1
|
Xian M, Li J, Liu T, Hou K, Sun L, Wei J. β-Synuclein Intermediates α-Synuclein Neurotoxicity in Parkinson's Disease. ACS Chem Neurosci 2024; 15:2445-2453. [PMID: 38905183 DOI: 10.1021/acschemneuro.4c00263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024] Open
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disease in the world, and synuclein is closely related to the onset and progression of PD. Synuclein is considered a therapeutic target for PD. Recent studies have found that abnormal aggregation of α-synuclein (α-Syn) in the brains of PD patients leads to mitochondrial dysfunction and neuroinflammation. Research in the field of neuroscience has confirmed that β-synuclein (β-Syn) also plays a role in Parkinson's disease. However, there has been little research on the role mechanisms and interactions between β-Syn and α-Syn in PD. Therefore, the purpose of this study is to clarify the relationship between α-Syn, β-Syn, and PD and to explore the roles and interactions of β-Syn and α-Syn in PD.
Collapse
Affiliation(s)
- Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Jingwen Li
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Kaiying Hou
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| |
Collapse
|
2
|
Sun Z, Kantor B, Chiba-Falek O. Neuronal-type-specific epigenome editing to decrease SNCA expression: Implications for precision medicine in synucleinopathies. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102084. [PMID: 38130373 PMCID: PMC10732167 DOI: 10.1016/j.omtn.2023.102084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
Overexpression of SNCA has been implicated in the pathogenesis of synucleinopathies, particularly Parkinson's disease (PD) and dementia with Lewy bodies (DLB). While PD and DLB share some clinical and pathological similarities, each disease presents distinct characteristics, including the primary affected brain region and neuronal type. We aimed to develop neuronal-type-specific SNCA-targeted epigenome therapies for synucleinopathies. The system is based on an all-in-one lentiviral vector comprised of CRISPR-dSaCas9 and guide RNA (gRNA) targeted at SNCA intron 1 fused with a synthetic repressor molecule of Krüppel-associated box (KRAB)/ methyl CpG binding protein 2 (MeCp2) transcription repression domain (TRD). To achieve neuronal-type specificity for dopaminergic and cholinergic neurons, the system was driven by tyrosine hydroxylase (TH) and choline acetyltransferase (ChAT) promoters, respectively. Delivering the system into human induced pluripotent stem cell (hiPSC)-derived dopaminergic and cholinergic neurons from a patient with the SNCA triplication resulted in efficient and neuronal-type-specific downregulation of SNCA-mRNA and protein. Furthermore, the reduction in SNCA levels by the gRNA-dSaCas9-repressor system rescued disease-related cellular phenotypes including Ser129-phophorylated α-synuclein, neuronal viability, and mitochondrial dysfunction. We established a novel neuronal-type-specific SNCA-targeted epigenome therapy and provided in vitro proof of concept using human-based disease models. Our results support the therapeutic potential of our system for PD and DLB and provide the foundation for further preclinical studies in animal models toward investigational new drug (IND) enablement and clinical trials.
Collapse
Affiliation(s)
| | - Boris Kantor
- Viral Vector Core, Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
3
|
Kon T, Forrest SL, Lee S, Martinez-Valbuena I, Li J, Nassir N, Uddin MJ, Lang AE, Kovacs GG. Neuronal SNCA transcription during Lewy body formation. Acta Neuropathol Commun 2023; 11:185. [PMID: 37996943 PMCID: PMC10666428 DOI: 10.1186/s40478-023-01687-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Misfolded α-synuclein (α-syn) is believed to contribute to neurodegeneration in Lewy body disease (LBD) based on considerable evidence including a gene-dosage effect observed in relation to point mutations and multiplication of SNCA in familial Parkinson's disease. A contradictory concept proposes early loss of the physiological α-syn as the major driver of neurodegeneration. There is a paucity of data on SNCA transcripts in various α-syn immunoreactive cytopathologies. Here, the total cell body, nuclear, and cytoplasmic area density of SNCA transcripts in neurons without and with various α-syn immunoreactive cytopathologies in the substantia nigra and amygdala in autopsy cases of LBD (n = 5) were evaluated using RNAscope combined with immunofluorescence for disease-associated α-syn. Single-nucleus RNA sequencing was performed to elucidate cell-type specific SNCA expression in non-diseased frontal cortex (n = 3). SNCA transcripts were observed in the neuronal nucleus and cytoplasm in neurons without α-syn, those containing punctate α-syn immunoreactivity, irregular-shaped compact inclusion, and brainstem-type and cortical-type LBs. However, SNCA transcripts were only rarely found in the α-syn immunoreactive LB areas. The total cell body SNCA transcript area densities in neurons with punctate α-syn immunoreactivity were preserved but were significantly reduced in neurons with compact α-syn inclusions both in the substantia nigra and amygdala. This reduction was also observed in the cytoplasm but not in the nucleus. Only single SNCA transcripts were detected in astrocytes with or without disease-associated α-syn immunoreactivity in the amygdala. Single-nucleus RNA sequencing revealed that excitatory and inhibitory neurons, oligodendrocyte progenitor cells, oligodendrocytes, and homeostatic microglia expressed SNCA transcripts, while expression was largely absent in astrocytes and microglia. The preserved cellular SNCA expression in the more abundant non-Lewy body type α-syn cytopathologies might provide a pool for local protein production that can aggregate and serve as a seed for misfolded α-syn. Successful segregation of disease-associated α-syn is associated with the exhaustion of SNCA production in the terminal cytopathology, the Lewy body. Our observations inform therapy development focusing on targeting SNCA transcription in LBD.
Collapse
Affiliation(s)
- Tomoya Kon
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Rm 6KD414, Tanz CRND, Krembil Discovery Tower, Toronto, ON, M5T 0S8, Canada
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shelley L Forrest
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Rm 6KD414, Tanz CRND, Krembil Discovery Tower, Toronto, ON, M5T 0S8, Canada
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Seojin Lee
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Rm 6KD414, Tanz CRND, Krembil Discovery Tower, Toronto, ON, M5T 0S8, Canada
| | - Ivan Martinez-Valbuena
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Rm 6KD414, Tanz CRND, Krembil Discovery Tower, Toronto, ON, M5T 0S8, Canada
| | - Jun Li
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Rm 6KD414, Tanz CRND, Krembil Discovery Tower, Toronto, ON, M5T 0S8, Canada
| | | | - Mohammed J Uddin
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
- GenomeArc Inc, Toronto, ON, Canada
| | - Anthony E Lang
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Rm 6KD414, Tanz CRND, Krembil Discovery Tower, Toronto, ON, M5T 0S8, Canada
- Edmund J Safra Program in Parkinson's Disease and Rossy Progressive Supranuclear Palsy Centre, Toronto Western Hospital, Toronto, ON, Canada
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, ON, Canada
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Rm 6KD414, Tanz CRND, Krembil Discovery Tower, Toronto, ON, M5T 0S8, Canada.
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada.
- Edmund J Safra Program in Parkinson's Disease and Rossy Progressive Supranuclear Palsy Centre, Toronto Western Hospital, Toronto, ON, Canada.
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Brown JL, Hart DW, Boyle GE, Brown TG, LaCroix M, Baraibar AM, Pelzel R, Kim M, Sherman MA, Boes S, Sung M, Cole T, Lee MK, Araque A, Lesné SE. SNCA genetic lowering reveals differential cognitive function of alpha-synuclein dependent on sex. Acta Neuropathol Commun 2022; 10:180. [PMID: 36517890 PMCID: PMC9749314 DOI: 10.1186/s40478-022-01480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Antisense oligonucleotide (ASO) therapy for neurological disease has been successful in clinical settings and its potential has generated hope for Alzheimer's disease (AD). We previously described that ablating SNCA encoding for α-synuclein (αSyn) in a mouse model of AD was beneficial. Here, we sought to demonstrate whether transient reduction of αSyn expression using ASOSNCA could be therapeutic in a mouse model of AD. The efficacy of the ASOSNCA was measured via immunocytochemistry, RT-qPCR and western blotting. To assess spatial learning and memory, ASOSNCA or PBS-injected APP and non-transgenic (NTG) mice, and separate groups of SNCA-null mice, were tested on the Barnes circular maze. Hippocampal slice electrophysiology and transcriptomic profiling were used to explore synaptic function and differential gene expression between groups. Reduction of SNCA transcripts alleviated cognitive deficits in male transgenic animals, but surprisingly, not in females. To determine the functional cause of this differential effect, we assessed memory function in SNCA-null mice. Learning and memory were intact in male mice but impaired in female animals, revealing that the role of αSyn on cognitive function is sex-specific. Transcriptional analyses identified a differentially expressed gene network centered around EGR1, a central modulator of learning and memory, in the hippocampi of SNCA-null mice. Thus, these novel results demonstrate that the function of αSyn on memory differs between male and female brains.
Collapse
Affiliation(s)
- Jennifer L Brown
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Damyan W Hart
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Gabriel E Boyle
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA, 98195, USA
| | - Taylor G Brown
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Michael LaCroix
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
- Medical Scientist Training Program, University of Texas Southwestern Medical School, Dallas, TX, 75390, USA
| | - Andrés M Baraibar
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Ross Pelzel
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Minwoo Kim
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Mathew A Sherman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Samuel Boes
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Michelle Sung
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Tracy Cole
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
- n-Lorem Foundation, Carlsbad, CA, 92010, USA
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Alfonso Araque
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Sylvain E Lesné
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA.
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA.
| |
Collapse
|
5
|
Hayashi J, Carver JA. β-Synuclein: An Enigmatic Protein with Diverse Functionality. Biomolecules 2022; 12:142. [PMID: 35053291 PMCID: PMC8773819 DOI: 10.3390/biom12010142] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 12/24/2022] Open
Abstract
α-Synuclein (αS) is a small, unstructured, presynaptic protein expressed in the brain. Its aggregated form is a major component of Lewy bodies, the large proteinaceous deposits in Parkinson's disease. The closely related protein, β-Synuclein (βS), is co-expressed with αS. In vitro, βS acts as a molecular chaperone to inhibit αS aggregation. As a result of this assignation, βS has been largely understudied in comparison to αS. However, recent reports suggest that βS promotes neurotoxicity, implying that βS is involved in other cellular pathways with functions independent of αS. Here, we review the current literature pertaining to human βS in order to understand better the role of βS in homeostasis and pathology. Firstly, the structure of βS is discussed. Secondly, the ability of βS to (i) act as a molecular chaperone; (ii) regulate synaptic function, lipid binding, and the nigrostriatal dopaminergic system; (iii) mediate apoptosis; (iv) participate in protein degradation pathways; (v) modulate intracellular metal levels; and (vi) promote cellular toxicity and protein aggregation is explored. Thirdly, the P123H and V70M mutations of βS, which are associated with dementia with Lewy bodies, are discussed. Finally, the importance of post-translational modifications on the structure and function of βS is reviewed. Overall, it is concluded that βS has both synergistic and antagonistic interactions with αS, but it may also possess important cellular functions independent of αS.
Collapse
Affiliation(s)
| | - John A. Carver
- Research School of Chemistry, The Australian National University, Acton, ACT 2601, Australia;
| |
Collapse
|
6
|
Wei J, Ho G, Takamatsu Y, Masliah E, Hashimoto M. Therapeutic Potential of α-Synuclein Evolvability for Autosomal Recessive Parkinson's Disease. PARKINSON'S DISEASE 2021; 2021:6318067. [PMID: 34858569 PMCID: PMC8632460 DOI: 10.1155/2021/6318067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/20/2021] [Accepted: 11/03/2021] [Indexed: 06/13/2023]
Abstract
The majority of Parkinson's disease (PD) is sporadic in elderly and is characterized by α-synuclein (αS) aggregation and other alterations involving mitochondria, ubiquitin-proteasome, and autophagy. The remaining are familial PD associated with gene mutations of either autosomal dominant or recessive inheritances. However, the former ones are similar to sporadic PD, and the latter ones are accompanied by impaired mitophagy during the reproductive stage. Since no radical therapies are available for PD, the objective of this paper is to discuss a mechanistic role for amyloidogenic evolvability, a putative physiological function of αS, among PD subtypes, and the potential relevance to therapy. Presumably, αS evolvability might benefit familial PD due to autosomal dominant genes and also sporadic PD during reproduction, which may manifest as neurodegenerative diseases through antagonistic pleiotropy mechanism in aging. Indeed, there are some reports describing that αS prevents apoptosis and mitochondrial alteration under the oxidative stress conditions, notwithstanding myriads of papers on the neuropathology of αS. Importantly, β-synuclein (βS), the nonamyloidogenic homologue of αS, might buffer against evolvability of αS protofibrils associated with neurotoxicity. Finally, it is intriguing to predict that increased αS evolvability through suppression of βS expression might protect against autosomal recessive PD. Collectively, further studies are warranted to better understand αS evolvability in PD pathogenesis, leading to rational therapy development.
Collapse
Affiliation(s)
- Jianshe Wei
- Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Gilbert Ho
- PCND Neuroscience Research Institute, Poway 92064, CA, USA
| | - Yoshiki Takamatsu
- Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Eliezer Masliah
- Division of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Makoto Hashimoto
- Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
7
|
Hernandez SM, Tikhonova EB, Baca KR, Zhao F, Zhu X, Karamyshev AL. Unexpected Implication of SRP and AGO2 in Parkinson's Disease: Involvement in Alpha-Synuclein Biogenesis. Cells 2021; 10:2792. [PMID: 34685771 PMCID: PMC8534902 DOI: 10.3390/cells10102792] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 01/21/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder classified by the loss of dopaminergic neurons in the substantia nigra pars compacta, the region of the brain that is responsible for motor control. Surviving neurons in this region contain aggregated protein alpha-Synuclein (αSyn) in the form of cytoplasmic inclusions, referred to as Lewy bodies. Changes in αSyn expression are also associated with PD and its progression. Previously, we demonstrated that signal recognition particle (SRP) and Argonaute 2 (AGO2) proteins are involved in protein quality control at the ribosome during translation. We also demonstrated that SRP has an mRNA protection function in addition to a protein targeting function, thus controlling mRNA and protein expression. In this study, we tested involvement of these factors in αSyn biogenesis. We hypothesize that loss of these factors may interfere with αSyn expression, and subsequently, be associated with PD. Using depletion assays in human cell culture and analysis of these proteins in the brains of deceased PD patients, we demonstrate that SRP and AGO2 are involved in the control of αSyn expression and AGO2 has reduced expression in PD. We show for the first time that SRP is involved in mRNA protection of αSyn, a protein that does not have a signal sequence or transmembrane span. Our findings suggest that SRP may interact with a hydrophobic domain in the middle of αSyn during translation. Understanding the molecular mechanisms controlling αSyn biogenesis in cells is vital to developing preventative therapies against PD.
Collapse
Affiliation(s)
- Sarah M. Hernandez
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.M.H.); (E.B.T.); (K.R.B.)
| | - Elena B. Tikhonova
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.M.H.); (E.B.T.); (K.R.B.)
| | - Kristen R. Baca
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.M.H.); (E.B.T.); (K.R.B.)
- Center for the Integration of STEM Education and Research (CISER), Texas Tech University, Lubbock, TX 79409, USA
| | - Fanpeng Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (F.Z.); (X.Z.)
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (F.Z.); (X.Z.)
| | - Andrey L. Karamyshev
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.M.H.); (E.B.T.); (K.R.B.)
| |
Collapse
|
8
|
Tanudjojo B, Shaikh SS, Fenyi A, Bousset L, Agarwal D, Marsh J, Zois C, Heman-Ackah S, Fischer R, Sims D, Melki R, Tofaris GK. Phenotypic manifestation of α-synuclein strains derived from Parkinson's disease and multiple system atrophy in human dopaminergic neurons. Nat Commun 2021; 12:3817. [PMID: 34155194 PMCID: PMC8217249 DOI: 10.1038/s41467-021-23682-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
α-Synuclein is critical in the pathogenesis of Parkinson's disease and related disorders, yet it remains unclear how its aggregation causes degeneration of human dopaminergic neurons. In this study, we induced α-synuclein aggregation in human iPSC-derived dopaminergic neurons using fibrils generated de novo or amplified in the presence of brain homogenates from Parkinson's disease or multiple system atrophy. Increased α-synuclein monomer levels promote seeded aggregation in a dose and time-dependent manner, which is associated with a further increase in α-synuclein gene expression. Progressive neuronal death is observed with brain-amplified fibrils and reversed by reduction of intraneuronal α-synuclein abundance. We identified 56 proteins differentially interacting with aggregates triggered by brain-amplified fibrils, including evasion of Parkinson's disease-associated deglycase DJ-1. Knockout of DJ-1 in iPSC-derived dopaminergic neurons enhance fibril-induced aggregation and neuronal death. Taken together, our results show that the toxicity of α-synuclein strains depends on aggregate burden, which is determined by monomer levels and conformation which dictates differential interactomes. Our study demonstrates how Parkinson's disease-associated genes influence the phenotypic manifestation of strains in human neurons.
Collapse
Affiliation(s)
- Benedict Tanudjojo
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Samiha S Shaikh
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alexis Fenyi
- CEA, Institut François Jacob (MIRCen) and CNRS, Laboratory of Neurodegenerative Diseases, Fontenay-aux-Roses, France
| | - Luc Bousset
- CEA, Institut François Jacob (MIRCen) and CNRS, Laboratory of Neurodegenerative Diseases, Fontenay-aux-Roses, France
| | - Devika Agarwal
- MRC Centre for Computational Biology, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jade Marsh
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Christos Zois
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Sabrina Heman-Ackah
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - David Sims
- MRC Centre for Computational Biology, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Ronald Melki
- CEA, Institut François Jacob (MIRCen) and CNRS, Laboratory of Neurodegenerative Diseases, Fontenay-aux-Roses, France
| | - George K Tofaris
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| |
Collapse
|
9
|
Sharma K, Mehra S, Sawner AS, Markam PS, Panigrahi R, Navalkar A, Chatterjee D, Kumar R, Kadu P, Patel K, Ray S, Kumar A, Maji SK. Effect of Disease-Associated P123H and V70M Mutations on β-Synuclein Fibrillation. ACS Chem Neurosci 2020; 11:2836-2848. [PMID: 32833434 DOI: 10.1021/acschemneuro.0c00405] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Synucleinopathies are a class of neurodegenerative diseases, including Parkinson's disease (PD), Dementia with Lewy bodies (DLB), and Multiple System Atrophy (MSA). The common pathological hallmark of synucleinopathies is the filamentous α-synuclein (α-Syn) aggregates along with membrane components in cytoplasmic inclusions in the brain. β-Synuclein (β-Syn), an isoform of α-Syn, inhibits α-Syn aggregation and prevents its neurotoxicity, suggesting the neuroprotective nature of β-Syn. However, this notion changed with the discovery of disease-associated β-Syn mutations, V70M and P123H, in patients with DLB. It is still unclear how these missense mutations alter the structural and amyloidogenic properties of β-Syn, leading to neurodegeneration. Here, we characterized the biophysical properties and investigated the effect of mutations on β-Syn fibrillation under different conditions. V70M and P123H show high membrane binding affinity compared to wild-type β-Syn, suggesting their potential role in membrane interactions. β-Syn and its mutants do not aggregate under normal physiological conditions; however, the proteins undergo self-polymerization in a slightly acidic microenvironment and/or in the presence of an inducer, forming long unbranched amyloid fibrils similar to α-Syn. Strikingly, V70M and P123H mutants exhibit accelerated fibrillation compared to native β-Syn under these conditions. NMR study further revealed that these point mutations induce local perturbations at the site of mutation in β-Syn. Overall, our data provide insight into the biophysical properties of disease-associated β-Syn mutations and demonstrate that these mutants make the native protein more susceptible to aggregation in an altered microenvironment.
Collapse
Affiliation(s)
- Karan Sharma
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Surabhi Mehra
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Ajay S. Sawner
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Pratap S. Markam
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Rajlaxmi Panigrahi
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Debdeep Chatterjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Rakesh Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Komal Patel
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Soumik Ray
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Samir K. Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| |
Collapse
|
10
|
Hydrogen Sulfide and β-Synuclein Are Involved and Interlinked in the Aging Glaucomatous Retina. J Ophthalmol 2020; 2020:8642135. [PMID: 32351728 PMCID: PMC7178476 DOI: 10.1155/2020/8642135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/14/2020] [Accepted: 03/11/2020] [Indexed: 11/18/2022] Open
Abstract
Purpose Glaucoma, one of the leading causes of irreversible blindness worldwide, is a group of disorders characterized by progressive retinal ganglion cell (RGC) loss. Synucleins, a family of small proteins, have been of interest in studies of neurodegeneration and CNS. However, their roles and functions in glaucoma are still not completely understood and remain to be explored. Our previous studies showed that α-synuclein and H2S play a pivotal role in glaucoma. This study aims to (1) elucidate the potential roles and functions of synucleins in glaucoma throughout aging, (2) investigate the interaction between the synucleins and H2S, and better understand the mechanism of H2S in neuroprotection. Methods The chronic IOP elevation model was carried out in 12 animals at different ages (3 months and 14 months), and RGCs were quantified by Brn3a staining. Mass spectrometric-assisted proteomics analysis was employed to measure synuclein levels and H2S producing proteins in retina. Secondly, the acute IOP elevation model was carried out in 12 juvenile animals, with or without intravitreal injection of GYY4137 (a H2S donor). RGCs were quantified along with the abundancy of synucleins. Results RGCs and β-synuclein (SNCB) are significantly changed in old animals. Under chronic IOP elevation, there is a significant RGC loss in old animals, whereas no significant change in young animals; SNCB is significantly downregulated and 3MST is significantly upregulated in young animals due to IOP, while no significant changes in old ones are notable. Under acute IOP elevation (approx. 55 mmHg), a significant RGC loss is observed; exogenous H2S significantly reduced RGC loss and downregulated SNCB levels. Conclusion The present study indicates a strong link between ageing and SNCB regulation. In young animals SNCB is downregulated going along with less RGC loss. Furthermore, increasing endogenous H2S is effective to downregulate SNCB and is neuroprotective against acute IOP elevation.
Collapse
|
11
|
Pathways of protein synthesis and degradation in PD pathogenesis. PROGRESS IN BRAIN RESEARCH 2020; 252:217-270. [PMID: 32247365 DOI: 10.1016/bs.pbr.2020.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the discovery of protein aggregates in the brains of individuals with Parkinson's disease (PD) in the early 20th century, the scientific community has been interested in the role of dysfunctional protein metabolism in PD etiology. Recent advances in the field have implicated defective protein handling underlying PD through genetic, in vitro, and in vivo studies incorporating many disease models alongside neuropathological evidence. Here, we discuss the existing body of research focused on understanding cellular pathways of protein synthesis and degradation, and how aberrations in either system could engender PD pathology with special attention to α-synuclein-related consequences. We consider transcription, translation, and post-translational modification to constitute protein synthesis, and protein degradation to encompass proteasome-, lysosome- and endoplasmic reticulum-dependent mechanisms. Novel findings connecting each of these steps in protein metabolism to development of PD indicate that deregulation of protein production and turnover remains an exciting area in PD research.
Collapse
|
12
|
Yang X, Williams JK, Yan R, Mouradian MM, Baum J. Increased Dynamics of α-Synuclein Fibrils by β-Synuclein Leads to Reduced Seeding and Cytotoxicity. Sci Rep 2019; 9:17579. [PMID: 31772376 PMCID: PMC6879756 DOI: 10.1038/s41598-019-54063-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022] Open
Abstract
Alpha-synuclein (αS) fibrils are toxic to cells and contribute to the pathogenesis and progression of Parkinson's disease and other synucleinopathies. β-Synuclein (βS), which co-localizes with αS, has been shown to provide a neuroprotective effect, but the molecular mechanism by which this occurs remains elusive. Here we show that αS fibrils formed in the presence of βS are less cytotoxic, exhibit reduced cell seeding capacity and are more resistant to fibril shedding compared to αS fibrils alone. Using solid-state NMR, we found that the overall structure of the core of αS fibrils when co-incubated with βS is minimally perturbed, however, the dynamics of Lys and Thr residues, located primarily in the imperfect KTKEGV repeats of the αS N-terminus, are increased. Our results suggest that amyloid fibril dynamics may play a key role in modulating toxicity and seeding. Thus, enhancing the dynamics of amyloid fibrils may be a strategy for future therapeutic targeting of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xue Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, 08854, USA
| | - Jonathan K Williams
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, 08854, USA
| | - Run Yan
- RWJMS Institute for Neurological Therapeutics, Rutgers Biomedical and Health Sciences, and Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, 08854, USA
| | - M Maral Mouradian
- RWJMS Institute for Neurological Therapeutics, Rutgers Biomedical and Health Sciences, and Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, 08854, USA
| | - Jean Baum
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, 08854, USA.
| |
Collapse
|
13
|
Wihan J, Grosch J, Kalinichenko LS, Müller CP, Winkler J, Kohl Z. Layer-specific axonal degeneration of serotonergic fibers in the prefrontal cortex of aged A53T α-synuclein–expressing mice. Neurobiol Aging 2019; 80:29-37. [DOI: 10.1016/j.neurobiolaging.2019.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 02/25/2019] [Accepted: 03/23/2019] [Indexed: 01/07/2023]
|
14
|
Gámez-Valero A, Canet-Pons J, Urbizu A, Anillo A, Santos C, Ariza A, Beyer K. INDEL Length and Haplotypes in the β-Synuclein Gene: A Key to Differentiate Dementia with Lewy Bodies? J Alzheimers Dis 2019; 65:207-219. [PMID: 30040713 DOI: 10.3233/jad-180074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lewy body diseases (LBD) include Parkinson's disease (PD) and dementia with Lewy bodies (DLB) and together with Alzheimer's disease (AD) they show an important neuropathological and clinical overlap. The human alpha- and beta-synuclein genes (SNCA and SNCB) are key factors for the development of Lewy body diseases. Here, we aimed to analyze the genotype distribution of potentially functional SNPs in SNCA and SNCB, perform haplotype analysis for SNCB, and to identify functional insertion and deletion (INDEL) variations within the regulatory region of SNCB which might be responsible for the drastically diminished beta-synuclein levels reported for pure DLB. Thus, we genotyped brain samples from AD, DLB, PD, and healthy controls for two SNCA and four SNCB SNPs. We also analyzed INDEL variations upstream of SNCB, determined SNCB expression levels, and correlated INDEL lengths with expression levels. Applying Fisher's exact, chi-square, ANOVA tests, and the ΔΔCt method, we found disease-specific genotype distribution of SNCA and SNCB SNPs. Additionally, we identified three INDEL variations upstream of SNCB and showed that the INDEL allele lengths were associated with SNCB expression levels. INDEL alleles associated with low SNCB expression were accumulated in pure DLB. Finally, one major and four minor DLB specific SNCB haplotypes were identified with Haploview and Arlequin. In summary, our study showed that different SNCA and SNCB genotypes are associated with the development of either PD or DLB, and that the frequencies of genotypes associated with low SNCB expression are elevated in DLB.
Collapse
Affiliation(s)
- Ana Gámez-Valero
- Department of Pathology, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain.,REMAR-IVECAT group, Health Sciences Research Institute Germans Trias i Pujol, Barcelona, Spain
| | - Julia Canet-Pons
- Department of Pathology, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain
| | - Aintzane Urbizu
- Department of Pathology, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain
| | - Ana Anillo
- Department of Pathology, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain
| | - Cristina Santos
- Unitat d'Antropologia Biològica, Departament de Biologia Animal, Biologia Vegetal i Ecologia, Universitat Autònoma de Barcelona, Spain
| | - Aurelio Ariza
- Department of Pathology, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain
| | - Katrin Beyer
- Department of Pathology, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain
| |
Collapse
|
15
|
Profiling of Alzheimer’s disease related genes in mild to moderate vitamin D hypovitaminosis. J Nutr Biochem 2019; 67:123-137. [DOI: 10.1016/j.jnutbio.2019.01.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/13/2018] [Accepted: 01/29/2019] [Indexed: 02/01/2023]
|
16
|
Hategan A, Masliah E, Nath A. HIV and Alzheimer's disease: complex interactions of HIV-Tat with amyloid β peptide and Tau protein. J Neurovirol 2019; 25:648-660. [PMID: 31016584 DOI: 10.1007/s13365-019-00736-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/15/2019] [Accepted: 02/14/2019] [Indexed: 12/21/2022]
Abstract
In patients infected with the human immunodeficiency virus (HIV), the HIV-Tat protein may be continually produced despite adequate antiretroviral therapy. As the HIV-infected population is aging, it is becoming increasingly important to understand how HIV-Tat may interact with proteins such as amyloid β and Tau which accumulate in the aging brain and eventually result in Alzheimer's disease. In this review, we examine the in vivo data from HIV-infected patients and animal models and the in vitro experiments that show how protein complexes between HIV-Tat and amyloid β occur through novel protein-protein interactions and how HIV-Tat may influence the pathways for amyloid β production, degradation, phagocytosis, and transport. HIV-Tat may also induce Tau phosphorylation through a cascade of cellular processes that lead to the formation of neurofibrillary tangles, another hallmark of Alzheimer's disease. We also identify gaps in knowledge and future directions for research. Available evidence suggests that HIV-Tat may accelerate Alzheimer-like pathology in patients with HIV infection which cannot be impacted by current antiretroviral therapy.
Collapse
Affiliation(s)
- Alina Hategan
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bldg 10; Room 7C-103, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Eliezer Masliah
- Division of Neuroscience, National Institute of Aging, National Institutes of Health, 7201 Wisconsin Ave, Bethesda, MD, 20892, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bldg 10; Room 7C-103, 10 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
Sanjeev A, Mattaparthi VSK. Computational Study on the Role of γ-Synuclein in Inhibiting the α-Synuclein Aggregation. Cent Nerv Syst Agents Med Chem 2019; 19:24-30. [PMID: 30318002 DOI: 10.2174/1871524918666181012160439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/28/2018] [Accepted: 10/05/2018] [Indexed: 05/27/2023]
Abstract
BACKGROUND α-Synuclein (αS) is the precursor protein present in Lewy Bodies that helps in the formation of highly ordered amyloid fibrils that is associated with the occurrence of Parkinson's disease, a neuro-degenerative disorder. Many reports have now been focused on finding the probable targets to weaken this debilitating disease. Recently γ-synuclein (γS), a presynaptic protein, was highlighted to inhibit the aggregation propensity of αS both in vivo and in vitro. However the nature, location and specificity of molecular interactions existing between the αS and γS is not known in spite of the potential importance of γS as an inhibitor of αS. OBJECTIVE To understand the inhibition of αS aggregation by γS at the molecular level. METHODS Umbrella sampling method was used along with molecular dynamics simulation to investigate the conformational dynamics, degree of association and molecular interaction between the monomeric units in the αS/γS hetero-dimer. RESULTS AND DISCUSSION The dissociation energy barrier for αS/γS hetero-dimer was found to be higher than αS/αS homo-dimer. αS can therefore readily form a hetero-dimer by combining with γS than forming a homo-dimer. We also observed strong transient interactions involving hydrogen bonds, salt-bridges and non-bonded contacts between the monomeric units in αS/γS hetero-dimer. CONCLUSION Our findings suggest that γS may inhibit the aggregation propensity of αS.
Collapse
Affiliation(s)
- Airy Sanjeev
- Molecular Modelling and Simulation Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| | - Venkata S K Mattaparthi
- Molecular Modelling and Simulation Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| |
Collapse
|
18
|
Deba F, Peterson S, Hamouda AK. An Animal Model to Test Reversal of Cognitive Decline Associated with Beta-Amyloid Pathologies. Methods Mol Biol 2019; 2011:393-412. [PMID: 31273712 DOI: 10.1007/978-1-4939-9554-7_23] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Disposition of beta-amyloid peptide 1-42 (Aβ1-42) in the space around the synapses and formation of Aβ-containing aggregates known as neuritic or senile plaques are hallmark features of neurodegenerative pathologies associated with Alzheimer's disease (AD). While AD is a multifactorial disease that includes other proteinopathies (e.g., hyperphosphorylated tau aggregates) and neurotransmitter disturbances (e.g., loss of cortical cholinergic innervation), Aβ (soluble or in senile plaques) remains the major undisputed factor that contributes to the pathological and behavior presentation of AD. Overproduction of Aβ and mutations in Aβ precursor (amyloid precursor protein) or enzymes involved in Aβ1-42 production and removal (γ secretase/presenilins) have been shown in cases of early onset of AD and produced AD-like pathologies in animal models. In addition, the level of soluble Aβ1-42 has been shown to correlate with cognitive impairment in animal models before the presence of senile plaques or other histological features of AD. However, much still is unknown about the biochemical processes leading to amyloid formation and its relation to the pathogenesis, neuronal damage/dysfunction, and behavioral changes associated with AD. In this article, we review animal models that have been developed to study AD-like pathologies and then provide detailed methodology to develop an acute rat model of Aβ-induced cognitive impairment. We use this model to examine the cognitive-enhancing effect of novel pharmacological interventions targeting nicotinic acetylcholine receptors.
Collapse
Affiliation(s)
- Farah Deba
- Department of Pharmaceutical Science, Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, USA
| | - Steven Peterson
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX, USA.
| | - Ayman K Hamouda
- Department of Pharmaceutical Science, Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, USA.
| |
Collapse
|
19
|
Iyer A, Claessens MMAE. Disruptive membrane interactions of alpha-synuclein aggregates. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1867:468-482. [PMID: 30315896 DOI: 10.1016/j.bbapap.2018.10.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/14/2018] [Accepted: 10/04/2018] [Indexed: 12/17/2022]
Abstract
Alpha synuclein (αS) is a ~14 kDa intrinsically disordered protein. Decades of research have increased our knowledge on αS yet its physiological function remains largely elusive. The conversion of monomeric αS into oligomers and amyloid fibrils is believed to play a central role of the pathology of Parkinson's disease (PD). It is becoming increasingly clear that the interactions of αS with cellular membranes are important for both αS's functional and pathogenic actions. Therefore, understanding interactions of αS with membranes seems critical to uncover functional or pathological mechanisms. This review summarizes our current knowledge of how physicochemical properties of phospholipid membranes affect the binding and aggregation of αS species and gives an overview of how post-translational modifications and point mutations in αS affect phospholipid membrane binding and protein aggregation. We discuss the disruptive effects resulting from the interaction of αS aggregate species with membranes and highlight current approaches and hypotheses that seek to understand the pathogenic and/or protective role of αS in PD.
Collapse
Affiliation(s)
- Aditya Iyer
- Membrane Enzymology Group, University of Groningen, Groningen 9747 AG, The Netherlands
| | | |
Collapse
|
20
|
Atrián-Blasco E, Gonzalez P, Santoro A, Alies B, Faller P, Hureau C. Cu and Zn coordination to amyloid peptides: From fascinating chemistry to debated pathological relevance. Coord Chem Rev 2018; 375:38-55. [PMID: 30262932 DOI: 10.1016/j.ccr.2018.04.007] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Several diseases share misfolding of different peptides and proteins as a key feature for their development. This is the case of important neurodegenerative diseases such as Alzheimer's and Parkinson's diseases and type II diabetes mellitus. Even more, metal ions such as copper and zinc might play an important role upon interaction with amyloidogenic peptides and proteins, which could impact their aggregation and toxicity abilities. In this review, the different coordination modes proposed for copper and zinc with amyloid-β, α-synuclein and IAPP will be reviewed as well as their impact on the aggregation, and ROS production in the case of copper. In addition, a special focus will be given to the mutations that affect metal binding and lead to familial cases of the diseases. Different modifications of the peptides that have been observed in vivo and could be relevant for the coordination of metal ions are also described.
Collapse
Affiliation(s)
- Elena Atrián-Blasco
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4, France
- University of Toulouse, UPS, INPT, 31077 Toulouse Cedex 4, France
| | - Paulina Gonzalez
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR7177), Université de Strasbourg, 4 rue B. Pascal, 67081 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| | - Alice Santoro
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR7177), Université de Strasbourg, 4 rue B. Pascal, 67081 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| | - Bruno Alies
- Université de Bordeaux, ChemBioPharm INSERM U1212 CNRS UMR 5320, Bordeaux, France
| | - Peter Faller
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR7177), Université de Strasbourg, 4 rue B. Pascal, 67081 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4, France
- University of Toulouse, UPS, INPT, 31077 Toulouse Cedex 4, France
| |
Collapse
|
21
|
Li N, Hu P, Xu T, Chen H, Chen X, Hu J, Yang X, Shi L, Luo JH, Xu J. iTRAQ-based Proteomic Analysis of APPSw,Ind Mice Provides Insights into the Early Changes in Alzheimer's Disease. Curr Alzheimer Res 2018; 14:1109-1122. [PMID: 28730955 PMCID: PMC5676024 DOI: 10.2174/1567205014666170719165745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/13/2017] [Accepted: 06/29/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Several proteins have been identified as potential diagnostic biomarkers in imaging, genetic, or proteomic studies in Alzheimer disease (AD) patients and mouse models. However, biomarkers for presymptom diagnosis of AD are still under investigation, as are the presymptom molecular changes in AD pathogenesis. OBJECTIVE In this study, we aim to analyzed the early proteomic changes in APPSw,Ind mice and to conduct further functional studies on interesting proteins. METHODS We used the isobaric tags for relative and absolute quantitation (iTRAQ) approach combined with mass spectrometry to examine the early proteomic changes in hippocampi of APPSw,Ind mice. Quantitative reverse transcription polymerase chain reaction (RT-PCR) and immuno-blotting were performed for further validation. Finally, the functions of interesting proteins β-spectrin and Rab3a in APP trafficking and processing were tested by shRNA knockdown, in N2A cells stably expressing β-amyloid precursor protein (APP). RESULTS The iTRAQ and RT-PCR results revealed the detailed molecular changes in oxidative stress, myelination, astrocyte activation, mTOR signaling and Rab3-dependent APP trafficking in the early stage of AD progression. Knock down of β -spectrin and Rab3a finally led to increased APP fragment production, indicating key roles of β-spectrin and Rab3a in regulating APP processing. CONCLUSION Our study provides the first insights into the proteomic changes that occur in the hippocampus in the early stages of the AD mouse model. In addition to improving the understanding of molecular alterations and functional cascades involved in early AD pathogenesis, our findings raise the possibility of developing potential biomarkers and therapeutic targets for early AD.
Collapse
Affiliation(s)
- Nan Li
- Center of Neuroscience, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058. China
| | - Pinghong Hu
- Center of Neuroscience, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058. China
| | - Tiantian Xu
- Center of Neuroscience, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058. China
| | - Huan Chen
- Center of Neuroscience, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058. China
| | - Xiaoying Chen
- Center of Neuroscience, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058. China
| | - Jianwen Hu
- Shanghai Applied Protein Technology Co., Ltd., Shanghai. China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Medical Key Laboratory of Guangdong Province, Medical Key Laboratory of Health Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen. China
| | - Lei Shi
- Key Laboratory of Modern Toxicology of Shenzhen, Medical Key Laboratory of Guangdong Province, Medical Key Laboratory of Health Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen. China
| | - Jian-Hong Luo
- Center of Neuroscience, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058. China
| | - Junyu Xu
- Center of Neuroscience, Zhejiang University, 866 Yuhangtang Road, Hangzhou. China
| |
Collapse
|
22
|
Afek A, Tagliafierro L, Glenn OC, Lukatsky DB, Gordan R, Chiba-Falek O. Toward deciphering the mechanistic role of variations in the Rep1 repeat site in the transcription regulation of SNCA gene. Neurogenetics 2018; 19:135-144. [PMID: 29730780 DOI: 10.1007/s10048-018-0546-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/25/2018] [Indexed: 12/01/2022]
Abstract
Short structural variants-variants other than single nucleotide polymorphisms-are hypothesized to contribute to many complex diseases, possibly by modulating gene expression. However, the molecular mechanisms by which noncoding short structural variants exert their effects on gene regulation have not been discovered. Here, we study simple sequence repeats (SSRs), a common class of short structural variants. Previously, we showed that repetitive sequences can directly influence the binding of transcription factors to their proximate recognition sites, a mechanism we termed non-consensus binding. In this study, we focus on the SSR termed Rep1, which was associated with Parkinson's disease (PD) and has been implicated in the cis-regulation of the PD-risk SNCA gene. We show that Rep1 acts via the non-consensus binding mechanism to affect the binding of transcription factors from the GATA and ELK families to their specific sites located right next to the Rep1 repeat. Next, we performed an expression analysis to further our understanding regarding the GATA and ELK family members that are potentially relevant for SNCA transcriptional regulation in health and disease. Our analysis indicates a potential role for GATA2, consistent with previous reports. Our study proposes non-consensus transcription factor binding as a potential mechanism through which noncoding repeat variants could exert their pathogenic effects by regulating gene expression.
Collapse
Affiliation(s)
- A Afek
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, 27710, USA.,Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - L Tagliafierro
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - O C Glenn
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - D B Lukatsky
- Department of Chemistry, Ben-Gurion University of the Negev, 8410501, Beersheba, Israel
| | - R Gordan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, 27710, USA. .,Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27710, USA. .,Department of Computer Science, Duke University, Durham, NC, 27708, USA.
| | - O Chiba-Falek
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27710, USA. .,Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
23
|
Angelova DM, Jones HBL, Brown DR. Levels of α- and β-synuclein regulate cellular susceptibility to toxicity from α-synuclein oligomers. FASEB J 2018; 32:995-1006. [PMID: 29054856 DOI: 10.1096/fj.201700675r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
α-Synuclein (α-syn) is associated with a range of diseases, including Parkinson disease. In disease, α-syn is known to aggregate and has the potential to be neurotoxic. The association between copper and α-syn results in the formation of stellate toxic oligomers that are highly toxic to cultured neurons. We further investigated the mechanism of toxicity of α-syn oligomers. Cells that overexpress α-syn showed increased susceptibility to the toxicity of the oligomers, while those that overexpressed β-syn showed increased resistance to the toxic oligomers. Elevated α-syn expression caused an increase in expression of the transcription factor Forkhead box O3a (FoxO3a). Inhibition of FoxO3a activity by the overexpression of DNA binding domain of FoxO3a resulted in significant protection from α-syn oligomer toxicity. Increased FoxO3a expression in cells was shown to be caused by increased ferrireductase activity and Fe(II) levels. These results suggest that α-syn increases FoxO3a expression as a result of its intrinsic ferrireductase activity. The results also suggest that FoxO3a plays a pivotal role in the toxicity of both Fe(II) and toxic α-syn species to neuronal cells.-Angelova, D. M., Jones, H. B. L., Brown, D. R. Levels of α- and β-synuclein regulate cellular susceptibility to toxicity from α-synuclein oligomers.
Collapse
Affiliation(s)
- Dafina M Angelova
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Hannah B L Jones
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - David R Brown
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| |
Collapse
|
24
|
Gámez-Valero A, Beyer K. Alternative Splicing of Alpha- and Beta-Synuclein Genes Plays Differential Roles in Synucleinopathies. Genes (Basel) 2018; 9:genes9020063. [PMID: 29370097 PMCID: PMC5852559 DOI: 10.3390/genes9020063] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 11/16/2022] Open
Abstract
The synuclein family is composed of three members, two of which, α- and β-synuclein, play a major role in the development of synucleinopathies, including Parkinson’s disease (PD) as most important movement disorder, dementia with Lewy bodies (DLB) as the second most frequent cause of dementia after Alzheimer’s disease and multiple system atrophy. Whereas abnormal oligomerization and fibrillation of α-synuclein are now well recognized as initial steps in the development of synucleinopathies, β-synuclein is thought to be a natural α-synuclein anti-aggregant. α-synuclein is encoded by the SNCA gene, and β-synuclein by SNCB. Both genes are homologous and undergo complex splicing events. On one hand, in-frame splicing of coding exons gives rise to at least three shorter transcripts, and the functional properties of the corresponding protein isoforms are different. Another type of alternative splicing is the alternative inclusion of at least four initial exons in the case of SNCA, and two in the case of SNCB. Finally, different lengths of 3’ untranslated regions have been also reported for both genes. SNCB only expresses in the brain, but some of the numerous SNCA transcripts are also brain-specific. With the present article, we aim to provide a systematic review of disease related changes in the differential expression of the various SNCA and SNCB transcript variants in brain, blood, and non-neuronal tissue of synucleinopathies, but especially PD and DLB as major neurodegenerative disorders.
Collapse
Affiliation(s)
- Ana Gámez-Valero
- Department of Pathology, Germans Trias i Pujol Research Institute, Badalona, 08916 Barcelona, Spain.
| | - Katrin Beyer
- Department of Pathology, Germans Trias i Pujol Research Institute, Badalona, 08916 Barcelona, Spain.
| |
Collapse
|
25
|
Yoshino Y, Mori T, Yoshida T, Yamazaki K, Ozaki Y, Sao T, Funahashi Y, Iga JI, Ueno SI. Elevated mRNA Expression and Low Methylation of SNCA in Japanese Alzheimer's Disease Subjects. J Alzheimers Dis 2018; 54:1349-1357. [PMID: 27567856 DOI: 10.3233/jad-160430] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Despite the continuing debate about the amyloid hypothesis in Alzheimer's disease (AD), the precise pathogenesis is still unclear. Mixed pathology is common and multiple different protein aggregates are seen in human postmortem brains. Aggregates consisting of the alpha-synuclein protein encoded by the Synuclein Alpha gene (SCNA) are common in both dementia with Lewy bodies and AD. We examined SNCA mRNA expression and methylation rates of the CpG island at intron 1 of SNCA in peripheral leukocytes in 50 AD and age- and sex-matched control subjects to verify whether alpha-synuclein pathology affects the AD pathogenesis. SNCA mRNA expression in AD subjects was significantly higher than that in control subjects (1.62±0.73 versus 0.98±0.50, p < 0.001). We found significant differences between AD and control subjects at seven CpG sites (average rate; 8.8±2.7 versus 9.5±2.5, respectively: p = 0.027). The methylation rates tended to be lower in AD subjects at all CpG sites. We conclude that mRNA expression and methylation of SNCA intron 1 are altered in AD, which may be caused by Lewy body pathology in AD.
Collapse
|
26
|
Phosphorylated α-Synuclein-Copper Complex Formation in the Pathogenesis of Parkinson's Disease. PARKINSONS DISEASE 2017; 2017:9164754. [PMID: 29333317 PMCID: PMC5733240 DOI: 10.1155/2017/9164754] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/11/2017] [Indexed: 11/17/2022]
Abstract
Parkinson's disease is the second most important neurodegenerative disorder worldwide. It is characterized by the presence of Lewy bodies, which are mainly composed of α-synuclein and ubiquitin-bound proteins. Both the ubiquitin proteasome system (UPS) and autophagy-lysosomal pathway (ALS) are altered in Parkinson's disease, leading to aggregation of proteins, particularly α-synuclein. Interestingly, it has been observed that copper promotes the protein aggregation process. Additionally, phosphorylation of α-synuclein along with copper also affects the protein aggregation process. The interrelation among α-synuclein phosphorylation and its capability to interact with copper, with the subsequent disruption of the protein degradation systems in the neurodegenerative process of Parkinson's disease, will be analyzed in detail in this review.
Collapse
|
27
|
Miotto MC, Pavese MD, Quintanar L, Zweckstetter M, Griesinger C, Fernández CO. Bioinorganic Chemistry of Parkinson’s Disease: Affinity and Structural Features of Cu(I) Binding to the Full-Length β-Synuclein Protein. Inorg Chem 2017; 56:10387-10395. [DOI: 10.1021/acs.inorgchem.7b01292] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Marco C. Miotto
- Max Planck Laboratory
for Structural Biology, Chemistry and Molecular Biophysics of Rosario
and Instituto de Investigaciones para el Descubrimiento de Fármacos
de Rosario, Universidad Nacional de Rosario, Ocampo y Esmeralda, S2002LRK Rosario, Argentina
| | - Mayra D. Pavese
- Max Planck Laboratory
for Structural Biology, Chemistry and Molecular Biophysics of Rosario
and Instituto de Investigaciones para el Descubrimiento de Fármacos
de Rosario, Universidad Nacional de Rosario, Ocampo y Esmeralda, S2002LRK Rosario, Argentina
| | - Liliana Quintanar
- Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico
Nacional 2508, 07360 D.F., México
| | - Markus Zweckstetter
- Department of NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, D-37077 Göttingen, Germany
- Deutches Zentrum für Neurodegenerative Erkrankungen, von-Siebold-Str. 3a, 37075 Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073 Göttingen, Germany
| | - Christian Griesinger
- Department of NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, D-37077 Göttingen, Germany
| | - Claudio O. Fernández
- Max Planck Laboratory
for Structural Biology, Chemistry and Molecular Biophysics of Rosario
and Instituto de Investigaciones para el Descubrimiento de Fármacos
de Rosario, Universidad Nacional de Rosario, Ocampo y Esmeralda, S2002LRK Rosario, Argentina
- Department of NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, D-37077 Göttingen, Germany
| |
Collapse
|
28
|
Chintalapudi SR, Jablonski MM. Systems Genetics Analysis to Identify the Genetic Modulation of a Glaucoma-Associated Gene. Methods Mol Biol 2017; 1488:391-417. [PMID: 27933535 DOI: 10.1007/978-1-4939-6427-7_18] [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: 12/10/2022]
Abstract
Loss of retinal ganglion cells (RGCs) is one of the hallmarks of retinal neurodegenerative diseases, glaucoma being one of the most common. Recently, γ-synuclein (SNCG) was shown to be highly expressed in the somas and axons of RGCs. In various mouse models of glaucoma, downregulation of Sncg gene expression correlates with RGC loss. To investigate the regulation of Sncg in RGCs, we used a systems genetics approach to identify a gene that modulates the expression of Sncg, followed by confirmatory studies in both healthy and diseased retinas. We found that chromosome 1 harbors an eQTL that modulates the expression of Sncg in the mouse retina and identified Pfdn2 as the candidate upstream modulator of Sncg expression. Downregulation of Pfdn2 in enriched RGCs causes a concomitant reduction in Sncg. In this chapter, we describe our strategy and methods for identifying and confirming a genetic modulation of a glaucoma-associated gene. A similar method can be applied to other genes expressed in other tissues.
Collapse
Affiliation(s)
- Sumana R Chintalapudi
- Department of Anatomy and Neurobiology, Hamilton Eye Institute, The University of Tennessee Health Science Center, 930 Madison Ave., Suite 710, Memphis, TN, 38163, USA
| | - Monica M Jablonski
- Department of Ophthalmology, Hamilton Eye Institute, The University of Tennessee Health Science Center, 930 Madison Ave., Suite 710, Memphis, TN, 38163, USA. .,Department of Anatomy and Neurobiology, Hamilton Eye Institute, The University of Tennessee Health Science Center, 930 Madison Ave., Suite 710, Memphis, TN, 38163, USA. .,Department of Pharmaceutical Sciences, Hamilton Eye Institute, The University of Tennessee Health Science Center, 930 Madison Ave., Suite 710, Memphis, TN, 38163, USA.
| |
Collapse
|
29
|
β-Synuclein suppresses both the initiation and amplification steps of α-synuclein aggregation via competitive binding to surfaces. Sci Rep 2016; 6:36010. [PMID: 27808107 PMCID: PMC5093550 DOI: 10.1038/srep36010] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/04/2016] [Indexed: 11/08/2022] Open
Abstract
α-Synuclein is an intrinsically disordered protein that is associated with the pathogenesis of Parkinson's disease through the processes involved in the formation of amyloid fibrils. α and β-synuclein are homologous proteins found at comparable levels in presynaptic terminals but β-synuclein has a greatly reduced propensity to aggregate and indeed has been found to inhibit α-synuclein aggregation. In this paper, we describe how sequence differences between α- and β-synuclein affect individual microscopic processes in amyloid formation. In particular, we show that β-synuclein strongly suppresses both lipid-induced aggregation and secondary nucleation of α-synuclein by competing for binding sites at the surfaces of lipid vesicles and fibrils, respectively. These results suggest that β-synuclein can act as a natural inhibitor of α-synuclein aggregation by reducing both the initiation of its self-assembly and the proliferation of its aggregates.
Collapse
|
30
|
Wang Q, Tian Q, Song X, Liu Y, Li W. SNCA Gene Polymorphism may Contribute to an Increased Risk of Alzheimer's Disease. J Clin Lab Anal 2016; 30:1092-1099. [PMID: 27184464 PMCID: PMC6807080 DOI: 10.1002/jcla.21986] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/31/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The purpose of this study is to elucidate the association between α-synuclein (SNCA) polymorphisms and the risk of Alzheimer's disease (AD). METHODS The PCR-RFLP was applied to detect SNCA gene rs6532190, rs3775430, and rs10516846 polymorphisms in 98 AD patients and 105 healthy elderly. RESULTS The GG frequency of rs10516846 was evidently increased in AD group than control group (P < 0.05). There was a significant difference in SNCA level between the AD and control groups (P < 0.01). In the AD group, the SNCA level in cerebrospinal fluid of GG (rs10516846) carriers was increased as compared with AA carriers (P < 0.05). The GG (rs10516846) frequency of the early-onset AD group is significantly higher than that of the late-onset AD group (P < 0.05). The frequency of rs3775430 GG was lower in the early-onset group than that in the late-onset group (0% vs. 16.7%). The SNCA level in cerebrospinal fluid of GG (rs10516846) carriers in the early-onset AD group is higher than that of AA carriers (P < 0.05). CONCLUSION SNCA gene polymorphism may be associated with an increased risk of AD and GG genotype of rs10516846 and elevated SNCA level in CSF may increase the risk of early-onset AD.
Collapse
Affiliation(s)
- Quanbao Wang
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong Province, China
| | - Qian Tian
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong Province, China
| | - Xiaojie Song
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong Province, China
| | - Yunyong Liu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong Province, China
| | - Wei Li
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong Province, China.
| |
Collapse
|
31
|
Protection against neurodegenerative disease on Earth and in space. NPJ Microgravity 2016; 2:16013. [PMID: 28725728 PMCID: PMC5515513 DOI: 10.1038/npjmgrav.2016.13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/19/2016] [Accepted: 03/06/2016] [Indexed: 01/07/2023] Open
Abstract
All living organisms have evolutionarily adapted themselves to the Earth’s gravity, and failure to adapt to gravity changes may lead to pathological conditions. This perspective may also apply to abnormal aging observed in bedridden elderly patients with aging-associated diseases such as osteoporosis and sarcopenia. Given that bedridden elderly patients are partially analogous to astronauts in that both cannot experience the beneficial effects of gravity on the skeletal system and may suffer from bone loss and muscle weakness, one may wonder whether there are gravity-related mechanisms underlying diseases among the elderly. In contrast to numerous studies of the relevance of microgravity in skeletal disorders, little attention has been paid to neurodegenerative diseases. Therefore, the objective of this paper is to discuss the possible relevance of microgravity in these diseases. We particularly noted a proteomics paper showing that levels of hippocampal proteins, including β-synuclein and carboxyl-terminal ubiquitin hydrolase L1, which have been linked to familial neurodegenerative diseases, were significantly decreased in the hippocampus of mice subjected to hindlimb suspension, a model of microgravity. We suggest that microgravity-induced neurodegeneration may be further exacerbated by diabetes and other factors. On the basis of this view, prevention of neurodegenerative diseases through ‘anti-diabetes’ and ‘hypergravity’ approaches may be important as a common therapeutic approach on Earth and in space. Collectively, neurodegenerative diseases and space medicine may be linked to each other more strongly than previously thought.
Collapse
|
32
|
Tagliafierro L, Chiba-Falek O. Up-regulation of SNCA gene expression: implications to synucleinopathies. Neurogenetics 2016; 17:145-57. [PMID: 26948950 DOI: 10.1007/s10048-016-0478-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/27/2016] [Indexed: 01/06/2023]
Abstract
Synucleinopathies are a group of neurodegenerative diseases that share a common pathological lesion of intracellular protein inclusions largely composed by aggregates of alpha-synuclein protein. Accumulating evidence, including genome wide association studies, has implicated alpha-synuclein (SNCA) gene in the etiology of synucleinopathies. However, the precise variants within SNCA gene that contribute to the sporadic forms of Parkinson's disease (PD), dementia with Lewy bodies (DLB), multiple system atrophy (MSA), and other synucleinopathies and their molecular mechanisms of action remain elusive. It has been suggested that SNCA expression levels are critical for the development of these diseases. Here, we review several model systems that have been developed to advance the understanding of the role of SNCA expression levels in the etiology of synucleinopathies. We also describe different molecular mechanisms that regulate SNCA gene expression and discuss possible strategies for SNCA down-regulation as means for therapeutic approaches. Finally, we highlight some examples that underscore the relationships between the genetic association findings and the regulatory mechanisms of SNCA expression, which suggest that genetic variability in SNCA locus is directly responsible, at least in part, to the changes in gene expression and explain the reported associations of SNCA with synucleinopathies. Future studies utilizing induced pluripotent stem cells (iPSCs)-derived neuronal lines and genome editing by CRISPR/Cas9, will allow us to validate, characterize, and manipulate the effects of particular cis-genetic variants on SNCA expression. Moreover, this model system will enable us to compare different neuronal and glial lineages involved in synucleinopathies representing an attractive strategy to elucidate-common and specific-SNCA-genetic variants, regulatory mechanisms, and vulnerable expression levels underlying synucleinopathy spectrum disorders. This forthcoming knowledge will support the development of precision medicine for synucleinopathies.
Collapse
Affiliation(s)
- L Tagliafierro
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - O Chiba-Falek
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
33
|
Dermentzaki G, Paschalidis N, Politis PK, Stefanis L. Complex Effects of the ZSCAN21 Transcription Factor on Transcriptional Regulation of α-Synuclein in Primary Neuronal Cultures and in Vivo. J Biol Chem 2016; 291:8756-72. [PMID: 26907683 DOI: 10.1074/jbc.m115.704973] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Indexed: 11/06/2022] Open
Abstract
α-Synuclein, a presynaptic neuronal protein encoded by the SNCA gene, is strongly implicated in Parkinson disease (PD). PD pathogenesis is linked to increased SNCA levels; however, the transcriptional elements that control SNCA expression are still elusive. Previous experiments in PC12 cells demonstrated that the transcription factor zinc finger and SCAN domain containing 21 (ZSCAN21) plays an important regulatory role in SNCA transcription. Currently, we characterized the role of ZSCAN21 in SNCA transcription in primary neuronal cultures and in vivo We found that ZSCAN21 is developmentally expressed in neurons in different rat brain regions. We confirmed its binding in the intron 1 region of SNCA in rat cortical cultures. Lentivirus-mediated silencing of ZSCAN21 increased significantly SNCA promoter activity, mRNA, and protein levels in such cultures. In contrast, ZSCAN21 silencing reduced SNCA in neurosphere cultures. Interestingly, ZSCAN21 overexpression in cortical neurons led to robust mRNA but negligible protein expression, suggesting that ZSCAN21 protein levels are tightly regulated post-transcriptionally and/or post-translationally in primary neurons. Efficient adeno-associated virus-mediated knockdown of ZSCAN21 in the postnatal and adult hippocampus, an area linked with non-motor PD symptoms, revealed no significant alterations in SNCA levels. Overall, our study demonstrates that ZSCAN21 is involved in the transcriptional regulation of SNCA in primary neuronal cultures, but the direction of the effect is variable, likely depending on neuronal maturation. However, the unaltered SNCA levels observed following ZSCAN21 down-regulation in the rat brain, possibly due to compensatory mechanisms, imply that ZSCAN21 is not a master regulator of SNCA in vivo.
Collapse
Affiliation(s)
- Georgia Dermentzaki
- From the Biomedical Research Foundation of the Academy of Athens, Athens 11527 and
| | - Nikolaos Paschalidis
- From the Biomedical Research Foundation of the Academy of Athens, Athens 11527 and
| | - Panagiotis K Politis
- From the Biomedical Research Foundation of the Academy of Athens, Athens 11527 and
| | - Leonidas Stefanis
- From the Biomedical Research Foundation of the Academy of Athens, Athens 11527 and the Second Department of Neurology, National and Kapodistrian University of Athens Medical School, Hospital Attikon, Athens 12462, Greece,
| |
Collapse
|
34
|
Chintalapudi SR, Morales-Tirado VM, Williams RW, Jablonski MM. Multipronged approach to identify and validate a novel upstream regulator of Sncg in mouse retinal ganglion cells. FEBS J 2016; 283:678-93. [PMID: 26663874 DOI: 10.1111/febs.13620] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/22/2015] [Accepted: 12/03/2015] [Indexed: 11/26/2022]
Abstract
Loss of retinal ganglion cells (RGCs) is one of the hallmarks of retinal neurodegenerative diseases, glaucoma being one of the most common. Mechanistic studies on RGCs are hindered by the lack of sufficient primary cells and consensus regarding their signature markers. Recently, γ-synuclein (SNCG) has been shown to be highly expressed in the somas and axons of RGCs. In various mouse models of glaucoma, downregulation of Sncg gene expression correlates with RGC loss. To investigate the role of Sncg in RGCs, we used a novel systems genetics approach to identify a gene that modulates Sncg expression, followed by confirmatory studies in both healthy and diseased retinae. We found that chromosome 1 harbors an expression quantitative trait locus that modulates Sncg expression in the mouse retina, and identified the prefoldin-2 (PFDN2) gene as the candidate upstream modulator of Sncg expression. Our immunohistochemical analyses revealed similar expression patterns in both mouse and human healthy retinae, with PFDN2 colocalizing with SNCG in RGCs and their axons. In contrast, in retinae from glaucoma subjects, SNCG levels were significantly reduced, although PFDN2 levels were maintained. Using a novel flow cytometry-based RGC isolation method, we obtained viable populations of murine RGCs. Knocking down Pfdn2 expression in primary murine RGCs significantly reduced Sncg expression, confirming that Pfdn2 regulates Sncg expression in murine RGCs. Gene Ontology analysis indicated shared mitochondrial function associated with Sncg and Pfdn2. These data solidify the relationship between Sncg and Pfdn2 in RGCs, and provide a novel mechanism for maintaining RGC health.
Collapse
Affiliation(s)
- Sumana R Chintalapudi
- Department of Ophthalmology, The Hamilton Eye Institute, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Vanessa M Morales-Tirado
- Department of Ophthalmology, The Hamilton Eye Institute, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Robert W Williams
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Monica M Jablonski
- Department of Ophthalmology, The Hamilton Eye Institute, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
35
|
McDowall JS, Brown DR. Alpha-synuclein: relating metals to structure, function and inhibition. Metallomics 2016; 8:385-97. [DOI: 10.1039/c6mt00026f] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
36
|
Janowska MK, Wu KP, Baum J. Unveiling transient protein-protein interactions that modulate inhibition of alpha-synuclein aggregation by beta-synuclein, a pre-synaptic protein that co-localizes with alpha-synuclein. Sci Rep 2015; 5:15164. [PMID: 26477939 PMCID: PMC4609965 DOI: 10.1038/srep15164] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/11/2015] [Indexed: 01/18/2023] Open
Abstract
Pathology in Parkinson’s disease is linked to self-association of α-Synuclein (αS) into pathogenic oligomeric species and highly ordered amyloid fibrils. Developing effective therapeutic strategies against this debilitating disease is critical and βS, a pre-synaptic protein that co-localizes with αS, can act as an inhibitor of αS assembly. Despite the potential importance of βS as an inhibitor of αS, the nature, location and specificity of the molecular interactions between these two proteins is unknown. Here we use NMR paramagnetic relaxation enhancement experiments, to demonstrate that βS interacts directly with αS in a transient dimer complex with high specificity and weak affinity. Inhibition of αS by βS arises from transient αS/βS heterodimer species that exist primarily in head- to- tail configurations while αS aggregation arises from a more heterogeneous and weaker range of transient interactions that include both head-to-head and head-to-tail configurations. Our results highlight that intrinsically disordered proteins can interact directly with one another at low affinity and that the transient interactions that drive inhibition versus aggregation are distinct by virtue of their plasticity and specificity.
Collapse
Affiliation(s)
- Maria K Janowska
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey 08854
| | - Kuen-Phon Wu
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey 08854
| | - Jean Baum
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey 08854
| |
Collapse
|
37
|
Krassnig S, Schweinzer C, Taub N, Havas D, Auer E, Flunkert S, Schreibmayer W, Hutter-Paier B, Windisch M. Influence of Lentiviral β-Synuclein Overexpression in the Hippocampus of a Transgenic Mouse Model of Alzheimer's Disease on Amyloid Precursor Protein Metabolism and Pathology. NEURODEGENER DIS 2015; 15:243-57. [PMID: 26111745 DOI: 10.1159/000430952] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 04/26/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND β-Synuclein (β-Syn) is a member of the highly homologous synuclein protein family. The most prominent family member, α-synuclein (α-Syn), abnormally accumulates in so-called Lewy bodies, one of the major pathological hallmarks of α-synucleinopathies. Notably, parts of the peptide backbone, called the nonamyloid component, are also found in amyloid plaques. However, β-Syn seems to have beneficial effects by reducing α-Syn aggregation, and amyloid antiaggregatory activity has been described. OBJECTIVE The aim of the study was to analyze if wild-type β-Syn can counteract functional and pathological changes in a murine Alzheimer model over different time periods. METHODS At the onset of pathology, lentiviral particles expressing human β-Syn were injected into the hippocampus of transgenic mice overexpressing human amyloid precursor protein with Swedish and London mutations (APPSL). An empty vector served as the control. Behavioral analyses were performed 1, 3 and 6 months after injection followed by biochemical and histological examinations of brain samples. RESULTS β-Syn expression was locally concentrated and rather modest, but nevertheless changed its effect on APP expression and plaque load in a time- and concentration-dependent manner. Interestingly, the phosphorylation of glycogen synthase kinase 3 beta was enhanced in APPSL mice expressing human β-Syn, but an inverse trend was observed in wild-type animals. CONCLUSION The initially reported beneficial effects of β-Syn could be partially reproduced, but locally elevated levels of β-Syn might also cause neurodegeneration. To enlighten the controversial pathological mechanism of β-Syn, further examinations considering the relationship between concentration and exposure time of β-Syn are needed.
Collapse
|
38
|
Liu X, Zuo H, Wang D, Peng R, Song T, Wang S, Xu X, Gao Y, Li Y, Wang S, Wang L, Zhao L. Improvement of spatial memory disorder and hippocampal damage by exposure to electromagnetic fields in an Alzheimer's disease rat model. PLoS One 2015; 10:e0126963. [PMID: 25978363 PMCID: PMC4433192 DOI: 10.1371/journal.pone.0126963] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 04/09/2015] [Indexed: 01/05/2023] Open
Abstract
Although some epidemiological investigations showed a potential association between long-term exposure of extremely low frequency electromagnetic fields (ELF-EMF) and Alzheimer’s disease (AD), no reasonable mechanism can explain this association, and the related animal experiments are rare. In this study, ELF-EMF exposure (50Hz 400µT 60d) combined with D-galactose intraperitoneal (50mg/kg, q.d., 42d) and Aβ25–35 hippocampal (5μl/unilateral, bilateral, single-dose) injection was implemented to establish a complex rat model. Then the effects of ELF-EMF exposure on AD development was studied by using the Morris water maze, pathological analysis, and comparative proteomics. The results showed that ELF-EMF exposure delayed the weight gain of rats, and partially improved cognitive and clinicopathologic symptoms of AD rats. The differential proteomic analysis results suggest that synaptic transmission, oxidative stress, protein degradation, energy metabolism, Tau aggregation, and inflammation involved in the effects mentioned above. Therefore, our findings indicate that certain conditions of ELF-EMF exposure could delay the development of AD in rats.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Hongyan Zuo
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
- * E-mail: (HZ); (DW)
| | - Dewen Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
- * E-mail: (HZ); (DW)
| | - Ruiyun Peng
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, 6 North Second Street, Zhongguancun, Beijing, China
| | - Shuiming Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Xinping Xu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Yabing Gao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Yang Li
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Shaoxia Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Lifeng Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Li Zhao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| |
Collapse
|
39
|
De Ricco R, Valensin D, Dell’Acqua S, Casella L, Dorlet P, Faller P, Hureau C. Remote His50 Acts as a Coordination Switch in the High-Affinity N-Terminal Centered Copper(II) Site of α-Synuclein. Inorg Chem 2015; 54:4744-51. [DOI: 10.1021/acs.inorgchem.5b00120] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Riccardo De Ricco
- CNRS, LCC (Laboratoire
de Chimie de Coordination), 205 route
de Narbonne, BP 44099, F-31077 Toulouse Cedex 4, France
- Université
de Toulouse, UPS, INPT, F-31077 Toulouse Cedex 4, France
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Daniela Valensin
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Simone Dell’Acqua
- Department
of Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Luigi Casella
- Department
of Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Pierre Dorlet
- Institute
for Integrative Biology of the Cell (I2BC), Laboratoire Stress Oxydant
et Détoxication, CNRS UMR9198, Université Paris-Saclay, 91191 Gif sur Yvette Cedex, France
| | - Peter Faller
- CNRS, LCC (Laboratoire
de Chimie de Coordination), 205 route
de Narbonne, BP 44099, F-31077 Toulouse Cedex 4, France
- Université
de Toulouse, UPS, INPT, F-31077 Toulouse Cedex 4, France
| | - Christelle Hureau
- CNRS, LCC (Laboratoire
de Chimie de Coordination), 205 route
de Narbonne, BP 44099, F-31077 Toulouse Cedex 4, France
- Université
de Toulouse, UPS, INPT, F-31077 Toulouse Cedex 4, France
| |
Collapse
|
40
|
The Role of α-Synuclein and LRRK2 in Tau Phosphorylation. PARKINSONS DISEASE 2015; 2015:734746. [PMID: 25977830 PMCID: PMC4419261 DOI: 10.1155/2015/734746] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 04/02/2015] [Accepted: 04/02/2015] [Indexed: 01/08/2023]
Abstract
There is now a considerable body of experimental evidence that Parkinson's disease arises through physiological interaction of causative molecules, leading to tau pathology. In this review, we discuss the physiological role of α-synuclein and LRRK2 in the abnormal phosphorylation of tau. In addition, as recent reports have indicated that heat shock proteins- (HSPs-) inducing drugs can help to ameliorate neurodegenerative diseases associated with tau pathology, we also discuss therapeutic strategies for PD focusing on inhibition of α-synuclein- and LRRK2-associated tau phosphorylation by HSPs.
Collapse
|
41
|
Abstract
α-Synuclein is an abundant neuronal protein which localizes predominantly to presynaptic terminals, and is strongly linked genetically and pathologically to Parkinson's disease and other neurodegenerative diseases. While the accumulation of α-synuclein in the form of misfolded oligomers and large aggregates defines multiple neurodegenerative diseases called "synucleinopathies", its cellular function has remained largely unclear, and is the subject of intense investigation. In this review, I focus on the structural characteristics of α-synuclein, its cellular and subcellular localization, and discuss how this relates to its function in neurons, in particular at the neuronal synapse.
Collapse
Affiliation(s)
- Jacqueline Burré
- Appel Institute for Alzheimer’s Disease Research, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
42
|
De Ricco R, Valensin D, Dell'Acqua S, Casella L, Gaggelli E, Valensin G, Bubacco L, Mangani S. Differences in the binding of copper(I) to α- and β-synuclein. Inorg Chem 2014; 54:265-72. [PMID: 25495902 DOI: 10.1021/ic502407w] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the presence of abnormal α-synuclein (αS) deposits in the brain. Alterations in homeostasis and metal-induced oxidative stress may play a crucial role in the progression of αS amyloid assembly and pathogenesis of PD. Contrary to αS, β-synuclein (βS) is not involved in the PD etiology. However, it has been suggested that the βS/αS ratio is altered in PD, indicating that a correct balance of these two proteins is implicated in the inhibition of αS aggregation. αS and βS share similar abilities to coordinate Cu(II). In this study, we investigated and compared the interaction of Cu(I) with the N-terminal portion of βS and αS by means of NMR, circular dichroism, and X-ray absorption spectroscopies. Our data show the importance of M10K mutation, which induces different Cu(I) chemical environments. Coordination modes 3S1O and 2S2O were identified for βS and αS, respectively. These new insights into the bioinorganic chemistry of copper and synuclein proteins are a basis to understand the molecular mechanism by which βS might inhibit αS aggregation.
Collapse
Affiliation(s)
- Riccardo De Ricco
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena , Via Aldo Moro 2, Siena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Many proteins associated with neurodegenerative diseases have poorly defined or unknown functions. α-Synuclein is one such protein which is associated with a range of diseases including Parkinson's disease. Now accepted as a metal-binding protein, α-synuclein's function could possibly be defined in relation to the binding of cofactors. It has been suggested recently that α-synuclein is able to reduce iron using copper as its catalytic centre. The consequence of this is that possibly the function of α-synuclein can now be defined. The evidence for this and the consequences for Parkinson's disease are discussed in the present review.
Collapse
|
44
|
Abstract
Human genetics has indicated a causal role for the protein α-synuclein in the pathogenesis of familial Parkinson's disease (PD), and the aggregation of synuclein in essentially all patients with PD suggests a central role for this protein in the sporadic disorder. Indeed, the accumulation of misfolded α-synuclein now defines multiple forms of neural degeneration. Like many of the proteins that accumulate in other neurodegenerative disorders, however, the normal function of synuclein remains poorly understood. In this article, we review the role of synuclein at the nerve terminal and in membrane remodeling. We also consider the prion-like propagation of misfolded synuclein as a mechanism for the spread of degeneration through the neuraxis.
Collapse
|
45
|
Wright JA, McHugh PC, Pan S, Cunningham A, Brown DR. Counter-regulation of alpha- and beta-synuclein expression at the transcriptional level. Mol Cell Neurosci 2013; 57:33-41. [DOI: 10.1016/j.mcn.2013.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/22/2013] [Accepted: 09/20/2013] [Indexed: 12/15/2022] Open
|
46
|
Opposing actions of environmental enrichment and Alzheimer's disease on the expression of hippocampal microRNAs in mouse models. Transl Psychiatry 2013; 3:e304. [PMID: 24022509 PMCID: PMC3784766 DOI: 10.1038/tp.2013.77] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 07/19/2013] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly. Although there are no drugs that modify the disease process, exposure to an enriched environment (EE) can slow the disease progression. Here, we characterize the effects of AD and EE on the post-transcriptional regulators, microRNAs (miRNAs), which may contribute to the detrimental and beneficial effects of AD and EE, respectively, on synaptic plasticity-related proteins and AD pathology. We found for the first time miRNAs that were inversely regulated in AD and EE, and may affect synaptic proteins and modulators, molecular factors associated with AD pathology, and survival and neuroprotective factors. MiRNAs that were upregulated only in 3xTgAD mice model of AD compared with their control mice were localized to synapses, predicted to downregulate essential synaptic proteins and are highly associated with regulating apoptosis, AD-associated processes and axon guidance. Studying the progressive change in miRNAs modulation during aging of 3xTgAD mice, we identified miRNAs that were regulated in earlier stages of AD, suggesting them as potential AD biomarkers. Last, we characterized AD- and EE-related effects in the mouse hippocampus on tomosyn protein levels, an inhibitor of the synaptic transmission machinery. While EE reduced tomosyn levels, tomosyn levels were increased in old 3xTgAD mice, suggesting a role for tomosyn in the impairment of synaptic transmission in AD. Interestingly, we found that miR-325 regulates the expression levels of tomosyn as demonstrated by a luciferase reporter assay, and that miR-325 was downregulated in AD and upregulated following EE. These findings improve our understanding of the molecular and cellular processes in AD pathology, following EE, and the interplay between the two processes, and open new avenues for the studies of understanding and controlling AD.
Collapse
|
47
|
Chaput D, Kirouac LH, Bell-Temin H, Stevens SM, Padmanabhan J. SILAC-based proteomic analysis to investigate the impact of amyloid precursor protein expression in neuronal-like B103 cells. Electrophoresis 2013; 33:3728-37. [PMID: 23161580 DOI: 10.1002/elps.201200251] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 08/27/2012] [Accepted: 08/27/2012] [Indexed: 11/05/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly. Amyloid plaque formation through aggregation of the amyloid beta peptide derived from amyloid precursor protein (APP) is considered one of the hallmark processes leading to AD pathology; however, the precise role of APP in plaque formation and AD pathogenesis is yet to be determined. Using stable isotope labeling by amino acids in cell culture (SILAC) and MS, protein expression profiles of APP null, rat neuronal-like B103 cells were compared to B103-695 cells that express the APP isoform, APP-695. A total of 2979 unique protein groups were identified among three biological replicates and significant protein expression changes were identified in a total of 102 nonredundant proteins. Some of the top biological functions associated with the differentially expressed proteins identified include cellular assembly, organization and morphology, cell cycle, lipid metabolism, protein folding, and PTMs. We report several novel biological pathways influenced by APP-695 expression in neuronal-like cells and provide additional framework for investigating altered molecular mechanisms associated with APP expression and processing and contribution to AD pathology.
Collapse
Affiliation(s)
- Dale Chaput
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
48
|
Brown DR. Gene regulation as a potential avenue for the treatment of neurodegenerative disorders. Expert Opin Drug Discov 2013; 4:515-24. [PMID: 23485084 DOI: 10.1517/17460440902849237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND As more people live to an older age, the frequency of diseases associated with longer life begins to increase. Neurodegenerative disorders are the worst of these in that there is now no treatment that offers any real improvement. For this reason, any new avenue of research that could lead to a treatment needs to be rigorously pursued. In many cases, neurodegenerative diseases are associated with the expression of a protein with an altered conformation or that generates a breakdown product associated with the cause. Clearly, the prevention of this process is a key therapeutic target. OBJECTIVE In this review, the potential for regulating gene expression to prevent or reverse neurodegenerative disease is explored. CONCLUSIONS Whereas much research has been directed at the proteins associated with neurodegeneration, understanding what controls their expression presents a new way this issue could be studied.
Collapse
Affiliation(s)
- David R Brown
- University of Bath, Department of Biology and Biochemistry, Bath, BA2 7AY, UK +44 1225 383133 ; +44 1225 386779 ;
| |
Collapse
|
49
|
Synaptic Proteins and Choline Acetyltransferase Loss in Visual Cortex in Dementia With Lewy Bodies. J Neuropathol Exp Neurol 2013; 72:53-60. [DOI: 10.1097/nen.0b013e31827c5710] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
50
|
Canela L, Selga E, García-Martínez JM, Amaral OB, Fernández-Dueñas V, Alberch J, Canela EI, Franco R, Noé V, Lluís C, Ciudad CJ, Ciruela F. Transcriptional profiling of striatal neurons in response to single or concurrent activation of dopamine D2, adenosine A(2A) and metabotropic glutamate type 5 receptors: focus on beta-synuclein expression. Gene 2012; 508:199-205. [PMID: 22892378 DOI: 10.1016/j.gene.2012.07.074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 07/30/2012] [Indexed: 11/28/2022]
Abstract
G protein-coupled receptor oligomerization is a concept which is changing the understanding of classical pharmacology. Both, oligomerization and functional interaction between adenosine A(2A,) dopamine D(2) and metabotropic glutamate type 5 receptors have been demonstrated in the striatum. However, the transcriptional consequences of receptors co-activation are still unexplored. We aim here to determine the changes in gene expression of striatal primary cultured neurons upon isolated or simultaneous receptor activation. Interestingly, we found that 95 genes of the total analyzed (15,866 transcripts and variants) changed their expression in response to simultaneous stimulation of all three receptors. Among these genes, we focused on the β-synuclein (β-Syn) gene (SCNB). Quantitative PCR verified the magnitude and direction of change in expression of SCNB. Since β-Syn belongs to the homologous synuclein family and may be considered a natural regulator of α-synuclein (α-Syn), it has been proposed that β-Syn might act protectively against α-Syn neuropathology.
Collapse
Affiliation(s)
- Laia Canela
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|