1
|
Ali N, Sane MS, Tang H, Compher J, McLaughlin Q, Jones CD, Maffi SK. 6-hydroxydopamine affects multiple pathways to induce cytotoxicity in differentiated LUHMES dopaminergic neurons. Neurochem Int 2023; 170:105608. [PMID: 37678429 DOI: 10.1016/j.neuint.2023.105608] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/09/2023]
Abstract
The debilitating effects of Parkinson's disease (PD) progress over time and are pathophysiologically characterized by the formation of Lewy bodies due to the accumulation of α-synuclein aggregates resulting in the death of dopaminergic neurons. In the present study, we determined cell death pathways activated by acute exposure to 6-hydroxydopamine (6-OHDA) in differentiated LUHMES cells empirically followed by a 24 h toxin free interval, henceforth termed as washout/recovery period. Acute 6-OHDA exposure led to morphological changes in LUHMES cells and resulted in significant loss of neurite length and neurite thickness. Generation of reactive oxygen species and loss of mitochondrial membrane potential in the neuronal processes were persistent even after the recovery period. Our results show that 6-OHDA exposure leads to significant reduction in expression of mitochondrial OXPHOS complexes I, II, and IV and activation of caspase mediated apoptotic cell death cascade as observed by enhanced protein expression of cleaved-PARP-1 and cleaved-Caspase-3. Immunofluorescence microscopy approach confirmed that cell death occurs independent of the AIF translocation to the nucleus. Our experimental model, led to a ∼5-fold lower α-synuclein monomer expression and, interestingly, resulted in loss of protein ubiquitination in whole cell lysates. Altogether, this work provides evidence of multiple pathways targeted by 6-OHDA in differentiated LUHMES cells and expands research avenues for addressing the knowledge gap regarding the effect of 6-OHDA in the ubiquitin proteasome system for PD therapies.
Collapse
Affiliation(s)
- Nilufar Ali
- Department of Research and Development, Azymus Therapeutics Inc., Boise, ID, USA; Department of Biological Science, Boise State University, Boise, ID, USA.
| | - Mukta S Sane
- Department of Research and Development, Azymus Therapeutics Inc., Boise, ID, USA
| | - Huiyuan Tang
- Department of Research and Development, Azymus Therapeutics Inc., Boise, ID, USA
| | - Jadon Compher
- Department of Research and Development, Azymus Therapeutics Inc., Boise, ID, USA
| | - Quinlan McLaughlin
- Department of Research and Development, Azymus Therapeutics Inc., Boise, ID, USA
| | - Christopher D Jones
- Department of Regulatory and Medical Affairs, Azymus Therapeutics Inc., Boise, ID, USA.
| | | |
Collapse
|
2
|
Wang P, Kljavin N, Nguyen TTT, Storm EE, Marsh B, Jiang J, Lin W, Menon H, Piskol R, de Sauvage FJ. Adrenergic nerves regulate intestinal regeneration through IL-22 signaling from type 3 innate lymphoid cells. Cell Stem Cell 2023; 30:1166-1178.e8. [PMID: 37597516 DOI: 10.1016/j.stem.2023.07.013] [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: 09/29/2022] [Revised: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
The intestinal epithelium has high intrinsic turnover rate, and the precise renewal of the epithelium is dependent on the microenvironment. The intestine is innervated by a dense network of peripheral nerves that controls various aspects of intestinal physiology. However, the role of neurons in regulating epithelial cell regeneration remains largely unknown. Here, we investigated the effects of gut-innervating adrenergic nerves on epithelial cell repair following irradiation (IR)-induced injury. We observed that adrenergic nerve density in the small intestine increased post IR, while chemical adrenergic denervation impaired epithelial regeneration. Single-cell RNA sequencing experiments revealed a decrease in IL-22 signaling post IR in denervated animals. Combining pharmacologic and genetic tools, we demonstrate that β-adrenergic receptor signaling drives IL-22 production from type 3 innate lymphoid cells (ILC3s) post IR, which in turn promotes epithelial regeneration. These results define an adrenergic-ILC3 axis important for intestinal regeneration.
Collapse
Affiliation(s)
- Putianqi Wang
- Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Noelyn Kljavin
- Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Thi Thu Thao Nguyen
- Oncology Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Elaine E Storm
- Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Bryan Marsh
- Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jian Jiang
- Research Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - William Lin
- Research Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hari Menon
- Microchemistry, Proteomics, Lipidomics and Next Generation Sequencing, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Robert Piskol
- Oncology Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | |
Collapse
|
3
|
Unravelling the neuroprotective mechanisms of carotenes in differentiated human neural cells: Biochemical and proteomic approaches. FOOD CHEMISTRY. MOLECULAR SCIENCES 2022; 4:100088. [PMID: 35415676 PMCID: PMC8991711 DOI: 10.1016/j.fochms.2022.100088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 12/20/2022]
Abstract
Total mixed carotenes (TMC) protect differentiated human neural cells against 6-hydroxydopamine-induced toxicity. TMC elevated the antioxidant enzymes activities and suppressed generation of reactive oxygen species. TMC augmented the dopamine and tyrosine hydroxylase levels. TMC exerted differential protein expression in human neural cells.
Carotenoids, fat-soluble pigments found ubiquitously in plants and fruits, have been reported to exert significant neuroprotective effects against free radicals. However, the neuroprotective effects of total mixed carotenes complex (TMC) derived from virgin crude palm oil have not been studied extensively. Therefore, the present study was designed to establish the neuroprotective role of TMC on differentiated human neural cells against 6-hydroxydopamine (6-OHDA)-induced cytotoxicity. The human neural cells were differentiated using retinoic acid for six days. Then, the differentiated neural cells were pre-treated for 24 hr with TMC before exposure to 6-OHDA. TMC pre-treated neurons showed significant alleviation of 6-OHDA-induced cytotoxicity as evidenced by enhanced activity of the superoxide dismutase (SOD) and catalase (CAT) enzymes. Furthermore, TMC elevated the levels of intra-neuronal dopamine and tyrosine hydroxylase (TH) in differentiated neural cells. The 6-OHDA induced overexpression of α-synuclein was significantly hindered in neural cells pre-treated with TMC. In proteomic analysis, TMC altered the expression of ribosomal proteins, α/β isotypes of tubulins, protein disulphide isomerases (PDI) and heat shock proteins (HSP) in differentiated human neural cells. The natural palm phytonutrient TMC is a potent antioxidant with significant neuroprotective effects against free radical-induced oxidative stress.
Collapse
Key Words
- 6-OHDA, 6-hydroxydopamine
- 6-hydroxydopamine
- AD, Alzheimer’s disease
- BCM, beta-carotene-15,15′-monooxygenase
- CAT, catalase
- DRD2, dopamine receptor D2
- Dopamine
- ER, endoplasmic reticulum
- GO, gene ontology
- HSP, Heat shock protein
- HSPA9, Heat shock protein family A (HSP70) member 9
- HSPD1, Heat shock protein family D (HSP60) member 1
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- LC-MS/MS, liquid chromatography-double mass spectrometry
- LDH, lactate dehydrogenase
- MCODE, minimal common oncology data elements
- MS, mass spectrometry
- Mixed carotene
- PD, Parkinson's disease
- PDI, protein disulphide isomerases
- PHB2, prohibitin 2
- PPI, protein–protein interaction
- RAN, Ras-related nuclear protein
- ROS, reactive oxygen species
- RPs, ribosomal proteins
- SH-SY5Y neuroblastoma cells
- SOD, superoxide dismutase
- TH, tyrosine hydroxylase
- TMC, total mixed carotene complex
Collapse
|
4
|
The Protective Effect of a Unique Mix of Polyphenols and Micronutrients against Neurodegeneration Induced by an In Vitro Model of Parkinson’s Disease. Int J Mol Sci 2022; 23:ijms23063110. [PMID: 35328530 PMCID: PMC8955775 DOI: 10.3390/ijms23063110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 12/04/2022] Open
Abstract
Parkinson’s disease (PD) is second-most common disabling neurological disorder worldwide, and unfortunately, there is not yet a definitive way to prevent it. Polyphenols have been widely shown protective efficacy against various PD symptoms. However, data on their effect on physio-pathological mechanisms underlying this disease are still lacking. In the present work, we evaluated the activity of a mixture of polyphenols and micronutrients, named A5+, in the murine neuroblastoma cell line N1E115 treated with 6-Hydroxydopamine (6-OHDA), an established neurotoxic stimulus used to induce an in vitro PD model. We demonstrate that a pretreatment of these cells with A5+ causes significant reduction of inflammation, resulting in a decrease in pro-inflammatory cytokines (IFN-γ, IL-6, TNF-α, and CXCL1), a reduction in ROS production and activation of extracellular signal-regulated kinases (ERK)1/2, and a decrease in apoptotic mechanisms with the related increase in cell viability. Intriguingly, A5+ treatment promoted cellular differentiation into dopaminergic neurons, as evident by the enhancement in the expression of tyrosine hydroxylase, a well-established dopaminergic neuronal marker. Overall, these results demonstrate the synergic and innovative efficacy of A5+ mixture against PD cellular pathological processes, although further studies are needed to clarify the mechanisms underlying its beneficial effect.
Collapse
|
5
|
Magalingam KB, Somanath SD, Ramdas P, Haleagrahara N, Radhakrishnan AK. 6-Hydroxydopamine Induces Neurodegeneration in Terminally Differentiated SH-SY5Y Neuroblastoma Cells via Enrichment of the Nucleosomal Degradation Pathway: a Global Proteomics Approach. J Mol Neurosci 2022; 72:1026-1046. [PMID: 35258800 PMCID: PMC9064865 DOI: 10.1007/s12031-021-01962-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 12/22/2021] [Indexed: 01/07/2023]
Abstract
The SH-SY5Y human neuroblastoma cells have been used for decades as a cell-based model of dopaminergic neurons to explore the underlying science of cellular and molecular mechanisms of neurodegeneration in Parkinson’s disease (PD). However, data revealing the protein expression changes in 6-OHDA induced cytotoxicity in differentiated SH-SY5Y cells remain void. Therefore, we investigated the differentially regulated proteins expressed in terminally differentiated SH-SY5Y cells (differ-SH-SY5Y neural cells) exposed to 6-hydroxydopamine (6-OHDA) using the LC–MS/MS technology and construed the data using the online bioinformatics databases such as PANTHER, STRING, and KEGG. Our studies demonstrated that the neuronal development in differ-SH-SY5Y neural cells was indicated by the overexpression of proteins responsible for neurite formations such as calnexin (CANX) and calreticulin (CALR) besides significant downregulation of ribosomal proteins. The enrichment of the KEGG ribosome pathway was detected with significant downregulation (p < 0.05) of all the 21 ribosomal proteins in differ-SH-SY5Y neural cells compared with undifferentiated cells. Whereas in the PD model, the pathological changes induced by 6-OHDA were indicated by the presence of unfolded and misfolded proteins, which triggered the response of 10 kDa heat shock proteins (HSP), namely HSPE1 and HSPA9. Moreover, the 6-OHDA-induced neurodegeneration in differ-SH-SY5Y neural cells also upregulated the voltage-dependent anion-selective channel protein 1 (VDAC1) protein and enriched the KEGG systemic lupus erythematosus (SLE) pathway that was regulated by 17 histone proteins (p < 0.05) in differ-SH-SY5Y neural cells. These results suggest that the nucleosomal degradation pathway may have regulated the 6-OHDA induced neurodegeneration in PD cell-based model, which is reflected by increased apoptosis and histone release in differ-SH-SY5Y neural cells.
Collapse
Affiliation(s)
- Kasthuri Bai Magalingam
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Malaysia, Bandar Sunway, Malaysia
| | - Sushela Devi Somanath
- Pathology Division, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Premdass Ramdas
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Nagaraja Haleagrahara
- College of Public Health, Medicine and Veterinary Sciences, James Cook University, Townsville, QLD, 4811, Australia
| | - Ammu Kutty Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Malaysia, Bandar Sunway, Malaysia.
- Monash-Industry Palm Oil Education and Research Platform (MIPO), Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
6
|
The Cyclic Nitroxide TEMPOL Ameliorates Oxidative Stress but Not Inflammation in a Cell Model of Parkinson’s Disease. Antioxidants (Basel) 2022; 11:antiox11020257. [PMID: 35204139 PMCID: PMC8868255 DOI: 10.3390/antiox11020257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/25/2022] Open
Abstract
The cyclic nitroxide TEMPOL exerts anti-oxidative and anti-inflammatory effects, and thus may provide therapeutic benefit in Parkinson’s disease (PD), in which mitochondrial dysfunction, oxidative damage and inflammation have been implicated as pathophysiological mechanisms underlying the selective loss of dopaminergic neurons. Markers of oxidative stress and inflammation were investigated in a cell model of differentiated human neuroblastoma (SH-SY5Y) cells treated with the neurotoxin, 6-hydroxydopamine (6-OHDA). Treatment with TEMPOL ameliorated 6-OHDA-mediated cytotoxicity and attenuated biomarkers of oxidative stress including: mitochondrial superoxide anion free radical production, lipid peroxidation, induction of heme oxygenase 1 (HO-1) protein expression and NFκB activation. Treatment with TEMPOL abated decreased gene expression of DRD2S and DRD2L induced by 6-OHDA indicating that TEMPOL may prevent mitochondrial dysfunction and activation of pathways that result in receptor desensitization. 6-OHDA insult decreased gene expression of the antioxidant, SOD-1, and this diminution was also mitigated by TEMPOL. Activation of NFκB increased pro-inflammatory IFNy and decreased IL-6, however, TEMPOL had no effect on these inflammation mediators. Overall, this data suggests that cyclic nitroxides may preserve dopaminergic neuronal cell viability by attenuating oxidative stress and mitochondrial dysfunction, but are unable to affect inflammatory mediators that propagate cellular damage and neurodegeneration in PD.
Collapse
|
7
|
1,5-Benzodiazepin-2(3H)-ones: In Vitro Evaluation as Antiparkinsonian Agents. Antioxidants (Basel) 2021; 10:antiox10101584. [PMID: 34679721 PMCID: PMC8533176 DOI: 10.3390/antiox10101584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
A new series of twenty-three 1,5-benzodiazepin-2(3H)-ones were synthesized and evaluated in the 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid (ABTS), ferric reducing antioxidant power (FRAP), and 2,2-diphenyl-1-picrylhydrazyl (DPPH) assays as a new chemotype with antioxidant and good drug-like properties. All of the derivatives showed low cytotoxicity in comparison to curcumin against the human neuroblastoma SH-SY5Y and the human hepatoma HepG2 cell lines. Experimental solubility in bio-relevant media showed a good relationship with melting points in this series. Five compounds with the best antioxidant properties showed neuroprotectant activity against H2O2-induced oxidative stress in the SH-SY5Y cell line. From them, derivatives 4-phenyl-1H-1,5-benzodiazepin-2(3H)-one (18) and 4-(3,4,5-trimethoxyphenyl)-1H-1,5-benzodiazepin-2(3H)-one (20) yielded good neuroprotection activity in the same neuronal cell line under 6-OHD and MPP+ insults as in vitro models of mitochondrial dysfunction and oxidative stress in Parkinson’s disease (PD). Both compounds also demonstrated a significant reduction of intracellular Reactive Oxygen Species (ROS) and superoxide levels, in parallel with a good improvement of the Mitochondrial Membrane Potential (ΔΨm). Compared with curcumin, compound 18 better reduced lipid peroxidation levels, malondialdehyde (MDA), in SH-SY5Y cells under oxidative stress pressure and recovered intracellular glutathione synthetase (GSH) levels. Apoptosis and caspase-3 levels of SH-SY5Y under H2O2 pressure were also reduced after treatment with 18. Neuroprotection in neuron-like differentiated SH-SY5Y cells was also achieved with 18. In summary, this family of 1,5-benzodiazepin-2-ones with an interesting antioxidant and drug-like profile, with low cytotoxic and good neuroprotectant activity, constitutes a new promising chemical class with high potential for the development of new therapeutic agents against PD.
Collapse
|
8
|
Trapani A, Guerra L, Corbo F, Castellani S, Sanna E, Capobianco L, Monteduro AG, Manno DE, Mandracchia D, Di Gioia S, Conese M. Cyto/Biocompatibility of Dopamine Combined with the Antioxidant Grape Seed-Derived Polyphenol Compounds in Solid Lipid Nanoparticles. Molecules 2021; 26:916. [PMID: 33572331 PMCID: PMC7916151 DOI: 10.3390/molecules26040916] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/15/2023] Open
Abstract
Background: The loss of nigrostriatal neurons containing dopamine (DA) together with the "mitochondrial dysfunction" in midbrain represent the two main causes related to the symptoms of Parkinson's disease (PD). Hence, the aim of this investigation is to co-administer the missing DA and the antioxidant grape seed-derived proanthocyanidins (grape seed extract, GSE) in order to increase the levels of the neurotransmitter (which is unable to cross the Blood Brain Barrier) and reducing the oxidative stress (OS) related to PD, respectively. Methods: For this purpose, we chose Solid Lipid Nanoparticles (SLN), because they have been already proven to increase DA uptake in the brain. DA-SLN adsorbing GSE (GSE/DA-SLN) were formulated and subjected to physico-chemical characterization, and their cytocompatibility and protection against OS were examined. Results: GSE was found on SLN surface and release studies evidenced the efficiency of GSE in preventing DA autoxidation. Furthermore, SLN showed high mucoadhesive strength and were found not cytotoxic to both primary Olfactory Ensheathing and neuroblastoma SH-SY5Y cells by MTT test. Co-administration of GSE/DA-SLN and the OS-inducing neurotoxin 6-hydroxydopamine (100 μM) resulted in an increase of SH-SY5Y cell viability. Conclusions: Hence, SLN formulations containing DA and GSE may constitute interesting candidates for non-invasive nose-to-brain delivery.
Collapse
Affiliation(s)
- Adriana Trapani
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Filomena Corbo
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Stefano Castellani
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Enrico Sanna
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, Faculty of Biology and Pharmacy, University of Cagliari, Cittadella Universitaria, 09042 Monserrato (Cagliari), Italy;
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.C.); (A.G.M.); (D.E.M.)
| | - Anna Grazia Monteduro
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.C.); (A.G.M.); (D.E.M.)
| | - Daniela Erminia Manno
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.C.); (A.G.M.); (D.E.M.)
| | - Delia Mandracchia
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy;
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| |
Collapse
|
9
|
Karunaratne TB, Okereke C, Seamon M, Purohit S, Wakade C, Sharma A. Niacin and Butyrate: Nutraceuticals Targeting Dysbiosis and Intestinal Permeability in Parkinson's Disease. Nutrients 2020; 13:E28. [PMID: 33374784 PMCID: PMC7824468 DOI: 10.3390/nu13010028] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Dysbiosis is implicated by many studies in the pathogenesis of Parkinson's disease (PD). Advances in sequencing technology and computing have resulted in confounding data regarding pathogenic bacterial profiles in conditions such as PD. Changes in the microbiome with reductions in short-chain fatty acid (SCFA)-producing bacteria and increases in endotoxin-producing bacteria likely contribute to the pathogenesis of PD. GPR109A, a G-protein coupled receptor found on the surface of the intestinal epithelium and immune cells, plays a key role in controlling intestinal permeability and the inflammatory cascade. The absence of GPR109A receptors is associated with decreased concentration of tight junction proteins, leading to increased intestinal permeability and susceptibility to inflammation. In inflammatory states, butyrate acts via GPR109A to increase concentrations of tight junction proteins and improve intestinal permeability. Niacin deficiency is exacerbated in PD by dopaminergic medications. Niacin supplementation has been shown to shift macrophage polarization from pro-inflammatory to an anti-inflammatory profile. Niacin and butyrate, promising nutrients and unique ligands for the G protein-coupled receptor GPR109A, are reviewed in this paper in detail.
Collapse
Affiliation(s)
- Tennekoon B. Karunaratne
- Digestive Health Clinical Research Center, Division of Gastroenterology/Hepatology, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA; (T.B.K.); (C.O.)
| | - Chijioke Okereke
- Digestive Health Clinical Research Center, Division of Gastroenterology/Hepatology, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA; (T.B.K.); (C.O.)
| | - Marissa Seamon
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA; (M.S.); (S.P.); (C.W.)
- Department of Neuroscience, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA
| | - Sharad Purohit
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA; (M.S.); (S.P.); (C.W.)
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA
- Department of Undergraduate Health Professionals, College of Allied Health Sciences, Augusta University, 1120, 15th St, Augusta, GA 30912, USA
| | - Chandramohan Wakade
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA; (M.S.); (S.P.); (C.W.)
- Department of Neuroscience, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 1120, 15th St, Augusta, GA 30912, USA
| | - Amol Sharma
- Digestive Health Clinical Research Center, Division of Gastroenterology/Hepatology, Medical College of Georgia, Augusta University, 1120, 15th St, Augusta, GA 30912, USA; (T.B.K.); (C.O.)
| |
Collapse
|
10
|
Travaglione S, Loizzo S, Vona R, Ballan G, Rivabene R, Giordani D, Guidotti M, Dupuis ML, Maroccia Z, Baiula M, Rimondini R, Campana G, Fiorentini C. The Bacterial Toxin CNF1 Protects Human Neuroblastoma SH-SY5Y Cells against 6-Hydroxydopamine-Induced Cell Damage: The Hypothesis of CNF1-Promoted Autophagy as an Antioxidant Strategy. Int J Mol Sci 2020; 21:ijms21093390. [PMID: 32403292 PMCID: PMC7247702 DOI: 10.3390/ijms21093390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/29/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
Several chronic neuroinflammatory diseases, including Parkinson’s disease (PD), have the so-called ‘redox imbalance’ in common, a dynamic system modulated by various factors. Among them, alteration of the mitochondrial functionality can cause overproduction of reactive oxygen species (ROS) with the consequent induction of oxidative DNA damage and apoptosis. Considering the failure of clinical trials with drugs that eliminate ROS directly, research currently focuses on approaches that counteract redox imbalance, thus restoring normal physiology in a neuroinflammatory condition. Herein, we used SH-SY5Y cells treated with 6-hydroxydopamine (6-OHDA), a neurotoxin broadly employed to generate experimental models of PD. Cells were pre-treated with the Rho-modulating Escherichia coli cytotoxic necrotizing factor 1 (CNF1), before the addition of 6-OHDA. Then, cell viability, mitochondrial morphology and dynamics, redox profile as well as autophagic markers expression were assessed. We found that CNF1 preserves cell viability and counteracts oxidative stress induced by 6-OHDA. These effects are accompanied by modulation of the mitochondrial network and an increase in macroautophagic markers. Our results confirm the Rho GTPases as suitable pharmacological targets to counteract neuroinflammatory diseases and evidence the potentiality of CNF1, whose beneficial effects on pathological animal models have been already proven to act against oxidative stress through an autophagic strategy.
Collapse
Affiliation(s)
- Sara Travaglione
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
- Correspondence: ; Tel.: +39-06-49903692
| | - Stefano Loizzo
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Rosa Vona
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Giulia Ballan
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Roberto Rivabene
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Danila Giordani
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Marco Guidotti
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Maria Luisa Dupuis
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Zaira Maroccia
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Monica Baiula
- University of Bologna, 40126 Bologna, Italy; (M.B.); (R.Rim); (G.C.)
| | - Roberto Rimondini
- University of Bologna, 40126 Bologna, Italy; (M.B.); (R.Rim); (G.C.)
| | - Gabriele Campana
- University of Bologna, 40126 Bologna, Italy; (M.B.); (R.Rim); (G.C.)
| | - Carla Fiorentini
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
- Association for Research on Integrative Oncology Therapies (ARTOI), 00165 Rome, Italy
| |
Collapse
|
11
|
Nitroxide Radical-Containing Redox Nanoparticles Protect Neuroblastoma SH-SY5Y Cells against 6-Hydroxydopamine Toxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9260748. [PMID: 32377313 PMCID: PMC7196160 DOI: 10.1155/2020/9260748] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/06/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) patients can benefit from antioxidant supplementation, and new efficient antioxidants are needed. The aim of this study was to evaluate the protective effect of selected nitroxide-containing redox nanoparticles (NRNPs) in a cellular model of PD. Antioxidant properties of NRNPs were studied in cell-free systems by protection of dihydrorhodamine 123 against oxidation by 3-morpholino-sydnonimine and protection of fluorescein against bleaching by 2,2-azobis(2-amidinopropane) hydrochloride and sodium hypochlorite. Model blood-brain barrier penetration was studied using hCMEC/D3 cells. Human neuroblastoma SH-SY5Y cells, exposed to 6-hydroxydopamine (6-OHDA), were used as an in vitro model of PD. Cells were preexposed to NRNPs or free nitroxides (TEMPO or 4-amino-TEMPO) for 2 h and treated with 6-OHDA for 1 h and 24 h. The reactive oxygen species (ROS) level was estimated with dihydroethidine 123 and Fluorimetric Mitochondrial Superoxide Activity Assay Kit. Glutathione level (GSH) was measured with ortho-phtalaldehyde, ATP by luminometry, changes in mitochondrial membrane potential with JC-1, and mitochondrial mass with 10-Nonyl-Acridine Orange. NRNP1, TEMPO, and 4-amino-TEMPO (25-150 μM) protected SH-SY5Y cells from 6-OHDA-induced viability loss; the protection was much higher for NRNP1 than for free nitroxides. NRNP1 were better antioxidants in vitro and permeated better the model BBB than free nitroxides. Exposure to 6-OHDA decreased the GSH level after 1 h and increased it considerably after 24 h (apparently a compensatory overresponse); NRNPs and free nitroxides prevented this increase. NRNP1 and free nitroxides prevented the decrease in ATP level after 1 h and increased it after 24 h. 6-OHDA increased the intracellular ROS level and mitochondrial superoxide level. Studied antioxidants mostly decreased ROS and superoxide levels. 6-OHDA decreased the mitochondrial potential and mitochondrial mass; both effects were prevented by NRNP1 and nitroxides. These results suggest that the mitochondria are the main site of 6-OHDA-induced cellular damage and demonstrate a protective effect of NRNP1 in a cellular model of PD.
Collapse
|
12
|
Abstract
Toxic peripheral neuropathies are an important form of acquired polyneuropathy produced by a variety of xenobiotics and different exposure scenarios. Delineating the mechanisms of neurotoxicants and determining the degenerative biological pathways triggered by peripheral neurotoxicants will facilitate the development of sensitive and specific biochemical-based methods for identifying neurotoxicants, designing therapeutic interventions, and developing structure-activity relationships for predicting potential neurotoxicants. This review presents an overview of the general concepts of toxic peripheral neuropathies with the goal of providing insight into why certain agents target the peripheral nervous system and produce their associated lesions. Experimental data and the main hypotheses for the mechanisms of selected agents that produce neuronopathies, axonopathies, or myelinopathies including covalent or noncovalent modifications, compromised energy or protein biosynthesis, and oxidative injury and disruption of ionic gradients across membranes are presented. The relevance of signaling between the main components of peripheral nerve, that is, glia, neuronal perikaryon, and axon, as a target for neurotoxicants and the contribution of active programmed degenerative pathways to the lesions observed in toxic peripheral neuropathies is also discussed.
Collapse
|
13
|
Varešlija D, Tipton KF, Davey GP, McDonald AG. 6-Hydroxydopamine: a far from simple neurotoxin. J Neural Transm (Vienna) 2020; 127:213-230. [DOI: 10.1007/s00702-019-02133-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/21/2019] [Indexed: 12/13/2022]
|
14
|
Ko KR, Tam NW, Teixeira AG, Frampton JP. SH-SY5Y and LUHMES cells display differential sensitivity to MPP+, tunicamycin, and epoxomicin in 2D and 3D cell culture. Biotechnol Prog 2019; 36:e2942. [PMID: 31756288 DOI: 10.1002/btpr.2942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/25/2019] [Accepted: 11/15/2019] [Indexed: 02/03/2023]
Abstract
SH-SY5Y and LUHMES cell lines are widely used as model systems for studying neurotoxicity. Most of the existing data regarding the sensitivity of these cell lines to neurotoxicants have been recorded from cells growing as two-dimensional (2D) cultures on the surface of glass or plastic. With the emergence of 3D culture platforms designed to better represent native tissue, there is a growing need to compare the toxicology of neurons grown in 3D environments to those grown in 2D to better understand the impact that culture environment has on toxicant sensitivity. Here, a simple 3D culture method was used to assess the impact of growth environment on the sensitivity of SH-SY5Y cells and LUHMES cells to MPP+, tunicamycin, and epoxomicin, three neurotoxicants that have been previously used to generate experimental models for studying Parkinson's disease pathogenesis. SH-SY5Y cell viability following treatment with these three toxicants was significantly lower in 2D cultures as compared to 3D cultures. On the contrary, LUHMES cells did not show significant differences between growth conditions for any of the toxicants examined. However, LUHMES cells were more sensitive to MPP+, tunicamycin, and epoxomicin than SH-SY5Y cells. Thus, both the choice of cell line and the choice of growth environment must be considered when interpreting in vitro neurotoxicity data.
Collapse
Affiliation(s)
- Kristin Robin Ko
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Nicky W Tam
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alyne G Teixeira
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - John P Frampton
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
15
|
Callizot N, Combes M, Henriques A, Poindron P. Necrosis, apoptosis, necroptosis, three modes of action of dopaminergic neuron neurotoxins. PLoS One 2019; 14:e0215277. [PMID: 31022188 PMCID: PMC6483187 DOI: 10.1371/journal.pone.0215277] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/31/2019] [Indexed: 12/21/2022] Open
Abstract
Most of the Parkinson’s disease (PD) cases are sporadic, although several genes are directly related to PD. Several pathways are central in PD pathogenesis: protein aggregation linked to proteasomal impairments, mitochondrial dysfunctions and impairment in dopamine (DA) release. Here we studied the close crossing of mitochondrial dysfunction and aggregation of α-synuclein (α-syn) and in the extension in the dopaminergic neuronal death. Here, using rat primary cultures of mesencephalic neurons, we induced the mitochondrial impairments using “DA-toxins” (MPP+, 6OHDA, rotenone). We showed that the DA-Toxins induced dopaminergic cell death through different pathways: caspase-dependent cell death for 6OHDA; MPP+ stimulated caspase-independent cell death, and rotenone activated both pathways. In addition, a decrease in energy production and/or a development of oxidative stress were observed and were linked to α-syn aggregation with generation of Lewy body-like inclusions (found inside and outside the dopaminergic neurons). We demonstrated that any of induced mitochondrial disturbances and processes of death led to α-syn protein aggregation and finally to cell death. Our study depicts the cell death mechanisms taking place in in vitro models of Parkinson’s disease and how mitochondrial dysfunctions is at the cross road of the pathologies of this disease.
Collapse
Affiliation(s)
- Noëlle Callizot
- Department of Pharmacology, Neuro-Sys SAS, Gardanne, France
- * E-mail:
| | - Maud Combes
- Department of Pharmacology, Neuro-Sys SAS, Gardanne, France
| | | | | |
Collapse
|
16
|
Tamtaji OR, Taghizadeh M, Aghadavod E, Mafi A, Dadgostar E, Daneshvar Kakhaki R, Abolhassani J, Asemi Z. The effects of omega-3 fatty acids and vitamin E co-supplementation on gene expression related to inflammation, insulin and lipid in patients with Parkinson's disease: A randomized, double-blind, placebo-controlled trial. Clin Neurol Neurosurg 2018; 176:116-121. [PMID: 30554092 DOI: 10.1016/j.clineuro.2018.12.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 12/01/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study was conducted to evaluate the effects of omega-3 fatty acids and vitamin E co-supplementation on gene expression related to inflammation, insulin and lipid in subjects with Parkinson's disease (PD). PATIENTS AND METHODS This randomized, double-blind, placebo-controlled clinical trial was performed in 40 subjects with PD. Participants were randomly allocated into two groups to take either 1000 mg/day of omega-3 fatty acids from flaxseed oil plus 400 IU/day of vitamin E supplements or placebo (n = 20 each group) for 12 weeks. Gene expression related to inflammation, insulin and lipid were quantified in peripheral blood mononuclear cells (PBMC) of PD patients with RT-PCR method. RESULTS After the 12-week intervention, compared with the placebo, omega-3 fatty acids and vitamin E co-supplementation downregulated gene expression of tumor necrosis factor alpha (TNF-α) (P = 0.002) in PBMC of subjects with PD. In addition, omega-3 fatty acids and vitamin E co-supplementation upregulated peroxisome proliferator-activated receptor gamma (PPAR-γ) (P = 0.03), and downregulated oxidized low-density lipoprotein receptor (LDLR) (P = 0.002) in PBMC of subjects with PD compared with the placebo. We did not observe any significant effect of omega-3 fatty acids and vitamin E co-supplementation on gene expression of interleukin-1 (IL-1) and IL-8 in PBMC of patients with PD. CONCLUSIONS Overall, omega-3 fatty acids and vitamin E co-supplementation for 12 weeks in PD patients significantly improved gene expression of TNF-α, PPAR-γ and LDLR, but did not affect IL-1 and IL-8.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Alireza Mafi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Ehsan Dadgostar
- Halal Research Center of IRI, FDA, Tehran, Islamic Republic of Iran
| | - Reza Daneshvar Kakhaki
- Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Javad Abolhassani
- Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
17
|
Parga JA, Rodriguez-Perez AI, Garcia-Garrote M, Rodriguez-Pallares J, Labandeira-Garcia JL. Angiotensin II induces oxidative stress and upregulates neuroprotective signaling from the NRF2 and KLF9 pathway in dopaminergic cells. Free Radic Biol Med 2018; 129:394-406. [PMID: 30315936 DOI: 10.1016/j.freeradbiomed.2018.10.409] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/30/2018] [Accepted: 10/05/2018] [Indexed: 12/19/2022]
Abstract
Nuclear factor-E2-related factor 2 (NRF2) is a transcription factor that activates the antioxidant cellular defense in response to oxidative stress, leading to neuroprotective effects in Parkinson's disease (PD) models. We have previously shown that Angiotensin II (AngII) induces an increase in reactive oxygen species (ROS) via AngII receptor type 1 and NADPH oxidase (NOX), which may activate the NRF2 pathway. However, controversial data suggest that AngII induces a decrease in NRF2 signaling leading to an increase in oxidative stress. We analyzed the effect of AngII and the dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA) in culture and in vivo, and examined the effects on the expression of NRF2-related genes. Treatment of neuronal cell lines Mes23.5, N27 and SH-SY5Y with AngII, 6-OHDA or a combination of both increased ROS production and reduced cell viability. Simultaneously, these treatments induced an increase in expression in the NRF2-regulated genes heme oxygenase 1 (Hmox1), NAD(P)H quinone dehydrogenase 1 (Nqo1) and Kruppel like factor 9 (Klf9). Moreover, overexpression of KLF9 transcription factor caused a reduction in the production of ROS induced by treatment with AngII or 6-OHDA and improved the survival of these neuronal cells. Rats treated with AngII, 6-OHDA or a combination of both also showed an increased expression of NRF2 related genes and KLF9. In conclusion, our data indicate that AngII induces a damaging effect in neuronal cells, but also acts as a signaling molecule to activate NRF2 and KLF9 neuroprotective pathways in cellular and animal models of PD.
Collapse
Affiliation(s)
- Juan A Parga
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria Garcia-Garrote
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jannette Rodriguez-Pallares
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
18
|
Kim H, Ham S, Lee JY, Jo A, Lee GH, Lee YS, Cho M, Shin HM, Kim D, Pletnikova O, Troncoso JC, Shin JH, Lee YI, Lee Y. Estrogen receptor activation contributes to RNF146 expression and neuroprotection in Parkinson's disease models. Oncotarget 2017; 8:106721-106739. [PMID: 29290984 PMCID: PMC5739769 DOI: 10.18632/oncotarget.21828] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/23/2017] [Indexed: 11/30/2022] Open
Abstract
RNF146 is an E3 ubiquitin ligase that specifically recognizes and polyubiquitinates poly (ADP-ribose) (PAR)-conjugated substrates for proteasomal degradation. RNF146 has been shown to be neuroprotective against PAR polymerase-1 (PARP1)-induced cell death during stroke. Here we report that RNF146 expression and RNF146 inducers can prevent cell death elicited by Parkinson’s disease (PD)-associated and PARP1-activating stimuli. In SH-SY5Y cells, RNF146 expression conferred resistance to toxic stimuli that lead to PARP1 activation. High-throughput screen using a luciferase construct harboring the RNF146 promoter identified liquiritigenin as an RNF146 inducer. We found that RNF146 expression by liquiritigenin was mediated by estrogen receptor activation and contributed to cytoprotective effect of liquiritigenin. Finally, RNF146 expression by liquiritigenin in mouse brains provided dopaminergic neuroprotection in a 6-hydroxydopamine PD mouse model. Given the presence of PARP1 activity and RNF146 deficits in PD, it could be a potential therapeutic strategy to restore RNF146 expression by natural compounds or estrogen receptor activation.
Collapse
Affiliation(s)
- Hyojung Kim
- Division of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 440-746, Republic of Korea
| | - Sangwoo Ham
- Division of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 440-746, Republic of Korea
| | - Joon Yeop Lee
- National Development Institute of Korean Medicine, Gyeongsan 38540, Republic of Korea
| | - Areum Jo
- Division of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 440-746, Republic of Korea
| | - Gum Hwa Lee
- College of Pharmacy, Chosun University, Gwangju 501-759, Republic of Korea
| | - Yun-Song Lee
- Division of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 440-746, Republic of Korea
| | - MyoungLae Cho
- National Development Institute of Korean Medicine, Gyeongsan 38540, Republic of Korea
| | - Heung-Mook Shin
- National Development Institute of Korean Medicine, Gyeongsan 38540, Republic of Korea
| | - Donghoon Kim
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olga Pletnikova
- Department of Pathology, Division of Neuropathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Juan C Troncoso
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pathology, Division of Neuropathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joo-Ho Shin
- Division of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 440-746, Republic of Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-Do 440-746, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, Daegu Geongbuk Institute of Science and Technology, Daegu 42988, South Korea.,Companion Diagnostics and Medical Technology Research Group, Daegu Geongbuk Institute of Science and Technology, Daegu 42988, South Korea
| | - Yunjong Lee
- Division of Pharmacology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 440-746, Republic of Korea
| |
Collapse
|
19
|
Patel M, McElroy PB. Mitochondrial Dysfunction in Parkinson’s Disease. OXIDATIVE STRESS AND REDOX SIGNALLING IN PARKINSON’S DISEASE 2017. [DOI: 10.1039/9781782622888-00061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders where oxidative stress and mitochondrial dysfunction have been implicated as etiological factors. Mitochondria are the major producers of reactive oxygen species (ROS) that can have damaging effects to cellular macromolecules leading to neurodegeneration. The most compelling evidence for the role of mitochondria in the pathogenesis of PD has been derived from toxicant-induced models of parkinsonism. Over the years, epidemiological studies have suggested a link between exposure to environmental toxins such as pesticides and the risk of developing PD. Data from human and experimental studies involving the use of chemical agents like paraquat, diquat, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, rotenone and maneb have provided valuable insight into the underlying mitochondrial mechanisms contributing to PD and associated neurodegeneration. In this review, we have discussed the role of mitochondrial ROS and dysfunction in the pathogenesis of PD with a special focus on environmental agent-induced parkinsonism. We have described the various mitochondrial mechanisms by which such chemicals exert neurotoxicity, highlighting some landmark epidemiological and experimental studies that support the role of mitochondrial ROS and oxidative stress in contributing to these effects. Finally, we have discussed the significance of these studies in understanding the mechanistic underpinnings of PD-related dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| | - Pallavi Bhuyan McElroy
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| |
Collapse
|
20
|
Lopes FM, Bristot IJ, da Motta LL, Parsons RB, Klamt F. Mimicking Parkinson's Disease in a Dish: Merits and Pitfalls of the Most Commonly used Dopaminergic In Vitro Models. Neuromolecular Med 2017; 19:241-255. [PMID: 28721669 DOI: 10.1007/s12017-017-8454-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/12/2017] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and has both unknown etiology and non-curative therapeutic options. Patients begin to present the classic motor symptoms of PD-tremor at rest, bradykinesia and rigidity-once 50-70% of the dopaminergic neurons of the nigrostriatal pathway have degenerated. As a consequence of this, it is difficult to investigate the early-stage events of disease pathogenesis. In vitro experimental models are used extensively in PD research because they present a controlled environment that enables the direct investigation of the early molecular mechanisms that are potentially involved with dopaminergic degeneration, as well as for the screening of potential therapeutic drugs. However, the establishment of PD in vitro models is a controversial issue for neuroscience research not only because it is challenging to mimic, in isolated cell systems, the physiological neuronal environment, but also the pathophysiological conditions experienced by human dopaminergic cells in vivo during the progression of the disease. Since no previous work has attempted to systematically review the literature regarding the establishment of an optimal in vitro model, and/or the features presented by available models used in the PD field, this review aims to summarize the merits and limitations of the most widely used dopaminergic in vitro models in PD research, which may help the PD researcher to choose the most appropriate model for studies directed at the elucidation of the early-stage molecular events underlying PD onset and progression.
Collapse
Affiliation(s)
- Fernanda Martins Lopes
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil. .,Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| | - Ivi Juliana Bristot
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Leonardo Lisbôa da Motta
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Richard B Parsons
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Fabio Klamt
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
21
|
Hu J, Ferchmin PA, Hemmerle AM, Seroogy KB, Eterovic VA, Hao J. 4R-Cembranoid Improves Outcomes after 6-Hydroxydopamine Challenge in Both In vitro and In vivo Models of Parkinson's Disease. Front Neurosci 2017; 11:272. [PMID: 28611572 PMCID: PMC5447022 DOI: 10.3389/fnins.2017.00272] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/28/2017] [Indexed: 11/13/2022] Open
Abstract
(1S, 2E, 4R, 6R,-7E, 11E)-2, 7, 11-cembratriene-4, 6-diol (4R) is one of the cembranoids found in tobacco leaves. Previous studies have found that 4R protected acute rat hippocampal slices against neurotoxicity induced by N-methyl-D-aspartate (NMDA) and against the toxic organophosphorus compounds paraoxon and diisopropylfluorophosphate (DFP). Furthermore, in vivo, 4R reduced the infarct size in a rodent ischemic stroke model and neurodegeneration caused by DFP. The present study expanded our previous study by focusing on the effect of 4R in Parkinson's disease (PD) and elucidating its underlying mechanisms using 6-hydroxydopamine (6-OHDA)-induced injury models. We found that 4R exhibited significant neuroprotective activity in the rat unilateral 6-OHDA-induced PD model in vivo. The therapeutic effect was evident both at morphological and behavioral levels. 4R (6 and 12 mg/kg) treatments significantly improved outcomes of 6-OHDA-induced PD in vivo as indicated by reducing forelimb asymmetry scores and corner test scores 4 weeks after injection of 6-OHDA (p < 0.05). The therapeutic effect of 4R was also reflected by decreased depletion of tyrosine hydroxylase (TH) in the striatum and substantia nigra (SN) on the side injected with 6-OHDA. TH expression was 70.3 and 62.8% of the contralateral side in striatum and SN, respectively, after 6 mg/kg 4R treatment; furthermore, it was 80.1 and 79.3% after treatment with 12 mg/kg of 4R. In the control group, it was 51.9 and 23.6% of the contralateral striatum and SN (p < 0.05). Moreover, 4R also protected differentiated neuro-2a cells from 6-OHDA-induced cytotoxicity in vitro. The activation of p-AKT and HAX-1, and inhibition of caspase-3 and endothelial inflammation, were involved in 4R-mediated protection against 6-OHDA-induced injury. In conclusion, the present study indicates that 4R shows a therapeutic effect in the rat 6-OHDA-induced PD model in vivo and in 6-OHDA-challenged neuro-2a cells in vitro.
Collapse
Affiliation(s)
- Jing Hu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of CincinnatiCincinnati, OH, United States
| | - P A Ferchmin
- Department of Neurosciences, School of Medicine, Universidad Central del CaribeBayamón, Puerto Rico
| | - Ann M Hemmerle
- Department of Neurology and Rehabilitation Medicine, University of CincinnatiCincinnati, OH, United States
| | - Kim B Seroogy
- Department of Neurology and Rehabilitation Medicine, University of CincinnatiCincinnati, OH, United States
| | - Vesna A Eterovic
- Department of Neurosciences, School of Medicine, Universidad Central del CaribeBayamón, Puerto Rico
| | - Jiukuan Hao
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of CincinnatiCincinnati, OH, United States
| |
Collapse
|
22
|
Parashar A, Udayabanu M. Gut microbiota: Implications in Parkinson's disease. Parkinsonism Relat Disord 2017; 38:1-7. [PMID: 28202372 PMCID: PMC7108450 DOI: 10.1016/j.parkreldis.2017.02.002] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 12/24/2016] [Accepted: 02/04/2017] [Indexed: 12/22/2022]
Abstract
Gut microbiota (GM) can influence various neurological outcomes, like cognition, learning, and memory. Commensal GM modulates brain development and behavior and has been implicated in several neurological disorders like Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, anxiety, stress and much more. A recent study has shown that Parkinson's disease patients suffer from GM dysbiosis, but whether it is a cause or an effect is yet to be understood. In this review, we try to connect the dots between GM and PD pathology using direct and indirect evidence.
Collapse
Affiliation(s)
- Arun Parashar
- Jaypee University of Information Technology, Waknaghat, District- Solan, Himachal Pradesh, PIN-173234, India
| | - Malairaman Udayabanu
- Jaypee University of Information Technology, Waknaghat, District- Solan, Himachal Pradesh, PIN-173234, India.
| |
Collapse
|
23
|
Taghizadeh M, Tamtaji OR, Dadgostar E, Daneshvar Kakhaki R, Bahmani F, Abolhassani J, Aarabi MH, Kouchaki E, Memarzadeh MR, Asemi Z. The effects of omega-3 fatty acids and vitamin E co-supplementation on clinical and metabolic status in patients with Parkinson's disease: A randomized, double-blind, placebo-controlled trial. Neurochem Int 2017; 108:183-189. [PMID: 28342967 DOI: 10.1016/j.neuint.2017.03.014] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/14/2017] [Accepted: 03/21/2017] [Indexed: 12/23/2022]
Abstract
The current research was performed to evaluate the effects of omega-3 fatty acids and vitamin E co-supplementation on clinical signs and metabolic status in people with Parkinson's disease (PD). This randomized double-blind placebo-controlled clinical trial was conducted in 60 patients with PD. Participants were randomly assigned into two groups to receive either 1000 mg omega-3 fatty acids from flaxseed oil plus 400 IU vitamin E supplements (n = 30) or placebo (n = 30) for 12 weeks. Unified Parkinson's disease rating stage (UPDRS) were recorded at baseline and the after 3-month intervention. After 12 weeks' intervention, compared with the placebo, omega-3 fatty acids and vitamin E co-supplementation led to a significant improve in UPDRS (-3.3 ± 10.0 vs. +4.4 ± 14.9, P = 0.02). Furthermore, co-supplementation decreased high-sensitivity C-reactive protein (hs-CRP) (-0.3 ± 0.6 vs. +0.3 ± 0.3 μg/mL, P < 0.001), and increased total antioxidant capacity (TAC) (+65.2 ± 68.7 vs. +16 ± 52.4 μmol/L, P = 0.003) and glutathione (GSH) concentrations (+41.4 ± 80.6 vs. -19.6 ± 55.9 μmol/L, P = 0.001) compared with the placebo. Additionally, co-supplementation meaningfully decreased insulin (-2.1 ± 4.9 vs. +1.4 ± 6.2 μIU/mL, P = 0.01), homeostasis model of assessment-estimated insulin resistance (-0.7 ± 1.8 vs.+0.3 ± 1.6, P = 0.02) and Beta cell function (-5.9 ± 13.9 vs. +5.7 ± 25.5, P = 0.03), and increased quantitative insulin sensitivity check index (+0.009 ± 0.02 vs. -0.006 ± 0.03, P = 0.03) compared with the placebo. Overall, our study demonstrated that omega-3 fatty acids and vitamin E co-supplementation in people with PD had favorable effects on UPDRS, hs-CRP, TAC, GSH and markers of insulin metabolism.
Collapse
Affiliation(s)
- Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Omid Reza Tamtaji
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Ehsan Dadgostar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Reza Daneshvar Kakhaki
- Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Fereshteh Bahmani
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Javad Abolhassani
- Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Mohammad Hossein Aarabi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Ebrahim Kouchaki
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran; Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
24
|
RA Differentiation Enhances Dopaminergic Features, Changes Redox Parameters, and Increases Dopamine Transporter Dependency in 6-Hydroxydopamine-Induced Neurotoxicity in SH-SY5Y Cells. Neurotox Res 2017; 31:545-559. [DOI: 10.1007/s12640-016-9699-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/28/2016] [Accepted: 12/30/2016] [Indexed: 12/19/2022]
|
25
|
Xicoy H, Wieringa B, Martens GJM. The SH-SY5Y cell line in Parkinson's disease research: a systematic review. Mol Neurodegener 2017; 12:10. [PMID: 28118852 PMCID: PMC5259880 DOI: 10.1186/s13024-017-0149-0] [Citation(s) in RCA: 585] [Impact Index Per Article: 83.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/05/2017] [Indexed: 12/29/2022] Open
Abstract
Parkinson’s disease (PD) is a devastating and highly prevalent neurodegenerative disease for which only symptomatic treatment is available. In order to develop a truly effective disease-modifying therapy, improvement of our current understanding of the molecular and cellular mechanisms underlying PD pathogenesis and progression is crucial. For this purpose, standardization of research protocols and disease models is necessary. As human dopaminergic neurons, the cells mainly affected in PD, are difficult to obtain and maintain as primary cells, current PD research is mostly performed with permanently established neuronal cell models, in particular the neuroblastoma SH-SY5Y lineage. This cell line is frequently chosen because of its human origin, catecholaminergic (though not strictly dopaminergic) neuronal properties, and ease of maintenance. However, there is no consensus on many fundamental aspects that are associated with its use, such as the effects of culture media composition and of variations in differentiation protocols. Here we present the outcome of a systematic review of scientific articles that have used SH-SY5Y cells to explore PD. We describe the cell source, culture conditions, differentiation protocols, methods/approaches used to mimic PD and the preclinical validation of the SH-SY5Y findings by employing alternative cellular and animal models. Thus, this overview may help to standardize the use of the SH-SY5Y cell line in PD research and serve as a future user’s guide.
Collapse
Affiliation(s)
- Helena Xicoy
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboudumc, Nijmegen, The Netherlands.,Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboudumc, Nijmegen, The Netherlands
| | - Gerard J M Martens
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Stępkowski TM, Męczyńska-Wielgosz S, Kruszewski M. mitoLUHMES: An Engineered Neuronal Cell Line for the Analysis of the Motility of Mitochondria. Cell Mol Neurobiol 2016; 37:1055-1066. [PMID: 27832395 PMCID: PMC5494036 DOI: 10.1007/s10571-016-0438-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/22/2016] [Indexed: 01/21/2023]
Abstract
Perturbations in the transport of mitochondria and their quality control in neuronal cells underlie many types of neurological pathologies, whereas systems enabling convenient analysis of mitochondria behavior in cellular models of neurodegenerative diseases are limited. In this study, we present a modified version of lund human mesencephalic cells, mitoLUHMES, expressing GFP and mitochondrially targeted DsRed2 fluorescent proteins, intended for in vitro analysis of mitochondria trafficking by real-time fluorescence microscopy. This cell line can be easily differentiated into neuronal phenotype and allows us to observe movements of single mitochondria in single cells grown in high-density cultures. We quantified the perturbations in mitochondria morphology and dynamics in cells treated with model neurotoxins: carbonyl cyanide m-chlorophenylhydrazone and 6-hydroxydopamine. For the first time we filmed the processes of fission, fusion, pausing, and reversal of mitochondria movement direction in LUHMES cells. We present a detailed analysis of mitochondria length, velocity, and frequency of movement for static, anterograde, and retrograde motile mitochondria. The observed neurotoxin treatment-mediated decreases in morphological and kinetic parameters of mitochondria provide foundation for the future studies exploiting mitoLUHMES as a new model for neurobiology.
Collapse
Affiliation(s)
- Tomasz M Stępkowski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna16, 03-195, Warsaw, Poland.
| | - Sylwia Męczyńska-Wielgosz
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna16, 03-195, Warsaw, Poland
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna16, 03-195, Warsaw, Poland.,Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090, Lublin, Poland.,Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, ul. Sucharskiego 2, 35-225, Rzeszow, Poland
| |
Collapse
|
27
|
Tunesi M, Fusco F, Fiordaliso F, Corbelli A, Biella G, Raimondi MT. Optimization of a 3D Dynamic Culturing System for In Vitro Modeling of Frontotemporal Neurodegeneration-Relevant Pathologic Features. Front Aging Neurosci 2016; 8:146. [PMID: 27445790 PMCID: PMC4916174 DOI: 10.3389/fnagi.2016.00146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/07/2016] [Indexed: 12/23/2022] Open
Abstract
Frontotemporal lobar degeneration (FTLD) is a severe neurodegenerative disorder that is diagnosed with increasing frequency in clinical setting. Currently, no therapy is available and in addition the molecular basis of the disease are far from being elucidated. Consequently, it is of pivotal importance to develop reliable and cost-effective in vitro models for basic research purposes and drug screening. To this respect, recent results in the field of Alzheimer’s disease have suggested that a tridimensional (3D) environment is an added value to better model key pathologic features of the disease. Here, we have tried to add complexity to the 3D cell culturing concept by using a microfluidic bioreactor, where cells are cultured under a continuous flow of medium, thus mimicking the interstitial fluid movement that actually perfuses the body tissues, including the brain. We have implemented this model using a neuronal-like cell line (SH-SY5Y), a widely exploited cell model for neurodegenerative disorders that shows some basic features relevant for FTLD modeling, such as the release of the FTLD-related protein progranulin (PRGN) in specific vesicles (exosomes). We have efficiently seeded the cells on 3D scaffolds, optimized a disease-relevant oxidative stress experiment (by targeting mitochondrial function that is one of the possible FTLD-involved pathological mechanisms) and evaluated cell metabolic activity in dynamic culture in comparison to static conditions, finding that SH-SY5Y cells cultured in 3D scaffold are susceptible to the oxidative damage triggered by a mitochondrial-targeting toxin (6-OHDA) and that the same cells cultured in dynamic conditions kept their basic capacity to secrete PRGN in exosomes once recovered from the bioreactor and plated in standard 2D conditions. We think that a further improvement of our microfluidic system may help in providing a full device where assessing basic FTLD-related features (including PRGN dynamic secretion) that may be useful for monitoring disease progression over time or evaluating therapeutic interventions.
Collapse
Affiliation(s)
- Marta Tunesi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di MilanoMilan, Italy; Unità di Ricerca Consorzio INSTM, Politecnico di MilanoMilan, Italy
| | - Federica Fusco
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Fabio Fiordaliso
- Unit of Bio-imaging, Department of Cardiovascular Research, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Alessandro Corbelli
- Unit of Bio-imaging, Department of Cardiovascular Research, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Gloria Biella
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Manuela T Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano Milan, Italy
| |
Collapse
|
28
|
Pahrudin Arrozi A, Wan Ngah WZ, Mohd Yusof YA, Ahmad Damanhuri MH, Makpol S. Antioxidant modulation in restoring mitochondrial function in neurodegeneration. Int J Neurosci 2016; 127:218-235. [PMID: 27074540 DOI: 10.1080/00207454.2016.1178261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the leading causes of disability associated with neurodegeneration worldwide. These diseases are influenced by multiple genetic and environmental factors and share similar mechanisms as both are characterized by accumulation and aggregation of misfolded proteins - amyloid-beta (Aβ) in AD and α-synuclein in PD. Over the past decade, increasing evidence has shown that mitochondrial dysfunction and the generation of reactive oxygen species (ROS) are involved in the pathology of these diseases, and the contributions of these defects to the cellular and molecular changes that eventually cause neuronal death have been explored. Using mitochondrial protective agents, such as antioxidants, to combat ROS provides a new strategy for neurodegenerative treatment. In this review, we highlight the potential of multiple types of antioxidants, including vitamins, phytochemicals, fatty acids and minerals, as well as synthetic antioxidants specifically targeting the mitochondria, which can restore mitochondrial function, in the treatment of neurodegenerative disorders at both the pre-clinical and clinical stages by focusing on AD and PD.
Collapse
Affiliation(s)
- Aslina Pahrudin Arrozi
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | - Wan Zurinah Wan Ngah
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | - Yasmin Anum Mohd Yusof
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | | | - Suzana Makpol
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| |
Collapse
|
29
|
Yew MY, Koh RY, Chye SM, Othman I, Ng KY. Edible bird's nest ameliorates oxidative stress-induced apoptosis in SH-SY5Y human neuroblastoma cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:391. [PMID: 25308934 PMCID: PMC4210536 DOI: 10.1186/1472-6882-14-391] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 10/01/2014] [Indexed: 12/21/2022]
Abstract
Background Parkinson’s disease (PD) is the second most common neurodegenerative disorder affecting the senile population with manifestation of motor disability and cognitive impairment. Reactive oxygen species (ROS) is implicated in the progression of oxidative stress-related apoptosis and cell death of the midbrain dopaminergic neurons. Its interplay with mitochondrial functionality constitutes an important aspect of neuronal survival in the perspective of PD. Edible bird’s nest (EBN) is an animal-derived natural food product made of saliva secreted by swiftlets from the Aerodamus genus. It contains bioactive compounds which might confer neuroprotective effects to the neurons. Hence this study aims to investigate the neuroprotective effect of EBN extracts in the neurotoxin-induced in vitro PD model. Methods EBN was first prepared into pancreatin-digested crude extract and water extract. In vitro PD model was generated by exposing SH-SY5Y cells to neurotoxin 6-hydroxydopamine (6-OHDA). Cytotoxicity of the extracts on SH-SY5Y cells was tested using MTT assay. Then, microscopic morphological and nuclear examination, cell viability test and ROS assay were performed to assess the protective effect of EBN extracts against 6-OHDA-induced cellular injury. Apoptotic event was later analysed with Annexin V-propidium iodide flow cytometry. To understand whether the mechanism underlying the neuroprotective effect of EBN was mediated via mitochondrial or caspase-dependent pathway, mitochondrial membrane potential (MMP) measurement and caspase-3 quantification were carried out. Results Cytotoxicity results showed that crude EBN extract did not cause SH-SY5Y cell death at concentrations up to 75 μg/ml while the maximum non-toxic dose (MNTD) of water extract was double of that of crude extract. Morphological observation and nuclear staining suggested that EBN treatment reduced the level of 6-OHDA-induced apoptotic changes in SH-SY5Y cells. MTT study further confirmed that cell viability was better improved with crude EBN extract. However, water extract exhibited higher efficacy in ameliorating ROS build up, early apoptotic membrane phosphatidylserine externalization as well as inhibition of caspase-3 cleavage. None of the EBN treatment had any effect on MMP. Conclusions Current findings suggest that EBN extracts might confer neuroprotective effect against 6-OHDA-induced degeneration of dopaminergic neurons, particularly through inhibition of apoptosis. Thus EBN may be a viable nutraceutical option to protect against oxidative stress-related neurodegenerative disorders such as PD.
Collapse
|
30
|
BAG1 is Neuroprotective in In Vivo and In Vitro Models of Parkinson’s Disease. J Mol Neurosci 2014; 55:587-95. [DOI: 10.1007/s12031-014-0396-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/29/2014] [Indexed: 11/27/2022]
|
31
|
Seidl SE, Santiago JA, Bilyk H, Potashkin JA. The emerging role of nutrition in Parkinson's disease. Front Aging Neurosci 2014; 6:36. [PMID: 24639650 PMCID: PMC3945400 DOI: 10.3389/fnagi.2014.00036] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/20/2014] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease in ageing individuals. It is now clear that genetic susceptibility and environmental factors play a role in disease etiology and progression. Because environmental factors are involved with the majority of the cases of PD, it is important to understand the role nutrition plays in both neuroprotection and neurodegeneration. Recent epidemiological studies have revealed the promise of some nutrients in reducing the risk of PD. In contrast, other nutrients may be involved with the etiology of neurodegeneration or exacerbate disease progression. This review summarizes the studies that have addressed these issues and describes in detail the nutrients and their putative mechanisms of action in PD.
Collapse
Affiliation(s)
- Stacey E Seidl
- The Cellular and Molecular Pharmacology Department, The Chicago Medical School, Rosalind Franklin University of Medicine and Science North Chicago, IL, USA
| | - Jose A Santiago
- The Cellular and Molecular Pharmacology Department, The Chicago Medical School, Rosalind Franklin University of Medicine and Science North Chicago, IL, USA
| | - Hope Bilyk
- The Nutrition Department, The College of Health Professions, Rosalind Franklin University of Medicine and Science North Chicago, IL, USA
| | - Judith A Potashkin
- The Cellular and Molecular Pharmacology Department, The Chicago Medical School, Rosalind Franklin University of Medicine and Science North Chicago, IL, USA
| |
Collapse
|
32
|
Dagda RK, Das Banerjee T, Janda E. How Parkinsonian toxins dysregulate the autophagy machinery. Int J Mol Sci 2013; 14:22163-89. [PMID: 24217228 PMCID: PMC3856058 DOI: 10.3390/ijms141122163] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 10/28/2013] [Accepted: 10/28/2013] [Indexed: 12/21/2022] Open
Abstract
Since their discovery, Parkinsonian toxins (6-hydroxydopamine, MPP+, paraquat, and rotenone) have been widely employed as in vivo and in vitro chemical models of Parkinson's disease (PD). Alterations in mitochondrial homeostasis, protein quality control pathways, and more recently, autophagy/mitophagy have been implicated in neurotoxin models of PD. Here, we highlight the molecular mechanisms by which different PD toxins dysregulate autophagy/mitophagy and how alterations of these pathways play beneficial or detrimental roles in dopamine neurons. The convergent and divergent effects of PD toxins on mitochondrial function and autophagy/mitophagy are also discussed in this review. Furthermore, we propose new diagnostic tools and discuss how pharmacological modulators of autophagy/mitophagy can be developed as disease-modifying treatments for PD. Finally, we discuss the critical need to identify endogenous and synthetic forms of PD toxins and develop efficient health preventive programs to mitigate the risk of developing PD.
Collapse
Affiliation(s)
- Ruben K. Dagda
- Department of Pharmacology, University of Nevada School of Medicine, Manville Building 18A, Reno, NV 89557, USA; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-775-784-4121; Fax: +1-775-784-1620
| | - Tania Das Banerjee
- Department of Pharmacology, University of Nevada School of Medicine, Manville Building 18A, Reno, NV 89557, USA; E-Mail:
| | - Elzbieta Janda
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, 88100 Cantazaro, Italy; E-Mail:
| |
Collapse
|
33
|
Tovilovic G, Zogovic N, Soskic V, Schrattenholz A, Kostic-Rajacic S, Misirkic-Marjanovic M, Janjetovic K, Vucicevic L, Arsikin K, Harhaji-Trajkovic L, Trajkovic V. Arylpiperazine-mediated activation of Akt protects SH-SY5Y neuroblastoma cells from 6-hydroxydopamine-induced apoptotic and autophagic death. Neuropharmacology 2013; 72:224-35. [DOI: 10.1016/j.neuropharm.2013.04.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 04/11/2013] [Accepted: 04/22/2013] [Indexed: 12/21/2022]
|
34
|
Tobón-Velasco JC, Limón-Pacheco JH, Orozco-Ibarra M, Macías-Silva M, Vázquez-Victorio G, Cuevas E, Ali SF, Cuadrado A, Pedraza-Chaverrí J, Santamaría A. RETRACTED: 6-OHDA-induced apoptosis and mitochondrial dysfunction are mediated by early modulation of intracellular signals and interaction of Nrf2 and NF-κB factors. Toxicology 2013; 304:109-19. [DOI: 10.1016/j.tox.2012.12.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 12/02/2012] [Accepted: 12/17/2012] [Indexed: 11/26/2022]
|
35
|
Weng SJ, Shiue CY, Huang WS, Cheng CY, Huang SY, Li IH, Tao CC, Chou TK, Liao MH, Chang YP, Ma KH. PET imaging of serotonin transporters with 4-[18F]-ADAM in a Parkinsonian rat model. Cell Transplant 2012; 22:1295-305. [PMID: 23127756 DOI: 10.3727/096368912x658683] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This study was undertaken to address the effects of fetal mesencephalic tissue transplantation on the serotonin system in a rat model of Parkinson's disease (PD) while also investigating the usefulness of 4-[18F]-ADAM (a serotonin transporter imaging agent) coupled with micro-PET for imaging serotonin transporters (SERTs). A PD model was induced by unilateral injection of 6-hydroxydopamine (6-OHDA) into the right medial forebrain bundle of the nigrostriatal pathway, while cell transplantation was performed via intrastriatal injection of mesencephalic brain tissue dissected from embryonic (E14) rats. The 4-[18F]-ADAM/micro-PET scanning was performed following both 6-OHDA lesioning and transplantation. Immunohistochemistry (IHC) studies were also performed following the final PET scan, and the results were compared to show a 17-43% decrease in the specific uptake ratio (SUR) and a 23-52% decrease in serotonin transporter immunoreactivity (SERT-ir) within various brain regions on the lesioned side. The number of methamphetamine-induced rotations also decreased significantly at the 4th week postgraft. In addition, striatal SUR and the SERT-ir levels were restored to 77% and 83% 5 weeks postgraft. These results suggest that Parkinson's disease also affects the serotonergic system, while both the dopaminergic and serotonergic systems can be partially restored in a rat model of PD after E14 mesencephalic tissue transplantation. In addition, we have also determined that 4-[18F]-ADAM/micro-PET can be used to detect serotonergic neuron loss, monitor the progress of Parkinson's disease, and oversee the effectiveness of therapy.
Collapse
Affiliation(s)
- Shao-Ju Weng
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Iglesias-González J, Sánchez-Iglesias S, Méndez-Álvarez E, Rose S, Hikima A, Jenner P, Soto-Otero R. Differential toxicity of 6-hydroxydopamine in SH-SY5Y human neuroblastoma cells and rat brain mitochondria: protective role of catalase and superoxide dismutase. Neurochem Res 2012; 37:2150-60. [PMID: 22821477 DOI: 10.1007/s11064-012-0838-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 06/22/2012] [Accepted: 07/12/2012] [Indexed: 11/24/2022]
Abstract
Oxidative stress and mitochondrial dysfunction are two pathophysiological factors often associated with the neurodegenerative process involved in Parkinson's disease (PD). Although, 6-hydroxydopamine (6-OHDA) is able to cause dopaminergic neurodegeneration in experimental models of PD by an oxidative stress-mediated process, the underlying molecular mechanism remains unclear. It has been established that some antioxidant enzymes such as catalase (CAT) and superoxide dismutase (SOD) are often altered in PD, which suggests a potential role of these enzymes in the onset and/or development of this multifactorial syndrome. In this study we have used high-resolution respirometry to evaluate the effect of 6-OHDA on mitochondrial respiration of isolated rat brain mitochondria and the lactate dehydrogenase cytotoxicity assay to assess the percentage of cell death induced by 6-OHDA in human neuroblastoma cell line SH-SY5Y. Our results show that 6-OHDA affects mitochondrial respiration by causing a reduction in both respiratory control ratio (IC(50) = 200 ± 15 nM) and state 3 respiration (IC(50) = 192 ± 17 nM), with no significant effects on state 4(o). An inhibition in the activity of both complex I and V was also observed. 6-OHDA also caused cellular death in human neuroblastoma SH-SY5Y cells (IC(50) = 100 ± 9 μM). Both SOD and CAT have been shown to protect against the toxic effects caused by 6-OHDA on mitochondrial respiration. However, whereas SOD protects against 6-OHDA-induced cellular death, CAT enhances its cytotoxicity. The here reported data suggest that both superoxide anion and hydroperoxyl radical could account for 6-OHDA toxicity. Furthermore, factors reducing the rate of 6-OHDA autoxidation to its p-quinone appear to enhance its cytotoxicity.
Collapse
Affiliation(s)
- Javier Iglesias-González
- Group of Neurochemistry for Parkinson's Disease, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Santiago de Compostela, San Francisco 1, 15782 Santiago de Compostela, Spain
| | | | | | | | | | | | | |
Collapse
|
37
|
Toulouse A, Collins GC, Sullivan AM. Neurotrophic effects of growth/differentiation factor 5 in a neuronal cell line. Neurotox Res 2011; 21:256-65. [PMID: 21858606 DOI: 10.1007/s12640-011-9266-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 08/02/2011] [Accepted: 08/04/2011] [Indexed: 10/17/2022]
Abstract
The neurotrophin growth/differentiation factor 5 (GDF5) is studied as a potential therapeutic agent for Parkinson's disease as it is believed to play a role in the development and maintenance of the nigrostriatal system. Progress in understanding the effects of GDF5 on dopaminergic neurones has been hindered by the use of mixed cell populations derived from primary cultures or in vivo experiments, making it difficult to differentiate between direct and indirect effects of GDF5 treatment on neurones. In an attempt to establish an useful model to study the direct neuronal influence of GDF5, we have characterised the effects of GDF5 on a human neuronal cell line, SH-SY5Y. Our results show that GDF5 has the capability to promote neuronal but not dopaminergic differentiation. We also show that it promotes neuronal survival in vitro following a 6-hydroxydopamine insult. Our results show that application of GDF5 to SH-SY5Y cultures induces the SMAD pathway which could potentially be implicated in the intracellular transmission of GDF5's neurotrophic effects. Overall, our study shows that the SH-SY5Y neuroblastoma cell line provides an excellent neuronal model to study the neurotrophic effects of GDF5.
Collapse
Affiliation(s)
- André Toulouse
- Department of Anatomy and Neuroscience, University College Cork, College Road, Cork, Ireland.
| | | | | |
Collapse
|
38
|
The dopamine transporter is differentially regulated after dopaminergic lesion. Neurobiol Dis 2010; 40:518-30. [DOI: 10.1016/j.nbd.2010.07.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 07/09/2010] [Accepted: 07/22/2010] [Indexed: 11/22/2022] Open
|
39
|
Shi ZH, Nie G, Duan XL, Rouault T, Wu WS, Ning B, Zhang N, Chang YZ, Zhao BL. Neuroprotective mechanism of mitochondrial ferritin on 6-hydroxydopamine-induced dopaminergic cell damage: implication for neuroprotection in Parkinson's disease. Antioxid Redox Signal 2010; 13:783-96. [PMID: 20121342 PMCID: PMC6777976 DOI: 10.1089/ars.2009.3018] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Neuronal iron homeostasis disruption and oxidative stress are closely related to the pathogenesis of Parkinson's disease (PD). Adult iron-regulatory protein 2 knockout (Ireb2(-/-)) mice develop iron accumulation in white matter tracts and nuclei in different brain area and display severe neurodegeneration in Purkinje cells of the cerebrum. Mitochondrial ferritin (MtFt), a newly discovered ferritin, specifically expresses in high energy-consuming cells, including neurons of brain and spinal cord. Interestingly, the decreased expression of MtFt in cerebrum, but not in striatum, matches the differential neurodegeneration pattern in these Ireb2(-/-) mice. To explore its effect on neurodegeneration, the effects of MtFt expression on 6-hydrodopamine (6-OHDA)-induced neuronal damage was examined. The overexpression of MtFt led to a cytosolic iron deficiency in the neuronal cells and significantly prevented the alteration of iron redistribution induced by 6-OHDA. Importantly, MtFt strongly inhibited mitochondrial damage, decreased production of the reactive oxygen species and lipid peroxidation, and dramatically rescued apoptosis by regulating Bcl-2, Bax and caspase-3 pathways. In conclusion, this study demonstrates that MtFt plays an important role in preventing neuronal damage in an 6-OHDA-induced parkinsonian phenotype by maintaining iron homeostasis. Regulation of MtFt expression in neuronal cells may provide a new neuroprotective strategy for PD.
Collapse
Affiliation(s)
- Zhen-Hua Shi
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Habisch HJ, Liebau S, Lenk T, Ludolph AC, Brenner R, Storch A. Neuroectodermally converted human mesenchymal stromal cells provide cytoprotective effects on neural stem cells and inhibit their glial differentiation. Cytotherapy 2010; 12:491-504. [DOI: 10.3109/14653241003649502] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
41
|
Rodriguez-Pallares J, Parga JA, Joglar B, Guerra MJ, Labandeira-Garcia JL. The Mitochondrial ATP-Sensitive Potassium Channel Blocker 5-Hydroxydecanoate Inhibits Toxicity of 6-Hydroxydopamine on Dopaminergic Neurons. Neurotox Res 2009; 15:82-95. [DOI: 10.1007/s12640-009-9010-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Revised: 12/12/2008] [Accepted: 12/17/2008] [Indexed: 12/21/2022]
|
42
|
Effects of calpain inhibition on dopaminergic markers and motor function following intrastriatal 6-hydroxydopamine administration in rats. Neuroscience 2008; 158:558-69. [PMID: 19007862 DOI: 10.1016/j.neuroscience.2008.10.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2008] [Revised: 10/15/2008] [Accepted: 10/18/2008] [Indexed: 11/21/2022]
Abstract
The neurotoxin 6-hydroxydopamine has been widely used to model aspects of Parkinson's disease in rodents, but the mechanisms underlying toxin-induced dopaminergic degeneration and functional impairment have not been fully elucidated. The main aim of the present study was to assess a possible role for calpains in neurochemical and behavioral deficits following unilateral infusion of intrastriatal 6-hydroxydopamine in adult rats. Toxin administration produced a profound dopaminergic denervation, as indicated by a 90-95% reduction in dopamine transporter radiolabeling measured in the caudate-putamen at 2 weeks post-lesion. Treatment with 6-hydroxydopamine also resulted in calpain activation in both caudate-putamen and substantia nigra, as measured by the appearance of calpain-specific spectrin breakdown products. Calpain activation peaked at 24 h after 6-hydroxydopamine infusion and remained elevated at later time points. In contrast, caspase-3-mediated spectrin cleavage subsided within 48 h in both brain areas. In a subsequent experiment, calpain inhibition was achieved by intrastriatal infusion of an adenovirus expressing the endogenous calpain inhibitor, calpastatin. Calpastatin delivery abolished the lesion-induced calpain-mediated spectrin cleavage and alleviated forelimb asymmetries resulting from unilateral intrastriatal 6-hydroxydopamine. Unexpectedly, dopamine transporter and tyrosine hydroxylase labeling revealed significant neuroprotection, not in the nigrostriatal pathway but rather in the ventral tegmental area. These findings support a role for calpain activation in 6-hydroxydopamine-induced degeneration of dopaminergic neurons. However, after near-total dopaminergic depletion, the primary benefit of calpain inhibition may not occur within the nigrostriatal dopaminergic pathway itself.
Collapse
|
43
|
Ma KH, Huang WS, Huang SY, Cheng CY, Chen CY, Shen LH, Liu JC, Fu YK. Imaging serotonin transporters using [123I]ADAM SPECT in a parkinsonian primate model. Appl Radiat Isot 2008; 66:1799-803. [PMID: 18703341 DOI: 10.1016/j.apradiso.2008.06.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 06/02/2008] [Accepted: 06/16/2008] [Indexed: 10/21/2022]
Abstract
Parkinson's disease (PD) affects multiple neurotransmitter systems. The purpose of this study was to investigate differences in the serotonin transport system between normal and parkinsonian monkeys using 2-([2-([di-methylamino]methyl)phenyl]thio)-5-[(123)I] iodophenyl-amine([(123)I]ADAM), a serotonin transporters (SERT) radioligand. The brain single photon emission computed tomography (SPECT) was performed on two normal and one parkinsonian monkey. The parkinsonian monkey was induced by bilateral injection of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle under magnetic resonance imaging (MRI) guidance. Each monkey underwent two [(99m)Tc] TRODAT-1 (a dopamine transporters imaging agent) and two [(123)I] ADAM brain SPECT scans. After a bolus injection of the radioligand, the SPECT data were acquired over 4h using a dual-head gamma camera equipped with ultra-high resolution fan-beam collimators. The striatal uptake of [(99m)Tc]TRODAT-1 was 46% lower in the parkinsonian monkey than those of normal monkeys at 210-240 min post-injection. [(123)I]ADAM uptake in the midbrain of the parkinsonian monkey was comparable to those of the controls. The uptakes of [(123)I]ADAM in the striatum, thalamus, and frontal cortex of the parkinsonian monkey, were 31%, 31%, and 23% lower than those of normal monkeys at 210-240 min post-injection, respectively. Our results suggest that [(123)I]ADAM SPECT has potential for evaluating the serotonin transporter changes in human PD.
Collapse
Affiliation(s)
- Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Neihu, Taipei, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Nagel F, Dohm CP, Bähr M, Wouters FS, Dietz GP. Quantitative evaluation of chaperone activity and neuroprotection by different preparations of a cell-penetrating Hsp70. J Neurosci Methods 2008; 171:226-32. [DOI: 10.1016/j.jneumeth.2008.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 03/12/2008] [Accepted: 03/13/2008] [Indexed: 01/01/2023]
|
45
|
Sabolek M, Mieskes I, Lenk T, Lehmensiek V, Hermann A, Schwarz J, Storch A. Stage-dependent vulnerability of fetal mesencephalic neuroprogenitors towards dopaminergic neurotoxins. Neurotoxicology 2008; 29:714-21. [PMID: 18513801 DOI: 10.1016/j.neuro.2008.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 04/10/2008] [Accepted: 04/16/2008] [Indexed: 12/17/2022]
Abstract
Although extensive knowledge exists on selective vulnerability of dopaminergic neurons against parkinsonism-inducing neurotoxins, there is a complete lack of such data on immature neuroprogenitors. Here we investigated the toxicity of 1-methyl-4-phenylpyridinium (MPP+), 6-hydroxydopamine (6-OHDA) and the free radical generator H2O2 on various developmental stages of predopaminergic mesencephalic neuroprogenitors (mNPCs) to evaluate stage-dependency of selective dopaminergic neurotoxicity. Striatal NPCs (sNPCs) without dopaminergic differentiation potential served as controls. Exposure of both undifferentiated NPCs to MPP+ resulted in concentration-dependent cell death at concentrations of >10 microM after 72 h without differences between both cell types, while 6-OHDA led to relevant cell death at 1000 microM after 24h with significant higher sensitivity of mNPCs compared to sNPCs. H2O2 did not induce relevant cell death in all cell types. In NPC cultures differentiated for 14 days, MPP+ showed enhanced toxicity compared to the undifferentiated counterparts, but no significant differences between both NPC type and differentiation conditions. 6-OHDA showed similar toxicity pattern in differentiated compared to undifferentiated NPCs. By evaluating the toxicity of MPP+ on MAP2ab+ neurons derived from both mNPCs and sNPCs as well as tyrosine hydroxylase (TH)+ dopaminergic cells from mNPCs, we found concentration-dependent cell death of all cell types with no increased vulnerability of TH+ cells. Primary TH+ neurons showed significantly higher vulnerability to MPP+. Together, we demonstrated stage-dependent vulnerability of NPCs towards dopaminergic neurotoxins, but no selective vulnerability of NPC-derived TH+ dopaminergic cells towards MPP+. This cell system seems not suitable as a screening tool for selective dopaminergic toxicity.
Collapse
|
46
|
Abstract
The neurotoxin 6-hydroxydopamine (6-OHDA) continues to constitute a valuable topical tool used chiefly in modeling Parkinson's disease in the rat. The classical method of intracerebral infusion of 6-OHDA involving a massive destruction of nigrostriatal dopaminergic neurons, is largely used to investigate motor and biochemical dysfunctions in Parkinson's disease. Subsequently, more subtle models of partial dopaminergic degeneration have been developed with the aim of revealing finer motor deficits. The present review will examine the main features of 6-OHDA models, namely the mechanisms of neurotoxin-induced neurodegeneration as well as several behavioural deficits and motor dysfunctions, including the priming model, modeled by this means. An overview of the most recent morphological and biochemical findings obtained with the 6-OHDA model will also be provided, particular attention being focused on the newly investigated intracellular mechanisms at the striatal level (e.g., A(2A) and NMDA receptors, PKA, CaMKII, ERK kinases, as well as immediate early genes, GAD67 and peptides). Thanks to studies performed in the 6-OHDA model, all these mechanisms have now been hypothesised to represent the site of pathological dysfunction at cellular level in Parkinson's disease.
Collapse
Affiliation(s)
- Nicola Simola
- Department of Toxicology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | | | | |
Collapse
|
47
|
Kulich SM, Horbinski C, Patel M, Chu CT. 6-Hydroxydopamine induces mitochondrial ERK activation. Free Radic Biol Med 2007; 43:372-83. [PMID: 17602953 PMCID: PMC2023873 DOI: 10.1016/j.freeradbiomed.2007.04.028] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 04/12/2007] [Accepted: 04/13/2007] [Indexed: 01/24/2023]
Abstract
Reactive oxygen species (ROS) are implicated in 6-hydroxydopamine (6-OHDA) injury to catecholaminergic neurons; however, the mechanism(s) are unclear. In addition to ROS generated during autoxidation, 6-OHDA may initiate secondary cellular sources of ROS that contribute to toxicity. Using a neuronal cell line, we found that catalytic metalloporphyrin antioxidants conferred protection if added 1 h after exposure to 6-OHDA, whereas the hydrogen peroxide scavenger catalase failed to protect if added more than 15 min after 6-OHDA. There was a temporal correspondence between loss of protection and loss of the ability of the antioxidant to inhibit 6-OHDA-induced ERK phosphorylation. Time course studies of aconitase inactivation, an indicator of intracellular superoxide, and MitoSOX red, a mitochondria targeted ROS indicator, demonstrate early intracellular ROS followed by a delayed phase of mitochondrial ROS production, associated with phosphorylation of a mitochondrial pool of ERK. Furthermore, on initiation of mitochondrial ROS and ERK activation, 6-OHDA-injured cells became refractory to rescue by metalloporphyrin antioxidants. Together with previous studies showing that inhibition of the ERK pathway confers protection from 6-OHDA toxicity, and that phosphorylated ERK accumulates in mitochondria of degenerating human Parkinson's disease neurons, these studies implicate mitochondrial ERK activation in Parkinsonian oxidative neuronal injury.
Collapse
Affiliation(s)
- Scott M Kulich
- Department of Pathology, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA.
| | | | | | | |
Collapse
|
48
|
Toda T, Nakamura M, Morisawa H, Hirota M. Proteomic identification of oxidative-stress-reporting biomarkers differentially secreted from human neuroblastoma SH-SY5Y cells. ACTA ACUST UNITED AC 2007. [DOI: 10.2198/jelectroph.51.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
49
|
Nakamura M, Yamada M, Ohsawa T, Morisawa H, Nishine T, Nishimura O, Toda T. Phosphoproteomic profiling of human SH-SY5Y neuroblastoma cells during response to 6-hydroxydopamine-induced oxidative stress. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1763:977-89. [PMID: 16949164 DOI: 10.1016/j.bbamcr.2006.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Revised: 07/14/2006] [Accepted: 07/17/2006] [Indexed: 11/19/2022]
Abstract
Dopaminergic neurons are known to be vulnerable to age-related neuronal disorders due to reactive oxygen species (ROS) generated during dopamine metabolism. However, it remains unclear what kinds of proteins are involved in the response to oxidative stress. We examined changes in whole proteins and phosphoproteins in the human dopaminergic neuroblastoma cell line SH-SY5Y under oxidative stress induced by the dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA). Proteins of SH-SY5Y cells at various stages of oxidative stress were separated by two-dimensional gel electrophoresis for comparative analysis. Increase in glutathione-S-transferase pi was detected on SYPRO Ruby-stained gels by computer-aided image analysis. Stress-induced alterations in phosphoproteins were detected by Pro-Q Diamond staining. Elongation factor 2, lamin A/C, T-complex protein 1, and heterogeneous nuclear ribonucleoprotein H3 were identified by MALDI-TOF mass spectrometry as stress-responsive elements.
Collapse
Affiliation(s)
- Megumi Nakamura
- Research Team for Molecular Biomarkers, Proteomic Gerontology Research Unit, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | | | | | | | | | | | | |
Collapse
|
50
|
Storch A, Hwang YI, Bringmann G, Feineis D, Ott S, Brückner R, Schwarz J. Cytotoxicity of chloral-derived β-carbolines is not specific towards neuronal nor dopaminergic cells. J Neural Transm (Vienna) 2006; 113:1895-901. [PMID: 16868795 DOI: 10.1007/s00702-006-0495-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2005] [Accepted: 03/13/2006] [Indexed: 10/24/2022]
Abstract
beta-Carbolines structurally related to the selective dopaminergic neurotoxin 1-methyl-4- phenylpyridinium (MPP(+)) may contribute to dopaminergic neurodegeneration in Parkinson's disease. The chloral-derived mammalian alkaloid derivative 1-trichloromethyl-1,2,3,4-tetrahydro-beta-carboline (TaClo) is formed endogenously by a Pictet-Spengler condensation from the biogenic amine tryptamine (Ta) and the hypnotic aldehyde chloral (Clo). Here we examine the dopaminergic toxicity of TaClo and related compounds by testing their differential cytotoxicities in dopaminergic SH-SY5Y and non-dopaminergic murine Neuro2A neuroblastoma cell lines as well as in heterologous expression systems of the dopamine transporter (DAT) using both HEK-293 and Neuro2A cells. All TaClo derivatives showed significant cytotoxicity in all cell lines after 72 hours with the following rank order of toxic potency: 1-Tribromomethyl-1,2,3,4-tetrahydro-beta-carboline (TaBro) > TaClo > MPP(+) > 1,2,3,4-tetrahydro-beta-carboline (THbetaC) > 2[N]-methyl-TaClo > 2[N]-methyl-THbetaC. In contrast to MPP(+), there was no selectivity towards dopaminergic cells or cells ectopically expressing the DAT in vitro. Our results suggest that TaClo and related analogs are strong cytotoxins without selectivity towards dopaminergic cells.
Collapse
Affiliation(s)
- A Storch
- Department of Neurology, Technical University of Dresden, Dresden, Germany.
| | | | | | | | | | | | | |
Collapse
|