scholarly journals Bidirectional modulation of Alzheimer phenotype by alpha-synuclein in mice and primary neurons

2018 ◽  
Vol 136 (4) ◽  
pp. 589-605 ◽  
Author(s):  
Shahzad S. Khan ◽  
Michael LaCroix ◽  
Gabriel Boyle ◽  
Mathew A. Sherman ◽  
Jennifer L. Brown ◽  
...  
2021 ◽  
Author(s):  
Éva M. Szegõ ◽  
Eva M. Szegö ◽  
Chris Van den Haute ◽  
Lennart Höfs ◽  
Veerle Baekelandt ◽  
...  

Abstract BackgroundDuring the pathogenesis of Parkinson’s disease (PD), aggregation of alpha-synuclein (αSyn) induces a vicious cycle of cellular impairments that lead to neurodegeneration. Consequently, removing toxic αSyn aggregates constitutes a plausible strategy against PD. In this work, we tested whether stimulating the autolysosomal degradation of αSyn aggregates through the Ras-related in brain 7 (Rab7) pathway can reverse αSyn-induced cellular impairment and prevent neurodegeneration in vivo.MethodsThe disease-related A53T mutant of αSyn was expressed in primary neurons and in dopaminergic neurons of the rat brain simultaneously with wild type (WT) Rab7 or its dominant-negative T22N mutant as a control. The cellular integrity was quantified by morphological and biochemical analyses.ResultsIn primary neurons, WT Rab7 rescued the αSyn -induced loss of neurons and neurites. Furthermore, Rab7 decreased the amount of reactive oxygen species and the amount of Triton X-100 insoluble αSyn. In rat brain, WT Rab7 reduced αSyn -induced loss of dopaminergic axon terminals in the striatum and the loss of dopaminergic dendrites in the substantia nigra pars reticulata. Further, WT Rab7 lowered αSyn pathology as quantified by phosphorylated αSyn staining. Finally, WT Rab7 attenuated αSyn-induced DNA damage in primary neurons and rat brain.ConclusionRab7 reduced αSyn-induced pathology, ameliorated αSyn-induced neuronal degeneration, oxidative stress and DNA damage. These findings indicate that Rab7 is able to disrupt the vicious cycle of cellular impairment, αSyn pathology and neurodegeneration present in PD. Stimulation of Rab7 and the autolysosomal degradation pathway could therefore constitute a beneficial strategy for PD.


2013 ◽  
Vol 120 (9) ◽  
pp. 1331-1343 ◽  
Author(s):  
Guangwei Liu ◽  
Peng Wang ◽  
Xin Li ◽  
Yaohua Li ◽  
Shengli Xu ◽  
...  

2018 ◽  
Vol 9 ◽  
Author(s):  
Leire Almandoz-Gil ◽  
Emma Persson ◽  
Veronica Lindström ◽  
Martin Ingelsson ◽  
Anna Erlandsson ◽  
...  

2018 ◽  
Author(s):  
Anne-Laure Mahul-Mellier ◽  
Firat Altay ◽  
Johannes Burtscher ◽  
Niran Maharjan ◽  
Nadine Ait Bouziad ◽  
...  

Although converging evidence point to alpha-synuclein (a-syn) aggregation and Lewy body (LB) formation as central events in Parkinson's disease (PD), the molecular mechanisms that regulate these processes and their role in disease pathogenesis remain poorly understood. Herein, we applied an integrative biochemical, structural and imaging approach to elucidate the sequence, molecular and cellular mechanisms that regulate LB formation in primary neurons. Our results establish that post-fibrillization C-terminal truncation mediated by calpains 1 and 2 and potentially other enzymes, plays critical roles in regulating a-syn seeding, fibrillization and orchestrates many of the events associated with LB formation and maturation. These findings combined with the abundance of a-syn truncated species in LBs and pathological a-syn aggregates have significant implications for ongoing efforts to develop therapeutic strategies based on targeting the C-terminus of a-syn or proteolytic processing of this region.


2021 ◽  
Author(s):  
Jiang Zhu ◽  
Sara Pittman ◽  
Dhruva Dhavale ◽  
Rachel French ◽  
Jessica N. Patterson ◽  
...  

Abstract Background: Neuronal uptake and subsequent spread of proteopathic seeds, such as αS (alpha-synuclein), tau, and TDP-43, contribute to neurodegeneration and disease progression. The cellular machinery necessary for this process is poorly understood. Methods: Cas9 expressing αS FRET biosensors were transduced with a whole-genome guide RNA (gRNA) library, seeded with αS fibrils, and flow-sorted. Candidate genes protective against αS seeding were identified following gRNA sequencing of FRET+ and FRET- cell populations. Secondary validation of the high probability candidate suppressor VCP, utilized VCP inhibitors or gene knockdown in αS biosensors and primary neurons. In vivo validation was performed in VCP disease mutation mice following intrastriatal injection of αS seeds. TDP-43 seeding was performed in primary neurons from control or VCP mutant mice.Results: We devised a genome-wide CRISPR-Cas9 screen to identify suppressors of αS seeding. This approach identified Valosin Containing Protein (VCP) as a suppressor of αS seeding. Dominant mutations in VCP cause multisystem proteinopathy (MSP) a phenotypically and pathologically variable neurodegeneraive disease characterized by myopathy, motor neuron disease and dementia with TDP-43, αS and tau inclusions. VCP inhibition or MSP disease mutations increased αS seeding in cells and primary cultured neurons. This was similar to treatment with the lysosomal damaging agent, LLoMe or knockdown of the endolysosomal damage response associated VCP cofactor, UBXD1. Intrastriatal injection of αS seeds into VCP disease mice demonstrated enhanced seeding efficiency as compared with controls. Finally, this phenomenon was not specific to αS since VCP disease mutant expression increased TDP-43 seeding in neurons.Conclusion: VCP surveillance of permeabilized late endosomes protects neurons against the proteopathic spread of pathogenic protein aggregates. The spread of distinct aggregate species may dictate the pleiotropic phenotypes and pathologies in VCP associated MSP.


2018 ◽  
Vol 111 ◽  
pp. 36-47 ◽  
Author(s):  
Emanuela Colla ◽  
Giulia Panattoni ◽  
Alessio Ricci ◽  
Caterina Rizzi ◽  
Lucia Rota ◽  
...  

2019 ◽  
Vol 116 ◽  
pp. 37-45 ◽  
Author(s):  
Zheng-Cun Wu ◽  
Jia-Hong Gao ◽  
Ting-Fu Du ◽  
Dong-Hong Tang ◽  
Nai-Hong Chen ◽  
...  

2019 ◽  
Author(s):  
Gregor Bieri ◽  
Michel Brahic ◽  
Luc Bousset ◽  
Julien Couthouis ◽  
Nicholas Kramer ◽  
...  

AbstractProgressive aggregation of the protein alpha-synuclein (α-syn) and loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) are key histopathological hallmarks of Parkinson’s disease (PD). Accruing evidence suggests that α-syn pathology can propagate through neuronal circuits in the brain, contributing to the progressive nature of the disease. Thus, it is therapeutically pertinent to identify modifiers of α-syn transmission and aggregation as potential targets to slow down disease progression. A growing number of genetic mutations and risk factors have been identified in studies of familial and sporadic forms of PD. However, how these genes affect α-syn aggregation and pathological transmission, and whether they can be targeted for therapeutic interventions, remains unclear. We performed a targeted genetic screen of risk genes associated with PD and parkinsonism for modifiers of α-syn aggregation, using an α-syn preformed-fibril (PFF) induction assay. We found that decreased expression of Lrrk2 and Gba modulated α-syn aggregation in mouse primary neurons. Conversely, α-syn aggregation increased in primary neurons from mice expressing the PD-linked LRRK2 G2019S mutation. In vivo, using LRRK2 G2019S transgenic mice, we observed acceleration of α-syn aggregation and degeneration of dopaminergic neurons in the SNpc, exacerbated degeneration-associated neuroinflammation and behavioral deficits. To validate our findings in a human context, we established a novel human α-syn transmission model using induced pluripotent stem cell (iPS)-derived neurons (iNs), where human α-syn PFFs triggered aggregation of endogenous α-syn in a time-dependent manner. In PD subject-derived iNs, the G2019S mutation enhanced α-syn aggregation, whereas loss of LRRK2 decreased aggregation. Collectively, these findings establish a strong interaction between the PD risk gene LRRK2 and α-syn transmission across mouse and human models. Since clinical trials of LRRK2 inhibitors in PD are currently underway, our findings raise the possibility that these may be effective in PD broadly, beyond cases caused by LRRK2 mutations.


Sign in / Sign up

Export Citation Format

Share Document