151
|
Giorgianni F, Beranova-Giorgianni S. Phosphoproteome Discovery in Human Biological Fluids. Proteomes 2016; 4:proteomes4040037. [PMID: 28248247 PMCID: PMC5260970 DOI: 10.3390/proteomes4040037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 11/11/2016] [Accepted: 11/23/2016] [Indexed: 01/07/2023] Open
Abstract
Phosphorylation plays a critical role in regulating protein function and thus influences a vast spectrum of cellular processes. With the advent of modern bioanalytical technologies, examination of protein phosphorylation on a global scale has become one of the major research areas. Phosphoproteins are found in biological fluids and interrogation of the phosphoproteome in biological fluids presents an exciting opportunity for discoveries that hold great potential for novel mechanistic insights into protein function in health and disease, and for translation to improved diagnostic and therapeutic approaches for the clinical setting. This review focuses on phosphoproteome discovery in selected human biological fluids: serum/plasma, urine, cerebrospinal fluid, saliva, and bronchoalveolar lavage fluid. Bioanalytical workflows pertinent to phosphoproteomics of biological fluids are discussed with emphasis on mass spectrometry-based approaches, and summaries of studies on phosphoproteome discovery in major fluids are presented.
Collapse
Affiliation(s)
- Francesco Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sarka Beranova-Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
152
|
Zahratka JA, Shao Y, Shaw M, Todd K, Formica SV, Khrestian M, Montine T, Leverenz JB, Bekris LM. Regulatory region genetic variation is associated with FYN expression in Alzheimer's disease. Neurobiol Aging 2016; 51:43-53. [PMID: 28033507 DOI: 10.1016/j.neurobiolaging.2016.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 10/26/2016] [Accepted: 11/08/2016] [Indexed: 12/15/2022]
Abstract
Neurofibrillary tangles (NFTs), composed of hyperphosphorylated tau, are a key pathologic feature of Alzheimer's disease (AD). Tau phosphorylation is under the control of multiple kinases and phosphatases, including Fyn. Previously, our group found an association between 2 regulatory single nucleotide polymorphisms in the FYN gene with increased tau levels in the cerebrospinal fluid. In this study, we hypothesized that Fyn expression in the brain is influenced by AD status and genetic content. We found that Fyn protein, but not messenger RNA, levels were increased in AD patients compared to cognitively normal controls and are associated with regulatory region single nucleotide polymorphisms. In addition, the expression of the FYN 3'UTR can decrease expression in multiple cell lines, suggesting this regulatory region plays an important role in FYN expression. Taken together, these data suggest that FYN expression is regulated according to AD status and regulatory region haplotype, and genetic variants may be instrumental in the development of neurofibrillary tangles in AD and other tauopathies.
Collapse
Affiliation(s)
- Jeffrey A Zahratka
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Yvonne Shao
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - McKenzie Shaw
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Kaitlin Todd
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Shane V Formica
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Maria Khrestian
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Thomas Montine
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lynn M Bekris
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
153
|
Ghosh D, Mehra S, Sahay S, Singh PK, Maji SK. α-synuclein aggregation and its modulation. Int J Biol Macromol 2016; 100:37-54. [PMID: 27737778 DOI: 10.1016/j.ijbiomac.2016.10.021] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 10/06/2016] [Accepted: 10/09/2016] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is a neurological disorder marked by the presence of cytoplasmic inclusions, Lewy bodies (LBs) and Lewy neurites (LNs) as well as the degeneration of dopamine producing neurons in the substantia nigra region of the brain. The LBs and LNs in PD are mainly composed of aggregated form of a presynaptic protein, α-synuclein (α-Syn). However, the mechanisms of α-Syn aggregation and actual aggregated species responsible for the degeneration of dopaminergic neurons have not yet been resolved. Despite the fact that α-Syn aggregation in LBs and LNs is crucial and mutations of α-Syn are associated with early onset PD, it is really a challenging task to establish a correlation between α-Syn aggregation rate and PD pathogenesis. Regardless of strong genetic contribution, PD is mostly sporadic and familial forms of the disease represent only a minor part (<10%) of all cases. The complexity in PD further increases due to the involvement of several cellular factors in the pathogenesis of the disease as well as the environmental factors associated with the risk of developing PD. Therefore, effect of these factors on α-Syn aggregation pathway and how these factors modulate the properties of wild type (WT) as well as mutated α-Syn should be collectively taken into account. The present review specifically provides an overview of recent research on α-Syn aggregation pathways and its modulation by several cellular factors potentially relevant to PD pathogenesis. We also briefly discuss about effect of environmental risk factors on α-Syn aggregation.
Collapse
Affiliation(s)
- Dhiman Ghosh
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India.
| | - Surabhi Mehra
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Shruti Sahay
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India.
| | - Pradeep K Singh
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India.
| |
Collapse
|
154
|
Sun W, Lee S, Huang X, Liu S, Inayathullah M, Kim KM, Tang H, Ashford JW, Rajadas J. Attenuation of synaptic toxicity and MARK4/PAR1-mediated Tau phosphorylation by methylene blue for Alzheimer's disease treatment. Sci Rep 2016; 6:34784. [PMID: 27708431 PMCID: PMC5052533 DOI: 10.1038/srep34784] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 09/19/2016] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by genotypic and phenotypic heterogeneity. Critical components of the two AD pathological pathways, Aβ-amyloidosis and Tauopathy, have been considered as therapeutic targets. Among them, much effort is focused on aberrant Tau phosphorylation and targeting Tau-phosphorylating kinases. Methylene blue (MB), a phenothiazine dye that crosses the blood-brain barrier, has been shown to hit multiple molecular targets involved in AD and have beneficial effects in clinical studies. Here we present evidence that microtubule affinity-regulating kinase (MARK4) is a novel target of MB. MB partially rescued the synaptic toxicity in Drosophila larva overexpressing PAR1 (MARK analog). In 293T culture, MB decreased MARK4-mediated Tau phosphorylation in a dose dependent manner. Further studies revealed a two-fold mechanism by MB including down-regulation of MARK4 protein level through ubiquitin-proteasome pathway and inhibition of MARK4 kinase activity in vitro. This study highlights the importance of MARK4 as a viable target for Tauopathy and provides fresh insight into the complex mechanism used by MB to treat AD.
Collapse
Affiliation(s)
- Wenchao Sun
- Biomaterial and Advanced Drug Delivery Lab, Stanford University School of Medicine, Stanford, California, USA
| | - Seongsoo Lee
- Biomaterial and Advanced Drug Delivery Lab, Stanford University School of Medicine, Stanford, California, USA.,Gwangju Center, Korea Basic Science Institute, Gwangju 61186, Korea
| | - Xiaoran Huang
- Biomaterial and Advanced Drug Delivery Lab, Stanford University School of Medicine, Stanford, California, USA
| | - Song Liu
- Biomaterial and Advanced Drug Delivery Lab, Stanford University School of Medicine, Stanford, California, USA
| | - Mohammed Inayathullah
- Biomaterial and Advanced Drug Delivery Lab, Stanford University School of Medicine, Stanford, California, USA
| | - Kwang-Min Kim
- Biomaterial and Advanced Drug Delivery Lab, Stanford University School of Medicine, Stanford, California, USA
| | - Hongxiang Tang
- Biomaterial and Advanced Drug Delivery Lab, Stanford University School of Medicine, Stanford, California, USA
| | - J Wesson Ashford
- War Related Illness and Injury Study Center (WRIISC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Jayakumar Rajadas
- Biomaterial and Advanced Drug Delivery Lab, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
155
|
Tyson T, Steiner JA, Brundin P. Sorting out release, uptake and processing of alpha-synuclein during prion-like spread of pathology. J Neurochem 2016; 139 Suppl 1:275-289. [PMID: 26617280 PMCID: PMC4958606 DOI: 10.1111/jnc.13449] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 12/17/2022]
Abstract
Parkinson's disease is a progressive neurological disorder that is characterized by the formation of intracellular protein inclusion bodies composed primarily of a misfolded and aggregated form of the protein α-synuclein. There is growing evidence that supports the prion-like hypothesis of α-synuclein progression. This hypothesis postulates that α-synuclein is a prion-like pathological agent and is responsible for the progression of Parkinson pathology in the brain. Potential misfolding or aggregation of α-synuclein that might occur in the peripheral nervous system as a result of some insult, environmental or genetic (or more likely a combination of both) that might spread into the midbrain, eventually causing degeneration of the neurons in the substantia nigra. As the disease progresses further, it is likely that α-synuclein pathology continues to spread throughout the brain, including the cortex, leading to deterioration of cognition and higher brain functions. While it is unknown why α-synuclein initially misfolds and aggregates, a great deal has been learned about how the cell handles aberrant α-synuclein assemblies. In this review, we focus on these mechanisms and discuss them in an attempt to define the role that they might play in the propagation of misfolded α-synuclein from cell-to-cell. The prion-like hypothesis of α-synuclein pathology suggests a method for the transmission of misfolded α-synuclein from one neuron to another. This hypothesis postulates that misfolded α-synuclein becomes aggregation prone and when released and taken up by neighboring cells, seeds further misfolding and aggregation. In this review we examine the cellular mechanisms that are involved in the processing of α-synuclein and how these may contribute to the prion-like propagation of α-synuclein pathology. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Trevor Tyson
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jennifer A Steiner
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA.
| |
Collapse
|
156
|
Loss of Tau protein affects the structure, transcription and repair of neuronal pericentromeric heterochromatin. Sci Rep 2016; 6:33047. [PMID: 27605042 PMCID: PMC5015075 DOI: 10.1038/srep33047] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/04/2016] [Indexed: 12/27/2022] Open
Abstract
Pericentromeric heterochromatin (PCH) gives rise to highly dense chromatin sub-structures rich in the epigenetic mark corresponding to the trimethylated form of lysine 9 of histone H3 (H3K9me3) and in heterochromatin protein 1α (HP1α), which regulate genome expression and stability. We demonstrate that Tau, a protein involved in a number of neurodegenerative diseases including Alzheimer's disease (AD), binds to and localizes within or next to neuronal PCH in primary neuronal cultures from wild-type mice. Concomitantly, we show that the clustered distribution of H3K9me3 and HP1α, two hallmarks of PCH, is disrupted in neurons from Tau-deficient mice (KOTau). Such altered distribution of H3K9me3 that could be rescued by overexpressing nuclear Tau protein was also observed in neurons from AD brains. Moreover, the expression of PCH non-coding RNAs, involved in PCH organization, was disrupted in KOTau neurons that displayed an abnormal accumulation of stress-induced PCH DNA breaks. Altogether, our results demonstrate a new physiological function of Tau in directly regulating neuronal PCH integrity that appears disrupted in AD neurons.
Collapse
|
157
|
Nevzglyadova OV, Artemov AV, Mikhailova EV, Lyublinskaya OG, Ozerova YE, Ivanova PA, Kostyleva EI, Soidla TR. The effect of yeast Saccharomyces cerevisiae red pigment on the expression of cloned human α-synuclein. ACTA ACUST UNITED AC 2016. [DOI: 10.1134/s1990519x16040106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
158
|
Kleinknecht A, Popova B, Lázaro DF, Pinho R, Valerius O, Outeiro TF, Braus GH. C-Terminal Tyrosine Residue Modifications Modulate the Protective Phosphorylation of Serine 129 of α-Synuclein in a Yeast Model of Parkinson's Disease. PLoS Genet 2016; 12:e1006098. [PMID: 27341336 PMCID: PMC4920419 DOI: 10.1371/journal.pgen.1006098] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 05/10/2016] [Indexed: 12/15/2022] Open
Abstract
Parkinson´s disease (PD) is characterized by the presence of proteinaceous inclusions called Lewy bodies that are mainly composed of α-synuclein (αSyn). Elevated levels of oxidative or nitrative stresses have been implicated in αSyn related toxicity. Phosphorylation of αSyn on serine 129 (S129) modulates autophagic clearance of inclusions and is prominently found in Lewy bodies. The neighboring tyrosine residues Y125, Y133 and Y136 are phosphorylation and nitration sites. Using a yeast model of PD, we found that Y133 is required for protective S129 phosphorylation and for S129-independent proteasome clearance. αSyn can be nitrated and form stable covalent dimers originating from covalent crosslinking of two tyrosine residues. Nitrated tyrosine residues, but not di-tyrosine-crosslinked dimers, contributed to αSyn cytotoxicity and aggregation. Analysis of tyrosine residues involved in nitration and crosslinking revealed that the C-terminus, rather than the N-terminus of αSyn, is modified by nitration and di-tyrosine formation. The nitration level of wild-type αSyn was higher compared to that of A30P mutant that is non-toxic in yeast. A30P formed more dimers than wild-type αSyn, suggesting that dimer formation represents a cellular detoxification pathway in yeast. Deletion of the yeast flavohemoglobin gene YHB1 resulted in an increase of cellular nitrative stress and cytotoxicity leading to enhanced aggregation of A30P αSyn. Yhb1 protected yeast from A30P-induced mitochondrial fragmentation and peroxynitrite-induced nitrative stress. Strikingly, overexpression of neuroglobin, the human homolog of YHB1, protected against αSyn inclusion formation in mammalian cells. In total, our data suggest that C-terminal Y133 plays a major role in αSyn aggregate clearance by supporting the protective S129 phosphorylation for autophagy and by promoting proteasome clearance. C-terminal tyrosine nitration increases pathogenicity and can only be partially detoxified by αSyn di-tyrosine dimers. Our findings uncover a complex interplay between S129 phosphorylation and C-terminal tyrosine modifications of αSyn that likely participates in PD pathology. Parkinson’s disease is characterized by loss of dopaminergic neurons in midbrain and the presence of αSyn protein inclusions. Human αSyn mimics the disease pathology in yeast resulting in cytotoxicity and aggregate formation. αSyn is abundantly phosphorylated at serine S129 and possesses four tyrosines (Y39, Y125, Y133, and Y136) that can be posttranslationally modified by nitration or phosphorylation. The consequence of each of these possible modifications is still unclear. Nitration as consequence of oxidative stress is a hallmark for neurodegenerative diseases. Here, we addressed the molecular mechanism, how tyrosine posttranslational modifications affect αSyn cytotoxicity. Tyrosine nitration can contribute to αSyn toxicity or can be part of a cellular salvage pathway when di-tyrosine-crosslinked dimers are formed. The Y133 residue, which can be either phosphorylated or nitrated, determines whether S129 is protectively phosphorylated and αSyn inclusions are cleared. This interplay with S129 phosphorylation demonstrates a dual role for C-terminal tyrosine residues. Yeast flavohemoglobin Yhb1 and its human counterpart neuroglobin NGB protect cells against cytotoxicity and aggregate formation. These novel insights into the molecular pathways responsible for αSyn cytotoxicity indicate NGB as a potential target for therapeutic intervention in PD.
Collapse
Affiliation(s)
- Alexandra Kleinknecht
- Department of Molecular Microbiology and Genetics and Göttingen Center for Molecular Biosciences (GZMB), Institute of Microbiology and Genetics, Georg-August-Universität, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Blagovesta Popova
- Department of Molecular Microbiology and Genetics and Göttingen Center for Molecular Biosciences (GZMB), Institute of Microbiology and Genetics, Georg-August-Universität, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Diana F. Lázaro
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- Department of NeuroDegeneration and Restorative Research, University of Göttingen Medical School, Göttingen, Germany
| | - Raquel Pinho
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- Department of NeuroDegeneration and Restorative Research, University of Göttingen Medical School, Göttingen, Germany
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Oliver Valerius
- Department of Molecular Microbiology and Genetics and Göttingen Center for Molecular Biosciences (GZMB), Institute of Microbiology and Genetics, Georg-August-Universität, Göttingen, Germany
| | - Tiago F. Outeiro
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- Department of NeuroDegeneration and Restorative Research, University of Göttingen Medical School, Göttingen, Germany
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Gerhard H. Braus
- Department of Molecular Microbiology and Genetics and Göttingen Center for Molecular Biosciences (GZMB), Institute of Microbiology and Genetics, Georg-August-Universität, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- * E-mail:
| |
Collapse
|
159
|
Rezaei-Ghaleh N, Kumar S, Walter J, Zweckstetter M. Phosphorylation Interferes with Maturation of Amyloid-β Fibrillar Structure in the N Terminus. J Biol Chem 2016; 291:16059-67. [PMID: 27252381 DOI: 10.1074/jbc.m116.728956] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Indexed: 11/06/2022] Open
Abstract
Neurodegeneration is characterized by the ubiquitous presence of modifications in protein deposits. Despite their potential significance in the initiation and progression of neurodegenerative diseases, the effects of posttranslational modifications on the molecular properties of protein aggregates are largely unknown. Here, we study the Alzheimer disease-related amyloid-β (Aβ) peptide and investigate how phosphorylation at serine 8 affects the structure of Aβ aggregates. Serine 8 is shown to be located in a region of high conformational flexibility in monomeric Aβ, which upon phosphorylation undergoes changes in local conformational dynamics. Using hydrogen-deuterium exchange NMR and fluorescence quenching techniques, we demonstrate that Aβ phosphorylation at serine 8 causes structural changes in the N-terminal region of Aβ aggregates in favor of less compact conformations. Structural changes induced by serine 8 phosphorylation can provide a mechanistic link between phosphorylation and other biological events that involve the N-terminal region of Aβ aggregates. Our data therefore support an important role of posttranslational modifications in the structural polymorphism of amyloid aggregates and their modulatory effect on neurodegeneration.
Collapse
Affiliation(s)
- Nasrollah Rezaei-Ghaleh
- From the German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Strasse 3a, 37075 Göttingen, Germany, Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany,
| | - Sathish Kumar
- Department of Neurology, University of Bonn, 53127 Bonn, Germany, and
| | - Jochen Walter
- Department of Neurology, University of Bonn, 53127 Bonn, Germany, and
| | - Markus Zweckstetter
- From the German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Strasse 3a, 37075 Göttingen, Germany, Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany, Department of Neurology, University Medical Center, University of Göttingen, Göttingen, Waldweg 33, 37073 Göttingen, Germany
| |
Collapse
|
160
|
Cao Y, Liang L, Xu J, Wu J, Yan Y, Lin P, Chen Q, Zheng F, Wang Q, Ren Q, Gou Z, Du Y. The effect of Scutellaria baicalensis stem-leaf flavonoids on spatial learning and memory in chronic cerebral ischemia-induced vascular dementia of rats. Acta Biochim Biophys Sin (Shanghai) 2016; 48:437-46. [PMID: 27118553 DOI: 10.1093/abbs/gmw024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 01/23/2016] [Indexed: 12/19/2022] Open
Abstract
Flavonoids have been shown to improve cognitive function and delay the dementia progression. However, the underlying mechanisms remain elusive. In the present study, we examined the effect of Scutellaria baicalensis stem-leaf total flavonoids (SSTFs) extracted from S. baicalensis Georgi on spatial learning and memory in a vascular dementia (VaD) rat model and explored its molecular mechanisms. The VaD rats were developed by permanent bilateral occlusion of the common carotid artery. Seven days after recovery, the VaD rats were treated with either 50 or 100 mg/kg of SSTF for 60 days. The spatial learning and memory was evaluated in the Morris water maze (MWM) test. The tau hyperphosphorylation and the levels of the related protein kinases or phosphatases were examined by western blot analysis. In VaD rats, SSTF treatment at 100 mg/kg significantly reduced the escape latency in training trial in MWM test. In the probe trial, SSTF treatment increased the searching time and travel distance in the target quadrant. SSTF treatment inhibited the tau phosphorylation in both cortex and hippocampus in VaD rats. Meanwhile, SSTF reduced the activity of glycogen synthase kinase 3β and cyclin-dependent kinase 5 in VaD rats. In contrast, SSTF treatment increased the level of the protein phosphatase 2A subunit B in VaD rats. SSTF treatment significantly improved the spatial cognition in VaD rats. Our results suggest that SSTF may alleviate tau-hyperphosphorylation-induced neurotoxicity through coordinating the activity of kinases and phosphatase after a stroke. SSTF may be developed into promising novel therapeutics for VaD.
Collapse
Affiliation(s)
- Yanjing Cao
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan 250013, China Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Lizhen Liang
- Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Jian Xu
- Department of Histology and Embryology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiali Wu
- Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Yongxing Yan
- Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Ping Lin
- Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Qiang Chen
- Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Fengming Zheng
- Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Qin Wang
- Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Qian Ren
- Department of Neurology, Third Hospital of Hangzhou, Hangzhou 310009, China
| | - Zengmei Gou
- Department of Neurology, The Second People's Hospital of Weifang, Weifang 261041, China
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan 250013, China
| |
Collapse
|
161
|
Rezaei-Ghaleh N, Amininasab M, Kumar S, Walter J, Zweckstetter M. Phosphorylation modifies the molecular stability of β-amyloid deposits. Nat Commun 2016; 7:11359. [PMID: 27072999 PMCID: PMC4833870 DOI: 10.1038/ncomms11359] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 03/16/2016] [Indexed: 11/17/2022] Open
Abstract
Protein aggregation plays a crucial role in neurodegenerative diseases. A key feature of protein aggregates is their ubiquitous modification by phosphorylation. Little is known, however, about the molecular consequences of phosphorylation of protein aggregates. Here we show that phosphorylation of β-amyloid at serine 8 increases the stability of its pathogenic aggregates against high-pressure and SDS-induced dissociation. We further demonstrate that phosphorylation results in an elevated number of hydrogen bonds at the N terminus of β-amyloid, the region that is critically regulated by a variety of post-translational modifications. Because of the increased lifetime of phosphorylated β-amyloid aggregates, phosphorylation can promote the spreading of β-amyloid in Alzheimer pathogenesis. Our study suggests that regulation of the molecular stability of protein aggregates by post-translational modifications is a crucial factor for disease progression in the brain. Protein aggregation plays a crucial role in several neurodegenerative diseases. Here the authors demonstrate that phosphorylation of β-amyloid aggregates—the pathological hallmark of Alzheimer's disease—can change the molecular properties of aggregates, suggesting how phosphorylation contributes to disease progression.
Collapse
Affiliation(s)
| | - Mehriar Amininasab
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, 1417466191 Tehran, Iran
| | - Sathish Kumar
- Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Jochen Walter
- Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany.,Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany.,Department of Neurology, University Medical Center Göttingen, University of Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
162
|
Vilas D, Shaw LM, Taylor P, Berg D, Brockmann K, Aasly J, Marras C, Pont-Sunyer C, Ríos J, Marek K, Tolosa E. Cerebrospinal fluid biomarkers and clinical features in leucine-rich repeat kinase 2 (LRRK2) mutation carriers. Mov Disord 2016; 31:906-14. [DOI: 10.1002/mds.26591] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/22/2022] Open
Affiliation(s)
- Dolores Vilas
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Institut de Neurociències Hospital Clínic, University of Barcelona; Catalonia Spain
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine; Perelman School of Medicine, University of Pennsylvania; Philadelphia Pennsylvania USA
| | | | - Daniela Berg
- Department of Neurodegenerative Diseases and Hertie Institute for Clinical Brain Research; University of Tübingen; Tübingen Germany
| | - Kathrin Brockmann
- Department of Neurodegenerative Diseases and Hertie Institute for Clinical Brain Research; University of Tübingen; Tübingen Germany
| | - Jan Aasly
- Department of Neuroscience; Norwegian University of Science and Technology; Trondheim Norway
- Department of Neurology; St. Olav's Hospital; Trondheim Norway
| | - Connie Marras
- Morton and Gloria Shulman Movement Disorders Centre and the Edmond J Safra Program in Parkinson's Disease, Toronto Western Hospital; Toronto Ontario Canada
| | - Claustre Pont-Sunyer
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Institut de Neurociències Hospital Clínic, University of Barcelona; Catalonia Spain
| | - José Ríos
- Biostatistics and Data Management Core Facility, IDIBAPS (Hospital Clinic); Barcelona Spain
- Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona; Catalonia Spain
| | - Ken Marek
- Institute for Neurodegenerative Disorders; New Haven Connecticut USA
| | - Eduardo Tolosa
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Institut de Neurociències Hospital Clínic, University of Barcelona; Catalonia Spain
- Centro en Red para la Investigación de Enfermedades Neurodegenerativas CIBERNED; Spain
| |
Collapse
|
163
|
Goudsmit J. The incubation period of Alzheimer's disease and the timing of tau versus amyloid misfolding and spreading within the brain. Eur J Epidemiol 2016; 31:99-105. [PMID: 27017509 PMCID: PMC4819914 DOI: 10.1007/s10654-016-0144-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/11/2016] [Indexed: 02/04/2023]
Affiliation(s)
- Jaap Goudsmit
- Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands. .,Janssen Prevention Center, Leiden, The Netherlands.
| |
Collapse
|
164
|
Intracellular repair of oxidation-damaged α-synuclein fails to target C-terminal modification sites. Nat Commun 2016; 7:10251. [PMID: 26807843 PMCID: PMC4737712 DOI: 10.1038/ncomms10251] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/18/2015] [Indexed: 12/31/2022] Open
Abstract
Cellular oxidative stress serves as a common denominator in many neurodegenerative disorders, including Parkinson's disease. Here we use in-cell NMR spectroscopy to study the fate of the oxidation-damaged Parkinson's disease protein alpha-synuclein (α-Syn) in non-neuronal and neuronal mammalian cells. Specifically, we deliver methionine-oxidized, isotope-enriched α-Syn into cultured cells and follow intracellular protein repair by endogenous enzymes at atomic resolution. We show that N-terminal α-Syn methionines Met1 and Met5 are processed in a stepwise manner, with Met5 being exclusively repaired before Met1. By contrast, C-terminal methionines Met116 and Met127 remain oxidized and are not targeted by cellular enzymes. In turn, persisting oxidative damage in the C-terminus of α-Syn diminishes phosphorylation of Tyr125 by Fyn kinase, which ablates the necessary priming event for Ser129 modification by CK1. These results establish that oxidative stress can lead to the accumulation of chemically and functionally altered α-Syn in cells. α-synuclein is a protein linked to the occurrence of Parkinson's disease. Here, the authors use time-resolved in-cell NMR spectroscopy to study the repair of methionine-oxidized α-synuclein by endogenous cellular enzymes.
Collapse
|
165
|
Bukar Maina M, Al-Hilaly YK, Serpell LC. Nuclear Tau and Its Potential Role in Alzheimer's Disease. Biomolecules 2016; 6:9. [PMID: 26751496 PMCID: PMC4808803 DOI: 10.3390/biom6010009] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/16/2015] [Accepted: 12/23/2015] [Indexed: 12/14/2022] Open
Abstract
Tau protein, found in both neuronal and non-neuronal cells, forms aggregates in neurons that constitutes one of the hallmarks of Alzheimer’s disease (AD). For nearly four decades, research efforts have focused more on tau’s role in physiology and pathology in the context of the microtubules, even though, for over three decades, tau has been localised in the nucleus and the nucleolus. Its nuclear and nucleolar localisation had stimulated many questions regarding its role in these compartments. Data from cell culture, mouse brain, and the human brain suggests that nuclear tau could be essential for genome defense against cellular distress. However, its nature of translocation to the nucleus, its nuclear conformation and interaction with the DNA and other nuclear proteins highly suggest it could play multiple roles in the nucleus. To find efficient tau-based therapies, there is a need to understand more about the functional relevance of the varied cellular distribution of tau, identify whether specific tau transcripts or isoforms could predict tau’s localisation and function and how they are altered in diseases like AD. Here, we explore the cellular distribution of tau, its nuclear localisation and function and its possible involvement in neurodegeneration.
Collapse
Affiliation(s)
- Mahmoud Bukar Maina
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, East Sussex, UK.
- Department of Human Anatomy, College of Medical Science, Gombe State University, Gombe 760, Nigeria.
| | - Youssra K Al-Hilaly
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, East Sussex, UK.
- Chemistry Department, College of Sciences, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Louise C Serpell
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, East Sussex, UK.
| |
Collapse
|
166
|
Tenreiro S, Rosado-Ramos R, Gerhardt E, Favretto F, Magalhães F, Popova B, Becker S, Zweckstetter M, Braus GH, Outeiro TF. Yeast reveals similar molecular mechanisms underlying alpha- and beta-synuclein toxicity. Hum Mol Genet 2015; 25:275-90. [PMID: 26586132 DOI: 10.1093/hmg/ddv470] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/10/2015] [Indexed: 02/05/2023] Open
Abstract
Synucleins belong to a family of intrinsically unstructured proteins that includes alpha-synuclein (aSyn), beta-synuclein (bSyn) and gamma-synuclein (gSyn). aSyn is the most studied member of the synuclein family due to its central role in genetic and sporadic forms of Parkinson's disease and other neurodegenerative disorders known as synucleionopathies. In contrast, bSyn and gSyn have been less studied, but recent reports also suggest that, unexpectedly, these proteins may also cause neurotoxicity. Here, we explored the yeast toolbox to investigate the cellular effects of bSyn and gSyn. We found that bSyn is toxic and forms cytosolic inclusions that are similar to those formed by aSyn. Moreover, we found that bSyn shares similar toxicity mechanisms with aSyn, including vesicular trafficking impairment and induction of oxidative stress. We demonstrate that co-expression of aSyn and bSyn exacerbates cytotoxicity, due to increased dosage of toxic synuclein forms, and that they are able to form heterodimers in both yeast and in human cells. In contrast, gSyn is not toxic and does not form inclusions in yeast cells. Altogether, our findings shed light into the question of whether bSyn can exert toxic effects and confirms the occurrence of aSyn/bSyn heterodimers, opening novel perspectives for the development of novel strategies for therapeutic intervention in synucleinopathies.
Collapse
Affiliation(s)
- Sandra Tenreiro
- Instituto de Medicina Molecular, Lisboa, Portugal, CEDOC - Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal,
| | | | - Ellen Gerhardt
- Department of NeuroDegeneration and Restorative Research, University Medical Center Göttingen, Göttingen, Germany
| | - Filippo Favretto
- German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen, Germany
| | | | - Blagovesta Popova
- Department of Molecular Microbiology and Genetics, Institute of Microbiology & Genetics, Georg-August-Universität Göttingen, Göttingen, Germany, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Stefan Becker
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany and
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen, Germany, Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany and DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Gerhard H Braus
- Department of Molecular Microbiology and Genetics, Institute of Microbiology & Genetics, Georg-August-Universität Göttingen, Göttingen, Germany, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Tiago Fleming Outeiro
- CEDOC - Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal, German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen, Germany, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany,
| |
Collapse
|
167
|
Sprenger FS, Stefanova N, Gelpi E, Seppi K, Navarro-Otano J, Offner F, Vilas D, Valldeoriola F, Pont-Sunyer C, Aldecoa I, Gaig C, Gines A, Cuatrecasas M, Högl B, Frauscher B, Iranzo A, Wenning GK, Vogel W, Tolosa E, Poewe W. Enteric nervous system α-synuclein immunoreactivity in idiopathic REM sleep behavior disorder. Neurology 2015; 85:1761-8. [PMID: 26475692 PMCID: PMC4653104 DOI: 10.1212/wnl.0000000000002126] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/23/2015] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To investigate the expression of α-synuclein in colonic biopsies of patients with idiopathic REM sleep behavior disorder (iRBD) and address if α-synuclein immunostaining of tissue obtained via colonic biopsies holds promise as a diagnostic biomarker for prodromal Parkinson disease (PD). METHODS Patients with iRBD, patients with PD, and healthy controls were prospectively recruited to undergo colonic biopsies for comparison of α-synuclein immunoreactivity patterns between the groups by using 2 different antibodies. RESULTS There was no difference in colonic mucosal and submucosal immunostaining between groups using the 15G7 α-synuclein antibody, which was found in almost all participants enrolled in this study. By contrast, immunostaining for serine 129-phosphorylated α-synuclein (pSyn) in submucosal nerve fibers or ganglia was found in none of 14 controls but was observed in 4 of 17 participants with iRBD and 1 out of 19 patients with PD. CONCLUSIONS The present findings of pSyn immunostaining of colonic biopsies in a substantial proportion of iRBD participants raise the possibility that this tissue marker may be a suitable candidate to study further as a prodromal PD marker in at-risk cohorts.
Collapse
Affiliation(s)
- Fabienne S Sprenger
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Nadia Stefanova
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Ellen Gelpi
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Klaus Seppi
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Judith Navarro-Otano
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Felix Offner
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Dolores Vilas
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Francesc Valldeoriola
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Claustre Pont-Sunyer
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Iban Aldecoa
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Carles Gaig
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Angels Gines
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Miriam Cuatrecasas
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Birgit Högl
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Birgit Frauscher
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Alex Iranzo
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Gregor K Wenning
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Wolfgang Vogel
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Eduardo Tolosa
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria
| | - Werner Poewe
- From the Departments of Neurology (F.S.S., N.S., K.S., B.H., B.F., G.K.W., W.P.) and Gastroenterology (W.V.), Medical University of Innsbruck, Austria; Neurological Tissue Bank of the Biobanc-Hospital Clinic de Barcelona (E.G.), IDIBAPS; the Departments of Neurology (J.N.-O., D.V., F.V., C.P.-S., C.G., A.I., E.T.), Pathology (I.A., M.C.), and Gastroenterology (A.G.), Hospital Clinic de Barcelona, Spain; and the Institute of Pathology (F.O.), Academic Teaching Hospital, Feldkirch, Austria.
| |
Collapse
|
168
|
Li P, Nie Y, Yu J. An Effective Method to Identify Shared Pathways and Common Factors among Neurodegenerative Diseases. PLoS One 2015; 10:e0143045. [PMID: 26575483 PMCID: PMC4648499 DOI: 10.1371/journal.pone.0143045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/29/2015] [Indexed: 11/18/2022] Open
Abstract
Groups of distinct but related diseases often share common symptoms, which suggest likely overlaps in underlying pathogenic mechanisms. Identifying the shared pathways and common factors among those disorders can be expected to deepen our understanding for them and help designing new treatment strategies effected on those diseases. Neurodegeneration diseases, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD), were taken as a case study in this research. Reported susceptibility genes for AD, PD and HD were collected and human protein-protein interaction network (hPPIN) was used to identify biological pathways related to neurodegeneration. 81 KEGG pathways were found to be correlated with neurodegenerative disorders. 36 out of the 81 are human disease pathways, and the remaining ones are involved in miscellaneous human functional pathways. Cancers and infectious diseases are two major subclasses within the disease group. Apoptosis is one of the most significant functional pathways. Most of those pathways found here are actually consistent with prior knowledge of neurodegenerative diseases except two cell communication pathways: adherens and tight junctions. Gene expression analysis showed a high probability that the two pathways were related to neurodegenerative diseases. A combination of common susceptibility genes and hPPIN is an effective method to study shared pathways involved in a group of closely related disorders. Common modules, which might play a bridging role in linking neurodegenerative disorders and the enriched pathways, were identified by clustering analysis. The identified shared pathways and common modules can be expected to yield clues for effective target discovery efforts on neurodegeneration.
Collapse
Affiliation(s)
- Ping Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaling Nie
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingkai Yu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- * E-mail:
| |
Collapse
|
169
|
Luca M, Luca A, Calandra C. The Role of Oxidative Damage in the Pathogenesis and Progression of Alzheimer's Disease and Vascular Dementia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:504678. [PMID: 26301043 PMCID: PMC4537746 DOI: 10.1155/2015/504678] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/08/2015] [Indexed: 01/12/2023]
Abstract
Oxidative stress (OS) has been demonstrated to be involved in the pathogenesis of the two major types of dementia: Alzheimer's disease (AD) and vascular dementia (VaD). Evidence of OS and OS-related damage in AD is largely reported in the literature. Moreover, OS is not only linked to VaD, but also to all its risk factors. Several researches have been conducted in order to investigate whether antioxidant therapy exerts a role in the prevention and treatment of AD and VaD. Another research field is that pertaining to the heat shock proteins (Hsps), that has provided promising findings. However, the role of OS antioxidant defence system and more generally stress responses is very complex. Hence, research on this topic should be improved in order to reach further knowledge and discover new therapeutic strategies to face a disorder with such a high burden which is dementia.
Collapse
Affiliation(s)
- Maria Luca
- Psychiatry Unit, Department of Medical and Surgical Sciences and Advanced Technologies, University Hospital Policlinico-Vittorio Emanuele, Santa Sofia Street 78, Catania, 95100 Sicily, Italy
| | - Antonina Luca
- Department of “G.F. Ingrassia”, University Hospital Policlinico-Vittorio Emanuele, Santa Sofia Street 78, Catania, 95100 Sicily, Italy
| | - Carmela Calandra
- Psychiatry Unit, Department of Medical and Surgical Sciences and Advanced Technologies, University Hospital Policlinico-Vittorio Emanuele, Santa Sofia Street 78, Catania, 95100 Sicily, Italy
| |
Collapse
|
170
|
Martins WC, Tasca CI, Cimarosti H. Battling Alzheimer's Disease: Targeting SUMOylation-Mediated Pathways. Neurochem Res 2015; 41:568-78. [PMID: 26227998 DOI: 10.1007/s11064-015-1681-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 06/29/2015] [Accepted: 07/22/2015] [Indexed: 01/10/2023]
Abstract
SUMO (small ubiquitin-like modifier) conjugation is a critically important control process in all eukaryotic cells, because it acts as a biochemical switch and regulates the function of hundreds of proteins in many different pathways. Although the diverse functional consequences and molecular targets of SUMOylation remain largely unknown, SUMOylation is becoming increasingly implicated in the pathophysiology of Alzheimer's disease (AD). Apart from the central SUMO-modified disease-associated proteins, such as amyloid precursor protein, amyloid β, and tau, SUMOylation also regulates several other processes underlying AD. These are involved in inflammation, mitochondrial dynamics, synaptic transmission and plasticity, as well as in protective responses to cell stress. Herein, we review current reports on the involvement of SUMOylation in AD, and present an overview of potential SUMO targets and pathways underlying AD pathogenesis.
Collapse
Affiliation(s)
- Wagner Carbolin Martins
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Carla Inês Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Helena Cimarosti
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| |
Collapse
|
171
|
Abstract
Based on own translational research of the biochemical and hormonal effects of cow's milk consumption in humans, this review presents milk as a signaling system of mammalian evolution that activates the nutrient-sensitive kinase mechanistic target of rapamycin complex 1 (mTORC1), the pivotal regulator of translation. Milk, a mammary gland-derived secretory product, is required for species-specific gene-nutrient interactions that promote appropriate growth and development of the newborn mammal. This signaling system is highly conserved and tightly controlled by the lactation genome. Milk is sufficient to activate mTORC1, the crucial regulator of protein, lipid, and nucleotide synthesis orchestrating anabolism, cell growth and proliferation. To fulfill its mTORC1-activating function, milk delivers four key metabolic messengers: (1) essential branched-chain amino acids (BCAAs); (2) glutamine; (3) palmitic acid; and (4) bioactive exosomal microRNAs, which in a synergistical fashion promote mTORC1-dependent translation. In all mammals except Neolithic humans, postnatal activation of mTORC1 by milk intake is restricted to the postnatal lactation period. It is of critical concern that persistent hyperactivation of mTORC1 is associated with aging and the development of age-related disorders such as obesity, type 2 diabetes mellitus, cancer, and neurodegenerative diseases. Persistent mTORC1 activation promotes endoplasmic reticulum (ER) stress and drives an aimless quasi-program, which promotes aging and age-related diseases.
Collapse
|
172
|
Ashraf GM, Greig NH, Khan TA, Hassan I, Tabrez S, Shakil S, Sheikh IA, Zaidi SK, Akram M, Jabir NR, Firoz CK, Naeem A, Alhazza IM, Damanhouri GA, Kamal MA. Protein misfolding and aggregation in Alzheimer's disease and type 2 diabetes mellitus. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2015; 13:1280-93. [PMID: 25230234 DOI: 10.2174/1871527313666140917095514] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 05/11/2014] [Accepted: 05/12/2014] [Indexed: 12/27/2022]
Abstract
In general, proteins can only execute their various biological functions when they are appropriately folded. Their amino acid sequence encodes the relevant information required for correct three-dimensional folding, with or without the assistance of chaperones. The challenge associated with understanding protein folding is currently one of the most important aspects of the biological sciences. Misfolded protein intermediates form large polymers of unwanted aggregates and are involved in the pathogenesis of many human diseases, including Alzheimer's disease (AD) and Type 2 diabetes mellitus (T2DM). AD is one of the most prevalent neurological disorders and has worldwide impact; whereas T2DM is considered a metabolic disease that detrementally influences numerous organs, afflicts some 8% of the adult population, and shares many risk factors with AD. Research data indicates that there is a widespread conformational change in the proteins involved in AD and T2DM that form β-sheet like motifs. Although conformation of these β-sheets is common to many functional proteins, the transition from α-helix to β-sheet is a typical characteristic of amyloid deposits. Any abnormality in this transition results in protein aggregation and generation of insoluble fibrils. The abnormal and toxic proteins can interact with other native proteins and consequently catalyze their transition into the toxic state. Both AD and T2DM are prevalent in the aged population. AD is characterized by the accumulation of amyloid-β (Aβ) in brain, while T2DM is characterized by the deposition of islet amyloid polypeptide (IAPP, also known as amylin) within beta-cells of the pancreas. T2DM increases pathological angiogenesis and immature vascularisation. This also leads to chronic cerebral hypoperfusion, which results in dysfunction and degeneration of neuroglial cells. With an abundance of common mechanisms underpinning both disorders, a significant question that can be posed is whether T2DM leads to AD in aged individuals and the associations between other protein misfolding diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mohammad A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Kingdom of Saudi Arabia.
| |
Collapse
|
173
|
Xu S, Chan P. Interaction between Neuromelanin and Alpha-Synuclein in Parkinson's Disease. Biomolecules 2015; 5:1122-42. [PMID: 26057626 PMCID: PMC4496713 DOI: 10.3390/biom5021122] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 04/29/2015] [Indexed: 12/27/2022] Open
Abstract
Parkinson's disease (PD) is a very common neurodegenerative disorder characterized by the accumulation of α-synuclein (α-syn) into Lewy body (LB) inclusions and the loss of neuronmelanin (NM) containing dopamine (DA) neurons in the substantia nigra (SN). Pathological α-syn and NM are two prominent hallmarks in this selective and progressive neurodegenerative disease. Pathological α-syn can induce dopaminergic neuron death by various mechanisms, such as inducing oxidative stress and inhibiting protein degradation systems. Therefore, to explore the factors that trigger α-syn to convert from a non-toxic protein to toxic one is a pivotal question to clarify the mechanisms of PD pathogenesis. Many triggers for pathological α-syn aggregation have been identified, including missense mutations in the α-syn gene, higher concentration, and posttranslational modifications of α-Syn. Recently, the role of NM in inducing α-syn expression and aggregation has been suggested as a mechanism for this pigment to modulate neuronal vulnerability in PD. NM may be responsible for PD and age-associated increase and aggregation in α-syn. Here, we reviewed our previous study and other recent findings in the area of interaction between NM and α-syn.
Collapse
Affiliation(s)
- Shengli Xu
- Beijing Institute of Geriatrics, Xuanwu Hospital of Capital University of Medical Sciences, No.45 changchun St., Xicheng District, Beijing 100053, China.
- Parkinson's disease Center of Beijing Institute for Brain Disorders, Beijing 100053, China.
| | - Piu Chan
- Beijing Institute of Geriatrics, Xuanwu Hospital of Capital University of Medical Sciences, No.45 changchun St., Xicheng District, Beijing 100053, China.
- Parkinson's disease Center of Beijing Institute for Brain Disorders, Beijing 100053, China.
| |
Collapse
|
174
|
Popova B, Kleinknecht A, Braus GH. Posttranslational Modifications and Clearing of α-Synuclein Aggregates in Yeast. Biomolecules 2015; 5:617-34. [PMID: 25915624 PMCID: PMC4496687 DOI: 10.3390/biom5020617] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/01/2015] [Accepted: 04/14/2015] [Indexed: 12/20/2022] Open
Abstract
The budding yeast Saccharomyces cerevisiae represents an established model system to study the molecular mechanisms associated to neurodegenerative disorders. A key-feature of Parkinson’s disease is the formation of Lewy bodies, which are cytoplasmic protein inclusions. Misfolded α-synuclein is one of their main constituents. Expression of α-synuclein protein in yeast leads to protein aggregation and cellular toxicity, which is reminiscent to Lewy body containing human cells. The molecular mechanism involved in clearance of α-synuclein aggregates is a central question for elucidating the α-synuclein-related toxicity. Cellular clearance mechanisms include ubiquitin mediated 26S proteasome function as well as lysosome/vacuole associated degradative pathways as autophagy. Various modifications change α-synuclein posttranslationally and alter its inclusion formation, cytotoxicity and the distribution to different clearance pathways. Several of these modification sites are conserved from yeast to human. In this review, we summarize recent findings on the effect of phosphorylation and sumoylation of α-synuclein to the enhanced channeling to either the autophagy or the proteasome degradation pathway in yeast model of Parkinson’s disease.
Collapse
Affiliation(s)
- Blagovesta Popova
- Department of Molecular Microbiology and Genetics, Institute of Microbiology and Genetics, Georg-August-Universität Göttingen, D-37077 Göttingen, Germany.
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), D-37077 Göttingen, Germany.
| | - Alexandra Kleinknecht
- Department of Molecular Microbiology and Genetics, Institute of Microbiology and Genetics, Georg-August-Universität Göttingen, D-37077 Göttingen, Germany.
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), D-37077 Göttingen, Germany.
| | - Gerhard H Braus
- Department of Molecular Microbiology and Genetics, Institute of Microbiology and Genetics, Georg-August-Universität Göttingen, D-37077 Göttingen, Germany.
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), D-37077 Göttingen, Germany.
| |
Collapse
|
175
|
Lopes da Fonseca T, Villar-Piqué A, Outeiro TF. The Interplay between Alpha-Synuclein Clearance and Spreading. Biomolecules 2015; 5:435-71. [PMID: 25874605 PMCID: PMC4496680 DOI: 10.3390/biom5020435] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 12/23/2022] Open
Abstract
Parkinson's Disease (PD) is a complex neurodegenerative disorder classically characterized by movement impairment. Pathologically, the most striking features of PD are the loss of dopaminergic neurons and the presence of intraneuronal protein inclusions primarily composed of alpha-synuclein (α-syn) that are known as Lewy bodies and Lewy neurites in surviving neurons. Though the mechanisms underlying the progression of PD pathology are unclear, accumulating evidence suggests a prion-like spreading of α-syn pathology. The intracellular homeostasis of α-syn requires the proper degradation of the protein by three mechanisms: chaperone-mediated autophagy, macroautophagy and ubiquitin-proteasome. Impairment of these pathways might drive the system towards an alternative clearance mechanism that could involve its release from the cell. This increased release to the extracellular space could be the basis for α-syn propagation to different brain areas and, ultimately, for the spreading of pathology and disease progression. Here, we review the interplay between α-syn degradation pathways and its intercellular spreading. The understanding of this interplay is indispensable for obtaining a better knowledge of the molecular basis of PD and, consequently, for the design of novel avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Tomás Lopes da Fonseca
- Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen 37073, Germany.
- Instituto de Fisiologia, Faculty of Medicine, University of Lisbon, Lisboa 1649-028, Portugal.
| | - Anna Villar-Piqué
- Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen 37073, Germany.
| | - Tiago Fleming Outeiro
- Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen 37073, Germany.
- Instituto de Fisiologia, Faculty of Medicine, University of Lisbon, Lisboa 1649-028, Portugal.
- CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa 1150, Portugal.
| |
Collapse
|
176
|
Bentea E, Van der Perren A, Van Liefferinge J, El Arfani A, Albertini G, Demuyser T, Merckx E, Michotte Y, Smolders I, Baekelandt V, Massie A. Nigral proteasome inhibition in mice leads to motor and non-motor deficits and increased expression of Ser129 phosphorylated α-synuclein. Front Behav Neurosci 2015; 9:68. [PMID: 25873870 PMCID: PMC4379937 DOI: 10.3389/fnbeh.2015.00068] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 02/27/2015] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease is a neurodegenerative disorder characterized by motor and non-motor disturbances. Various pathogenic pathways drive disease progression including oxidative stress, mitochondrial dysfunction, α-synuclein aggregation and impairment of protein degradation systems. Dysfunction of the ubiquitin-proteasome system in the substantia nigra of Parkinson's disease patients is believed to be one of the causes of protein aggregation and cell death associated with this disorder. Lactacystin, a potent inhibitor of the proteasome, was previously delivered to the nigrostriatal pathway of rodents to model nigrostriatal degeneration. Although lactacystin-treated animals develop parkinsonian motor impairment, it is currently unknown whether they also develop non-motor symptoms characteristic of this disorder. In order to further describe the proteasome inhibition model of Parkinson's disease, we characterized the unilateral lactacystin model, performed by stereotaxic injection of the toxin in the substantia nigra of mice. We studied the degree of neurodegeneration and the behavioral phenotype 1 and 3 weeks after lactacystin lesion both in terms of motor impairment, as well as non-motor symptoms. We report that unilateral administration of 3 μg lactacystin to the substantia nigra of mice leads to partial (~40%) dopaminergic cell loss and concurrent striatal dopamine depletion, accompanied by increased expression of Ser129-phosphorylated α-synuclein. Behavioral characterization of the model revealed parkinsonian motor impairment, as well as signs of non-motor disturbances resembling early stage Parkinson's disease including sensitive and somatosensory deficits, anxiety-like behavior, and perseverative behavior. The consistent finding of good face validity, together with relevant construct validity, warrant a further evaluation of proteasome inhibition models of Parkinson's disease in pre-clinical research and validation of therapeutic targets.
Collapse
Affiliation(s)
- Eduard Bentea
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Anke Van der Perren
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Joeri Van Liefferinge
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Anissa El Arfani
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Thomas Demuyser
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Ellen Merckx
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Yvette Michotte
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Ann Massie
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| |
Collapse
|
177
|
Kosten J, Binolfi A, Stuiver M, Verzini S, Theillet FX, Bekei B, van Rossum M, Selenko P. Efficient modification of alpha-synuclein serine 129 by protein kinase CK1 requires phosphorylation of tyrosine 125 as a priming event. ACS Chem Neurosci 2014; 5:1203-8. [PMID: 25320964 DOI: 10.1021/cn5002254] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
S129-phosphorylated alpha-synuclein (α-syn) is abundantly found in Lewy-body inclusions of Parkinson's disease patients. Residues neighboring S129 include the α-syn tyrosine phosphorylation sites Y125, Y133, and Y136. Here, we use time-resolved NMR spectroscopy to delineate atomic resolution insights into the modification behaviors of different serine and tyrosine kinases targeting these sites and show that Y125 phosphorylation constitutes a necessary priming event for the efficient modification of S129 by CK1, both in reconstituted kinase reactions and mammalian cell lysates. These results suggest that α-syn Y125 phosphorylation augments S129 modification under physiological in vivo conditions.
Collapse
Affiliation(s)
- Jonas Kosten
- In-Cell NMR Laboratory, Department
of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle Strasse 10, 13125 Berlin, Germany
| | - Andres Binolfi
- In-Cell NMR Laboratory, Department
of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle Strasse 10, 13125 Berlin, Germany
| | - Marchel Stuiver
- In-Cell NMR Laboratory, Department
of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle Strasse 10, 13125 Berlin, Germany
| | - Silvia Verzini
- In-Cell NMR Laboratory, Department
of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle Strasse 10, 13125 Berlin, Germany
| | - Francois-Xavier Theillet
- In-Cell NMR Laboratory, Department
of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle Strasse 10, 13125 Berlin, Germany
| | - Beata Bekei
- In-Cell NMR Laboratory, Department
of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle Strasse 10, 13125 Berlin, Germany
| | - Marleen van Rossum
- In-Cell NMR Laboratory, Department
of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle Strasse 10, 13125 Berlin, Germany
| | - Philipp Selenko
- In-Cell NMR Laboratory, Department
of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle Strasse 10, 13125 Berlin, Germany
| |
Collapse
|
178
|
Phosphoproteomic profiling of selenate-treated Alzheimer's disease model cells. PLoS One 2014; 9:e113307. [PMID: 25485856 PMCID: PMC4259334 DOI: 10.1371/journal.pone.0113307] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/21/2014] [Indexed: 11/19/2022] Open
Abstract
The reversible phosphorylation of proteins regulates most biological processes, while abnormal phosphorylation is a cause or consequence of many diseases including Alzheimer's disease (AD). One of the hallmarks of AD is the formation of neurofibrillary tangles (NFTs), which is composed of hyperphosphorylated tau proteins. Sodium selenate has been recently found to reduce tau hyperphosphorylation and NFTs formation, and to improve spatial learning and motor performance in AD mice. In the current study, the phosphoproteomics of N2aSW cells treated with selenate were investigated. To avoid missing low-abundance phosphoproteins, both the total proteins of cells and the phosphor-enriched proteins were extracted and subjected to the two-dimensional gel electrophoresis with Pro-Q diamond staining and then LC-MS/MS analysis. A total of 65 proteins were altered in phosphorylation level, of which 39 were up-regulated and 26 were down-regulated. All identified phosphoproteins were bioinformatically annotated according to their physiochemical features, subcellular location, and biological function. Most of these significantly changed phosphoproteins are involved in crucial neural processes such as protesome activity, oxidative stress, cysteine and methionine metabolism, and energy metabolism. Furthermore, decreases were found in homocysteine, phosphor-tau and amyloid β upon selenate treatment. Our results suggest that selenate may intervene in the pathological process of AD by altering the phosphorylation of some key proteins involved in oxidative stress, energy metabolism and protein degradation, thus play important roles in maintaining redox homeostasis, generating ATP, and clearing misfolded proteins and aggregates. The present paper provides some new clues to the mechanism of selenate in AD prevention.
Collapse
|
179
|
TNF-α regulates miRNA targeting mitochondrial complex-I and induces cell death in dopaminergic cells. Biochim Biophys Acta Mol Basis Dis 2014; 1852:451-61. [PMID: 25481834 DOI: 10.1016/j.bbadis.2014.11.019] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/22/2014] [Accepted: 11/26/2014] [Indexed: 01/06/2023]
Abstract
Parkinson's disease (PD) is a complex neurological disorder of the elderly population and majorly shows the selective loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta (SNpc) region of the brain. The mechanisms leading to increased cell death of DAergic neurons are not well understood. Tumor necrosis factor-alpha (TNF-α), a pro-inflammatory cytokine is elevated in blood, CSF and striatum region of the brain in PD patients. The increased level of TNF-α and its role in pathogenesis of PD are not well understood. In the current study, we investigated the role of TNF-α in the regulation of cell death and miRNA mediated mitochondrial functions using, DAergic cell line, SH-SY5Y (model of dopaminergic neuron degeneration akin to PD). The cells treated with low dose of TNF-α for prolonged period induce cell death which was rescued in the presence of zVAD.fmk, a caspase inhibitor and N-acetyl-cysteine (NAC), an antioxidant. TNF-α alters mitochondrial complex-I activity, decreases adenosine triphosphate (ATP) levels, increases reactive oxygen species levels and mitochondrial turnover through autophagy. TNF-α differentially regulates miRNA expression involved in pathogenesis of PD. Bioinformatics analysis revealed that the putative targets of altered miRNA included both pro/anti apoptotic genes and subunits of mitochondrial complex. The cells treated with TNF-α showed decreased level of nuclear encoded transcript of mitochondrial complexes, the target of miRNA. To our knowledge, the evidences in the current study demonstrated that TNF-α is a potential regulator of miRNAs which may regulate mitochondrial functions and neuronal cell death, having important implication in pathogenesis of PD.
Collapse
|
180
|
Gonfloni S. Defying c-Abl signaling circuits through small allosteric compounds. Front Genet 2014; 5:392. [PMID: 25429298 PMCID: PMC4228975 DOI: 10.3389/fgene.2014.00392] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 10/25/2014] [Indexed: 11/13/2022] Open
Abstract
Many extracellular and intracellular signals promote the c-Abl tyrosine kinase activity. c-Abl in turn triggers a multitude of changes either in protein phosphorylation or in gene expression in the cell. Yet, c-Abl takes part in diverse signaling routes because of several domains linked to its catalytic core. Complex conformational changes turn on and off its kinase activity. These changes affect surface features of the c-Abl kinase and likely its capability to bind actin and/or DNA. Two specific inhibitors (ATP-competitive or allosteric compounds) regulate the c-Abl kinase through different mechanisms. NMR studies show that a c-Abl fragment (SH3-SH2-linker-SH1) adopts different conformational states upon binding to each inhibitor. This supports an unconventional use for allosteric compounds to unraveling physiological c-Abl signaling circuits.
Collapse
|
181
|
Taymans JM, Baekelandt V. Phosphatases of α-synuclein, LRRK2, and tau: important players in the phosphorylation-dependent pathology of Parkinsonism. Front Genet 2014; 5:382. [PMID: 25426138 PMCID: PMC4224088 DOI: 10.3389/fgene.2014.00382] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022] Open
Abstract
An important challenge in the field of Parkinson’s disease (PD) is to develop disease modifying therapies capable of stalling or even halting disease progression. Coupled to this challenge is the need to identify disease biomarkers, in order to identify pre-symptomatic hallmarks of disease and monitor disease progression. The answer to these challenges lies in the elucidation of the molecular causes underlying PD, for which important leads are disease genes identified in studies investigating the underlying genetic causes of PD. LRRK2 and α-syn have been both linked to familial forms of PD as well as associated to sporadic PD. Another gene, microtubule associated protein tau (MAPT), has been genetically linked to a dominant form of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) and genome-wide association studies report a strong association between MAPT and sporadic PD. Interestingly, LRRK2, α-syn, and tau are all phosphorylated proteins, and their phosphorylation patterns are linked to disease. In this review, we provide an overview of the evidence linking LRRK2, α-syn, and tau phosphorylation to PD pathology and focus on studies which have identified phosphatases responsible for dephosphorylation of pathology-related phosphorylations. We also discuss how the LRRK2, α-syn, and tau phosphatases may point to separate or cross-talking pathological pathways in PD. Finally, we will discuss how the study of phosphatases of dominant Parkinsonism proteins opens perspectives for targeting pathological phosphorylation events.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven Leuven, Belgium
| | - Veerle Baekelandt
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven Leuven, Belgium
| |
Collapse
|
182
|
Taymans JM, Baekelandt V, Harvey K. Regulation and targeting of enzymes mediating Parkinson's disease pathogenesis: focus on Parkinson's disease kinases, GTPases, and ATPases. Front Mol Neurosci 2014; 7:71. [PMID: 25120428 PMCID: PMC4114254 DOI: 10.3389/fnmol.2014.00071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/09/2014] [Indexed: 12/25/2022] Open
Affiliation(s)
- Jean-Marc Taymans
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences KU Leuven, Leuven, Belgium
| | - Kirsten Harvey
- School of Pharmacy, University College London London, UK
| |
Collapse
|