1
|
Xie M, Derks MGN, Koch EHW, van Boven CB, Janlad M, Bagheri B, Xu Z, Kovryzhenko D, van Walree CA, Sobota A, Weingarth M, Wong-Ekkabut J, Karttunen M, Breukink E, Killian JA, Sonnen AFP, Lorent JH. Structure and pH Dependence of Membranolytic Mechanisms by Truncated Oxidized Phospholipids. J Am Chem Soc 2025; 147:9175-9189. [PMID: 40041982 DOI: 10.1021/jacs.4c12543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Membrane lipid oxidation is a universal process that occurs in situations of oxidative stress and is encountered in numerous physiological and pathological situations. Oxidized truncated phospholipids make up a large part of the oxidation products and alter the membrane properties in a way that can lead to cell death. However, the underlying mechanisms are not well understood nor is it clear whether environmental factors, such as pH, can modulate these effects. Using model membranes, we investigate how individual lipid aldehydes and carboxylic acids with truncated acyl chains alter the membrane structure. Our data shows that lipid aldehydes and carboxylic acids have different permeabilization efficiencies towards molecules of varying charge and size and that ΔC9 truncated lipids are usually more efficient in permeabilizing membranes than ΔC5. In terms of physical mechanisms, the ΔC9 truncated lipid carboxylic acid induces permeabilization and membrane curvature in a pH-dependent fashion. This is explained by ionization-dependent exposure of the carboxyl group to the water-bilayer interface, which increases the intrinsic molecular curvature of the oxidized lipid. Conversely, ΔC9 truncated lipid aldehydes and nonionized carboxyls do not induce curved structures but are more efficient in increasing permeability toward larger molecules. We further show that truncated lipids can escape the bilayer and accumulate at interfaces, implying that they might act on neighboring cells. This study indicates that oxidized phospholipids with truncated acyl chains disrupt membrane structure, depending on their specific molecular structure and the pH of the environment, opening a possible route for the design of lipid nanoparticles with pH-dependent drug release.
Collapse
Affiliation(s)
- Min Xie
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Maik G N Derks
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Nuclear Magnetic Resonance Spectroscopy, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Eveline H W Koch
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - C Bjorn van Boven
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Minchakarn Janlad
- Department of Physics, Faculty of Science, Kasetsart University, 50 Ngamwongwan Rd., Chatuchak, Bangkok 10900, Thailand
- Computational Biomodelling Laboratory for Agricultural Science and Technology (CBLAST), Faculty of Science, Kasetsart University, 50 Ngamwongwan Rd., Chatuchak, Bangkok 10900, Thailand
| | - Behnaz Bagheri
- Department of Applied Physics and Science Education, Technical University of Eindhoven, PO Box 513, Eindhoven 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, PO Box 513, Eindhoven 5600 MB, The Netherlands
| | - Zexi Xu
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Daria Kovryzhenko
- Cellular and Molecular Pharmacology, Translational Research from Experimental and Clinical Pharmacology to Treatment Optimization, Louvain Drug Research Institute, UCLouvain, Avenue Mounier 73+1, B1.73.05, B-1200 Brussels, Belgium
| | - Cornelis A van Walree
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- University College Utrecht, Campusplein 1, 3584 ED Utrecht, The Netherlands
| | - Ana Sobota
- Department of Applied Physics and Science Education, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Markus Weingarth
- Nuclear Magnetic Resonance Spectroscopy, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Jirasak Wong-Ekkabut
- Department of Physics, Faculty of Science, Kasetsart University, 50 Ngamwongwan Rd., Chatuchak, Bangkok 10900, Thailand
- Computational Biomodelling Laboratory for Agricultural Science and Technology (CBLAST), Faculty of Science, Kasetsart University, 50 Ngamwongwan Rd., Chatuchak, Bangkok 10900, Thailand
| | - Mikko Karttunen
- Department of Chemistry, Western University, 1151 Richmond Street, London, Ontario N6A 5B7, Canada
- Department of Physics and Astronomy, Western University, 1151 Richmond Street, London, Ontario N6A 3K7, Canada
| | - Eefjan Breukink
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - J Antoinette Killian
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Andreas F P Sonnen
- Pathology Department, University Medical Center Utrecht, Heidelberglaan 100, H04.312, Postbus 85500, 3508 GA Utrecht, The Netherlands
| | - Joseph H Lorent
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Cellular and Molecular Pharmacology, Translational Research from Experimental and Clinical Pharmacology to Treatment Optimization, Louvain Drug Research Institute, UCLouvain, Avenue Mounier 73+1, B1.73.05, B-1200 Brussels, Belgium
| |
Collapse
|
2
|
Xu M, Li T, Liu X, Islam B, Xiang Y, Zou X, Wang J. Mechanism and Clinical Application Prospects of Mitochondrial DNA Single Nucleotide Polymorphism in Neurodegenerative Diseases. Neurochem Res 2024; 50:61. [PMID: 39673588 DOI: 10.1007/s11064-024-04311-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/12/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Mitochondrial dysfunction is well recognized as a critical component of the complicated pathogenesis of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. This review investigates the influence of mitochondrial DNA single nucleotide polymorphisms on mitochondrial function, as well as their role in the onset and progression of these neurodegenerative diseases. Furthermore, the contemporary approaches to mitochondrial regulation in these disorders are discussed. Our objective is to uncover early diagnostic targets and formulate precision medicine strategies for neurodegenerative diseases, thereby offering new paths for preventing and treating these conditions.
Collapse
Affiliation(s)
- Mengying Xu
- Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Tianjiao Li
- Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Xuan Liu
- Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Binish Islam
- Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Yuyue Xiang
- Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Xiyan Zou
- Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Jianwu Wang
- Xiangya School of Public Health, Central South University, Changsha, 410078, China.
| |
Collapse
|
3
|
Wang J, Lu J, Wang Y, Shu Z, Cheng Y, Zhang X, Yu Y, Han J, Zhu Z, Yu N, Wu J. Decrease in walking ability with increased functional connectivity between multiple brain areas in Parkinson's disease: a functional near-infrared spectroscopy study. Front Aging Neurosci 2024; 16:1454598. [PMID: 39539460 PMCID: PMC11557353 DOI: 10.3389/fnagi.2024.1454598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Gait disturbances significantly impact the mobility and quality of life of individuals with Parkinson's disease (PD). This study aims to delve into the cortical mechanisms underlying gait disorders in PD, specifically focusing on the prefrontal cortex (PFC), premotor cortex (PMC), and primary somatosensory cortex (PSC). Objective To compare the functional connectivity of the PFC, PMC, and PSC regions during walking between individuals with PD and healthy controls. Methods The study included 30 individuals with PD (mean age 62.40 ± 7.16 years) and 22 healthy older adults (mean age 60.95 ± 6.34 years). All participants were requested to walk back and forth at a comfortable pace for 30 s over a 10-meter course three times. A mobile functional near-infrared spectroscopy (fNIRS) system was employed to evaluate the oxyhemoglobin concentration change (∆HbO2). To assess the interactions between the PFC, PMC, and PSC, the connectivity strength between different fNIRS channels was computed. Results Individuals with PD in the off-state exhibited significantly decreased walking speed and shorter stride length compared to the healthy controls. For six brain regions including the left (L) and right (R) PFC, PMC, and PSC, no significant differences in functional connectivity within each region were found between the PD and control groups. However, when it comes to the functional connectivity between every two regions, the PD group exhibited stronger functional connectivity than the control group in the LPFC-LPMC, LPFC-RPMC, LPFC-LPSC, RPFC-LPMC, RPFC-LPSC, LPMC-LPSC, LPMC-RPSC, and RPMC-RPSC. Positive correlations were found between gait performance (speed and stride length) and functional connectivity within the RPMC as well as between the RPMC and the RPSC. Conclusion Individuals with PD exhibit notable gait disturbances and increased functional connectivity in brain regions responsible for sensorimotor integration and motor function in their off-state. Strengthening the functional connectivity within the RPMC and between the RPMC and the RPSC could be a potential target for future treatments of gait impairments in PD.
Collapse
Affiliation(s)
- Jin Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Jiewei Lu
- College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Yue Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhilin Shu
- College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Yuanyuan Cheng
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Rehabilitation Medicine, Tianjin Huanhu Hospital, Tianjin, China
| | - Xinyuan Zhang
- College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Yang Yu
- Department of Rehabilitation Medicine, Tianjin Huanhu Hospital, Tianjin, China
| | - Jianda Han
- College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Zhizhong Zhu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Rehabilitation Medicine, Tianjin Huanhu Hospital, Tianjin, China
| | - Ningbo Yu
- College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Jialing Wu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin, China
| |
Collapse
|
4
|
Scholefield M, Church SJ, Xu J, Cooper GJS. Metallomic analysis of brain tissues distinguishes between cases of dementia with Lewy bodies, Alzheimer's disease, and Parkinson's disease dementia. Front Neurosci 2024; 18:1412356. [PMID: 38988772 PMCID: PMC11233441 DOI: 10.3389/fnins.2024.1412356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024] Open
Abstract
Background Dementia with Lewy bodies (DLB) can be difficult to distinguish from Alzheimer's disease (AD) and Parkinson's disease dementia (PDD) at different stages of its progression due to some overlaps in the clinical and neuropathological presentation of these conditions compared with DLB. Metallomic changes have already been observed in the AD and PDD brain-including widespread decreases in Cu levels and more localised alterations in Na, K, Mn, Fe, Zn, and Se. This study aimed to determine whether these metallomic changes appear in the DLB brain, and how the metallomic profile of the DLB brain appears in comparison to the AD and PDD brain. Methods Brain tissues from ten regions of 20 DLB cases and 19 controls were obtained. The concentrations of Na, Mg, K, Ca, Zn, Fe, Mn, Cu, and Se were determined using inductively coupled plasma-mass spectrometry (ICP-MS). Case-control differences were evaluated using Mann-Whitney U tests. Results were compared with those previously obtained from AD and PDD brain tissue, and principal component analysis (PCA) plots were created to determine whether cerebral metallomic profiles could distinguish DLB from AD or PDD metallomic profiles. Results Na was increased and Cu decreased in four and five DLB brain regions, respectively. More localised alterations in Mn, Ca, Fe, and Se were also identified. Despite similarities in Cu changes between all three diseases, PCA plots showed that DLB cases could be readily distinguished from AD cases using data from the middle temporal gyrus, primary visual cortex, and cingulate gyrus, whereas DLB and PDD cases could be clearly separated using data from the primary visual cortex alone. Conclusion Despite shared alterations in Cu levels, the post-mortem DLB brain shows very few other similarities with the metallomic profile of the AD or PDD brain. These findings suggest that while Cu deficiencies appear common to all three conditions, metal alterations otherwise differ between DLB and PDD/AD. These findings can contribute to our understanding of the underlying pathogenesis of these three diseases; if these changes can be observed in the living human brain, they may also contribute to the differential diagnosis of DLB from AD and/or PDD.
Collapse
Affiliation(s)
- Melissa Scholefield
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stephanie J Church
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Jingshu Xu
- Faculty of Science, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Garth J S Cooper
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Faculty of Science, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
5
|
Kelty TJ, Taylor CL, Wieschhaus NE, Thorne PK, Amin AR, Mueller CM, Olver TD, Tharp DL, Emter CA, Caulk AW, Rector RS. Western diet-induced obesity results in brain mitochondrial dysfunction in female Ossabaw swine. Front Mol Neurosci 2023; 16:1320879. [PMID: 38163062 PMCID: PMC10755880 DOI: 10.3389/fnmol.2023.1320879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
Diet-induced obesity is implicated in the development of a variety of neurodegenerative disorders. Concurrently, the loss of mitochondrial Complex I protein or function is emerging as a key phenotype across an array of neurodegenerative disorders. Therefore, the objective of this study was to determine if Western diet (WD) feeding in swine [carbohydrate = 40.8% kCal (17.8% of total calories from high fructose corn syrup), protein = 16.2% kcal, fat = 42.9% kCal, and 2% cholesterol] would result in Complex I syndrome pathology. To characterize the effects of WD-induced obesity on brain mitochondria in swine, high resolution respirometry measurements from isolated brain mitochondria, oxidative phosphorylation Complex expression, and indices of oxidative stress and mitochondrial biogenesis were assessed in female Ossabaw swine fed a WD for 6-months. In line with Complex I syndrome, WD feeding severely reduced State 3 Complex I, State 3 Complex I and II, and uncoupled mitochondrial respiration in the hippocampus and prefrontal cortex (PFC). State 3 Complex I mitochondrial respiration in the PFC inversely correlated with serum total cholesterol. WD feeding also significantly reduced protein expression of oxidative phosphorylation Complexes I-V in the PFC. WD feeding significantly increased markers of antioxidant defense and mitochondrial biogenesis in the hippocampi and PFC. These data suggest WD-induced obesity may contribute to Complex I syndrome pathology by increasing oxidative stress, decreasing oxidative phosphorylation Complex protein expression, and reducing brain mitochondrial respiration. Furthermore, these findings provide mechanistic insight into the clinical link between obesity and mitochondrial Complex I related neurodegenerative disorders.
Collapse
Affiliation(s)
- Taylor J. Kelty
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Chris L. Taylor
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | | | - Pamela K. Thorne
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Amira R. Amin
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Christina M. Mueller
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - T. Dylan Olver
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Darla L. Tharp
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Craig A. Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | | | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
6
|
Readman MR, Wan F, Fairman I, Linkenauger SA, Crawford TJ, Plack CJ. Is Hearing Loss a Risk Factor for Idiopathic Parkinson's Disease? An English Longitudinal Study of Ageing Analysis. Brain Sci 2023; 13:1196. [PMID: 37626551 PMCID: PMC10452744 DOI: 10.3390/brainsci13081196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Observations that hearing loss is a substantial risk factor for dementia may be accounted for by a common pathology. Mitochondrial oxidative stress and alterations in α-synuclein pathology may be common pathology candidates. Crucially, these candidate pathologies are implicated in Parkinson's disease (PD). Consequently, hearing loss may be a risk factor for PD. Subsequently, this prospective cohort study of the English Longitudinal Study of Ageing examines whether hearing loss is a risk factor for PD longitudinally. Participants reporting self-reported hearing capabilities and no PD diagnosis prior to entry (n = 14,340) were used. A joint longitudinal and survival model showed that during a median follow up of 10 years (SD = 4.67 years) increased PD risk (p < 0.001), but not self-reported hearing capability (p = 0.402). Additionally, an exploratory binary logistic regression modelling the influence of hearing loss identified using a screening test (n = 4812) on incident PD indicated that neither moderate (p = 0.794), nor moderately severe/severe hearing loss (p = 0.5210), increased PD risk, compared with normal hearing. Whilst discrepancies with prior literature may suggest a neurological link between hearing loss and PD, further large-scale analyses using clinically derived hearing loss are needed.
Collapse
Affiliation(s)
- Megan Rose Readman
- Department of Psychology, Lancaster University, Lancaster LA1 4YW, UK
- Department of Primary Care and Mental Health, The University of Liverpool, Liverpool L69 3BX, UK
- NIHR ARC NWC, Liverpool L7 8XP, UK
| | - Fang Wan
- Department of Mathematics and Statistics, Lancaster University, Lancaster LA1 4YW, UK
| | - Ian Fairman
- Public Advisor, Associated with Lancaster University Psychology Department, Lancaster LA1 4YF, UK
| | | | | | - Christopher J. Plack
- Department of Psychology, Lancaster University, Lancaster LA1 4YW, UK
- Manchester Centre for Audiology and Deafness, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
7
|
Llido JP, Jayanti S, Tiribelli C, Gazzin S. Bilirubin and Redox Stress in Age-Related Brain Diseases. Antioxidants (Basel) 2023; 12:1525. [PMID: 37627520 PMCID: PMC10451892 DOI: 10.3390/antiox12081525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Cellular redox status has a crucial role in brain physiology, as well as in pathologic conditions. Physiologic senescence, by dysregulating cellular redox homeostasis and decreasing antioxidant defenses, enhances the central nervous system's susceptibility to diseases. The reduction of free radical accumulation through lifestyle changes, and the supplementation of antioxidants as a prophylactic and therapeutic approach to increase brain health, are strongly suggested. Bilirubin is a powerful endogenous antioxidant, with more and more recognized roles as a biomarker of disease resistance, a predictor of all-cause mortality, and a molecule that may promote health in adults. The alteration of the expression and activity of the enzymes involved in bilirubin production, as well as an altered blood bilirubin level, are often reported in neurologic conditions and neurodegenerative diseases (together denoted NCDs) in aging. These changes may predict or contribute both positively and negatively to the diseases. Understanding the role of bilirubin in the onset and progression of NCDs will be functional to consider the benefits vs. the drawbacks and to hypothesize the best strategies for its manipulation for therapeutic purposes.
Collapse
Affiliation(s)
- John Paul Llido
- Liver Brain Unit “Rita Moretti”, Italian Liver Foundation, Bldg. Q, AREA Science Park, Basovizza, 34149 Trieste, Italy; (J.P.L.); or (S.J.); (S.G.)
- Department of Science and Technology, Philippine Council for Health Research and Development, Bicutan, Taguig City 1631, Philippines
- Department of Life Sciences, University of Trieste, 34139 Trieste, Italy
| | - Sri Jayanti
- Liver Brain Unit “Rita Moretti”, Italian Liver Foundation, Bldg. Q, AREA Science Park, Basovizza, 34149 Trieste, Italy; (J.P.L.); or (S.J.); (S.G.)
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16911, Indonesia
| | - Claudio Tiribelli
- Liver Brain Unit “Rita Moretti”, Italian Liver Foundation, Bldg. Q, AREA Science Park, Basovizza, 34149 Trieste, Italy; (J.P.L.); or (S.J.); (S.G.)
| | - Silvia Gazzin
- Liver Brain Unit “Rita Moretti”, Italian Liver Foundation, Bldg. Q, AREA Science Park, Basovizza, 34149 Trieste, Italy; (J.P.L.); or (S.J.); (S.G.)
| |
Collapse
|
8
|
Kamienieva I, Charzyńska A, Duszyński J, Malińska D, Szczepanowska J. In search for mitochondrial biomarkers of Parkinson's disease: Findings in parkin-mutant human fibroblasts. Biochim Biophys Acta Mol Basis Dis 2023:166787. [PMID: 37302428 DOI: 10.1016/j.bbadis.2023.166787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
Most cases of Parkinson's disease (PD) are idiopathic, with unknown aetiology and genetic background. However, approximately 10 % of cases are caused by defined genetic mutations, among which mutations in the parkin gene are the most common. There is increasing evidence of the involvement of mitochondrial dysfunction in the development of both idiopathic and genetic PD. However, the data on mitochondrial changes reported by different studies are inconsistent, which can reflect the variability in genetic background of the disease. Mitochondria, as a plastic and dynamic organelles, are the first place in the cell to respond to external and internal stress. In this work, we characterized mitochondrial function and dynamics (network morphology and turnover regulation) in primary fibroblasts from PD patients with parkin mutations. We performed clustering analysis of the obtained data to compare the profiles of mitochondrial parameters in PD patients and healthy donors. This allowed to extract the features characteristic for PD patients fibroblasts, which were a smaller and less complex mitochondrial network and decreased levels of mitochondrial biogenesis regulators and mitophagy mediators. The approach we used allowed a comprehensive characteristics of elements common for mitochondrial dynamics remodelling accompanying pathogenic mutation. This may be helpful in the deciphering key pathomechanisms of the PD disease.
Collapse
Affiliation(s)
- Iryna Kamienieva
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, ul. Pasteura 3, 02-093 Warszawa, Poland
| | - Agata Charzyńska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, ul. Pasteura 3, 02-093 Warszawa, Poland
| | - Jerzy Duszyński
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, ul. Pasteura 3, 02-093 Warszawa, Poland
| | - Dominika Malińska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, ul. Pasteura 3, 02-093 Warszawa, Poland.
| | - Joanna Szczepanowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, ul. Pasteura 3, 02-093 Warszawa, Poland.
| |
Collapse
|
9
|
Otsuka T, Matsui H. Fish Models for Exploring Mitochondrial Dysfunction Affecting Neurodegenerative Disorders. Int J Mol Sci 2023; 24:ijms24087079. [PMID: 37108237 PMCID: PMC10138900 DOI: 10.3390/ijms24087079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Neurodegenerative disorders are characterized by the progressive loss of neuronal structure or function, resulting in memory loss and movement disorders. Although the detailed pathogenic mechanism has not been elucidated, it is thought to be related to the loss of mitochondrial function in the process of aging. Animal models that mimic the pathology of a disease are essential for understanding human diseases. In recent years, small fish have become ideal vertebrate models for human disease due to their high genetic and histological homology to humans, ease of in vivo imaging, and ease of genetic manipulation. In this review, we first outline the impact of mitochondrial dysfunction on the progression of neurodegenerative diseases. Then, we highlight the advantages of small fish as model organisms, and present examples of previous studies regarding mitochondria-related neuronal disorders. Lastly, we discuss the applicability of the turquoise killifish, a unique model for aging research, as a model for neurodegenerative diseases. Small fish models are expected to advance our understanding of the mitochondrial function in vivo, the pathogenesis of neurodegenerative diseases, and be important tools for developing therapies to treat diseases.
Collapse
Affiliation(s)
- Takayoshi Otsuka
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| |
Collapse
|
10
|
Ahmad F, Ramamorthy S, Areeshi MY, Ashraf GM, Haque S. Isolated Mitochondrial Preparations and In organello Assays: A Powerful and Relevant Ex vivo Tool for Assessment of Brain (Patho)physiology. Curr Neuropharmacol 2023; 21:1433-1449. [PMID: 36872352 PMCID: PMC10324330 DOI: 10.2174/1570159x21666230303123555] [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: 05/16/2022] [Revised: 10/30/2022] [Accepted: 12/29/2022] [Indexed: 03/07/2023] Open
Abstract
Mitochondria regulate multiple aspects of neuronal development, physiology, plasticity, and pathology through their regulatory roles in bioenergetic, calcium, redox, and cell survival/death signalling. While several reviews have addressed these different aspects, a comprehensive discussion focussing on the relevance of isolated brain mitochondria and their utilities in neuroscience research has been lacking. This is relevant because the employment of isolated mitochondria rather than their in situ functional evaluation, offers definitive evidence of organelle-specificity, negating the interference from extra mitochondrial cellular factors/signals. This mini-review was designed primarily to explore the commonly employed in organello analytical assays for the assessment of mitochondrial physiology and its dysfunction, with a particular focus on neuroscience research. The authors briefly discuss the methodologies for biochemical isolation of mitochondria, their quality assessment, and cryopreservation. Further, the review attempts to accumulate the key biochemical protocols for in organello assessment of a multitude of mitochondrial functions critical for neurophysiology, including assays for bioenergetic activity, calcium and redox homeostasis, and mitochondrial protein translation. The purpose of this review is not to examine each and every method or study related to the functional assessment of isolated brain mitochondria, but rather to assemble the commonly used protocols of in organello mitochondrial research in a single publication. The hope is that this review will provide a suitable platform aiding neuroscientists to choose and apply the required protocols and tools to address their particular mechanistic, diagnostic, or therapeutic question dealing within the confines of the research area of mitochondrial patho-physiology in the neuronal perspective.
Collapse
Affiliation(s)
- Faraz Ahmad
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore, 632014, India
| | - Siva Ramamorthy
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore, 632014, India
| | - Mohammed Y. Areeshi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Jazan University, Jazan, 45142, Saudi Arabia
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Ghulam Md. Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
11
|
The Proteome of Neuromelanin Granules in Dementia with Lewy Bodies. Cells 2022; 11:cells11223538. [PMID: 36428966 PMCID: PMC9688080 DOI: 10.3390/cells11223538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
Neuromelanin granules (NMGs) are organelle-like structures present in the human substantia nigra pars compacta. In addition to neuromelanin, NMGs contain proteins, lipids and metals. As NMG-containing dopaminergic neurons are preferentially lost in Parkinson's disease and dementia with Lewy bodies (DLB), it is assumed that NMGs may play a role in neurodegenerative processes. Until now, this role is not completely understood and needs further investigation. We therefore set up an exploratory proteomic study to identify differences in the proteomic profile of NMGs from DLB patients (n = 5) compared to healthy controls (CTRL, n = 5). We applied a laser microdissection and mass-spectrometry-based approach, in which we used targeted mass spectrometric experiments for validation. In NMG-surrounding (SNSurr.) tissue of DLB patients, we found evidence for ongoing oxidative damage and an impairment of protein degradation. As a potentially disease-related mechanism, we found α-synuclein and protein S100A9 to be enriched in NMGs of DLB cases, while the abundance of several ribosomal proteins was significantly decreased. As S100A9 is known to be able to enhance the formation of toxic α-synuclein fibrils, this finding points towards an involvement of NMGs in pathogenesis, however the exact role of NMGs as either neuroprotective or neurotoxic needs to be further investigated. Nevertheless, our study provides evidence for an impairment of protein degradation, ongoing oxidative damage and accumulation of potentially neurotoxic protein aggregates to be central mechanisms of neurodegeneration in DLB.
Collapse
|
12
|
Schmidt S, Luecken MD, Trümbach D, Hembach S, Niedermeier KM, Wenck N, Pflügler K, Stautner C, Böttcher A, Lickert H, Ramirez-Suastegui C, Ahmad R, Ziller MJ, Fitzgerald JC, Ruf V, van de Berg WDJ, Jonker AJ, Gasser T, Winner B, Winkler J, Vogt Weisenhorn DM, Giesert F, Theis FJ, Wurst W. Primary cilia and SHH signaling impairments in human and mouse models of Parkinson's disease. Nat Commun 2022; 13:4819. [PMID: 35974013 PMCID: PMC9380673 DOI: 10.1038/s41467-022-32229-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 07/21/2022] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) as a progressive neurodegenerative disorder arises from multiple genetic and environmental factors. However, underlying pathological mechanisms remain poorly understood. Using multiplexed single-cell transcriptomics, we analyze human neural precursor cells (hNPCs) from sporadic PD (sPD) patients. Alterations in gene expression appear in pathways related to primary cilia (PC). Accordingly, in these hiPSC-derived hNPCs and neurons, we observe a shortening of PC. Additionally, we detect a shortening of PC in PINK1-deficient human cellular and mouse models of familial PD. Furthermore, in sPD models, the shortening of PC is accompanied by increased Sonic Hedgehog (SHH) signal transduction. Inhibition of this pathway rescues the alterations in PC morphology and mitochondrial dysfunction. Thus, increased SHH activity due to ciliary dysfunction may be required for the development of pathoetiological phenotypes observed in sPD like mitochondrial dysfunction. Inhibiting overactive SHH signaling may be a potential neuroprotective therapy for sPD.
Collapse
Affiliation(s)
- Sebastian Schmidt
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
| | - Malte D Luecken
- Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Sina Hembach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
| | - Kristina M Niedermeier
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
| | - Nicole Wenck
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
| | - Klaus Pflügler
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
| | - Constantin Stautner
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Ciro Ramirez-Suastegui
- Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Ruhel Ahmad
- Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Michael J Ziller
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
| | - Julia C Fitzgerald
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Viktoria Ruf
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität Munich, Feodor-Lynen-Str. 23, 81377, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Wilma D J van de Berg
- Section Clinical Neuroanatomy and Biobanking (CNAB), Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HV, Amsterdam, The Netherlands
| | - Allert J Jonker
- Section Clinical Neuroanatomy and Biobanking (CNAB), Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HV, Amsterdam, The Netherlands
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glückstrasse 6, 91054, Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054, Erlangen, Germany
| | - Daniela M Vogt Weisenhorn
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
| | - Florian Giesert
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- Department of Mathematics, Technische Universität München, Boltzmannstraße 3, 85748, Garching bei München, Germany.
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) site Munich, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
| |
Collapse
|
13
|
A New Zebrafish Model to Measure Neuronal α-Synuclein Clearance In Vivo. Genes (Basel) 2022; 13:genes13050868. [PMID: 35627253 PMCID: PMC9141618 DOI: 10.3390/genes13050868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023] Open
Abstract
The accumulation and aggregation of α-synuclein (α-SYN) is a common characteristic of synucleinopathies, such as Parkinson's Disease (PD), Dementia with Lewy Bodies (DLB) or Multiple System Atrophy (MSA). Multiplications of the wildtype gene of α-SYN (SNCA) and most point mutations make α-SYN more aggregate-prone, and are associated with mitochondrial defects, trafficking obstruction, and impaired proteostasis, which contribute to elevated neuronal death. Here, we present new zebrafish models expressing either human wildtype (wt), or A53T mutant, α-SYN that recapitulate the above-mentioned hallmarks of synucleinopathies. The appropriate clearance of toxic α-SYN has been previously shown to play a key role in maintaining cell homeostasis and survival. However, the paucity of models to investigate α-SYN degradation in vivo limits our understanding of this process. Based on our recently described imaging method for measuring tau protein clearance in neurons in living zebrafish, we fused human SNCA to the photoconvertible protein Dendra2 which enabled analyses of wt and A53T α-SYN clearance kinetics in vivo. Moreover, these zebrafish models can be used to investigate the kinetics of α-SYN aggregation and to study the mechanisms, and potential new targets, controlling the clearance of both soluble and aggregated α-SYN.
Collapse
|
14
|
Soman SK, Dagda RK. Role of Cleaved PINK1 in Neuronal Development, Synaptogenesis, and Plasticity: Implications for Parkinson's Disease. Front Neurosci 2021; 15:769331. [PMID: 34795558 PMCID: PMC8593325 DOI: 10.3389/fnins.2021.769331] [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: 09/01/2021] [Accepted: 10/11/2021] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial dysfunction plays a significant role in the pathogenesis of Parkinson's disease (PD). Consistent with this concept, loss of function mutations in the serine/threonine kinase- PINK1 (PTEN-induced putative kinase-1) causes autosomal recessive early onset PD. While the functional role of f-PINK1 (full-length PINK1) in clearing dysfunctional mitochondria via mitophagy is extensively documented, our understanding of specific physiological roles that the non-mitochondrial pool of PINK1 imparts in neurons is more limited. PINK1 is proteolytically processed in the intermembrane space and matrix of the mitochondria into functional cleaved products (c-PINK1) that are exported to the cytosol. While it is clear that posttranslational processing of PINK1 depends on the mitochondria's oxidative state and structural integrity, the functional roles of c-PINK1 in modulating neuronal functions are poorly understood. Here, we review the diverse roles played by c-PINK1 in modulating various neuronal functions. Specifically, we describe the non-canonical functional roles of PINK1, including but not limited to: governing mitochondrial movement, neuronal development, neuronal survival, and neurogenesis. We have published that c-PINK1 stimulates neuronal plasticity and differentiation via the PINK1-PKA-BDNF signaling cascade. In addition, we provide insight into how mitochondrial membrane potential-dependent processing of PINK1 confers conditional retrograde signaling functions to PINK1. Further studies delineating the role of c-PINK1 in neurons would increase our understanding regarding the role played by PINK1 in PD pathogenesis.
Collapse
Affiliation(s)
- Smijin K Soman
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, NV, United States
| | - Ruben K Dagda
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
15
|
Protective effects of chrysin against the neurotoxicity induced by aluminium: In vitro and in vivo studies. Toxicology 2021; 465:153033. [PMID: 34774662 DOI: 10.1016/j.tox.2021.153033] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/04/2021] [Accepted: 11/09/2021] [Indexed: 12/23/2022]
Abstract
Chronic exposure to aluminium (Al) can contribute to the progression of several neurological and neurodegenerative diseases. Al is a metal that promotes oxidative damage leading to neuronal death in different brain regions with behavior, cognition, and memory deficits. Chrysin is a flavonoid found mainly in honey, passion fruit, and propolis with antioxidant, anti-inflammatory, and cytoprotective properties. In this study, we used an integrated approach of in vitro and in vivo studies to evaluate the antioxidant and neuroprotective effects of chrysin against the neurotoxicity elicited by aluminium chloride (AlCl3). In in vitro studies, chrysin (5 μM) showed the ability to counteract the early oxidative stress elicited by tert-butyl hydroperoxide, an oxidant that mimics the lipid peroxidation and Fenton reaction in presence of AlCl3 as well as the late necrotic death triggered by AlCl3 in neuronal SH-SY5Y cells. In vivo studies in a mouse model of neurotoxicity induced by chronic exposure to AlCl3 (100 mg/kg/day) for ninety days then corroborated the antioxidant and neuroprotective effect of chrysin (10, 30, and 100 mg/kg/day) using the oral route. In particular, chrysin reduced the cognitive impairment induced by AlCl3 as well as normalized the acetylcholinesterase and butyrylcholinesterase activities in the hippocampus. In parallel, chrysin counteracted the oxidative damage, in terms of lipid peroxidation, protein carbonylation, catalase, and superoxide dismutase impairment, in the brain cortex and hippocampus. Lastly, necrotic cells frequency in the same brain regions was also decreased by chrysin. These results highlight the ability of chrysin to prevent the neurotoxic effects associated with chronic exposure to Al and suggest its potential use as a food supplement for brain health.
Collapse
|
16
|
Shin EJ, Jeong JH, Hwang Y, Sharma N, Dang DK, Nguyen BT, Nah SY, Jang CG, Bing G, Nabeshima T, Kim HC. Methamphetamine-induced dopaminergic neurotoxicity as a model of Parkinson's disease. Arch Pharm Res 2021; 44:668-688. [PMID: 34286473 DOI: 10.1007/s12272-021-01341-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/06/2021] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, 900000, Can Tho City, Vietnam
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, 05029, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Guoying Bing
- Department of Neuroscience, College of Medicine, University of Kentucky, KY, 40536, Lexington, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Science, Fujita Health University, 470-1192, Toyoake, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea. .,Neuropsychopharmacology & Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.
| |
Collapse
|
17
|
Gries M, Christmann A, Schulte S, Weyland M, Rommel S, Martin M, Baller M, Röth R, Schmitteckert S, Unger M, Liu Y, Sommer F, Mühlhaus T, Schroda M, Timmermans JP, Pintelon I, Rappold GA, Britschgi M, Lashuel H, Menger MD, Laschke MW, Niesler B, Schäfer KH. Parkinson mice show functional and molecular changes in the gut long before motoric disease onset. Mol Neurodegener 2021; 16:34. [PMID: 34078425 PMCID: PMC8170976 DOI: 10.1186/s13024-021-00439-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There is increasing evidence that Parkinson's disease (PD) might start in the gut, thus involving and compromising also the enteric nervous system (ENS). At the clinical onset of the disease the majority of dopaminergic neurons in the midbrain is already destroyed, so that the lack of early biomarkers for the disease represents a major challenge for developing timely treatment interventions. Here, we use a transgenic A30P-α-synuclein-overexpressing PD mouse model to identify appropriate candidate markers in the gut before hallmark symptoms begin to manifest. METHODS Based on a gait analysis and striatal dopamine levels, we defined 2-month-old A30P mice as pre-symptomatic (psA30P), since they are not showing any motoric impairments of the skeletal neuromuscular system and no reduced dopamine levels, but an intestinal α-synuclein pathology. Mice at this particular age were further used to analyze functional and molecular alterations in both, the gastrointestinal tract and the ENS, to identify early pathological changes. We examined the gastrointestinal motility, the molecular composition of the ENS, as well as the expression of regulating miRNAs. Moreover, we applied A30P-α-synuclein challenges in vitro to simulate PD in the ENS. RESULTS A retarded gut motility and early molecular dysregulations were found in the myenteric plexus of psA30P mice. We found that i.e. neurofilament light chain, vesicle-associated membrane protein 2 and calbindin 2, together with the miRNAs that regulate them, are significantly altered in the psA30P, thus representing potential biomarkers for early PD. Many of the dysregulated miRNAs found in the psA30P mice are reported to be changed in PD patients as well, either in blood, cerebrospinal fluid or brain tissue. Interestingly, the in vitro approaches delivered similar changes in the ENS cultures as seen in the transgenic animals, thus confirming the data from the mouse model. CONCLUSIONS These findings provide an interesting and novel approach for the identification of appropriate biomarkers in men.
Collapse
Affiliation(s)
- Manuela Gries
- Department of Informatics and Microsystems and Technology, University of Applied Science Kaiserslautern, Working Group Enteric Nervous System, 66482, Zweibrücken, Germany
| | - Anne Christmann
- Department of Informatics and Microsystems and Technology, University of Applied Science Kaiserslautern, Working Group Enteric Nervous System, 66482, Zweibrücken, Germany
| | - Steven Schulte
- Department of Informatics and Microsystems and Technology, University of Applied Science Kaiserslautern, Working Group Enteric Nervous System, 66482, Zweibrücken, Germany
| | - Maximilian Weyland
- Department of Informatics and Microsystems and Technology, University of Applied Science Kaiserslautern, Working Group Enteric Nervous System, 66482, Zweibrücken, Germany
| | - Stephanie Rommel
- Department of Informatics and Microsystems and Technology, University of Applied Science Kaiserslautern, Working Group Enteric Nervous System, 66482, Zweibrücken, Germany
| | - Monika Martin
- Department of Informatics and Microsystems and Technology, University of Applied Science Kaiserslautern, Working Group Enteric Nervous System, 66482, Zweibrücken, Germany
| | - Marko Baller
- Department of Informatics and Microsystems and Technology, University of Applied Science Kaiserslautern, Working Group Enteric Nervous System, 66482, Zweibrücken, Germany
| | - Ralph Röth
- Department of Human Molecular Genetics, University of Heidelberg, 69120, Heidelberg, Germany
| | - Stefanie Schmitteckert
- Department of Human Molecular Genetics, University of Heidelberg, 69120, Heidelberg, Germany
| | - Marcus Unger
- Department of Neurology, Saarland University, 66421, Homburg, Germany
| | - Yang Liu
- Department of Neurology, Saarland University, 66421, Homburg, Germany
| | - Frederik Sommer
- Molecular Biotechnology and Systems Biology, University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Timo Mühlhaus
- Computational Systems Biology, University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Michael Schroda
- Molecular Biotechnology and Systems Biology, University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610, Antwerp, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610, Antwerp, Belgium
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, University of Heidelberg, 69120, Heidelberg, Germany
- Interdisciplinary Center of Neuroscience, 69120, Heidelberg, Germany
| | - Markus Britschgi
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Medicine Area, Neuroscience Discovery, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Hilal Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, 66421, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, 66421, Homburg, Germany
| | - Beate Niesler
- Department of Human Molecular Genetics, University of Heidelberg, 69120, Heidelberg, Germany
| | - Karl-Herbert Schäfer
- Department of Informatics and Microsystems and Technology, University of Applied Science Kaiserslautern, Working Group Enteric Nervous System, 66482, Zweibrücken, Germany.
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany.
| |
Collapse
|
18
|
Subrahmanian N, LaVoie MJ. Is there a special relationship between complex I activity and nigral neuronal loss in Parkinson's disease? A critical reappraisal. Brain Res 2021; 1767:147434. [PMID: 33745923 PMCID: PMC9520341 DOI: 10.1016/j.brainres.2021.147434] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/25/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease manifesting both motor and non-motor symptoms. The motor features are generally ascribed to the selective loss of dopamine neurons within the substantia nigra pars compacta. While the precise etiology of PD remains elusive, multiple genetic and environmental elements have emerged as contributing factors. The discovery of MPTP-induced parkinsonism directed intense inquiry towards mitochondrial pathways, with a specific focus on mitochondrial complex I. Consisting of more than 40 subunits, complex I is the first enzyme of the electron transport chain that is required for mitochondrial ATP production. In this review, we present a critical analysis of studies assessing the prevalence and specificity of mitochondrial complex I deficiency in PD. In addition, we take the novel view of incorporating the features of genetically-defined bona fide complex I disorders and the prevalence of nigral involvement in such cases. Through this innovative bi-directional view, we consider both complex I changes in a disease of the substantia nigra and nigral changes in diseases of complex I. We assess the strength of association between nigral cell loss and complex I deficits, as well as the oft under-appreciated heterogeneity of complex I deficiency disorders and the variability of the PD data.
Collapse
Affiliation(s)
- Nitya Subrahmanian
- Department of Neurology, University of Florida, Gainesville, FL 32610, USA
| | - Matthew J LaVoie
- Department of Neurology, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
19
|
Modelling Parkinson's Disease: iPSCs towards Better Understanding of Human Pathology. Brain Sci 2021; 11:brainsci11030373. [PMID: 33799491 PMCID: PMC8000082 DOI: 10.3390/brainsci11030373] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s Disease (PD) is a chronic neurodegenerative disorder characterized by motor and non-motor symptoms, among which are bradykinesia, rigidity, tremor as well as mental symptoms such as dementia. The underlying cause of Parkinson disease is degeneration of dopaminergic neurons. It has been challenging to develop an efficient animal model to accurately represent the complex phenotypes found with PD. However, it has become possible to recapitulate the myriad of phenotypes underlying the PD pathology by using human induced pluripotent stem cell (iPSC) technology. Patient-specific iPSC-derived dopaminergic neurons are available and present an opportunity to study many aspects of the PD phenotypes in a dish. In this review, we report the available data on iPSC-derived neurons derived from PD patients with identified gene mutations. Specifically, we will report on the key phenotypes of the generated iPSC-derived neurons from PD patients with different genetic background. Furthermore, we discuss the relationship these cellular phenotypes have to PD pathology and future challenges and prospects for iPSC modelling and understanding of the pathogenesis of PD.
Collapse
|
20
|
Abstract
OBJECTIVES Lewy body dementia (LBD) is the second most prevalent neurodegenerative dementia and it causes more morbidity and mortality than Alzheimer's disease. Several genetic associations of LBD have been reported and their functional implications remain uncertain. Hence, we aimed to do a systematic review of all gene expression studies that investigated people with LBD for improving our understanding of LBD molecular pathology and for facilitating discovery of novel biomarkers and therapeutic targets for LBD. METHODS We systematically reviewed five online databases (PROSPERO protocol: CRD42017080647) and assessed the functional implications of all reported differentially expressed genes (DEGs) using Ingenuity Pathway Analyses. RESULTS We screened 3,809 articles and identified 31 eligible studies. In that, 1,242 statistically significant (p < 0.05) DEGs including 70 microRNAs have been reported in people with LBD. Expression levels of alternatively spliced transcripts of SNCA, SNCB, PRKN, APP, RELA, and ATXN2 significantly differ in LBD. Several mitochondrial genes and genes involved in ubiquitin proteasome system and autophagy-lysosomal pathway were significantly downregulated in LBD. Evidence supporting chronic neuroinflammation in LBD was inconsistent. Our functional analyses highlighted the importance of ribonucleic acid (RNA)-mediated gene silencing, neuregulin signalling, and neurotrophic factors in the molecular pathology of LBD. CONCLUSIONS α-synuclein aggregation, mitochondrial dysfunction, defects in molecular networks clearing misfolded proteins, and RNA-mediated gene silencing contribute to neurodegeneration in LBD. Larger longitudinal transcriptomic studies investigating biological fluids of people living with LBD are needed for molecular subtyping and staging of LBD. Diagnostic biomarker potential and therapeutic promise of identified DEGs warrant further research.
Collapse
|
21
|
Miranda-Díaz AG, García-Sánchez A, Cardona-Muñoz EG. Foods with Potential Prooxidant and Antioxidant Effects Involved in Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6281454. [PMID: 32832004 PMCID: PMC7424374 DOI: 10.1155/2020/6281454] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/02/2020] [Accepted: 07/18/2020] [Indexed: 12/20/2022]
Abstract
Oxidative stress plays a fundamental role in the pathogenesis of Parkinson's disease (PD). Oxidative stress appears to be responsible for the gradual dysfunction that manifests via numerous cellular pathways throughout PD progression. This review will describe the prooxidant effect of excessive consumption of processed food. Processed meat can affect health due to its high sodium content, advanced lipid oxidation end-products, cholesterol, and free fatty acids. During cooking, lipids can react with proteins to form advanced end-products of lipid oxidation. Excessive consumption of different types of carbohydrates is a risk factor for PD. The antioxidant effects of some foods in the regular diet provide an inconclusive interpretation of the environment's mechanisms with the modulation of oxidation stress-induced PD. Some antioxidant molecules are known whose primary mechanism is the neuroprotective effect. The melatonin mechanism consists of neutralizing reactive oxygen species (ROS) and inducing antioxidant enzyme's expression and activity. N-acetylcysteine protects against the development of PD by restoring levels of brain glutathione. The balanced administration of vitamin B3, ascorbic acid, vitamin D and the intake of caffeine every day seem beneficial for brain health in PD. Excessive chocolate intake could have adverse effects in PD patients. The findings reported to date do not provide clear benefits for a possible efficient therapeutic intervention by consuming the nutrients that are consumed regularly.
Collapse
Affiliation(s)
| | - Andrés García-Sánchez
- Department of Physiology, University Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Ernesto Germán Cardona-Muñoz
- Department of Physiology, University Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
22
|
Foster C, Steventon JJ, Helme D, Tomassini V, Wise RG. Assessment of the Effects of Aerobic Fitness on Cerebrovascular Function in Young Adults Using Multiple Inversion Time Arterial Spin Labeling MRI. Front Physiol 2020; 11:360. [PMID: 32372976 PMCID: PMC7187806 DOI: 10.3389/fphys.2020.00360] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/27/2020] [Indexed: 11/13/2022] Open
Abstract
This cross-sectional study investigated the effects of aerobic fitness on cerebrovascular function in the healthy brain. Gray matter cerebral blood flow (CBF) and cerebrovascular reactivity (CVR) were quantified in a sample of young adults within a normal fitness range. Based on existing Transcranial Doppler ultrasound and fMRI evidence, we predicted a positive relationship between fitness and resting gray matter CBF and CVR. Exploratory hypotheses that higher V . O2peak would be associated with higher GM volume and cognitive performance were also investigated. 20 adults underwent a V . O2peak test and a battery of cognitive tests. All subjects also underwent an MRI scan where multiple inversion time (MTI) pulsed arterial spin labeling (PASL) was used to quantify resting CBF and CVR to 5% CO2. Region of interest analysis showed a non-significant inverse correlation between whole-brain gray matter CBF and V . O2peak; r = -0.4, p = 0.08, corrected p (p') = 0.16 and a significant positive correlation between V . O2peak and whole-brain averaged gray matter CVR; r = 0.62, p = 0.003, p' = 0.006. Voxel-wise analysis revealed a significant inverse association between V . O2peak and resting CBF in the left and right thalamus, brainstem, right lateral occipital cortex, left intra-calcarine cortex and cerebellum. The results of this study suggest that aerobic fitness is associated with lower baseline CBF and greater CVR in young adults.
Collapse
Affiliation(s)
- Catherine Foster
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Jessica J. Steventon
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and Astronomy, Cardiff University, Cardiff, United Kingdom
- Neuroscience and Mental Health Research Institute (NMHRI), School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Daniel Helme
- Department of Anaesthetics and Intensive Care Medicine, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Valentina Tomassini
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Department of Neuroscience, Imaging and Clinical Sciences, “G. D’Annunzio University” of Chieti-Pescara, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. D’Annunzio University” of Chieti-Pescara, Chieti, Italy
| | - Richard G. Wise
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- Department of Neuroscience, Imaging and Clinical Sciences, “G. D’Annunzio University” of Chieti-Pescara, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. D’Annunzio University” of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
23
|
Ge P, Dawson VL, Dawson TM. PINK1 and Parkin mitochondrial quality control: a source of regional vulnerability in Parkinson's disease. Mol Neurodegener 2020; 15:20. [PMID: 32169097 PMCID: PMC7071653 DOI: 10.1186/s13024-020-00367-7] [Citation(s) in RCA: 298] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
That certain cell types in the central nervous system are more likely to undergo neurodegeneration in Parkinson's disease is a widely appreciated but poorly understood phenomenon. Many vulnerable subpopulations, including dopamine neurons in the substantia nigra pars compacta, have a shared phenotype of large, widely distributed axonal networks, dense synaptic connections, and high basal levels of neural activity. These features come at substantial bioenergetic cost, suggesting that these neurons experience a high degree of mitochondrial stress. In such a context, mechanisms of mitochondrial quality control play an especially important role in maintaining neuronal survival. In this review, we focus on understanding the unique challenges faced by the mitochondria in neurons vulnerable to neurodegeneration in Parkinson's and summarize evidence that mitochondrial dysfunction contributes to disease pathogenesis and to cell death in these subpopulations. We then review mechanisms of mitochondrial quality control mediated by activation of PINK1 and Parkin, two genes that carry mutations associated with autosomal recessive Parkinson's disease. We conclude by pinpointing critical gaps in our knowledge of PINK1 and Parkin function, and propose that understanding the connection between the mechanisms of sporadic Parkinson's and defects in mitochondrial quality control will lead us to greater insights into the question of selective vulnerability.
Collapse
Affiliation(s)
- Preston Ge
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
- Present address: Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
- Present address: Picower Institute for Learning and Memory, Cambridge, MA 02139 USA
- Present address: Harvard-MIT MD/PhD Program, Harvard Medical School, Boston, MA 02115 USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
| |
Collapse
|
24
|
Amin N, Tan X, Ren Q, Zhu N, Botchway BOA, Hu Z, Fang M. Recent advances of induced pluripotent stem cells application in neurodegenerative diseases. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109674. [PMID: 31255650 DOI: 10.1016/j.pnpbp.2019.109674] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/03/2019] [Accepted: 06/17/2019] [Indexed: 01/30/2023]
Abstract
Stem cell is defined by its ability to self-renewal and generates differentiated functional cell types, which are derived from the embryo and various sources of postnatal animal. These cells can be divided according to their potential development into totipotent, unipotent, multipotent andpluripotent. Pluripotent is considered as the most important type due to its advantageous capability to create different cell types of the body in a similar behavior as embryonic stem cell. Induced pluripotent stem cells (iPSCs) are adult cells that maintain the characteristics of embryonic stem cells because it can be genetically reprogrammed to an embryonic stem cell-like state via express genes and transcription factors. Such cells provide an efficient pathway to explorehuman diseases and their corresponding therapy, particularly, neurodevelopmental disorders. Consequently, iPSCs can be investigated to check the specific mutations of neurodegenerative disease due to their unique ability to differentiate into neural cell types and/or neural organoids. The current review addresses the different neurodegenerative diseases model by using iPSCs approach such as Alzheimer's diseases (AD), Parkinson diseases (PD),multiplesclerosis(MS) and psychiatric disorders. We also highlight the importance of autophagy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Nashwa Amin
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China; Department of Zoology, Faculty of Science, Aswan University, Egypt
| | - Xiaoning Tan
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiannan Ren
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Zhu
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China; Hebei North University,Zhangjiakou, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiying Hu
- Obstetrics & Gynecology Department, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, China.
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
25
|
Valdez LB, Zaobornyj T, Bandez MJ, López-Cepero JM, Boveris A, Navarro A. Complex I syndrome in striatum and frontal cortex in a rat model of Parkinson disease. Free Radic Biol Med 2019; 135:274-282. [PMID: 30862545 DOI: 10.1016/j.freeradbiomed.2019.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/27/2019] [Accepted: 03/05/2019] [Indexed: 12/21/2022]
Abstract
Mitochondrial dysfunction named complex I syndrome was observed in striatum mitochondria of rotenone treated rats (2 mg rotenone/kg, i. p., for 30 or 60 days) in an animal model of Parkinson disease. After 60 days of rotenone treatment, the animals showed: (a) 6-fold increased bradykinesia and 60% decreased locomotor activity; (b) 35-34% decreases in striatum O2 uptake and in state 3 mitochondrial respiration with malate-glutamate as substrate; (c) 43-57% diminished striatum complex I activity with 60-71% decreased striatum mitochondrial NOS activity, determined both as biochemical activity and as functional activity (by the NO inhibition of active respiration); (d) 34-40% increased rates of mitochondrial O2•- and H2O2 productions and 36-46% increased contents of the products of phospholipid peroxidation and of protein oxidation; and (e) 24% decreased striatum mitochondrial content, likely associated to decreased NO-dependent mitochondrial biogenesis. Intermediate values were observed after 30 days of rotenone treatment. Frontal cortex tissue and mitochondria showed similar but less marked changes. Rotenone-treated rats showed mitochondrial complex I syndrome associated with cellular oxidative stress in the dopaminergic brain areas of striatum and frontal cortex, a fact that describes the high sensitivity of mitochondrial complex I to inactivation by oxidative reactions.
Collapse
Affiliation(s)
- Laura B Valdez
- University of Buenos Aires, School of Pharmacy and Biochemistry, Physical Chemistry Division, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Buenos Aires, Argentina.
| | - Tamara Zaobornyj
- University of Buenos Aires, School of Pharmacy and Biochemistry, Physical Chemistry Division, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Buenos Aires, Argentina
| | - Manuel J Bandez
- University of Cadiz, School of Medicine, Department of Biochemistry and Molecular Biology, Cadiz, Spain
| | - José María López-Cepero
- University of Cadiz, School of Medicine, Department of Cell Biology and Histology, Cadiz, Spain
| | - Alberto Boveris
- University of Buenos Aires, School of Pharmacy and Biochemistry, Physical Chemistry Division, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Buenos Aires, Argentina
| | - Ana Navarro
- University of Cadiz, School of Medicine, Department of Biochemistry and Molecular Biology, Cadiz, Spain
| |
Collapse
|
26
|
Samokhvalov V, Jamieson KL, Darwesh AM, Keshavarz-Bahaghighat H, Lee TYT, Edin M, Lih F, Zeldin DC, Seubert JM. Deficiency of Soluble Epoxide Hydrolase Protects Cardiac Function Impaired by LPS-Induced Acute Inflammation. Front Pharmacol 2019; 9:1572. [PMID: 30692927 PMCID: PMC6339940 DOI: 10.3389/fphar.2018.01572] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/24/2018] [Indexed: 12/14/2022] Open
Abstract
Lipopolysaccharide (LPS) is a bacterial wall endotoxin producing many pathophysiological conditions including myocardial inflammation leading to cardiotoxicity. Linoleic acid (18:2n6, LA) is an essential n-6 PUFA which is converted to arachidonic acid (20:4n6, AA) by desaturation and elongation via enzyme systems within the body. Biological transformation of PUFA through CYP-mediated hydroxylation, epoxidation, and allylic oxidation produces lipid mediators, which may be subsequently hydrolyzed to corresponding diol metabolites by soluble epoxide hydrolase (sEH). In the current study, we investigate whether inhibition of sEH, which alters the PUFA metabolite profile, can influence LPS induced cardiotoxicity and mitochondrial function. Our data demonstrate that deletion of soluble epoxide hydrolase provides protective effects against LPS-induced cardiotoxicity by maintaining mitochondrial function. There was a marked alteration in the cardiac metabolite profile with notable increases in sEH-derived vicinal diols, 9,10- and 12,13-dihydroxyoctadecenoic acid (DiHOME) in WT hearts following LPS administration, which was absent in sEH null mice. We found that DiHOMEs triggered pronounced mitochondrial structural abnormalities, which also contributed to the development of extensive mitochondrial dysfunction in cardiac cells. Accumulation of DiHOMEs may represent an intermediate mechanism through which LPS-induced acute inflammation triggers deleterious alterations in the myocardium in vivo and cardiac cells in vitro. This study reveals novel research exploring the contribution of DiHOMEs in the progression of adverse inflammatory responses toward cardiac function in vitro and in vivo.
Collapse
Affiliation(s)
- Victor Samokhvalov
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - K Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | | | - Tim Y T Lee
- Department of Pharmacology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Matthew Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Fred Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Pharmacology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Iglesias DE, Bombicino SS, Boveris A, Valdez LB. (+)-Catechin inhibits heart mitochondrial complex I and nitric oxide synthase: functional consequences on membrane potential and hydrogen peroxide production. Food Funct 2019; 10:2528-2537. [DOI: 10.1039/c8fo01843j] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The aim was to study thein vitroeffect of nM to low μM concentration of (+)-catechin on the enzymatic activities of mitochondrial complex I and mtNOS, as well as the consequences on the membrane potential and H2O2production rate.
Collapse
Affiliation(s)
- Darío E. Iglesias
- University of Buenos Aires
- School of Pharmacy and Biochemistry
- Physical Chemistry Division
- Buenos Aires
- Argentina
| | - Silvina S. Bombicino
- University of Buenos Aires
- School of Pharmacy and Biochemistry
- Physical Chemistry Division
- Buenos Aires
- Argentina
| | - Alberto Boveris
- University of Buenos Aires
- School of Pharmacy and Biochemistry
- Physical Chemistry Division
- Buenos Aires
- Argentina
| | - Laura B. Valdez
- University of Buenos Aires
- School of Pharmacy and Biochemistry
- Physical Chemistry Division
- Buenos Aires
- Argentina
| |
Collapse
|
28
|
Docosahexaenoic acid protection in a rotenone induced Parkinson's model: Prevention of tubulin and synaptophysin loss, but no association with mitochondrial function. Neurochem Int 2018; 121:26-37. [DOI: 10.1016/j.neuint.2018.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/15/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
|
29
|
Marques NF, Massari CM, Tasca CI. Guanosine Protects Striatal Slices Against 6-OHDA-Induced Oxidative Damage, Mitochondrial Dysfunction, and ATP Depletion. Neurotox Res 2018; 35:475-483. [PMID: 30417317 DOI: 10.1007/s12640-018-9976-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 01/18/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by loss of dopaminergic neurons in substantia nigra pars compacta which induces severe motor symptoms. 6-OHDA is a neurotoxin widely used in PD animal models due to its high affinity by dopamine transporter, its rapid non-enzymatic auto-oxidation which generates reactive oxygen species (ROS), oxidative stress, and for induced mitochondrial dysfunction. We previously reported an in vitro protocol of 6-OHDA-induced toxicity in brain regions slices, as a simple and sensitive assay to screen for protective compounds related to PD. Guanosine (GUO), a guanine-based purine nucleoside, is a neuroprotective molecule that is showing promising effects as an antiparkinsonian agent. To investigate the mechanisms involved on GUO-induced neuroprotection, slices of cortex, striatum, and hippocampus were incubated with GUO in the presence of 6-OHDA (100 μM). 6-OHDA promoted a decrease in cellular viability and increased ROS generation in all brain regions. Disruption of mitochondrial potential, depletion in intracellular ATP levels, and increase in cell membrane permeabilization were evidenced in striatal slices. GUO prevented the increase in ROS generation, disruption in mitochondrial potential, and depletion of intracellular ATP induced by 6-OHDA in striatal slices. In conclusion, GUO was effective to prevent oxidative events before cell damage, such as mitochondrial disruption, intracellular ATP levels depletion, and ROS generation in striatal slices subjected to in vitro 6-OHDA-induced toxicity.
Collapse
Affiliation(s)
- Naiani Ferreira Marques
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Caio Marcos Massari
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Carla Inês Tasca
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil. .,Departamento de Bioquímica, CCB, UFSC, Trindade, Florianópolis, Santa Catarina, 88040-900, Brazil.
| |
Collapse
|
30
|
Zeinsteger PA, Barberón JL, Leaden PJ, Palacios A. Antioxidant properties of Calendula officinalis L. (Asteraceae) on Fe2+-initiated peroxidation of rat brain mitochondria. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2254-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
Sison SL, Vermilyea SC, Emborg ME, Ebert AD. Using Patient-Derived Induced Pluripotent Stem Cells to Identify Parkinson's Disease-Relevant Phenotypes. Curr Neurol Neurosci Rep 2018; 18:84. [PMID: 30284665 DOI: 10.1007/s11910-018-0893-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Parkinson's disease (PD) is the second most common neurodegenerative disorder affecting older individuals. The specific cause underlying dopaminergic (DA) neuron loss in the substantia nigra, a pathological hallmark of PD, remains elusive. Here, we highlight peer-reviewed reports using induced pluripotent stem cells (iPSCs) to model PD in vitro and discuss the potential disease-relevant phenotypes that may lead to a better understanding of PD etiology. Benefits of iPSCs are that they retain the genetic background of the donor individual and can be differentiated into specialized neurons to facilitate disease modeling. RECENT FINDINGS Mitochondrial dysfunction, oxidative stress, ER stress, and alpha-synuclein accumulation are common phenotypes observed in PD iPSC-derived neurons. New culturing technologies, such as directed reprogramming and midbrain organoids, offer innovative ways of investigating intraneuronal mechanisms of PD pathology. PD patient-derived iPSCs are an evolving resource to understand PD pathology and identify therapeutic targets.
Collapse
Affiliation(s)
- S L Sison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, BSB 409, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - S C Vermilyea
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - M E Emborg
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - A D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, BSB 409, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA.
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
32
|
Omura T, Sasaoka M, Hashimoto G, Imai S, Yamamoto J, Sato Y, Nakagawa S, Yonezawa A, Nakagawa T, Yano I, Tasaki Y, Matsubara K. Oxicam-derived non-steroidal anti-inflammatory drugs suppress 1-methyl-4-phenyl pyridinium-induced cell death via repression of endoplasmic reticulum stress response and mitochondrial dysfunction in SH-SY5Y cells. Biochem Biophys Res Commun 2018; 503:2963-2969. [DOI: 10.1016/j.bbrc.2018.08.078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/08/2018] [Indexed: 10/28/2022]
|
33
|
Ferrer I. Sisyphus in Neverland. J Alzheimers Dis 2018; 62:1023-1047. [PMID: 29154280 PMCID: PMC5870014 DOI: 10.3233/jad-170609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2017] [Indexed: 11/24/2022]
Abstract
The study of life and living organisms and the way in which these interact and organize to form social communities have been central to my career. I have been fascinated by biology, neurology, and neuropathology, but also by history, sociology, and art. Certain current historical, political, and social events, some occurring proximally but others affecting people in apparently distant places, have had an impact on me. Epicurus, Seneca, and Camus shared their philosophical positions which I learned from. Many scientists from various disciplines have been exciting sources of knowledge as well. I have created a world of hypothesis and experiments but I have also got carried away by serendipity following unexpected observations. It has not been an easy path; errors and wanderings are not uncommon, and opponents close to home much more abundant than one might imagine. Ambition, imagination, resilience, and endurance have been useful in moving ahead in response to setbacks. In the end, I have enjoyed my dedication to science and I am grateful to have glimpsed beauty in it. These are brief memories of a Spanish neuropathologist born and raised in Barcelona, EU.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Service of Pathological Anatomy, Bellvitge University Hospital; CIBERNED; Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
34
|
Jellinger KA. Dementia with Lewy bodies and Parkinson's disease-dementia: current concepts and controversies. J Neural Transm (Vienna) 2017; 125:615-650. [PMID: 29222591 DOI: 10.1007/s00702-017-1821-9] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022]
Abstract
Dementia with Lewy bodies (DLB) and Parkinson's disease-dementia (PDD), although sharing many clinical, neurochemical and morphological features, according to DSM-5, are two entities of major neurocognitive disorders with Lewy bodies of unknown etiology. Despite considerable clinical overlap, their diagnosis is based on an arbitrary distinction between the time of onset of motor and cognitive symptoms: dementia often preceding parkinsonism in DLB and onset of cognitive impairment after onset of motor symptoms in PDD. Both are characterized morphologically by widespread cortical and subcortical α-synuclein/Lewy body plus β-amyloid and tau pathologies. Based on recent publications, including the fourth consensus report of the DLB Consortium, a critical overview is given. The clinical features of DLB and PDD include cognitive impairment, parkinsonism, visual hallucinations, and fluctuating attention. Intravitam PET and post-mortem studies revealed more pronounced cortical atrophy, elevated cortical and limbic Lewy pathologies (with APOE ε4), apart from higher prevalence of Alzheimer pathology in DLB than PDD. These changes may account for earlier onset and greater severity of cognitive defects in DLB, while multitracer PET studies showed no differences in cholinergic and dopaminergic deficits. DLB and PDD sharing genetic, neurochemical, and morphologic factors are likely to represent two subtypes of an α-synuclein-associated disease spectrum (Lewy body diseases), beginning with incidental Lewy body disease-PD-nondemented-PDD-DLB (no parkinsonism)-DLB with Alzheimer's disease (DLB-AD) at the most severe end, although DLB does not begin with PD/PDD and does not always progress to DLB-AD, while others consider them as the same disease. Both DLB and PDD show heterogeneous pathology and neurochemistry, suggesting that they share important common underlying molecular pathogenesis with AD and other proteinopathies. Cognitive impairment is not only induced by α-synuclein-caused neurodegeneration but by multiple regional pathological scores. Recent animal models and human post-mortem studies have provided important insights into the pathophysiology of DLB/PDD showing some differences, e.g., different spreading patterns of α-synuclein pathology, but the basic pathogenic mechanisms leading to the heterogeneity between both disorders deserve further elucidation. In view of the controversies about the nosology and pathogenesis of both syndromes, there remains a pressing need to differentiate them more clearly and to understand the processes leading these synucleinopathies to cause one disorder or the other. Clinical management of both disorders includes cholinesterase inhibitors, other pharmacologic and nonpharmacologic strategies, but these have only a mild symptomatic effect. Currently, no disease-modifying therapies are available.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
35
|
Bombicino SS, Iglesias DE, Rukavina-Mikusic IA, Buchholz B, Gelpi RJ, Boveris A, Valdez LB. Hydrogen peroxide, nitric oxide and ATP are molecules involved in cardiac mitochondrial biogenesis in Diabetes. Free Radic Biol Med 2017; 112:267-276. [PMID: 28756312 DOI: 10.1016/j.freeradbiomed.2017.07.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/12/2017] [Accepted: 07/25/2017] [Indexed: 01/21/2023]
Abstract
This study, in an experimental model of type I Diabetes Mellitus in rats, deals with the mitochondrial production rates and steady-state concentrations of H2O2 and NO, and ATP levels as part of a network of signaling molecules involved in heart mitochondrial biogenesis. Sustained hyperglycemia leads to a cardiac compromise against a work overload, in the absence of changes in resting cardiac performance and of heart hypertrophy. Diabetes was induced in male Wistar rats by a single dose of Streptozotocin (STZ, 60mg × kg-1, ip.). After 28 days of STZ-injection, rats were sacrificed and hearts were isolated. The mitochondrial mass (mg mitochondrial protein × g heart-1), determined through cytochrome oxidase activity ratio, was 47% higher in heart from diabetic than from control animals. Stereological analysis of cardiac tissue microphotographs showed an increase in the cytosolic volume occupied by mitochondria (30%) and in the number of mitochondria per unit area (52%), and a decrease in the mean area of each mitochondrion (23%) in diabetic respect to control rats. Additionally, an enhancement (76%) in PGC-1α expression was observed in cardiac tissue of diabetic animals. Moreover, heart mitochondrial H2O2 (127%) and NO (23%) productions and mtNOS expression (132%) were higher, while mitochondrial ATP production rate was lower (~ 40%), concomitantly with a partial-mitochondrial depolarization, in diabetic than in control rats. Changes in mitochondrial H2O2 and NO steady-state concentrations and an imbalance between cellular energy demand and mitochondrial energy transduction could be involved in the signaling pathways that lead to the novo synthesis of mitochondria. However, this compensatory mechanism triggered to restore the mitochondrial and tissue normal activities, did not lead to competent mitochondria capable of supplying the energetic demands in diabetic pathological conditions.
Collapse
Affiliation(s)
- Silvina S Bombicino
- University of Buenos Aires, School of Pharmacy and Biochemistry, Physical Chemistry Division, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), University of Buenos Aires, Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires, Argentina
| | - Darío E Iglesias
- University of Buenos Aires, School of Pharmacy and Biochemistry, Physical Chemistry Division, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), University of Buenos Aires, Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires, Argentina
| | - Ivana A Rukavina-Mikusic
- University of Buenos Aires, School of Pharmacy and Biochemistry, Physical Chemistry Division, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), University of Buenos Aires, Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires, Argentina
| | - Bruno Buchholz
- University of Buenos Aires, Faculty of Medicine, Pathology Department, Cardiovascular Physiopathology Institute (INFICA), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), University of Buenos Aires, Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires, Argentina
| | - Ricardo J Gelpi
- University of Buenos Aires, Faculty of Medicine, Pathology Department, Cardiovascular Physiopathology Institute (INFICA), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), University of Buenos Aires, Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires, Argentina
| | - Alberto Boveris
- University of Buenos Aires, School of Pharmacy and Biochemistry, Physical Chemistry Division, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), University of Buenos Aires, Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires, Argentina
| | - Laura B Valdez
- University of Buenos Aires, School of Pharmacy and Biochemistry, Physical Chemistry Division, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), University of Buenos Aires, Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires, Argentina.
| |
Collapse
|
36
|
Huenchuguala S, Muñoz P, Segura-Aguilar J. The Importance of Mitophagy in Maintaining Mitochondrial Function in U373MG Cells. Bafilomycin A1 Restores Aminochrome-Induced Mitochondrial Damage. ACS Chem Neurosci 2017; 8:2247-2253. [PMID: 28763613 DOI: 10.1021/acschemneuro.7b00152] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aminochrome, an orthoquinone formed during the dopamine oxidation of neuromelanin, is neurotoxic because it induces mitochondria dysfunction, protein degradation dysfunction (both autophagy and proteasomal systems), α-synuclein aggregation to neurotoxic oligomers, neuroinflammation, and oxidative and endoplasmic reticulum stress. In this study, we investigated the relationship between aminochrome-induced autophagy/lysosome dysfunction and mitochondrial dysfunction in U373MGsiGST6 cells. Aminochrome (75 μM) induces mitochondrial dysfunction as determined by (i) a significant decrease in ATP levels (70%; P < 0.001) and (ii) a significant decrease in mitochondrial membrane potential (P < 0.001). Interestingly, the pretreatment of U373MGsiGST6 cells with 100 nM bafilomycin-A1, an inhibitor of lysosomal vacuolar-type H+-ATPase, restores ATP levels, mitochondrial membrane potential, and mitophagy, and decreases cell death. These results reveal (i) the importance of macroautophagy/the lysosomal degradation system for the normal functioning of mitochondria and for cell survival, and (ii) aminochrome-induced lysosomal dysfunction depends on the aminochrome-dependent inactivation of the vacuolar-type H+-ATPase, which pumps protons into the lysosomes. This study also supports the proposed protective role of glutathione transferase mu2-2 (GSTM2) in astrocytes against aminochrome toxicity, mediated by mitochondrial and lysosomal dysfunction.
Collapse
Affiliation(s)
- Sandro Huenchuguala
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Patricia Muñoz
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Segura-Aguilar
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
37
|
Salidroside Protects against MPP+-Induced Neuronal Injury through DJ-1-Nrf2 Antioxidant Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:5398542. [PMID: 29234413 PMCID: PMC5637855 DOI: 10.1155/2017/5398542] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/25/2017] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. We have found that salidroside (Sal) exhibited neuroprotective effects against MPP+ toxicity. However, the molecular mechanism is not fully understood. In this study, we found that Sal significantly prevented MPP+-induced decrease of mRNA and protein expression of Nrf2, GCLc, SOD1, and SOD2 in SH-SY5Y cells. Moreover, silencing of Nrf2 significantly inhibited Sal-induced increase in mRNA and protein expression of GCLc, SOD1, and SOD2. But Nrf2 silence did not significantly impact Sal-exhibited effects on DJ-1 expression. Silencing of Nrf2 significantly suppressed the decrease of apoptosis induced by Sal in MPP+-treated SH-SY5Y cells. Sal significantly prevented MPP+-induced decrease of the mRNA and protein expression of DJ-1 in SH-SY5Y cells. Moreover, silencing of DJ-1 significantly inhibited Sal-induced increase in mRNA and protein expression of Nrf2, GCLc, SOD1, and SOD2 in MPP+-treated SH-SY5Y cells. These results indicated that DJ-1 was an upstream regulator of Nrf2 in the neuroprotective effects of Sal. Furthermore, silencing of DJ-1 significantly suppressed the decrease of apoptosis induced by Sal in MPP+-treated SH-SY5Y cells. In conclusion, Sal prevented MPP+-induced neurotoxicity through upregulation of DJ-1-Nrf2-antioxidant pathway. Our findings provide novel insights into the neuroprotective effects of Sal against PD.
Collapse
|
38
|
Guo C, Ding P, Xie C, Ye C, Ye M, Pan C, Cao X, Zhang S, Zheng S. Potential application of the oxidative nucleic acid damage biomarkers in detection of diseases. Oncotarget 2017; 8:75767-75777. [PMID: 29088908 PMCID: PMC5650463 DOI: 10.18632/oncotarget.20801] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 08/27/2017] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) are generated after exposure to harmful environmental factors and during normal cellular metabolic processes. The balance of the generating and scavenging of ROS plays a significant role in living cells. The accumulation of ROS will lead to oxidative damage to biomolecules including nucleic acid. Although many types of oxidative nucleic acid damage products have been identified, 8-oxo-7,8-dihydro-2’-deoxyguanosine (8-oxodG) and 8-oxo-7,8-dihydroguanosine (8-oxoG) has been commonly chosen as the biomarkers of oxidative damage to DNA and RNA, respectively. It has been demonstrated that oxidative damage to nucleic acid is an initiator in pathogenesis of numerous diseases. Thus, oxidative nucleic acid damage biomarkers have the potential to be utilized for detection of diseases. Herein, we reviewed the relationship of oxidative nucleic acid damage and development of various diseases including cancers (colorectal cancer, gastrointestinal cancer, breast cancer, lung cancer, epithelial ovarian carcinoma, esophageal squamous cell carcinoma), neurodegenerative disorders and chronic diseases (diabetes and its complications, cardiovascular diseases). The potential of oxidative nucleic acid damage biomarkers for detection of diseases and drug development were described. Moreover, the approaches for detection of these biomarkers were also summarized.
Collapse
Affiliation(s)
- Cheng Guo
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Peili Ding
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Cong Xie
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Chenyang Ye
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Minfeng Ye
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, China
| | - Chi Pan
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Xiaoji Cao
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Suzhan Zhang
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Shu Zheng
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
39
|
Santpere G, Garcia-Esparcia P, Andres-Benito P, Lorente-Galdos B, Navarro A, Ferrer I. Transcriptional network analysis in frontal cortex in Lewy body diseases with focus on dementia with Lewy bodies. Brain Pathol 2017; 28:315-333. [PMID: 28321951 DOI: 10.1111/bpa.12511] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/15/2017] [Indexed: 12/13/2022] Open
Abstract
The present study investigates global transcriptional changes in frontal cortex area 8 in incidental Lewy Body disease (iLBD), Parkinson disease (PD) and Dementia with Lewy bodies (DLB). We identified different coexpressed gene sets associated with disease stages, and gene ontology categories enriched in gene modules and differentially expressed genes including modules or gene clusters correlated to iLBD comprising upregulated dynein genes and taste receptors, and downregulated innate inflammation. Focusing on DLB, we found modules with genes significantly enriched in functions related to RNA and DNA production, mitochondria and energy metabolism, purine metabolism, chaperone and protein folding system and synapses and neurotransmission (particularly the GABAergic system). The expression of more than fifty selected genes was assessed with real time quantitative polymerase chain reaction. Our findings provide, for the first time, evidence of molecular cortical alterations in iLBD and involvement of several key metabolic pathways and gene hubs in DLB which may underlie cognitive impairment and dementia.
Collapse
Affiliation(s)
- Gabriel Santpere
- Department of Neurobiology, Yale School of Medicine, New Haven, CT.,Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain
| | - Paula Garcia-Esparcia
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Pol Andres-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Belen Lorente-Galdos
- Department of Neurobiology, Yale School of Medicine, New Haven, CT.,Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain
| | - Arcadi Navarro
- Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain.,Institute of Science and Technology, Centre for Genomic Regulation (CRG), Barcelona, Spain.,National Institute for Bioinformatics (INB), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain.,Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain.,Institute of Neurosciences, University of Barcelona, Hospitalet de Llobregat, Spain.,CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain
| |
Collapse
|
40
|
Garcia-Esparcia P, Koneti A, Rodríguez-Oroz MC, Gago B, Del Rio JA, Ferrer I. Mitochondrial activity in the frontal cortex area 8 and angular gyrus in Parkinson's disease and Parkinson's disease with dementia. Brain Pathol 2017; 28:43-57. [PMID: 27984680 DOI: 10.1111/bpa.12474] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 12/01/2016] [Indexed: 11/30/2022] Open
Abstract
Altered mitochondrial function is characteristic in the substantia nigra in Parkinson's disease (PD). Information about mitochondria in other brain regions such as the cerebral cortex is conflicting mainly because most studies have not contemplated the possibility of variable involvement depending on the region, stage of disease progression and clinical symptoms such as the presence or absence of dementia. RT-qPCR of 18 nuclear mRNAs encoding subunits of mitochondrial complexes and 12 mRNAs encoding energy metabolism-related enzymes; western blotting of mitochondrial proteins; and analysis of enzymatic activities of complexes I, II, II, IV and V of the respiratory chain were assessed in frontal cortex area 8 and the angular gyrus of middle-aged individuals (MA), and those with incidental PD (iPD), long-lasting PD with parkinsonism without dementia (PD) and long-lasting PD with dementia (PDD). Up-regulation of several genes was found in frontal cortex area 8 in PD when compared with MA and in the angular gyrus in iPD when compared with MA. Marked down-regulation of genes encoding mitochondrial subunits and energy metabolism-related enzymes occurs in frontal cortex but only of genes coding for energy metabolism-related enzymes in the angular gyrus in PDD. Significant decrease in the protein expression levels of several mitochondrial subunits encoded by these genes occurs in frontal cortex area 8 and angular gyrus in PDD. Moreover, expression of MT-ND1 which is encoded by mitochondrial DNA is also reduced in PDD. Reduced enzymatic activity of complex III in frontal cortex area 8 and angular gyrus is observed in PD, but dramatic reduction in the activity of complexes I, II, II and IV in both regions characterizes PDD. Dementia in the context of PD is linked to region-specific deregulation of genomic genes encoding subunits of mitochondrial complexes and to marked reduction in the activity of mitochondrial complexes I, II, III and IV.
Collapse
Affiliation(s)
- Paula Garcia-Esparcia
- Institute of Neuropathology, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de LLobregat, Catalonia, Spain
| | - Anusha Koneti
- Institute of Neuropathology, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de LLobregat, Catalonia, Spain
| | - M Cruz Rodríguez-Oroz
- University Hospital Donostia, San Sebastián, Spain.,Ikerbasque (Basque Foundation for Science), Bilbao, Spain.,Biodonostia Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Belen Gago
- Biodonostia Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José Antonio Del Rio
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Isidro Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de LLobregat, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
41
|
Garcia-Esparcia P, López-González I, Grau-Rivera O, García-Garrido MF, Konetti A, Llorens F, Zafar S, Carmona M, Del Rio JA, Zerr I, Gelpi E, Ferrer I. Dementia with Lewy Bodies: Molecular Pathology in the Frontal Cortex in Typical and Rapidly Progressive Forms. Front Neurol 2017; 8:89. [PMID: 28348546 PMCID: PMC5346561 DOI: 10.3389/fneur.2017.00089] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/24/2017] [Indexed: 11/29/2022] Open
Abstract
Objectives The goal of this study was to assess mitochondrial function, energy, and purine metabolism, protein synthesis machinery from the nucleolus to the ribosome, inflammation, and expression of newly identified ectopic olfactory receptors (ORs) and taste receptors (TASRs) in the frontal cortex of typical cases of dementia with Lewy bodies (DLB) and cases with rapid clinical course (rpDLB: 2 years or less) compared with middle-aged non-affected individuals, in order to learn about the biochemical abnormalities underlying Lewy body pathology. Methods Real-time quantitative PCR, mitochondrial enzymatic assays, and analysis of β-amyloid, tau, and synuclein species were used. Results The main alterations in DLB and rpDLB, which are more marked in the rapidly progressive forms, include (i) deregulated expression of several mRNAs and proteins of mitochondrial subunits, and reduced activity of complexes I, II, III, and IV of the mitochondrial respiratory chain; (ii) reduced expression of selected molecules involved in energy metabolism and increased expression of enzymes involved in purine metabolism; (iii) abnormal expression of nucleolar proteins, rRNA18S, genes encoding ribosomal proteins, and initiation factors of the transcription at the ribosome; (iv) discrete inflammation; and (v) marked deregulation of brain ORs and TASRs, respectively. Severe mitochondrial dysfunction involving activity of four complexes, minimal inflammatory responses, and dramatic altered expression of ORs and TASRs discriminate DLB from Alzheimer’s disease. Altered solubility and aggregation of α-synuclein, increased β-amyloid bound to membranes, and absence of soluble tau oligomers are common in DLB and rpDLB. Low levels of soluble β-amyloid are found in DLB. However, increased soluble β-amyloid 1–40 and β-amyloid 1–42, and increased TNFα mRNA and protein expression, distinguish rpDLB. Conclusion Molecular alterations in frontal cortex in DLB involve key biochemical pathways such as mitochondria and energy metabolism, protein synthesis, purine metabolism, among others and are accompanied by discrete innate inflammatory response.
Collapse
Affiliation(s)
- Paula Garcia-Esparcia
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain; CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Madrid, Spain
| | - Irene López-González
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain; CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Madrid, Spain
| | - Oriol Grau-Rivera
- Neurological Tissue Bank of the Biobanc-Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS) , Barcelona , Spain
| | - María Francisca García-Garrido
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat , Barcelona , Spain
| | - Anusha Konetti
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat , Barcelona , Spain
| | - Franc Llorens
- Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University, German Center for Neurodegenerative Diseases (DZNE) , Göttingen , Germany
| | - Saima Zafar
- Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University, German Center for Neurodegenerative Diseases (DZNE) , Göttingen , Germany
| | - Margarita Carmona
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain; CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Madrid, Spain
| | - José Antonio Del Rio
- CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Madrid, Spain; Molecular and Cellular Neurobiotechnology, Department of Cell Biology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University, German Center for Neurodegenerative Diseases (DZNE) , Göttingen , Germany
| | - Ellen Gelpi
- Neurological Tissue Bank of the Biobanc-Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS) , Barcelona , Spain
| | - Isidro Ferrer
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain; CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Madrid, Spain; Department of Pathology and Experimental Therapeutics, L'Hospitalet de Llobregat, University of Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Ambrosi G, Milani P. Endoplasmic reticulum, oxidative stress and their complex crosstalk in neurodegeneration: proteostasis, signaling pathways and molecular chaperones. AIMS MOLECULAR SCIENCE 2017. [DOI: 10.3934/molsci.2017.4.424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
43
|
Toulorge D, Schapira AHV, Hajj R. Molecular changes in the postmortem parkinsonian brain. J Neurochem 2016; 139 Suppl 1:27-58. [PMID: 27381749 DOI: 10.1111/jnc.13696] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/14/2016] [Accepted: 05/27/2016] [Indexed: 12/16/2022]
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disease after Alzheimer disease. Although PD has a relatively narrow clinical phenotype, it has become clear that its etiological basis is broad. Post-mortem brain analysis, despite its limitations, has provided invaluable insights into relevant pathogenic pathways including mitochondrial dysfunction, oxidative stress and protein homeostasis dysregulation. Identification of the genetic causes of PD followed the discovery of these abnormalities, and reinforced the importance of the biochemical defects identified post-mortem. Recent genetic studies have highlighted the mitochondrial and lysosomal areas of cell function as particularly significant in mediating the neurodegeneration of PD. Thus the careful analysis of post-mortem PD brain biochemistry remains a crucial component of research, and one that offers considerable opportunity to pursue etiological factors either by 'reverse biochemistry' i.e. from defective pathway to mutant gene, or by the complex interplay between pathways e.g. mitochondrial turnover by lysosomes. In this review we have documented the spectrum of biochemical defects identified in PD post-mortem brain and explored their relevance to metabolic pathways involved in neurodegeneration. We have highlighted the complex interactions between these pathways and the gene mutations causing or increasing risk for PD. These pathways are becoming a focus for the development of disease modifying therapies for PD. Parkinson's is accompanied by multiple changes in the brain that are responsible for the progression of the disease. We describe here the molecular alterations occurring in postmortem brains and classify them as: Neurotransmitters and neurotrophic factors; Lewy bodies and Parkinson's-linked genes; Transition metals, calcium and calcium-binding proteins; Inflammation; Mitochondrial abnormalities and oxidative stress; Abnormal protein removal and degradation; Apoptosis and transduction pathways. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
| | | | - Rodolphe Hajj
- Department of Discovery, Pharnext, Issy-Les-Moulineaux, France.
| |
Collapse
|
44
|
Ansoleaga B, Garcia-Esparcia P, Llorens F, Hernández-Ortega K, Carmona Tech M, Antonio Del Rio J, Zerr I, Ferrer I. Altered Mitochondria, Protein Synthesis Machinery, and Purine Metabolism Are Molecular Contributors to the Pathogenesis of Creutzfeldt-Jakob Disease. J Neuropathol Exp Neurol 2016; 75:755-769. [PMID: 27297670 DOI: 10.1093/jnen/nlw048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Neuron loss, synaptic decline, and spongiform change are the hallmarks of sporadic Creutzfeldt-Jakob disease (sCJD), and may be related to deficiencies in mitochondria, energy metabolism, and protein synthesis. To investigate these relationships, we determined the expression levels of genes encoding subunits of the 5 protein complexes of the electron transport chain, proteins involved in energy metabolism, nucleolar and ribosomal proteins, and enzymes of purine metabolism in frontal cortex samples from 15 cases of sCJD MM1 and age-matched controls. We also assessed the protein expression levels of subunits of the respiratory chain, initiation and elongation translation factors of protein synthesis, and localization of selected mitochondrial components. We identified marked, generalized alterations of mRNA and protein expression of most subunits of all 5 mitochondrial respiratory chain complexes in sCJD cases. Expression of molecules involved in protein synthesis and purine metabolism were also altered in sCJD. These findings point to altered mRNA and protein expression of components of mitochondria, protein synthesis machinery, and purine metabolism as components of the pathogenesis of CJD.
Collapse
Affiliation(s)
- Belén Ansoleaga
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Paula Garcia-Esparcia
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Franc Llorens
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Karina Hernández-Ortega
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Margarita Carmona Tech
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - José Antonio Del Rio
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Inga Zerr
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Isidro Ferrer
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF).
| |
Collapse
|
45
|
Gatt AP, Duncan OF, Attems J, Francis PT, Ballard CG, Bateman JM. Dementia in Parkinson's disease is associated with enhanced mitochondrial complex I deficiency. Mov Disord 2016; 31:352-9. [PMID: 26853899 DOI: 10.1002/mds.26513] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/12/2015] [Accepted: 11/16/2015] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Dementia is a common feature of Parkinson's disease (PD), but the neuropathological changes associated with the development of Parkinson's disease dementia (PDD) are only partially understood. Mitochondrial dysfunction is a hallmark of PD but has not been studied in PDD. METHODS Molecular and biochemical approaches were used to study mitochondrial activity and quantity in postmortem prefrontal cortex tissue. Tissues from pathologically confirmed PD and PDD patients and from age-matched controls were used to analyze the activity of mitochondrial enzyme complex nicotinamide adenine dinucleotide:ubiquinone oxidoreductase, or complex I (the first enzyme in the mitochondrial respiratory chain), mitochondrial DNA levels, and the expression of mitochondrial proteins. RESULTS Complex I activity was significantly decreased (27% reduction; analysis of variance with Tukey's post hoc test; P < 0.05) in PDD patients, and mitochondrial DNA levels were also significantly decreased (18% reduction; Kruskal-Wallis analysis of variance with Dunn's multiple comparison test; P < 0.05) in PDD patients compared with controls, but neither was significantly reduced in PD patients. Overall, mitochondrial biogenesis was unaffected in PD or PDD, because the expression of mitochondrial proteins in patients was similar to that in controls. CONCLUSIONS Patients with PDD have a deficiency in mitochondrial complex I activity and reduced mitochondrial DNA levels in the prefrontal cortex without a change in mitochondrial protein quantity. Therefore, mitochondrial complex I deficiency and reduced mitochondrial DNA in the prefrontal cortex may be a hallmark of dementia in patients with PD.
Collapse
Affiliation(s)
- Ariana P Gatt
- Wolfson Center for Age-Related Diseases, King's College London, Guy's Campus, London, United Kingdom
| | - Olivia F Duncan
- Wolfson Center for Age-Related Diseases, King's College London, Guy's Campus, London, United Kingdom
| | - Johannes Attems
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | - Paul T Francis
- Wolfson Center for Age-Related Diseases, King's College London, Guy's Campus, London, United Kingdom
| | - Clive G Ballard
- Wolfson Center for Age-Related Diseases, King's College London, Guy's Campus, London, United Kingdom
| | - Joseph M Bateman
- Wolfson Center for Age-Related Diseases, King's College London, Guy's Campus, London, United Kingdom
| |
Collapse
|
46
|
Lin CL, Cheng YS, Li HH, Chiu PY, Chang YT, Ho YJ, Lai TJ. Amyloid-β suppresses AMP-activated protein kinase (AMPK) signaling and contributes to α-synuclein-induced cytotoxicity. Exp Neurol 2016; 275 Pt 1:84-98. [DOI: 10.1016/j.expneurol.2015.10.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 10/11/2015] [Accepted: 10/24/2015] [Indexed: 12/01/2022]
|
47
|
Garcia-Esparcia P, Hernández-Ortega K, Koneti A, Gil L, Delgado-Morales R, Castaño E, Carmona M, Ferrer I. Altered machinery of protein synthesis is region- and stage-dependent and is associated with α-synuclein oligomers in Parkinson's disease. Acta Neuropathol Commun 2015; 3:76. [PMID: 26621506 PMCID: PMC4666041 DOI: 10.1186/s40478-015-0257-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/14/2015] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Parkinson's disease (PD) is characterized by the accumulation of abnormal α-synuclein in selected regions of the brain following a gradient of severity with disease progression. Whether this is accompanied by globally altered protein synthesis is poorly documented. The present study was carried out in PD stages 1-6 of Braak and middle-aged (MA) individuals without alterations in brain in the substantia nigra, frontal cortex area 8, angular gyrus, precuneus and putamen. RESULTS Reduced mRNA expression of nucleolar proteins nucleolin (NCL), nucleophosmin (NPM1), nucleoplasmin 3 (NPM3) and upstream binding transcription factor (UBF), decreased NPM1 but not NPM3 nucleolar protein immunostaining in remaining neurons; diminished 18S rRNA, 28S rRNA; reduced expression of several mRNAs encoding ribosomal protein (RP) subunits; and altered protein levels of initiation factor eIF3 and elongation factor eEF2 of protein synthesis was found in the substantia nigra in PD along with disease progression. Although many of these changes can be related to neuron loss in the substantia nigra, selective alteration of certain factors indicates variable degree of vulnerability of mRNAs, rRNAs and proteins in degenerating sustantia nigra. NPM1 mRNA and 18S rRNA was increased in the frontal cortex area 8 at stage 5-6; modifications were less marked and region-dependent in the angular gyrus and precuneus. Several RPs were abnormally regulated in the frontal cortex area 8 and precuneus, but only one RP in the angular gyrus, in PD. Altered levels of eIF3 and eIF1, and decrease eEF1A and eEF2 protein levels were observed in the frontal cortex in PD. No modifications were found in the putamen at any time of the study except transient modifications in 28S rRNA and only one RP mRNA at stages 5-6. These observations further indicate marked region-dependent and stage-dependent alterations in the cerebral cortex in PD. Altered solubility and α-synuclein oligomer formation, assessed in total homogenate fractions blotted with anti-α-synuclein oligomer-specific antibody, was demonstrated in the substantia nigra and frontal cortex, but not in the putamen, in PD. Dramatic increase in α-synuclein oligomers was also seen in fluorescent-activated cell sorter (FACS)-isolated nuclei in the frontal cortex in PD. CONCLUSIONS Altered machinery of protein synthesis is altered in the substantia nigra and cerebral cortex in PD being the frontal cortex area 8 more affected than the angular gyrus and precuneus; in contrast, pathways of protein synthesis are apparently preserved in the putamen. This is associated with the presence of α-synuclein oligomeric species in total homogenates; substantia nigra and frontal cortex are enriched, albeit with different band patterns, in α-synuclein oligomeric species, whereas α-synuclein oligomers are not detected in the putamen.
Collapse
Affiliation(s)
- Paula Garcia-Esparcia
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Karina Hernández-Ortega
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Anusha Koneti
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Laura Gil
- Department of Genetics, Medical School, Alfonso X el Sabio University, Villanueva de la Cañada, Madrid, Spain
| | - Raul Delgado-Morales
- Cancer Epigenetics and Biology Program, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Ester Castaño
- Biology-Bellvitge Unit, Scientific and Technological Centers-University of Barcelona (CCiTUB), Hospitalet de Llobregat, Barcelona, Spain
| | - Margarita Carmona
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Isidre Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.
- Institute of Neuropathology, Service of Pathologic Anatomy, Bellvitge University Hospital, carrer Feixa Llarga s/n, 08907, Hospitalet de Llobregat, Spain.
| |
Collapse
|
48
|
Ronsoni MF, Remor AP, Lopes MW, Hohl A, Troncoso IHZ, Leal RB, Boos GL, Kondageski C, Nunes JC, Linhares MN, Lin K, Latini AS, Walz R. Mitochondrial Respiration Chain Enzymatic Activities in the Human Brain: Methodological Implications for Tissue Sampling and Storage. Neurochem Res 2015; 41:880-91. [PMID: 26586405 DOI: 10.1007/s11064-015-1769-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/12/2015] [Accepted: 11/11/2015] [Indexed: 12/25/2022]
Abstract
Mitochondrial respiratory chain complexes enzymatic (MRCCE) activities were successfully evaluated in frozen brain samples. Epilepsy surgery offers an ethical opportunity to study human brain tissue surgically removed to treat drug resistant epilepsies. Epilepsy surgeries are done with hemodynamic and laboratory parameters to maintain physiology, but there are no studies analyzing the association among these parameters and MRCCE activities in the human brain tissue. We determined the intra-operative parameters independently associated with MRCCE activities in middle temporal neocortex (Cx), amygdala (AMY) and head of hippocampus (HIP) samples of patients (n = 23) who underwent temporal lobectomy using multiple linear regressions. MRCCE activities in Cx, AMY and HIP are differentially associated to trans-operative mean arterial blood pressure, O2 saturation, hemoglobin, and anesthesia duration to time of tissue sampling. The time-course between the last seizure occurrence and tissue sampling as well as the sample storage to biochemical assessments were also associated with enzyme activities. Linear regression models including these variables explain 13-17 % of MRCCE activities and show a moderate to strong effect (r = 0.37-0.82). Intraoperative hemodynamic and laboratory parameters as well as the time from last seizure to tissue sampling and storage time are associated with MRCCE activities in human samples from the Cx, AMYG and HIP. Careful control of these parameters is required to minimize confounding biases in studies using human brain samples collected from elective neurosurgery.
Collapse
Affiliation(s)
- Marcelo Fernando Ronsoni
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Aline Pertile Remor
- Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Mark William Lopes
- Laboratório de Transdução de Sinal no Sistema Nervoso Central, Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Alexandre Hohl
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Iris H Z Troncoso
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Rodrigo Bainy Leal
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Laboratório de Transdução de Sinal no Sistema Nervoso Central, Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Gustavo Luchi Boos
- Centro de Ensino e Treinamento Integrado de Anestesiologia, Hospital Governador Celso Ramos (HGCR), Florianópolis, SC, Brazil
| | - Charles Kondageski
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Divisão de Neurocirurgia, Departamento de Cirurgia, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Jean Costa Nunes
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Laboratório de Neuropatologia, Serviço de Patologia, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Marcelo Neves Linhares
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Serviço de Cirurgia de Epilepsia, Hospital Governador Celso Ramos (HGCR), Florianópolis, SC, Brazil
| | - Kátia Lin
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Serviço de Neurologia, Departamento de Clínica Médica, Hospital Universitário, 3 andar, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, 88.040-970, Brazil
| | - Alexandra Susana Latini
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Roger Walz
- Centro de Neurociências Aplicadas, Hospital Universitário (HU), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil. .,Serviço de Neurologia, Departamento de Clínica Médica, Hospital Universitário, 3 andar, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, 88.040-970, Brazil.
| |
Collapse
|
49
|
Caro AA, Bell M, Ejiofor S, Zurcher G, Petersen DR, Ronis MJJ. N-acetylcysteine inhibits the up-regulation of mitochondrial biogenesis genes in livers from rats fed ethanol chronically. Alcohol Clin Exp Res 2015; 38:2896-906. [PMID: 25581647 DOI: 10.1111/acer.12576] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 09/06/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Chronic ethanol (EtOH) administration to experimental animals induces hepatic oxidative stress and up-regulates mitochondrial biogenesis. The mechanisms by which chronic EtOH up-regulates mitochondrial biogenesis have not been fully explored. In this work, we hypothesized that oxidative stress is a factor that triggers mitochondrial biogenesis after chronic EtOH feeding. If our hypothesis is correct, co-administration of antioxidants should prevent up-regulation of mitochondrial biogenesis genes. METHODS Rats were fed an EtOH-containing diet intragastrically by total enteral nutrition for 150 days, in the absence or presence of the antioxidant N-acetylcysteine (NAC) at 1.7 g/kg/d; control rats were administered isocaloric diets where carbohydrates substituted for EtOH calories. RESULTS EtOH administration significantly increased hepatic oxidative stress, evidenced as decreased liver total glutathione and reduced glutathione/glutathione disulfide ratio. These effects were inhibited by co-administration of EtOH and NAC. Chronic EtOH increased the expression of mitochondrial biogenesis genes including peroxisome proliferator-activated receptor gamma-coactivator-1 alpha and mitochondrial transcription factor A, and mitochondrial DNA; co-administration of EtOH and NAC prevented these effects. Chronic EtOH administration was associated with decreased mitochondrial mass, inactivation and depletion of mitochondrial complex I and complex IV, and increased hepatic mitochondrial oxidative damage, effects that were not prevented by NAC. CONCLUSIONS These results suggest that oxidative stress caused by chronic EtOH triggered the up-regulation of mitochondrial biogenesis genes in rat liver, because an antioxidant such as NAC prevented both effects. Because NAC did not prevent liver mitochondrial oxidative damage, extra-mitochondrial effects of reactive oxygen species may regulate mitochondrial biogenesis. In spite of the induction of hepatic mitochondrial biogenesis genes by chronic EtOH, mitochondrial mass and function decreased probably in association with mitochondrial oxidative damage. These results also predict that the effectiveness of NAC as an antioxidant therapy for chronic alcoholism will be limited by its limited antioxidant effects in mitochondria, and its inhibitory effect on mitochondrial biogenesis.
Collapse
Affiliation(s)
- Andres A Caro
- Chemistry Department , Hendrix College, Conway, Arkansas
| | | | | | | | | | | |
Collapse
|
50
|
Endothelial proliferation modulates neuron-glia survival and differentiation in ischemic stress. Ann Neurosci 2015; 22:150-61. [PMID: 26130923 PMCID: PMC4481556 DOI: 10.5214/ans.0972.7531.220305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/06/2015] [Accepted: 02/06/2015] [Indexed: 12/03/2022] Open
Abstract
Background Recent studies have shown that endothelial proliferation and angiogenic response are characteristic of degenerative events, such that the magnitude of endothelial activation is reflective of the progression of neurodegeneration. Purpose This study sets out to, compare, the degenerative changes seen in the parietal cortex (PC) and periventricular zone (PVZ) after cyanide toxicity or vascular occlusion. Methods Global vascular occlusion (VO) and cyanide toxicity (CN) were induced in separate sets of male adult wistar rats for 10 days (treatment phase). Subsequently, the treatment was discontinued for another 10 days (withdrawal phase) (CN-I and VO-I). A separate group of control was maintained for 10 days and received normal saline for this duration. The animals were euthanized at day 10 (treatment and control) and day 20 (withdrawal) after which the tissue was processed for antigen retrieval immunohistochemistry to demonstrate; H&E (general histology) CD31/PECAM 1(endothelial proliferation), CD45 (monocyte/phagocyte), GFAP (glia), NSE (neuron), Ki-67 (cell proliferation) and NF (neurofilament). Total cell count, immunopositive cell counts, arterial wall thickness and lumen width were determined and plotted using ANOVA with significance set at P<0.05*. Results We observed an increase in endothelial proliferation (↑CD31), glia activation and a decrease in neuron count in vascular occlusion and cyanide toxicity after the treatment phase (degeneration). The neuron count increased (↑NSE) after withdrawal of cyanide treatment and vascular occlusion and was accompanied by a corresponding decrease in endothelial and glia activation (↓CD31/GFAP). Degenerative changes were more prominent in cyanide toxicity when compared with vascular occlusion. The increase in CD45 expression coupled with a reduced CD31/GFAP after the withdrawal phase was evident of vascular remodeling and neurosurvival. Conclusion We conclude that neuronal degeneration in cyanide toxicity or vascular occlusion is dependent on an increase in endothelial proliferation (↑CD31), glia activation (↑GFAP) and a decrease in monocyte expression (↓CD45); representing a pro-inflammatory response. Furthermore, cyanide toxicity induced more prominent degenerative changes when compared with the vascular occlusion due to a higher CD31/GFAP expression. Subsequent withdrawal of the ischemia facilitated a reduction in GFAP/CD31 with a corresponding increase in monocytes (↑CD45) for vascular remodeling and neurosurvival. The VO-I showed a significant increase in vascular remodelling than the CN-I due to a more significant increase in monocytic expression (CD45) after the withdrawal of the occlusion. Generally, we found that degeneration was prominent in the parietal cortex and less in the periventricular zone for both forms of ischemia.
Collapse
|