1
|
Riaz Z, Richardson GS, Jin H, Zenitsky G, Anantharam V, Kanthasamy A, Kanthasamy AG. Nuclear pore and nucleocytoplasmic transport impairment in oxidative stress-induced neurodegeneration: relevance to molecular mechanisms in Pathogenesis of Parkinson's and other related neurodegenerative diseases. Mol Neurodegener 2024; 19:87. [PMID: 39578912 PMCID: PMC11585115 DOI: 10.1186/s13024-024-00774-0] [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: 08/23/2024] [Accepted: 10/28/2024] [Indexed: 11/24/2024] Open
Abstract
Nuclear pore complexes (NPCs) are embedded in the nuclear envelope and facilitate the exchange of macromolecules between the nucleus and cytoplasm in eukaryotic cells. The dysfunction of the NPC and nuclear transport plays a significant role in aging and the pathogenesis of various neurodegenerative diseases. Common features among these neurodegenerative diseases, including Parkinson's disease (PD), encompass mitochondrial dysfunction, oxidative stress and the accumulation of insoluble protein aggregates in specific brain regions. The susceptibility of dopaminergic neurons to mitochondrial stress underscores the pivotal role of mitochondria in PD progression. Disruptions in mitochondrial-nuclear communication are exacerbated by aging and α-synuclein-induced oxidative stress in PD. The precise mechanisms underlying mitochondrial impairment-induced neurodegeneration in PD are still unclear. Evidence suggests that perturbations in dopaminergic neuronal nuclei are linked to PD-related neurodegeneration. These perturbations involve structural damage to the nuclear envelope and mislocalization of pivotal transcription factors, potentially driven by oxidative stress or α-synuclein pathology. The presence of protein aggregates, pathogenic mutations, and ongoing oxidative stress can exacerbate the dysfunction of NPCs, yet this mechanism remains understudied in the context of oxidative stress-induced PD. This review summarizes the link between mitochondrial dysfunction and dopaminergic neurodegeneration and outlines the current evidence for nuclear envelope and nuclear transport abnormalities in PD, particularly in oxidative stress. We highlight the potential role of nuclear pore and nucleocytoplasmic transport dysfunction in PD and stress the importance of systematically investigating NPC components in PD.
Collapse
Affiliation(s)
- Zainab Riaz
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Gabriel S Richardson
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Anumantha G Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
2
|
Ullah I, Uddin S, Zhao L, Wang X, Li H. Autophagy and UPS pathway contribute to nicotine-induced protection effect in Parkinson's disease. Exp Brain Res 2024:10.1007/s00221-023-06765-9. [PMID: 38430248 DOI: 10.1007/s00221-023-06765-9] [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: 10/21/2023] [Accepted: 12/11/2023] [Indexed: 03/03/2024]
Abstract
The gradual nature of age-related neurodegeneration causes Parkinson's disease (PD) and impairs movement, memory, intellectual ability, and social interaction. One of the most prevalent neurodegenerative conditions affecting the central nervous system (CNS) among the elderly is PD. PD affects both motor and cognitive functions. Degeneration of dopaminergic (DA) neurons and buildup of the protein α-synuclein (α-Syn) in the substantia nigra pars compacta (SNpc) are two major causes of this disorder. Both UPS and ALS systems serve to eliminate α-Syn. Autophagy and UPS deficits, shortened life duration, and lipofuscin buildup accelerate PD. This sickness has no cure. Innovative therapies are halting PD progression. Bioactive phytochemicals may provide older individuals with a natural substitute to help delay the onset of neurodegenerative illnesses. This study examines whether nicotine helps transgenic C. elegans PD models. According to numerous studies, nicotine enhances synaptic plasticity and dopaminergic neuronal survival. Upgrades UPS pathways, increases autophagy, and decreases oxidative stress and mitochondrial dysfunction. At 100, 150, and 200 µM nicotine levels, worms showed reduced α-Syn aggregation, repaired DA neurotoxicity after 6-OHDA intoxication, increased lifetime, and reduced lipofuscin accumulation. Furthermore, nicotine triggered autophagy and UPS. We revealed nicotine's potential as a UPS and autophagy activator to prevent PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Inam Ullah
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Shahab Uddin
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Longhe Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Xin Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China.
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, Lanzhou, China.
- School of Pharmacy, Lanzhou University, Lanzhou, China.
| |
Collapse
|
3
|
Mahbub NU, Islam MM, Hong ST, Chung HJ. Dysbiosis of the gut microbiota and its effect on α-synuclein and prion protein misfolding: consequences for neurodegeneration. Front Cell Infect Microbiol 2024; 14:1348279. [PMID: 38435303 PMCID: PMC10904658 DOI: 10.3389/fcimb.2024.1348279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
Abnormal behavior of α-synuclein and prion proteins is the hallmark of Parkinson's disease (PD) and prion illnesses, respectively, being complex neurological disorders. A primary cause of protein aggregation, brain injury, and cognitive loss in prion illnesses is the misfolding of normal cellular prion proteins (PrPC) into an infectious form (PrPSc). Aggregation of α-synuclein causes disruptions in cellular processes in Parkinson's disease (PD), leading to loss of dopamine-producing neurons and motor symptoms. Alteration in the composition or activity of gut microbes may weaken the intestinal barrier and make it possible for prions to go from the gut to the brain. The gut-brain axis is linked to neuroinflammation; the metabolites produced by the gut microbiota affect the aggregation of α-synuclein, regulate inflammation and immunological responses, and may influence the course of the disease and neurotoxicity of proteins, even if their primary targets are distinct proteins. This thorough analysis explores the complex interactions that exist between the gut microbiota and neurodegenerative illnesses, particularly Parkinson's disease (PD) and prion disorders. The involvement of the gut microbiota, a complex collection of bacteria, archaea, fungi, viruses etc., in various neurological illnesses is becoming increasingly recognized. The gut microbiome influences neuroinflammation, neurotransmitter synthesis, mitochondrial function, and intestinal barrier integrity through the gut-brain axis, which contributes to the development and progression of disease. The review delves into the molecular mechanisms that underlie these relationships, emphasizing the effects of microbial metabolites such as bacterial lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs) in regulating brain functioning. Additionally, it looks at how environmental influences and dietary decisions affect the gut microbiome and whether they could be risk factors for neurodegenerative illnesses. This study concludes by highlighting the critical role that the gut microbiota plays in the development of Parkinson's disease (PD) and prion disease. It also provides a promising direction for future research and possible treatment approaches. People afflicted by these difficult ailments may find hope in new preventive and therapeutic approaches if the role of the gut microbiota in these diseases is better understood.
Collapse
Affiliation(s)
- Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
4
|
Hou X, Chen TH, Koga S, Bredenberg JM, Faroqi AH, Delenclos M, Bu G, Wszolek ZK, Carr JA, Ross OA, McLean PJ, Murray ME, Dickson DW, Fiesel FC, Springer W. Alpha-synuclein-associated changes in PINK1-PRKN-mediated mitophagy are disease context dependent. Brain Pathol 2023; 33:e13175. [PMID: 37259617 PMCID: PMC10467041 DOI: 10.1111/bpa.13175] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/05/2023] [Indexed: 06/02/2023] Open
Abstract
Alpha-synuclein (αsyn) aggregates are pathological features of several neurodegenerative conditions including Parkinson disease (PD), dementia with Lewy bodies, and multiple system atrophy (MSA). Accumulating evidence suggests that mitochondrial dysfunction and impairments of the autophagic-lysosomal system can contribute to the deposition of αsyn, which in turn may interfere with health and function of these organelles in a potentially vicious cycle. Here we investigated a potential convergence of αsyn with the PINK1-PRKN-mediated mitochondrial autophagy pathway in cell models, αsyn transgenic mice, and human autopsy brain. PINK1 and PRKN identify and selectively label damaged mitochondria with phosphorylated ubiquitin (pS65-Ub) to mark them for degradation (mitophagy). We found that disease-causing multiplications of αsyn resulted in accumulation of the ubiquitin ligase PRKN in cells. This effect could be normalized by starvation-induced autophagy activation and by CRISPR/Cas9-mediated αsyn knockout. Upon acute mitochondrial damage, the increased levels of PRKN protein contributed to an enhanced pS65-Ub response. We further confirmed increased pS65-Ub-immunopositive signals in mouse brain with αsyn overexpression and in postmortem human disease brain. Of note, increased pS65-Ub was associated with neuronal Lewy body-type αsyn pathology, but not glial cytoplasmic inclusions of αsyn as seen in MSA. While our results add another layer of complexity to the crosstalk between αsyn and the PINK1-PRKN pathway, distinct mechanisms may underlie in cells and brain tissue despite similar outcomes. Notwithstanding, our finding suggests that pS65-Ub may be useful as a biomarker to discriminate different synucleinopathies and may serve as a potential therapeutic target for Lewy body disease.
Collapse
Affiliation(s)
- Xu Hou
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Shunsuke Koga
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Ayman H. Faroqi
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | | | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | | | - Jonathan A. Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Owen A. Ross
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Pamela J. McLean
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Melissa E. Murray
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Dennis W. Dickson
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Fabienne C. Fiesel
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Wolfdieter Springer
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| |
Collapse
|
5
|
Datta AK, Mukherjee A, Biswas A. Gastrointestinal, Respiratory, and Olfactory Neurotropism of Sars-Cov2 as a Possible Trigger of Parkinson's Disease: Is a Multi-Hit Multi-Step Process on the Cards. Ann Indian Acad Neurol 2023; 26:127-136. [PMID: 37179662 PMCID: PMC10171009 DOI: 10.4103/aian.aian_767_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/20/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
Since the first emergence of COVID-19 on the global stage, there has been a wealth of evidence to suggest that SARS-Cov2 is not merely a pulmonary pathogen. This virus is unique in its ability to disrupt cellular pathways related to protein homeostasis, mitochondrial function, stress response, and aging. Such effects raise concerns about the long-term fate of survivors of COVID-19 infection, particularly regarding neurodegenerative diseases. The concept of interaction between environmental factors and alpha-synuclein formation in the olfactory bulb and vagal autonomic terminals with subsequent caudo-cranial migration has received much attention in the context of PD pathogenesis. Anosmia and gastrointestinal symptoms are two well-known symptoms of COVID-19, with evidence of an olfactory bulb and vagal infiltration by SARS-CoV2. This raises the possibility of the spread of the viral particles to the brain along multiple cranial nerve routes. Neurotropism, coupled with the ability of the SARS-Cov2 virion to induce abnormal protein folding and stress responses in the central nervous system, in presence of an inflammatory milieu, reinforced by hypoxia, coagulopathy, and endothelial dysfunction, reverberates the intriguing possibility of activation of a neurodegenerative cascade leading to the development of pathological alpha-synuclein aggregates and thus, triggering the development of PD in survivors of COVID19. This review attempts to summarize and critically appraise existing evidence from basic science research and clinical reports of links between COVID-19 and PD and explores the prospect of a multi-hit pathophysiological process, induced by SARS-Cov2 infection, ultimately converging on perturbed cellular protein homeostasis, which although is intriguing, presently lacks robust evidence for confirmation.
Collapse
Affiliation(s)
- Amlan K. Datta
- Department of Neurology, Institute of Post Graduate of Medical Education and Research (IPGME&R) and Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Adreesh Mukherjee
- Department of Neurology, Institute of Post Graduate of Medical Education and Research (IPGME&R) and Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Atanu Biswas
- Department of Neurology, Institute of Post Graduate of Medical Education and Research (IPGME&R) and Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| |
Collapse
|
6
|
Ko TK, Tan DJY. Is Disrupted Mitophagy a Central Player to Parkinson's Disease Pathology? Cureus 2023; 15:e35458. [PMID: 36860818 PMCID: PMC9969326 DOI: 10.7759/cureus.35458] [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] [Accepted: 02/25/2023] [Indexed: 02/27/2023] Open
Abstract
Whilst the pathophysiology at a cellular level has been defined, the cause of Parkinson's disease (PD) remains poorly understood. This neurodegenerative disorder is associated with impaired dopamine transmission in the substantia nigra, and protein accumulations known as Lewy bodies are visible in affected neurons. Cell culture models of PD have indicated impaired mitochondrial function, so the focus of this paper is on the quality control processes involved in and around mitochondria. Mitochondrial autophagy (mitophagy) is the process through which defective mitochondria are removed from the cell by internalisation into autophagosomes which fuse with a lysosome. This process involves many proteins, notably including PINK1 and parkin, both of which are known to be coded on genes associated with PD. Normally in healthy individuals, PINK1 associates with the outer mitochondrial membrane, which then recruits parkin, activating it to attach ubiquitin proteins to the mitochondrial membrane. PINK1, parkin, and ubiquitin cooperate to form a positive feedback system which accelerates the deposition of ubiquitin on dysfunctional mitochondria, resulting in mitophagy. However, in hereditary PD, the genes encoding PINK1 and parkin are mutated, resulting in proteins that are less efficient at removing poorly performing mitochondria, leaving cells more vulnerable to oxidative stress and ubiquitinated inclusion bodies, such as Lewy bodies. Current research that looks into the connection between mitophagy and PD is promising, already yielding potentially therapeutic compounds; until now, pharmacological support for the mitophagy process has not been part of the therapeutic arsenal. Continued research in this area is warranted.
Collapse
Affiliation(s)
- Tsz Ki Ko
- Otolaryngology, College of Life Sciences, Leicester Medical School, George Davies Centre, Leicester, GBR
| | | |
Collapse
|
7
|
Toxicity of extracellular alpha-synuclein is independent of intracellular alpha-synuclein. Sci Rep 2022; 12:21951. [PMID: 36535974 PMCID: PMC9763379 DOI: 10.1038/s41598-022-25790-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Parkinson´s disease (PD) pathology progresses throughout the nervous system. Whereas motor symptoms are always present, there is a high variability in the prevalence of non-motor symptoms. It has been postulated that the progression of the pathology is based on a prion-like disease mechanism partly due to the seeding effect of endocytosed-alpha-synuclein (ASYN) on the endogenous ASYN. Here, we analyzed the role of endogenous ASYN in the progression of PD-like pathology in vivo and in vitro and compared the effect of endocytosed-ASYN as well as paraquat and rotenone on primary enteric, dopaminergic and cortical neurons from wild-type and ASYN-KO mice. Our results show that, in vivo, pathology progression did not occur in the absence of endogenous ASYN. Remarkably, the damage caused by endocytosed-ASYN, rotenone or paraquat was independent from endogenous ASYN and related to the alteration of the host´s mitochondrial membrane potential. Dopaminergic neurons were very sensitive to these noxae compared to other neuronal subtypes. These results suggest that ASYN-mitochondrial interactions play a major role in initiating the pathological process in the host neuron and endogenous ASYN is essential for the transsynaptical transmission of the pathology. Our results also suggest that protecting mitochondrial function is a valid primary therapeutic target.
Collapse
|
8
|
Neth BJ, Bauer BA, Benarroch EE, Savica R. The Role of Intermittent Fasting in Parkinson's Disease. Front Neurol 2021; 12:682184. [PMID: 34140926 PMCID: PMC8203905 DOI: 10.3389/fneur.2021.682184] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Bryan J Neth
- Departments of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Brent A Bauer
- Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | | | - Rodolfo Savica
- Departments of Neurology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
9
|
Alpha-Synuclein and Mitochondrial Dysfunction in Parkinson's Disease: The Emerging Role of VDAC. Biomolecules 2021; 11:biom11050718. [PMID: 34064816 PMCID: PMC8170894 DOI: 10.3390/biom11050718] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 12/12/2022] Open
Abstract
Alpha-Synuclein (αSyn) is a protein whose function is still debated, as well as its role in modulation of mitochondrial function in both physiological and pathological conditions. Mitochondrial porins or Voltage-Dependent Anion Channel (VDAC) proteins are the main gates for ADP/ATP and various substrates towards the organelle. Furthermore, they act as a mitochondrial hub for many cytosolic proteins, including αSyn. This review analyzes the main aspects of αSyn-mitochondria interaction, focusing on the role of VDAC and its emerging involvement in the pathological processes.
Collapse
|
10
|
Kwon O, Song J, Yang Y, Kim S, Kim JY, Seok M, Hwang I, Yu J, Karmacharya J, Maeng H, Kim J, Jho E, Ko SY, Son H, Chang M, Lee S. SGK1 inhibition in glia ameliorates pathologies and symptoms in Parkinson disease animal models. EMBO Mol Med 2021; 13:e13076. [PMID: 33646633 PMCID: PMC8033538 DOI: 10.15252/emmm.202013076] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 01/20/2021] [Accepted: 01/23/2021] [Indexed: 12/13/2022] Open
Abstract
Astrocytes and microglia are brain-resident glia that can establish harmful inflammatory environments in disease contexts and thereby contribute to the progression of neuronal loss in neurodegenerative disorders. Correcting the diseased properties of glia is therefore an appealing strategy for treating brain diseases. Previous studies have shown that serum/ glucocorticoid related kinase 1 (SGK1) is upregulated in the brains of patients with various neurodegenerative disorders, suggesting its involvement in the pathogenesis of those diseases. In this study, we show that inhibiting glial SGK1 corrects the pro-inflammatory properties of glia by suppressing the intracellular NFκB-, NLRP3-inflammasome-, and CGAS-STING-mediated inflammatory pathways. Furthermore, SGK1 inhibition potentiated glial activity to scavenge glutamate toxicity and prevented glial cell senescence and mitochondrial damage, which have recently been reported as critical pathologic features of and therapeutic targets in Parkinson disease (PD) and Alzheimer disease (AD). Along with those anti-inflammatory/neurotrophic functions, silencing and pharmacological inhibition of SGK1 protected midbrain dopamine neurons from degeneration and cured pathologic synuclein alpha (SNCA) aggregation and PD-associated behavioral deficits in multiple in vitro and in vivo PD models. Collectively, these findings suggest that SGK1 inhibition could be a useful strategy for treating PD and other neurodegenerative disorders that share the common pathology of glia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Oh‐Chan Kwon
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Jae‐Jin Song
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
| | - Yunseon Yang
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Seong‐Hoon Kim
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Ji Young Kim
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Min‐Jong Seok
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Inhwa Hwang
- Korea Department of Microbiology and ImmunologyInstitute for Immunology and Immunological DiseasesBrain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulSouth Korea
| | - Je‐Wook Yu
- Korea Department of Microbiology and ImmunologyInstitute for Immunology and Immunological DiseasesBrain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulSouth Korea
| | | | | | - Jiyoung Kim
- Department of Life ScienceUniversity of SeoulSeoulKorea
| | - Eek‐hoon Jho
- Department of Life ScienceUniversity of SeoulSeoulKorea
| | - Seung Yeon Ko
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Hyeon Son
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Mi‐Yoon Chang
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
| | - Sang‐Hun Lee
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| |
Collapse
|
11
|
Wulansari N, Darsono WHW, Woo HJ, Chang MY, Kim J, Bae EJ, Sun W, Lee JH, Cho IJ, Shin H, Lee SJ, Lee SH. Neurodevelopmental defects and neurodegenerative phenotypes in human brain organoids carrying Parkinson's disease-linked DNAJC6 mutations. SCIENCE ADVANCES 2021; 7:eabb1540. [PMID: 33597231 PMCID: PMC7888924 DOI: 10.1126/sciadv.abb1540] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 12/28/2020] [Indexed: 05/14/2023]
Abstract
Loss-of-function mutations of DNAJC6, encoding HSP40 auxilin, have recently been identified in patients with early-onset Parkinson's disease (PD). To study the roles of DNAJC6 in PD pathogenesis, we used human embryonic stem cells with CRISPR-Cas9-mediated gene editing. Here, we show that DNAJC6 mutations cause key PD pathologic features, i.e., midbrain-type dopamine (mDA) neuron degeneration, pathologic α-synuclein aggregation, increase of intrinsic neuronal firing frequency, and mitochondrial and lysosomal dysfunctions in human midbrain-like organoids (hMLOs). In addition, neurodevelopmental defects were also manifested in hMLOs carrying the mutations. Transcriptomic analyses followed by experimental validation revealed that defects in DNAJC6-mediated endocytosis impair the WNT-LMX1A signal during the mDA neuron development. Furthermore, reduced LMX1A expression during development caused the generation of vulnerable mDA neurons with the pathologic manifestations. These results suggest that the human model of DNAJC6-PD recapitulates disease phenotypes and reveals mechanisms underlying disease pathology, providing a platform for assessing therapeutic interventions.
Collapse
Affiliation(s)
- Noviana Wulansari
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Wahyu Handoko Wibowo Darsono
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hye-Ji Woo
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Mi-Yoon Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Jinil Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Eun-Jin Bae
- Department of Biomedical Sciences and Medicine, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ju-Hyun Lee
- Department of Anatomy, Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Il-Joo Cho
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Daejeon, Republic of Korea
- School of Electrical and Electronics Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyogeun Shin
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences and Medicine, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea.
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Polansky H, Lori G. How microcompetition with latent viruses can cause α synuclein aggregation, mitochondrial dysfunction, and eventually Parkinson's disease. J Neurovirol 2021; 27:52-57. [PMID: 33405201 DOI: 10.1007/s13365-020-00929-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 10/19/2020] [Accepted: 11/19/2020] [Indexed: 11/24/2022]
Abstract
The cause of most Parkinson's disease cases is unknown. However, it is well documented that mitochondrial dysfunction and misfolded α synuclein aggregation are important cellular abnormalities associated with the disease. In this paper, we use the microcompetition model to show how latent viruses, which infect the central and peripheral nervous systems, can cause the observed mitochondrial dysfunction and excess α synuclein aggregation, and eventually, Parkinson's disease.
Collapse
Affiliation(s)
- Hanan Polansky
- The Center for the Biology of Chronic Disease (CBCD), 3 Germay Dr, Wilmington, DE, 19804, USA.
| | - Gillad Lori
- The Center for the Biology of Chronic Disease (CBCD), 3 Germay Dr, Wilmington, DE, 19804, USA
| |
Collapse
|
13
|
Michalska P, León R. When It Comes to an End: Oxidative Stress Crosstalk with Protein Aggregation and Neuroinflammation Induce Neurodegeneration. Antioxidants (Basel) 2020; 9:antiox9080740. [PMID: 32806679 PMCID: PMC7463521 DOI: 10.3390/antiox9080740] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/27/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are characterized by a progressive loss of neurons in the brain or spinal cord that leads to a loss of function of the affected areas. The lack of effective treatments and the ever-increasing life expectancy is raising the number of individuals affected, having a tremendous social and economic impact. The brain is particularly vulnerable to oxidative damage given the high energy demand, low levels of antioxidant defenses, and high levels of metal ions. Driven by age-related changes, neurodegeneration is characterized by increased oxidative stress leading to irreversible neuronal damage, followed by cell death. Nevertheless, neurodegenerative diseases are known as complex pathologies where several mechanisms drive neuronal death. Herein we discuss the interplay among oxidative stress, proteinopathy, and neuroinflammation at the early stages of neurodegenerative diseases. Finally, we discuss the use of the Nrf2-ARE pathway as a potential therapeutic strategy based on these molecular mechanisms to develop transformative medicines.
Collapse
Affiliation(s)
- Patrycja Michalska
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
- Correspondence: (P.M.); (R.L.); Tel.: +34-91-497-27-66 (P.M. & R.L.)
| | - Rafael León
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28006 Madrid, Spain
- Correspondence: (P.M.); (R.L.); Tel.: +34-91-497-27-66 (P.M. & R.L.)
| |
Collapse
|
14
|
Mashima K, Takahashi S, Minami K, Izawa Y, Abe T, Tsukada N, Hishiki T, Suematsu M, Kajimura M, Suzuki N. Neuroprotective Role of Astroglia in Parkinson Disease by Reducing Oxidative Stress Through Dopamine-Induced Activation of Pentose-Phosphate Pathway. ASN Neuro 2019; 10:1759091418775562. [PMID: 29768946 PMCID: PMC5960859 DOI: 10.1177/1759091418775562] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress plays an important role in the onset and progression of Parkinson disease. Although released dopamine at the synaptic terminal is mostly reabsorbed by dopaminergic neurons, some dopamine is presumably taken up by astroglia. This study examined the dopamine-induced astroglial protective function through the activation of the pentose-phosphate pathway (PPP) to reduce reactive oxygen species (ROS). In vitro experiments were performed using striatal neurons and cortical or striatal astroglia prepared from Sprague-Dawley rats or C57BL/6 mice. The rates of glucose phosphorylation in astroglia were evaluated using the [14C]deoxyglucose method. PPP activity was measured using [1-14C]glucose and [6-14C]glucose after acute (60 min) or chronic (15 hr) exposure to dopamine. ROS production was measured using 2',7'-dichlorodihydrofluorescein diacetate. The involvement of the Kelch-like ECH-associated protein 1 (Keap1) or nuclear factor-erythroid-2-related factor 2 (Nrf2) system was evaluated using Nrf2 gene knockout mice, immunohistochemistry, and quantitative reverse transcription polymerase chain reaction analysis for heme oxygenase-1. Acute exposure to dopamine elicited increases in astroglial glucose consumption with lactate release. PPP activity in astroglia was robustly enhanced independently of Na+-dependent monoamine transporters. In contrast, chronic exposure to dopamine induced moderate increases in PPP activity via the Keap1/Nrf2 system. ROS production from dopamine increased gradually over 12 hr. Dopamine induced neuronal cell damage that was prevented by coculturing with astroglia but not with Nrf2-deficient astroglia. Dopamine-enhanced astroglial PPP activity in both acute and chronic manners may possibly reduce neuronal oxidative stress.
Collapse
Affiliation(s)
- Kyoko Mashima
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Shinichi Takahashi
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kazushi Minami
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshikane Izawa
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Takato Abe
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan.,2 Department of Neurology, Graduate School of Medicine, Osaka City University, Japan
| | - Naoki Tsukada
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Takako Hishiki
- 3 Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan.,4 Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- 4 Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Kajimura
- 5 Department of Biology, Keio University School of Medicine, Yokohama, Japan
| | - Norihiro Suzuki
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Segura-Aguilar J. On the Role of Aminochrome in Mitochondrial Dysfunction and Endoplasmic Reticulum Stress in Parkinson's Disease. Front Neurosci 2019; 13:271. [PMID: 30983959 PMCID: PMC6449441 DOI: 10.3389/fnins.2019.00271] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/07/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology, Faculty of Medicine, ICBM, University of Chile, Santiago, Chile
| |
Collapse
|
16
|
Bussi C, Peralta Ramos JM, Arroyo DS, Gallea JI, Ronchi P, Kolovou A, Wang JM, Florey O, Celej MS, Schwab Y, Ktistakis NT, Iribarren P. Alpha-synuclein fibrils recruit TBK1 and OPTN to lysosomal damage sites and induce autophagy in microglial cells. J Cell Sci 2018; 131:jcs226241. [PMID: 30404831 PMCID: PMC6518333 DOI: 10.1242/jcs.226241] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 01/05/2023] Open
Abstract
Autophagic dysfunction and protein aggregation have been linked to several neurodegenerative disorders, but the exact mechanisms and causal connections are not clear and most previous work was done in neurons and not in microglial cells. Here, we report that exogenous fibrillary, but not monomeric, alpha-synuclein (AS, also known as SNCA) induces autophagy in microglial cells. We extensively studied the dynamics of this response using both live-cell imaging and correlative light-electron microscopy (CLEM), and found that it correlates with lysosomal damage and is characterised by the recruitment of the selective autophagy-associated proteins TANK-binding kinase 1 (TBK1) and optineurin (OPTN) to ubiquitylated lysosomes. In addition, we observed that LC3 (MAP1LC3B) recruitment to damaged lysosomes was dependent on TBK1 activity. In these fibrillar AS-treated cells, autophagy inhibition impairs mitochondrial function and leads to microglial cell death. Our results suggest that microglial autophagy is induced in response to lysosomal damage caused by persistent accumulation of AS fibrils. Importantly, triggering of the autophagic response appears to be an attempt at lysosomal quality control and not for engulfment of fibrillar AS.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Claudio Bussi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Javier M Peralta Ramos
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Daniela S Arroyo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Jose I Gallea
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC-CONICET), Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Paolo Ronchi
- EMBL, Electron Microscopy Core Facility, Heidelberg 69117, Germany
| | | | - Ji M Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 20982, USA
| | - Oliver Florey
- Babraham Institute, Signalling Programme, Cambridge CB22 3AT, UK
| | - Maria S Celej
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC-CONICET), Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Yannick Schwab
- EMBL, Electron Microscopy Core Facility, Heidelberg 69117, Germany
- EMBL, Cell Biology and Biophysics Unit, Heidelberg 69117, Germany
| | | | - Pablo Iribarren
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| |
Collapse
|
17
|
Nuñez MT, Chana-Cuevas P. New Perspectives in Iron Chelation Therapy for the Treatment of Neurodegenerative Diseases. Pharmaceuticals (Basel) 2018; 11:ph11040109. [PMID: 30347635 PMCID: PMC6316457 DOI: 10.3390/ph11040109] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023] Open
Abstract
Iron chelation has been introduced as a new therapeutic concept for the treatment of neurodegenerative diseases with features of iron overload. At difference with iron chelators used in systemic diseases, effective chelators for the treatment of neurodegenerative diseases must cross the blood–brain barrier. Given the promissory but still inconclusive results obtained in clinical trials of iron chelation therapy, it is reasonable to postulate that new compounds with properties that extend beyond chelation should significantly improve these results. Desirable properties of a new generation of chelators include mitochondrial destination, the center of iron-reactive oxygen species interaction, and the ability to quench free radicals produced by the Fenton reaction. In addition, these chelators should have moderate iron binding affinity, sufficient to chelate excessive increments of the labile iron pool, estimated in the micromolar range, but not high enough to disrupt physiological iron homeostasis. Moreover, candidate chelators should have selectivity for the targeted neuronal type, to lessen unwanted secondary effects during long-term treatment. Here, on the basis of a number of clinical trials, we discuss critically the current situation of iron chelation therapy for the treatment of neurodegenerative diseases with an iron accumulation component. The list includes Parkinson’s disease, Friedreich’s ataxia, pantothenate kinase-associated neurodegeneration, Huntington disease and Alzheimer’s disease. We also review the upsurge of new multifunctional iron chelators that in the future may replace the conventional types as therapeutic agents for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Marco T Nuñez
- Faculty of Sciences, Universidad de Chile, Las Palmeras 3425, Santiago 7800024, Chile.
| | - Pedro Chana-Cuevas
- Center for the Treatment of Movement Disorders, Universidad de Santiago de Chile, Belisario Prat 1597, Santiago 83800000, Chile.
| |
Collapse
|
18
|
Chiu CC, Lu CS, Weng YH, Chen YL, Huang YZ, Chen RS, Cheng YC, Huang YC, Liu YC, Lai SC, Lin KJ, Lin YW, Chen YJ, Chen CL, Yeh TH, Wang HL. PARK14 (D331Y) PLA2G6 Causes Early-Onset Degeneration of Substantia Nigra Dopaminergic Neurons by Inducing Mitochondrial Dysfunction, ER Stress, Mitophagy Impairment and Transcriptional Dysregulation in a Knockin Mouse Model. Mol Neurobiol 2018; 56:3835-3853. [PMID: 30088174 DOI: 10.1007/s12035-018-1118-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/11/2018] [Indexed: 12/27/2022]
Abstract
PARK14 patients with homozygous (D331Y) PLA2G6 mutation display motor deficits of pure early-onset Parkinson's disease (PD). The aim of this study is to investigate the pathogenic mechanism of mutant (D331Y) PLA2G6-induced PD. We generated knockin (KI) mouse model of PARK14 harboring homozygous (D331Y) PLA2G6 mutation. Then, we investigated neuropathological and neurological phenotypes of PLA2G6D331Y/D331Y KI mice and molecular pathogenic mechanisms of (D331Y) PLA2G6-induced degeneration of substantia nigra (SN) dopaminergic neurons. Six-or nine-month-old PLA2G6D331Y/D331Y KI mice displayed early-onset cell death of SNpc dopaminergic neurons. Lewy body pathology was found in the SN of PLA2G6D331Y/D331Y mice. Six-or nine-month-old PLA2G6D331Y/D331Y KI mice exhibited early-onset parkinsonism phenotypes. Disrupted cristae of mitochondria were found in SNpc dopaminergic neurons of PLA2G6D331Y/D331Y mice. PLA2G6D331Y/D331Y mice displayed mitochondrial dysfunction and upregulated ROS production, which may lead to activation of apoptotic cascade. Upregulated protein levels of Grp78, IRE1, PERK, and CHOP, which are involved in activation of ER stress, were found in the SN of PLA2G6D331Y/D331Y mice. Protein expression of mitophagic proteins, including parkin and BNIP3, was downregulated in the SN of PLA2G6D331Y/D331Y mice, suggesting that (D331Y) PLA2G6 mutation causes mitophagy dysfunction. In the SN of PLA2G6D331Y/D331Y mice, mRNA levels of eight genes that are involved in neuroprotection/neurogenesis were decreased, while mRNA levels of two genes that promote apoptotic death were increased. Our results suggest that PARK14 (D331Y) PLA2G6 mutation causes degeneration of SNpc dopaminergic neurons by causing mitochondrial dysfunction, elevated ER stress, mitophagy impairment, and transcriptional abnormality.
Collapse
Affiliation(s)
- Ching-Chi Chiu
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chin-Song Lu
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Hsin Weng
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying-Ling Chen
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Ying-Zu Huang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
| | - Rou-Shayn Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yin-Cheng Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yu-Chuan Liu
- Division of Sports Medicine, Taiwan Landseed Hospital, Taoyuan, Taiwan
| | - Szu-Chia Lai
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Jun Lin
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Molecular Imaging Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yan-Wei Lin
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yu-Jie Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan
| | - Chao-Lang Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan
| | - Tu-Hsueh Yeh
- Department of Neurology, Taipei Medical University Hospital, No. 252, Wuxing St, Xinyi District, Taipei City, 110, Taiwan. .,School of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Hung-Li Wang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan. .,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan. .,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan. .,Department of Physiology and Pharmacology, Chang Gung University College of Medicine, No. 259, Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan.
| |
Collapse
|
19
|
Segura-Aguilar J, Huenchuguala S. Aminochrome Induces Irreversible Mitochondrial Dysfunction by Inducing Autophagy Dysfunction in Parkinson's Disease. Front Neurosci 2018; 12:106. [PMID: 29593482 PMCID: PMC5859232 DOI: 10.3389/fnins.2018.00106] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/12/2018] [Indexed: 01/21/2023] Open
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), University of Chile, Santiago, Chile
| | - Sandro Huenchuguala
- Departamento de Ciencias Biológicas y Químicas, Facultad de Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| |
Collapse
|
20
|
Tas D, Stickley L, Miozzo F, Koch R, Loncle N, Sabado V, Gnägi B, Nagoshi E. Parallel roles of transcription factors dFOXO and FER2 in the development and maintenance of dopaminergic neurons. PLoS Genet 2018. [PMID: 29529025 PMCID: PMC5864087 DOI: 10.1371/journal.pgen.1007271] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Forkhead box (FOXO) proteins are evolutionarily conserved, stress-responsive transcription factors (TFs) that can promote or counteract cell death. Mutations in FOXO genes are implicated in numerous pathologies, including age-dependent neurodegenerative disorders, such as Parkinson’s disease (PD). However, the complex regulation and downstream mechanisms of FOXOs present a challenge in understanding their roles in the pathogenesis of PD. Here, we investigate the involvement of FOXO in the death of dopaminergic (DA) neurons, the key pathological feature of PD, in Drosophila. We show that dFOXO null mutants exhibit a selective loss of DA neurons in the subgroup crucial for locomotion, the protocerebral anterior medial (PAM) cluster, during development as well as in adulthood. PAM neuron-targeted adult-restricted knockdown demonstrates that dFOXO in adult PAM neurons tissue-autonomously promotes neuronal survival during aging. We further show that dFOXO and the bHLH-TF 48-related-2 (FER2) act in parallel to protect PAM neurons from different forms of cellular stress. Remarkably, however, dFOXO and FER2 share common downstream processes leading to the regulation of autophagy and mitochondrial morphology. Thus, overexpression of one can rescue the loss of function of the other. These results indicate a role of dFOXO in neuroprotection and highlight the notion that multiple genetic and environmental factors interact to increase the risk of DA neuron degeneration and the development of PD. PD, mainly characterized by a progressive loss of dopaminergic neurons in the substantia nigra (SN), is the most prevalent neurodegenerative movement disorder affecting more than 6 million people worldwide. Despite the discovery of several genes linked to familial PD, our understanding of its pathogenesis remains limited, as approximately 90% of the PD cases are sporadic with no apparent genetic linkage. Genome-wide expression studies have implicated the stress-responsive TF FOXO in PD. However, the exact role of FOXO in the survival of DA neurons and PD pathogenesis is still poorly understood. Here, we use fruit flies to address the role of FOXO in the maintenance of DA neurons. dFOXO (Drosophila FOXO) null mutants show a progressive loss of DA neurons in the subgroup essential for locomotion, a phenotype identical to that of Fer2 mutants. Remarkably, dFOXO and FER2 act in parallel pathways to protect PAM neurons from different cellular stressors, but both pathways contribute to the regulation of autophagy and mitochondrial biology. These results demonstrate that dFOXO is required for the maintenance of DA neurons important for locomotion and shed new light on the molecular mechanisms underpinning the complex gene-environment interactions affecting DA neuron survival and PD pathogenesis.
Collapse
Affiliation(s)
- Damla Tas
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Luca Stickley
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Federico Miozzo
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Rafael Koch
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Nicolas Loncle
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Virginie Sabado
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Bettina Gnägi
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, Bern, CH, Switzerland
| | - Emi Nagoshi
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
21
|
Bal-Price A, Meek MEB. Adverse outcome pathways: Application to enhance mechanistic understanding of neurotoxicity. Pharmacol Ther 2017; 179:84-95. [PMID: 28529068 PMCID: PMC5869951 DOI: 10.1016/j.pharmthera.2017.05.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent developments have prompted the transition of empirically based testing of late stage toxicity in animals for a range of different endpoints including neurotoxicity to more efficient and predictive mechanistically based approaches with greater emphasis on measurable key events early in the progression of disease. The adverse outcome pathway (AOP) has been proposed as a simplified organizational construct to contribute to this transition by linking molecular initiating events and earlier (more predictive) key events at lower levels of biological organization to disease outcomes. As such, AOPs are anticipated to facilitate the compilation of information to increase mechanistic understanding of pathophysiological pathways that are responsible for human disease. In this review, the sequence of key events resulting in adverse outcome (AO) defined as parkinsonian motor impairment and learning and memory deficit in children, triggered by exposure to environmental chemicals has been briefly described using the AOP framework. These AOPs follow convention adopted in an Organization for Economic Cooperation and Development (OECD) AOP development program, publically available, to permit tailored application of AOPs for a range of different purposes. Due to the complexity of disease pathways, including neurodegenerative disorders, a specific symptom of the disease (e.g. parkinsonian motor deficit) is considered as the AO in a developed AOP. Though the description is necessarily limited by the extent of current knowledge, additional characterization of involved pathways through description of related AOPs interlinked into networks for the same disease has potential to contribute to more holistic and mechanistic understanding of the pathophysiological pathways involved, possibly leading to the mechanism-based reclassification of diseases, thus facilitating more personalized treatment.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission Joint Research Centre, Directorate F - Health, Consumers and Reference Materials, Ispra, Italy.
| | - M E Bette Meek
- McLaughlin Centre for Risk Science, University of Ottawa, Ottawa, Canada
| |
Collapse
|
22
|
Low-Intensity Ultrasound Decreases α-Synuclein Aggregation via Attenuation of Mitochondrial Reactive Oxygen Species in MPP(+)-Treated PC12 Cells. Mol Neurobiol 2016; 54:6235-6244. [PMID: 27714630 DOI: 10.1007/s12035-016-0104-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/06/2016] [Indexed: 12/23/2022]
Abstract
Many studies have shown that mitochondrial dysfunction and the subsequent oxidative stress caused by excessive reactive oxygen species (ROS) generation play a central role in the pathogenesis of Parkinson's disease (PD). We have previously shown that low-intensity ultrasound (LIUS) could reduce ROS generation by L-buthionine-(S,R)-sulfoximine (BSO) in retinal pigment epithelial cells. In this study, we studied the effects of LIUS stimulation on the ROS-dependent α-synuclein aggregation in 1-methyl-4-phenylpyridinium ion (MPP+)-treated PC12 cells. We found that LIUS stimulation suppressed the MPP+-induced ROS generation and inhibition of mitochondrial complex I activity in PC12 cells in an intensity-dependent manner at 30, 50, and 100 mW/cm2. Furthermore, LIUS stimulation at 100 mW/cm2 suppressed inhibition of mitochondrial complex activity by MPP+ and actually resulted in a decrease of α-synuclein phosphorylation and aggregation induced by MMP+ treatment in PC12 cells. LIUS stimulation also inhibited expression of casein kinase 2 (CK2) that appears to mediate ROS-dependent α-synuclein aggregation. Finally, LIUS stimulation alleviated the death of PC12 cells by MPP+ treatment in an intensity-dependent manner. We, hence, suggest that LIUS stimulation inhibits ROS generation by MPP+ treatment, thereby suppressing α-synuclein aggregation in PC12 cells.
Collapse
|
23
|
M'Angale PG, Staveley BE. Bcl-2 homologue Debcl enhances α-synuclein-induced phenotypes in Drosophila. PeerJ 2016; 4:e2461. [PMID: 27672511 PMCID: PMC5028777 DOI: 10.7717/peerj.2461] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022] Open
Abstract
Background Parkinson disease (PD) is a debilitating movement disorder that afflicts 1–2% of the population over 50 years of age. The common hallmark for both sporadic and familial forms of PD is mitochondrial dysfunction. Mammals have at least twenty proapoptotic and antiapoptotic Bcl-2 family members, in contrast, only two Bcl-2 family genes have been identified in Drosophila melanogaster, the proapoptotic mitochondrial localized Debcl and the antiapoptotic Buffy. The expression of the human transgene α-synuclein, a gene that is strongly associated with inherited forms of PD, in dopaminergic neurons (DA) of Drosophila, results in loss of neurons and locomotor dysfunction to model PD in flies. The altered expression of Debcl in the DA neurons and neuron-rich eye and along with the expression of α-synuclein offers an opportunity to highlight the role of Debcl in mitochondrial-dependent neuronal degeneration and death. Results The directed overexpression of Debcl using the Ddc-Gal4 transgene in the DA of Drosophila resulted in flies with severely decreased survival and a premature age-dependent loss in climbing ability. The inhibition of Debcl resulted in enhanced survival and improved climbing ability whereas the overexpression of Debcl in the α-synuclein-induced Drosophila model of PD resulted in more severe phenotypes. In addition, the co-expression of Debcl along with Buffy partially counteracts the Debcl-induced phenotypes, to improve the lifespan and the associated loss of locomotor ability observed. In complementary experiments, the overexpression of Debcl along with the expression of α-synuclein in the eye, enhanced the eye ablation that results from the overexpression of Debcl. The co-expression of Buffy along with Debcl overexpression results in the rescue of the moderate developmental eye defects. The co-expression of Buffy along with inhibition of Debcl partially restores the eye to a roughened eye phenotype. Discussion The overexpression of Debcl in DA neurons produces flies with shortened lifespan and impaired locomotor ability, phenotypes that are strongly associated with models of PD in Drosophila. The co-expression of Debcl along with α-synuclein enhanced the PD-like phenotypes. The co-expression of Debcl along with Buffy suppresses these phenotypes. Complementary experiments in the Drosophila eye show similar trends during development. Taken all together these results suggest a role for Debcl in neurodegenerative disorders.
Collapse
Affiliation(s)
- P Githure M'Angale
- Department of Biology, Memorial University of Newfoundland , St. John's, Newfoundland and Labrador , Canada
| | - Brian E Staveley
- Department of Biology, Memorial University of Newfoundland , St. John's, Newfoundland and Labrador , Canada
| |
Collapse
|
24
|
Spano M, Signorelli M, Vitaliani R, Aguglia E, Giometto B. The possible involvement of mitochondrial dysfunctions in Lewy body dementia: a systematic review. FUNCTIONAL NEUROLOGY 2016; 30:151-8. [PMID: 26346695 DOI: 10.11138/fneur/2015.30.3.151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The hallmark of dementia with Lewy bodies (DLB) is the “Lewy body”, an abnormal aggregation of alpha-synuclein found in some areas of the brain. The brain is the organ/system that is most vulnerable to this oxidative damage, and reactive oxygen species can cause neurodegenerative diseases. Different models of mitochondrial deregulation have been compared in DLB. The results are consistent with the hypothesis that alpha-synuclein affects the mitochondria themselves, increasing their sensitivity or leading to cell death through protective (neurosin) and accelerating (cytochrome c) factors. This systematic review suggests that mitochondria play an important role in neurodegeneration and a crucial role in the formation of Lewy bodies. DLB is a disease characterized by abnormal accumulation of alpha-synuclein that could result in the release of cytochrome c and subsequent activation of the apoptotic cascade.
Collapse
|
25
|
M'Angale PG, Staveley BE. The Bcl-2 homologue Buffy rescues α-synuclein-induced Parkinson disease-like phenotypes in Drosophila. BMC Neurosci 2016; 17:24. [PMID: 27192974 PMCID: PMC4872331 DOI: 10.1186/s12868-016-0261-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/11/2016] [Indexed: 01/09/2023] Open
Abstract
Background In contrast to the complexity found in mammals, only two Bcl-2 family genes have been found in Drosophila melanogaster including the pro-cell survival, human Bok-related orthologue, Buffy. The directed expression of α-synuclein, the first gene identified to contribute to inherited forms of Parkinson disease (PD), in the dopaminergic neurons (DA) of flies has provided a robust and well-studied Drosophila model of PD complete with the loss of neurons and accompanying motor defects. To more fully understand the biological basis of Bcl-2 genes in PD, we altered the expression of Buffy in the dopamine producing neurons with and without the expression of α-synuclein, and in the developing neuron-rich eye. Results To alter the expression of Buffy in the dopaminergic neurons of Drosophila, the Ddc-Gal4 transgene was used. The directed expression of Buffy in the dopamine producing neurons resulted in flies with increased climbing ability and enhanced survival, while the inhibition of Buffy in the dopaminergic neurons reduced climbing ability over time prematurely, similar to the phenotype observed in the α-synuclein-induced Drosophila model of PD. Subsequently, the expression of Buffy was altered in the α-synuclein-induced Drosophila model of PD. Analysis revealed that Buffy acted to rescue the associated loss of locomotor ability observed in the α-synuclein-induced model of PD, while Buffy RNA interference resulted in an enhanced α-synuclein-induced loss of climbing ability. In complementary experiments the overexpression of Buffy in the developing eye suppressed the mild rough eye phenotype that results from Gal4 expression and from α-synuclein expression. When Buffy is inhibited the roughened eye phenotype is enhanced. Conclusions The inhibition of Buffy in DA neurons produces a novel model of PD in Drosophila. The directed expression of Buffy in DA neurons provide protection and counteracts the α-synuclein-induced Parkinson disease-like phenotypes. Taken all together this demonstrates a role for Buffy, a Bcl-2 pro-cell survival gene, in neuroprotection.
Collapse
Affiliation(s)
- P Githure M'Angale
- Department of Biology, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Brian E Staveley
- Department of Biology, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada.
| |
Collapse
|
26
|
Pahrudin Arrozi A, Wan Ngah WZ, Mohd Yusof YA, Ahmad Damanhuri MH, Makpol S. Antioxidant modulation in restoring mitochondrial function in neurodegeneration. Int J Neurosci 2016; 127:218-235. [PMID: 27074540 DOI: 10.1080/00207454.2016.1178261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the leading causes of disability associated with neurodegeneration worldwide. These diseases are influenced by multiple genetic and environmental factors and share similar mechanisms as both are characterized by accumulation and aggregation of misfolded proteins - amyloid-beta (Aβ) in AD and α-synuclein in PD. Over the past decade, increasing evidence has shown that mitochondrial dysfunction and the generation of reactive oxygen species (ROS) are involved in the pathology of these diseases, and the contributions of these defects to the cellular and molecular changes that eventually cause neuronal death have been explored. Using mitochondrial protective agents, such as antioxidants, to combat ROS provides a new strategy for neurodegenerative treatment. In this review, we highlight the potential of multiple types of antioxidants, including vitamins, phytochemicals, fatty acids and minerals, as well as synthetic antioxidants specifically targeting the mitochondria, which can restore mitochondrial function, in the treatment of neurodegenerative disorders at both the pre-clinical and clinical stages by focusing on AD and PD.
Collapse
Affiliation(s)
- Aslina Pahrudin Arrozi
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | - Wan Zurinah Wan Ngah
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | - Yasmin Anum Mohd Yusof
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | | | - Suzana Makpol
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| |
Collapse
|
27
|
Angione C, Costanza J, Carapezza G, Lió P, Nicosia G. Multi-Target Analysis and Design of Mitochondrial Metabolism. PLoS One 2015; 10:e0133825. [PMID: 26376088 PMCID: PMC4574446 DOI: 10.1371/journal.pone.0133825] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 07/02/2015] [Indexed: 12/30/2022] Open
Abstract
Analyzing and optimizing biological models is often identified as a research priority in biomedical engineering. An important feature of a model should be the ability to find the best condition in which an organism has to be grown in order to reach specific optimal output values chosen by the researcher. In this work, we take into account a mitochondrial model analyzed with flux-balance analysis. The optimal design and assessment of these models is achieved through single- and/or multi-objective optimization techniques driven by epsilon-dominance and identifiability analysis. Our optimization algorithm searches for the values of the flux rates that optimize multiple cellular functions simultaneously. The optimization of the fluxes of the metabolic network includes not only input fluxes, but also internal fluxes. A faster convergence process with robust candidate solutions is permitted by a relaxed Pareto dominance, regulating the granularity of the approximation of the desired Pareto front. We find that the maximum ATP production is linked to a total consumption of NADH, and reaching the maximum amount of NADH leads to an increasing request of NADH from the external environment. Furthermore, the identifiability analysis characterizes the type and the stage of three monogenic diseases. Finally, we propose a new methodology to extend any constraint-based model using protein abundances.
Collapse
Affiliation(s)
- Claudio Angione
- Computer Laboratory-University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| | - Jole Costanza
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, Milan, Italy
| | - Giovanni Carapezza
- Department of Mathematics and Computer Science-University of Catania, Catania, Italy
| | - Pietro Lió
- Computer Laboratory-University of Cambridge, Cambridge, United Kingdom
| | - Giuseppe Nicosia
- Department of Mathematics and Computer Science-University of Catania, Catania, Italy
| |
Collapse
|
28
|
Plotegher N, Stringari C, Jahid S, Veronesi M, Girotto S, Gratton E, Bubacco L. NADH fluorescence lifetime is an endogenous reporter of α-synuclein aggregation in live cells. FASEB J 2015; 29:2484-94. [PMID: 25713058 DOI: 10.1096/fj.14-260281] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 02/06/2015] [Indexed: 12/22/2022]
Abstract
α-Synuclein (aS) aggregation has been amply investigated for its involvement in Parkinson's disease because its amyloid fibrils are the main constituent of Lewy bodies, one of the hallmarks of the disease. aS aggregation was studied here in vitro and in cellular models to correlate aggregation products with toxicity mechanisms. Independent results published elsewhere suggested that aS overexpression and/or aggregation may impair cellular metabolism and cause mitochondrial damage. In this context, we report the characterization of changes in NADH fluorescence properties in vitro and in human embryonic kidney 293 cells upon aS aggregation. The application of the phasor approach to study NADH fluorescence lifetime and emission allowed us to identify changes that correlate with aS aggregation. In particular, the fraction of bound NADH, characterized by longer lifetimes in comparison to free NADH, is increased, and the maximum of the NADH emission is shifted toward shorter wavelengths in the presence of aggregating aS both in vitro and in cells. These data suggest that NADH binds to aggregated aS. NMR experiments in vitro substantiate such binding, which occurs during aggregation. NADH fluorescence is thus useful to detect aS aggregation and by extension the associated oxidative stress.
Collapse
Affiliation(s)
- Nicoletta Plotegher
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Chiara Stringari
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Sohail Jahid
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Marina Veronesi
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Stefania Girotto
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Enrico Gratton
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Luigi Bubacco
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
29
|
Rcom-H'cheo-Gauthier A, Goodwin J, Pountney DL. Interactions between calcium and alpha-synuclein in neurodegeneration. Biomolecules 2014; 4:795-811. [PMID: 25256602 PMCID: PMC4192672 DOI: 10.3390/biom4030795] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/25/2014] [Accepted: 07/14/2014] [Indexed: 12/01/2022] Open
Abstract
In Parkinson’s disease and some atypical Parkinson’s syndromes, aggregation of the α-synuclein protein (α-syn) has been linked to neurodegeneration. Many triggers for pathological α-syn aggregation have been identified, including port-translational modifications, oxidative stress and raised metal ions, such as Ca2+. Recently, it has been found using cell culture models that transient increases of intracellular Ca2+ induce cytoplasmic α-syn aggregates. Ca2+-dependent α-syn aggregation could be blocked by the Ca2+ buffering agent, BAPTA-AM, or by the Ca2+ channel blocker, Trimethadione. Furthermore, a greater proportion of cells positive for aggregates occurred when both raised Ca2+ and oxidative stress were combined, indicating that Ca2+ and oxidative stress cooperatively promote α-syn aggregation. Current on-going work using a unilateral mouse lesion model of Parkinson’s disease shows a greater proportion of calbindin-positive neurons survive the lesion, with intracellular α-syn aggregates almost exclusively occurring in calbindin-negative neurons. These and other recent findings are reviewed in the context of neurodegenerative pathologies and suggest an association between raised Ca2+, α-syn aggregation and neurotoxicity.
Collapse
Affiliation(s)
- Alex Rcom-H'cheo-Gauthier
- Griffith Health Institute, School of Medical Science, Griffith University, Gold Coast, Queensland 4222, Australia.
| | - Jacob Goodwin
- Griffith Health Institute, School of Medical Science, Griffith University, Gold Coast, Queensland 4222, Australia.
| | - Dean L Pountney
- Griffith Health Institute, School of Medical Science, Griffith University, Gold Coast, Queensland 4222, Australia.
| |
Collapse
|
30
|
Pan-Montojo F, Reichmann H. Considerations on the role of environmental toxins in idiopathic Parkinson's disease pathophysiology. Transl Neurodegener 2014; 3:10. [PMID: 24826210 PMCID: PMC4019355 DOI: 10.1186/2047-9158-3-10] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/29/2014] [Indexed: 12/21/2022] Open
Abstract
Neurodegenerative diseases are characterized by a progressive dysfunction of the nervous system. Often associated with atrophy of the affected central or peripheral nervous structures, they include diseases such as Parkinson’s Disease (PD), Alzheimer’s Disease and other dementias, Genetic Brain Disorders, Amyotrophic Lateral Sclerosis (ALS or Lou Gehrig’s Disease), Huntington’s Disease, Prion Diseases, and others. The prevalence of neurodegenerative diseases has increased over the last years. This has had a major impact both on patients and their families and has exponentially increased the medical bill by hundreds of billions of Euros. Therefore, understanding the role of environmental and genetic factors in the pathogenesis of PD is crucial to develop preventive strategies. While some authors believe that PD is mainly genetic and that the aging of the society is the principal cause for this increase, different studies suggest that PD may be due to an increased exposure to environmental toxins. In this article we review epidemiological, sociological and experimental studies to determine which hypothesis is more plausible. Our conclusion is that, at least in idiopathic PD (iPD), the exposure to toxic environmental substances could play an important role in its aetiology.
Collapse
Affiliation(s)
- Francisco Pan-Montojo
- Neurologische Klinik, Klinikum der Ludwig-Maximilians-Universität München, Marchioninistr. 15, 81377 Munich, Germany ; Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut Ludwig-Maximilians-Universität München, Schillerstr. 44, 80336 Munich, Germany
| | - Heinz Reichmann
- Klinik und Poliklinik für Neurologie, Carl Gustav Carus University Hospital, TU-Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| |
Collapse
|
31
|
Fares MB, Ait-Bouziad N, Dikiy I, Mbefo MK, Jovičić A, Kiely A, Holton JL, Lee SJ, Gitler AD, Eliezer D, Lashuel HA. The novel Parkinson's disease linked mutation G51D attenuates in vitro aggregation and membrane binding of α-synuclein, and enhances its secretion and nuclear localization in cells. Hum Mol Genet 2014; 23:4491-509. [PMID: 24728187 DOI: 10.1093/hmg/ddu165] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A novel mutation in the α-Synuclein (α-Syn) gene "G51D" was recently identified in two familial cases exhibiting features of Parkinson's disease (PD) and multiple system atrophy (MSA). In this study, we explored the impact of this novel mutation on the aggregation, cellular and biophysical properties of α-Syn, in an attempt to unravel how this mutant contributes to PD/MSA. Our results show that the G51D mutation significantly attenuates α-Syn aggregation in vitro. Moreover, it disrupts local helix formation in the presence of SDS, decreases binding to lipid vesicles C-terminal to the site of mutation and severely inhibits helical folding in the presence of acidic vesicles. When expressed in yeast, α-Syn(G51D) behaves similarly to α-Syn(A30P), as both exhibit impaired membrane association, form few inclusions and are non-toxic. In contrast, enhanced secreted and nuclear levels of the G51D mutant were observed in mammalian cells, as well as in primary neurons, where α-Syn(G51D) was enriched in the nuclear compartment, was hyper-phosphorylated at S129 and exacerbated α-Syn-induced mitochondrial fragmentation. Finally, post-mortem human brain tissues of α-Syn(G51D) cases were examined, and revealed only partial colocalization with nuclear membrane markers, probably due to post-mortem tissue delay and fixation. These findings suggest that the PD-linked mutations may cause neurodegeneration via different mechanisms, some of which may be independent of α-Syn aggregation.
Collapse
Affiliation(s)
- Mohamed-Bilal Fares
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015 Lausanne, Switzerland
| | - Nadine Ait-Bouziad
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015 Lausanne, Switzerland
| | - Igor Dikiy
- Department of Biochemistry and Program in Structural Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Martial K Mbefo
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015 Lausanne, Switzerland
| | - Ana Jovičić
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aoife Kiely
- Queen Square Brain Bank, UCL Institute of Neurology, London, UK
| | - Janice L Holton
- Queen Square Brain Bank, UCL Institute of Neurology, London, UK
| | - Seung-Jae Lee
- Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David Eliezer
- Department of Biochemistry and Program in Structural Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015 Lausanne, Switzerland
| |
Collapse
|
32
|
Inhibition of store-operated calcium entry attenuates MPP(+)-induced oxidative stress via preservation of mitochondrial function in PC12 cells: involvement of Homer1a. PLoS One 2013; 8:e83638. [PMID: 24358303 PMCID: PMC3866123 DOI: 10.1371/journal.pone.0083638] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 11/05/2013] [Indexed: 12/15/2022] Open
Abstract
The process of store-operated calcium entry (SOCE), whereby the release of intracellular Ca2+ from endoplasmic reticulum (ER) activates Ca2+ influx channels in the plasma membrane, has been demonstrated to impact a diverse range of cell functions. In the present study, we investigated the potential protective effect of SOCE inhibition against 1-methyl-4-phenylpyridinium (MPP+) injury by using pharmacological antagonists or specific small interfering RNA (siRNA) in PC12 cells. The results showed that both antagonists (15 μM MRS-1845 and 50 μM ML-9) and stromal interacting molecule-1 (STIM1) targeted siRNA (Si-STIM1) significantly increased cell viability, decreased apoptotic cell death and reduced intracellular reactive oxygen species (ROS) production and lipid peroxidation in MPP+ injured PC12 cells. SOCE inhibition also prevented MPP+ induced mitochondrial dysfunction and activation of mitochondrial related apoptotic factors, while had no effect on mitochondrial biogenesis. Moreover, inhibition of SOCE by antagonists and siRNA increased the expression levels of Homer1a mRNA and protein, and knockdown of Homer1a expression by specific siRNA partly reversed the protective effects induced by SOCE inhibition in PC12 cells. All these results indicated that SOCE inhibition protected PC12 cells against MPP+ insult through upregulation of Homer1a expression, and SOCE might be an ideal target for investigating therapeutic strategy against neuronal injury in PD patients.
Collapse
|
33
|
Uptake and mitochondrial dysfunction of alpha-synuclein in human astrocytes, cortical neurons and fibroblasts. Transl Neurodegener 2013; 2:20. [PMID: 24093918 PMCID: PMC3853407 DOI: 10.1186/2047-9158-2-20] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 10/01/2013] [Indexed: 12/14/2022] Open
Abstract
The accumulation and aggregation of alpha-synuclein (α-syn) in several tissue including the brain is a major pathological hallmark in Parkinson’s disease (PD). In this study, we show that α-syn can be taken up by primary human cortical neurons, astrocytes and skin-derived fibroblasts in vitro. Our findings that brain and peripheral cells exposed to α-syn can lead to impaired mitochondrial function, leading to cellular degeneration and cell death, provides additional evidence for the involvement of mitochondrial dysfunction as a mechanism of toxicity of α-syn in human cells.
Collapse
|
34
|
Alpha-synuclein transmission and mitochondrial toxicity in primary human foetal enteric neurons in vitro. Neurotox Res 2013; 25:170-82. [PMID: 24026637 DOI: 10.1007/s12640-013-9420-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 08/06/2013] [Accepted: 08/17/2013] [Indexed: 01/19/2023]
Abstract
Parkinson's disease (PD) is a multicentred neurodegenerative disorder characterised by the accumulation and aggregation of alpha-synuclein (α-syn) in several parts of the central nervous system. However, it is well established that PD can generate symptoms of constipation and other gastrointestinal problems and α-syn containing lesions have been identified in intestinal nerve cells. In this study, we show that α-syn can be taken up and accumulate in primary human foetal enteric neurons from the gastrointestinal tract and can be transferred between foetal enteric neurons. Impaired proteosomal/lysosomal degradation can promote the uptake and accumulation of α-syn in enteric neurons. Enteric neurons exposed to α-syn can also lead to impaired mitochondrial complex I activity, reduced mitochondrial function, and NAD(+) depletion culminating in cell death via energy restriction. These findings demonstrate neuron-to-neuron transmission of α-syn in enteric neurons, providing renewed evidence for Braak's hypothesis and the aetiology of PD.
Collapse
|
35
|
Surmeier DJ, Guzman JN, Sanchez J, Schumacker PT. Physiological phenotype and vulnerability in Parkinson's disease. Cold Spring Harb Perspect Med 2013; 2:a009290. [PMID: 22762023 DOI: 10.1101/cshperspect.a009290] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review will focus on the principles underlying the hypothesis that neuronal physiological phenotype-how a neuron generates and regulates action potentials-makes a significant contribution to its vulnerability in Parkinson's disease (PD) and aging. A cornerstone of this hypothesis is that the maintenance of ionic gradients underlying excitability can pose a significant energetic burden for neurons, particularly those that have sustained residence times at depolarized membrane potentials, broad action potentials, prominent Ca(2+) entry, and modest intrinsic Ca(2+) buffering capacity. This energetic burden is shouldered in neurons primarily by mitochondria, the sites of cellular respiration. Mitochondrial respiration increases the production of damaging superoxide and other reactive oxygen species (ROS) that have widely been postulated to contribute to cellular aging and PD. Many of the genetic mutations and toxins associated with PD compromise mitochondrial function, providing a mechanistic linkage between known risk factors and cellular physiology that could explain the pattern of pathology in PD. Because much of the mitochondrial burden created by this at-risk phenotype is created by Ca(2+) entry through L-type voltage-dependent channels for which there are antagonists approved for human use, a neuroprotective strategy to reduce this burden is feasible.
Collapse
Affiliation(s)
- D James Surmeier
- Department of Physiology, Northwestern University, Chicago, Illinois, USA.
| | | | | | | |
Collapse
|
36
|
Diversity of mitochondrial pathology in a mouse model of axonal degeneration in synucleinopathies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:817807. [PMID: 23577227 PMCID: PMC3612494 DOI: 10.1155/2013/817807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 02/18/2013] [Indexed: 11/17/2022]
Abstract
There is mounting evidence for a role of mitochondrial dysfunction in the pathogenesis of α-synucleinopathies such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB). In particular, recent studies have demonstrated that failure of mitochondrial quality control caused by loss of function of the PTEN-induced kinase 1 (PINK1, PARK6) Parkin (PARK2) pathway may be causative in some familial PD. In sporadic PD, α-synuclein aggregation may interfere with mitochondrial function, and this might be further exacerbated by leucine-rich repeat kinase 2 (LRRK2). The majority of these findings have been obtained in Drosophila and cell cultures, whereas the objective of this paper is to discuss our recent results on the axonal pathology of brains derived from transgenic mice expressing α-synuclein or DLB-linked P123H β-synuclein. In line with the current view of the pathogenesis of sporadic PD, mitochondria abnormally accumulated in α-synuclein/LRRK2-immunopositive axonal swellings in mice expressing α-synuclein. Curiously, neither mitochondria nor LRRK2 was present in the swellings of mice expressing P123H β-synuclein, suggesting that α- and β-synuclein might play differential roles in the mitochondrial pathology of α-synucleinopathies.
Collapse
|
37
|
Environmental toxins trigger PD-like progression via increased alpha-synuclein release from enteric neurons in mice. Sci Rep 2012. [PMID: 23205266 PMCID: PMC3510466 DOI: 10.1038/srep00898] [Citation(s) in RCA: 282] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pathological studies on Parkinson's disease (PD) patients suggest that PD pathology progresses from the enteric nervous system (ENS) and the olfactory bulb into the central nervous system. We have previously shown that environmental toxins acting locally on the ENS mimic this PD-like pathology progression pattern in mice. Here, we show for the first time that the resection of the autonomic nerves stops this progression. Moreover, our results show that an environmental toxin (i.e. rotenone) promotes the release of alpha-synuclein by enteric neurons and that released enteric alpha-synuclein is up-taken by presynaptic sympathetic neurites and retrogradely transported to the soma, where it accumulates. These results strongly suggest that pesticides can initiate the progression of PD pathology and that this progression is based on the transneuronal and retrograde axonal transport of alpha-synuclein. If confirmed in patients, this study would have crucial implications in the strategies used to prevent and treat PD.
Collapse
|
38
|
In vivo alterations in calcium buffering capacity in transgenic mouse model of synucleinopathy. J Neurosci 2012; 32:9992-8. [PMID: 22815513 DOI: 10.1523/jneurosci.1270-12.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abnormal accumulation of α-synuclein is centrally involved in the pathogenesis of many disorders with Parkinsonism and dementia. Previous in vitro studies suggest that α-synuclein dysregulates intracellular calcium. However, it is unclear whether these alterations occur in vivo. For this reason, we investigated calcium dynamics in transgenic mice expressing human WT α-synuclein using two-photon microscopy. We imaged spontaneous and stimulus-induced neuronal activity in the barrel cortex. Transgenic mice exhibited augmented, long-lasting calcium transients characterized by considerable deviation from the exponential decay. The most evident pathology was observed in response to a repetitive stimulation in which subsequent stimuli were presented before relaxation of calcium signal to the baseline. These alterations were detected in the absence of significant increase in neuronal spiking response compared with age-matched controls, supporting the possibility that α-synuclein promoted alterations in calcium dynamics via interference with intracellular buffering mechanisms. The characteristic shape of calcium decay and augmented response during repetitive stimulation can serve as in vivo imaging biomarkers in this model of neurodegeneration, to monitor progression of the disease and screen candidate treatment strategies.
Collapse
|
39
|
Martins-Branco D, Esteves AR, Santos D, Arduino DM, Swerdlow RH, Oliveira CR, Januario C, Cardoso SM. Ubiquitin proteasome system in Parkinson's disease: a keeper or a witness? Exp Neurol 2012; 238:89-99. [PMID: 22921536 DOI: 10.1016/j.expneurol.2012.08.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 07/26/2012] [Accepted: 08/07/2012] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The aim of this work was to evaluate the role of ubiquitin-proteasome system (UPS) on mitochondrial-driven alpha-synuclein (aSN) clearance in in vitro, ex vivo and in vivo Parkinson's disease (PD) cellular models. METHOD We used SH-SY5Y ndufa2 knock-down (KD) cells, PD cybrids and peripheral blood mononuclear cells (PBMC) from patients meeting the diagnostic criteria for PD. We quantified aSN aggregation, proteasome activity and protein ubiquitination levels. In PBMC of PD patient population we evaluated the aSN levels in the plasma and the influence of several demographic characteristics in the above mentioned determinations. RESULTS We found that ubiquitin-independent proteasome activity was up-regulated in SH-SY5Y ndufa2 KD cells while a downregulation was observed in PD cybrids and PBMC. Moreover, we observed an increase in protein ubiquitination that correlates with a decrease in ubiquitin-dependent proteasome activity. Accordingly, proteasome inhibition prevented ubiquitin-dependent aSN clearance. Ubiquitin-independent proteasome activity was positively correlated with ubiquitination in PBMC. We also report a negative correlation of chymotrypsin-like activity with age in control and late-onset PD groups. Total ubiquitin content is positively correlated with aSN oligomer levels, which leads to an age-dependent increase of aSN ubiquitination in LOPD. Moreover, aSN levels are increased in the plasma of PD patients. INTERPRETATION aSN oligomers are ubiquitinated and we identified a ubiquitin-dependent clearance insufficiency with the accumulation of both aSN and ubiquitin. However, SH-SY5Y ndufa2 KD cells showed a significant up-regulation of ubiquitin-independent proteasomal enzymatic activity that could mean a cell rescue attempt. Moreover, we identified that UPS function is age-dependent in PBMC.
Collapse
|
40
|
Gulati V, Wallace R. Rafts, Nanoparticles and Neural Disease. NANOMATERIALS (BASEL, SWITZERLAND) 2012; 2:217-250. [PMID: 28348305 PMCID: PMC5304588 DOI: 10.3390/nano2030217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 07/19/2012] [Accepted: 07/20/2012] [Indexed: 11/17/2022]
Abstract
This review examines the role of membrane rafts in neural disease as a rationale for drug targeting utilizing lipid-based nanoparticles. The article begins with an overview of methodological issues involving the existence, sizes, and lifetimes of rafts, and then examines raft function in the etiologies of three major neural diseases-epilepsy, Parkinson's disease, and Alzheimer's disease-selected as promising candidates for raft-based therapeutics. Raft-targeting drug delivery systems involving liposomes and solid lipid nanoparticles are then examined in detail.
Collapse
Affiliation(s)
- Vishal Gulati
- Ross University School of Medicine, Miami Beach Community Health Center, 11645 Biscayne Boulevard, North Miami, FL 33181, USA.
| | - Ron Wallace
- Department of Anthropology, University of Central Florida, Orlando, FL 32816, USA.
| |
Collapse
|
41
|
Batelli S, Peverelli E, Rodilossi S, Forloni G, Albani D. Macroautophagy and the proteasome are differently involved in the degradation of alpha-synuclein wild type and mutated A30P in an in vitro inducible model (PC12/TetOn). Neuroscience 2011; 195:128-37. [PMID: 21906659 PMCID: PMC3188703 DOI: 10.1016/j.neuroscience.2011.08.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 08/04/2011] [Accepted: 08/14/2011] [Indexed: 12/19/2022]
Abstract
Many data suggest that alpha synuclein (α-syn) aggregation is involved in Parkinson's disease (PD) neurotoxicity and is accelerated by the pathogenetic point mutation A30P. The triplication of α-syn gene has been linked to early-onset familial PD, suggesting that the cellular dosage of α-syn is an important modulator of its toxicity. To verify this point, we developed an inducible model of α-syn expression (both wild type [WT] and mutated A30P) in rat PC12/TetOn cells. At low expression level, both α-syn(WT) and (A30P) did not aggregate, were not toxic, and displayed a protective action against oxidative stress triggered by hydrogen peroxide (H2O2). By increasing α-syn expression, its antioxidant function was no longer detectable as for the A30P form, but again no aggregation and cell death were present both for the WT and the mutated protein. To clarify why α-syn did not accumulate at high expression level, we inhibited macroautophagy by 3-methyladenine (3-MA) and the proteasome by MG132. In presence of 3-MA, α-syn(WT) accumulated, A11 anti-oligomer antibody-positive aggregates were detectable, and cell toxicity was evident, while proteasome inhibition did not increase α-syn(WT) accumulation. Macroautophagy or proteasome inhibition slightly increased α-syn(A30P) toxicity, with no detectable aggregation. This model can provide useful details about α-syn function, aggregation, and degradation pathways.
Collapse
Affiliation(s)
- S Batelli
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | | | | | | | | |
Collapse
|