerative diseases such as Parkinson or Alzheimer, where oxidative stress is an important promoter of damage. In the present paper, we focus on the role of NOD as possible neuroprotective therapeutic agents for ischemia/reperfusion treatment.
Collapse
|
23
|
Fanelli F, Sepe S, D’Amelio M, Bernardi C, Cristiano L, Cimini A, Cecconi F, Ceru' MP, Moreno S. Age-dependent roles of peroxisomes in the hippocampus of a transgenic mouse model of Alzheimer's disease. Mol Neurodegener 2013; 8:8. [PMID: 23374228 PMCID: PMC3599312 DOI: 10.1186/1750-1326-8-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 01/29/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) is a progressive neurodegenerative disease, especially affecting the hippocampus. Impairment of cognitive and memory functions is associated with amyloid β-peptide-induced oxidative stress and alterations in lipid metabolism. In this scenario, the dual role of peroxisomes in producing and removing ROS, and their function in fatty acids β-oxidation, may be critical. This work aims to investigating the possible involvement of peroxisomes in AD onset and progression, as studied in a transgenic mouse model, harboring the human Swedish familial AD mutation. We therefore characterized the peroxisomal population in the hippocampus, focusing on early, advanced, and late stages of the disease (3, 6, 9, 12, 18 months of age). Several peroxisome-related markers in transgenic and wild-type hippocampal formation were comparatively studied, by a combined molecular/immunohistochemical/ultrastructural approach. RESULTS Our results demonstrate early and significant peroxisomal modifications in AD mice, compared to wild-type. Indeed, the peroxisomal membrane protein of 70 kDa and acyl-CoA oxidase 1 are induced at 3 months, possibly reflecting the need for efficient fatty acid β-oxidation, as a compensatory response to mitochondrial dysfunction. The concomitant presence of oxidative damage markers and the altered expression of antioxidant enzymes argue for early oxidative stress in AD. During physiological and pathological brain aging, important changes in the expression of peroxisome-related proteins, also correlating with ongoing gliosis, occur in the hippocampus. These age- and genotype-based alterations, strongly dependent on the specific marker considered, indicate metabolic and/or numerical remodeling of peroxisomal population. CONCLUSIONS Overall, our data support functional and biogenetic relationships linking peroxisomes to mitochondria and suggest peroxisomal proteins as biomarkers/therapeutic targets in pre-symptomatic AD.
Collapse
Affiliation(s)
- Francesca Fanelli
- Department of Biology-LIME, University “Roma Tre”, viale Marconi 446, 00146, Rome, Italy
- University Campus Bio-Medico, via Alvaro del Portillo 21, 00128, Rome, Italy
| | - Sara Sepe
- Department of Biology-LIME, University “Roma Tre”, viale Marconi 446, 00146, Rome, Italy
| | - Marcello D’Amelio
- IRCCS S. Lucia Foundation, via del Fosso di Fiorano 65, 00143, Rome, Italy
- University Campus Bio-Medico, via Alvaro del Portillo 21, 00128, Rome, Italy
| | - Cinzia Bernardi
- Department of Radiological Sciences and Laboratory Medicine, UOC Pathological Anatomy, San Filippo Neri Hospital, via Martinotti 20, 00135, Rome, Italy
| | - Loredana Cristiano
- Department of Life, Health and Environmental Sciences, University of L’Aquila, piazzale Salvatore Tommasi 1, 67100, Coppito, (AQ), Italy
| | - AnnaMaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, piazzale Salvatore Tommasi 1, 67100, Coppito, (AQ), Italy
| | - Francesco Cecconi
- IRCCS S. Lucia Foundation, via del Fosso di Fiorano 65, 00143, Rome, Italy
- Department of Biology, University of Rome ‘Tor Vergata’, via della Ricerca Scientifica, 00133, Rome, Italy
| | - Maria Paola Ceru'
- Department of Life, Health and Environmental Sciences, University of L’Aquila, piazzale Salvatore Tommasi 1, 67100, Coppito, (AQ), Italy
| | - Sandra Moreno
- Department of Biology-LIME, University “Roma Tre”, viale Marconi 446, 00146, Rome, Italy
| |
Collapse
|
24
|
Gresa-Arribas N, Viéitez C, Dentesano G, Serratosa J, Saura J, Solà C. Modelling neuroinflammation in vitro: a tool to test the potential neuroprotective effect of anti-inflammatory agents. PLoS One 2012; 7:e45227. [PMID: 23028862 PMCID: PMC3447933 DOI: 10.1371/journal.pone.0045227] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 08/16/2012] [Indexed: 02/02/2023] Open
Abstract
Neuron-microglia co-cultures treated with pro-inflammatory agents are a useful tool to study neuroinflammation in vitro, where to test the potential neuroprotective effect of anti-inflammatory compounds. However, a great diversity of experimental conditions can be found in the literature, making difficult to select the working conditions when considering this approach for the first time. We compared the use of neuron-primary microglia and neuron-BV2 cells (a microglial cell line) co-cultures, using different neuron:microglia ratios, treatments and time post-treatment to induce glial activation and derived neurotoxicity. We show that each model requires different experimental conditions, but that both neuron-BV2 and neuron-primary microglia LPS/IFN-γ-treated co-cultures are good to study the potential neuroprotective effect of anti-inflammatory agents. The contribution of different pro-inflammatory parameters in the neurotoxicity induced by reactive microglial cells was determined. IL-10 pre-treatment completely inhibited LPS/IFN-γ-induced TNF-α and IL-6 release, and COX-2 expression both in BV2 and primary microglial cultures, but not NO production and iNOS expression. However, LPS/IFN-γ induced neurotoxicity was not inhibited in IL-10 pre-treated co-cultures. The inhibition of NO production using the specific iNOS inhibitor 1400 W totally abolished the neurotoxic effect of LPS/IFN-γ, suggesting a major role for NO in the neurotoxic effect of activated microglia. Consequently, among the anti-inflammatory agents, special attention should be paid to compounds that inhibit NO production.
Collapse
Affiliation(s)
- Núria Gresa-Arribas
- Department of Cerebral Ischemia and Neurodegeneration, Institut d’Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d’Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Cristina Viéitez
- Department of Cerebral Ischemia and Neurodegeneration, Institut d’Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d’Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Guido Dentesano
- Department of Cerebral Ischemia and Neurodegeneration, Institut d’Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d’Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Joan Serratosa
- Department of Cerebral Ischemia and Neurodegeneration, Institut d’Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d’Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Josep Saura
- Biochemistry and Molecular Biology Unit, School of Medicine, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Carme Solà
- Department of Cerebral Ischemia and Neurodegeneration, Institut d’Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d’Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain
- * E-mail:
| |
Collapse
|
25
|
Gervois P, Mansouri RM. PPARα as a therapeutic target in inflammation-associated diseases. Expert Opin Ther Targets 2012; 16:1113-25. [PMID: 22925108 DOI: 10.1517/14728222.2012.715633] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The nuclear receptor peroxisome proliferator-activated receptor alpha (PPARα) plays a major regulatory function of genes involved in energy metabolism and is a therapeutic target for dyslipidemia. The last decade provided a constellation of findings demonstrating that PPARα behaves as a modulator of both acute and chronic inflammation. PPARα became a rational potential therapeutic target for the treatment of inflammatory disorders. AERAS COVERED The ability of PPARα to control inflammatory signaling pathways via a diversity of molecular mechanisms is discussed. This review is especially focused on the global action of PPARα on inflammation in several tissues from data obtained in numerous cell types and in vivo models exposed to inflammatory stimuli. EXPERT OPINION Available PPARα agonists currently used in clinic belong to the class of hypolipidemic drugs but were not expected and not designed to act as anti-inflammatory drugs. To date, accumulating preclinical suggest evidence promising benefits when considering PPARα as a drug target to treat inflammatory disorders. However, clinical studies are needed to validate this concept. Drug design should also be directed toward the elaboration of PPARα agonists more specifically active in the control inflammatory signaling.
Collapse
Affiliation(s)
- Philippe Gervois
- Laboratoire de Biochimie, Faculté des Sciences Pharmaceutiques et Biologiques, Université Lille Nord de France, 3, rue du professeur Laguesse, BP83 F-59006, Lille, France.
| | | |
Collapse
|
26
|
Gatta V, Granzotto A, Fincati K, Drago D, Bolognin S, Zatta P, Sensi SL. Microarray analysis of gene expression profiles in human neuroblastoma cells exposed to Aβ–Zn and Aβ–Cu complexes. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aims: Abnormal metal accumulation is associated with Alzheimer’s disease and plays a relevant role in affecting amyloid-β (Aβ) peptide aggregation and neurotoxicity. Material & Methods: In the present study, employing a microarray analysis of 35,129 genes, we analyzed gene expression profile changes due to exposure to Aβ1-42 –Zn or Aβ1-42 –Cu complexes in neuronal-like cells (SH-SY5Y). Results: Microarray data indicated that Aβ–Zn or Aβ–Cu complexes selectively alter expression of genes mainly related to cell death, inflammatory responses, cytoprotective mechanisms and apoptosis. Conclusions: Taken together, these findings indicate that Aβ1–42 –Zn or Aβ1–42 –Cu show some commonalities in affecting Alzheimer’s disease-related target functions. The overall modulatory activity on these genes supports the idea of a possible net effect resulting in the activation of pathways that counteract toxic effects of Aβ–Zn or Aβ–Cu.
Collapse
Affiliation(s)
- Valentina Gatta
- Department of Oral Health & Biotechnological Sciences, “G. D’Annunzio” University, Chieti-Pescara, Italy
- Functional Genetics Unit – Center of Excellence in Aging (Ce.S.I.), Chieti, Italy
| | | | | | - Denise Drago
- CNS Repair Unit – INSPE, Biological Mass Spectrometry Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Bolognin
- Department of Neurological, Neuropsychological, Morphological & Motor Sciences – Physiology & Psychology Unit, Verona, Italy
| | - Paolo Zatta
- National Research Council, Biomedical Technology Institute (CNR-ITB), Metalloproteins Unit, Department of Biology, University of Padua, Padua, Italy
| | - Stefano L Sensi
- Department of Neuroscience & Imaging, “G. D’Annunzio” University, Chieti, Italy
| |
Collapse
|
27
|
Rice CM, Sun M, Kemp K, Gray E, Wilkins A, Scolding NJ. Mitochondrial sirtuins - a new therapeutic target for repair and protection in multiple sclerosis. Eur J Neurosci 2012; 35:1887-93. [DOI: 10.1111/j.1460-9568.2012.08150.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
28
|
D'Agostino G, Russo R, Avagliano C, Cristiano C, Meli R, Calignano A. Palmitoylethanolamide protects against the amyloid-β25-35-induced learning and memory impairment in mice, an experimental model of Alzheimer disease. Neuropsychopharmacology 2012; 37:1784-92. [PMID: 22414817 PMCID: PMC3358748 DOI: 10.1038/npp.2012.25] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alzheimer disease (AD) is the most common form of neurodegenerative dementia. Amyloid-β deposition, neurofibrillary tangle formation, and neuro-inflammation are the major pathogenic mechanisms that in concert lead to memory dysfunction and decline of cognition. Palmitoylethanolamide (PEA) is the naturally occurring lipid amide between palmitic acid and ethanolamine. Despite its clear role in inflammation and pain control, only limited in vitro evidence exist about a role for PEA in neurodegenerative diseases. Here we describe the neuroprotective activities of PEA in mice injected intracerebroventricularly with amyloid-β 25-35 (Ab25-35) peptide (9 nmol). We used spatial and non-spatial memory tasks to evaluate learning and memory dysfunctions. Ab25-35 injection significantly impaired spontaneous alternation performances, water maze spatial reference and working-like memory, and novel object recognition test. PEA was administered once a day (3-30 mg/kg, subcutaneously), starting 3 h after Ab25-35, for 1 or 2 weeks. PEA reduced (10 mg/kg) or prevented (30 mg/kg) behavioral impairments induced by Ab25-35 injection. PEA failed to rescue memory deficits induced by Ab25-35 injection in peroxisome proliferator-activated receptor-α (PPAR-α) null mice. GW7647 (2-(4-(2-(1-cyclohexanebutyl)-3-cyclohexylureido)ethyl)phenylthio)-2-methylpropionic acid; 5 mg/kg per day), a synthetic PPAR-α agonist, mimicked the effect of PEA. Acute treatment with PEA was ineffective. According with the neuroprotective profile of PEA observed during behavioral studies, experimental molecular and biochemical markers induced by Ab25-35 injection, such as lipid peroxidation, protein nytrosylation, inducible nitric oxide synthase induction, and caspase3 activation, were reduced by PEA treatment. These data disclose novel findings about the therapeutic potential of PEA, unrevealing a previously unknown therapeutic possibility to treat memory deficits associated with AD.
Collapse
Affiliation(s)
- Giuseppe D'Agostino
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Roberto Russo
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Carmen Avagliano
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Claudia Cristiano
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Rosaria Meli
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Antonio Calignano
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples Federico II, Naples, Italy,Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy. Tel: +39 (0) 81678411; Fax: +39 (0) 81678403, E-mail:
| |
Collapse
|
29
|
Sehgal N, Kumawat KL, Basu A, Ravindranath V. Fenofibrate reduces mortality and precludes neurological deficits in survivors in murine model of Japanese encephalitis viral infection. PLoS One 2012; 7:e35427. [PMID: 22514742 PMCID: PMC3325984 DOI: 10.1371/journal.pone.0035427] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 03/16/2012] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Japanese encephalitis (JE), the most common form of viral encephalitis occurs periodically in endemic areas leading to high mortality and neurological deficits in survivors. It is caused by a flavivirus, Japanese encephalitis virus (JEV), which is transmitted to humans through mosquitoes. No effective cure exists for reducing mortality and morbidity caused by JEV infection, which is primarily due to excessive inflammatory response. Fenofibrate, a peroxisome proliferator-activated receptor-α (PPARα) agonist is known to resolve inflammation by repressing nuclear factor-κB (NF-κB) and enhancing transcription of anti-oxidant and anti-inflammatory genes. In addition, fenofibrate also up-regulates a class of proteins, cytochrome P4504Fs (Cyp4fs), which are involved in detoxification of the potent pro-inflammatory eicosanoid, leukotriene B(4) (LTB(4)) to 20-hydroxy LTB(4). METHODOLOGY/PRINCIPAL FINDINGS The neuroprotective effect of fenofibrate was examined using in vitro (BV-2 microglial cell line) and in vivo (BALB/c mice) models of JEV infection. Mice were treated with fenofibrate for 2 or 4 days prior to JEV exposure. Pretreatment with fenofibrate for 4 but not 2 days reduced mortality by 80% and brain LTB(4) levels decreased concomitantly with the induction of Cyp4f15 and 4f18, which catalyze detoxification of LTB(4) through hydroxylation. Expression of cytokines and chemokine decreased significantly as did microglial activation and replication of the JEV virus. CONCLUSIONS/SIGNIFICANCE Fenofibrate confers neuroprotection against Japanese encephalitis, in vivo, in mouse model of JEV infection. Thus, fenofibrate, a PPARα agonist that is commonly used as a hypolipidemic drug could potentially be used for prophylaxis during JE epidemics to reduce mortality and morbidity.
Collapse
Affiliation(s)
| | | | | | - Vijayalakshmi Ravindranath
- Division of Molecular and Cellular Neuroscience, National Brain Research Centre, Nainwal Mode, Manesar, Haryana, India
- * E-mail:
| |
Collapse
|
30
|
Gray E, Ginty M, Kemp K, Scolding N, Wilkins A. The PPAR-γ agonist pioglitazone protects cortical neurons from inflammatory mediators via improvement in peroxisomal function. J Neuroinflammation 2012; 9:63. [PMID: 22480361 PMCID: PMC3368767 DOI: 10.1186/1742-2094-9-63] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 04/05/2012] [Indexed: 11/10/2022] Open
Abstract
Background Inflammation is known to play a pivotal role in mediating neuronal damage and axonal injury in a variety of neurodegenerative disorders. Among the range of inflammatory mediators, nitric oxide and hydrogen peroxide are potent neurotoxic agents. Recent evidence has suggested that oligodendrocyte peroxisomes may play an important role in protecting neurons from inflammatory damage. Methods To assess the influence of peroxisomal activation on nitric oxide mediated neurotoxicity, we investigated the effects of the peroxisomal proliferator activated receptor (PPAR) gamma agonist, pioglitazone in primary cortical neurons that were either exposed to a nitric oxide donor or co-cultured with activated microglia. Results Pioglitazone protected neurons and axons against both nitric-oxide donor-induced and microglia-derived nitric oxide-induced toxicity. Moreover, cortical neurons treated with this compound showed a significant increase in the protein and gene expression of PPAR-gamma, which was associated with a concomitant increase in the enzymatic activity of catalase. In addition, the protection of neurons and axons against hydrogen peroxide-induced toxicity afforded by pioglitazone appeared to be dependent on catalase. Conclusions Collectively, these observations provide evidence that modulation of PPAR-gamma activity and peroxisomal function by pioglitazone attenuates both NO and hydrogen peroxide-mediated neuronal and axonal damage suggesting a new therapeutic approach to protect against neurodegenerative changes associated with neuroinflammation.
Collapse
Affiliation(s)
- Elizabeth Gray
- Multiple Sclerosis and Stem Cell Group, Burden Centre, Institute of Clinical Neurosciences, Frenchay Hospital, University of Bristol, Bristol BS16 1JB, UK.
| | | | | | | | | |
Collapse
|
31
|
Abstract
AbstractAlthough the pathophysiology of neurodegenerative diseases is distinct for each disease, considerable evidence suggests that a single manipulation, dietary restriction, is strikingly protective against a wide range of such diseases. Thus pharmacological mimetics of dietary restrictions could prove widely protective across a range of neurodegenerative diseases. The PPAR transcription complex functions to re-program gene expression in response to nutritional deprivation as well as in response to a wide variety of lipophilic compounds. In mammals there are three PPAR homologs, which dimerize with RXR homologs and recruit coactivators Pgc1-alpha and Creb-binding protein (Cbp). PPARs are currently of clinical interest mainly because PPAR activators are approved for use in humans to reduce lipidemia and to improve glucose control in Type 2 diabetic patients. However, pharmacological enhancement of the activity of the PPAR complex is neuroprotective across a wide variety of models for neuropathological processes, including stroke, Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. Conversely activity of the PPAR transcriptional complex is reduced in a variety of neuropathological processes. The main mechanisms mediating the neuroprotective effects of the PPAR transcription complex appear to be re-routing metabolism away from glucose metabolism and toward alternative subtrates, and reduction in inflammatory processes. Recent evidence suggests that the PPAR transcriptional complex may also mediate protective effects of dietary restriction on neuropathological processes. Thus this complex represents one of the most promising for the development of pharmacological treatment of neurodegenerative diseases.
Collapse
|