Abstract
Neurodegenerative disorders with alpha-synuclein (α-syn) accumulation (synucleinopathies) include Parkinson's disease, Parkinson's disease dementia, dementia with Lewy bodies and multiple system atrophy (MSA). Due to the involvement of toxic α-syn aggregates in the molecular origin of these disorders, developing effective therapies targeting α-syn is a priority as a disease-modifying alternative to current symptomatic treatments. Importantly, the clinical and pathological attributes of MSA make this disorder an excellent candidate as a synucleinopathy model for accelerated drug development. Recent therapeutic strategies targeting α-syn in in vivo and in vitro models of MSA, as well as in clinical trials, have been focused on the pathological mechanisms of α-syn synthesis, aggregation, clearance, and/or cell-to-cell propagation of its neurotoxic conformers. Here we summarize the most relevant approaches in this direction, with emphasis on their potential as general synucleinopathy modifiers, and enumerate research areas for potential improvement in MSA drug discovery.
Keywords: synucleinopathy, multiple system atrophy, alpha-synuclein, therapies, clinical trials
Introduction
In the US alone over 1.5 million people [1] are affected by Parkinson's disease (PD), PD dementia, dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). In all these neurodegenerative disorders the synaptic protein α-synuclein (α-syn) accumulates in the CNS and peripheral organs, and for that reason the term “synucleinopathies” is often used to refer to them [2, 3]. Despite the fact that synucleinopathies are nowadays the second leading cause of dementia and parkinsonism in the elder population, no significant strides have been made yet towards the development of effective disease-modifying therapies that significantly stop or delay the progression of the neurodegenerative pathology.
In synucleinopathies, α-syn aggregates can be observed within neurons in the form of Lewy bodies (LBs) or neuronal cytoplasmic inclusions (NCIs), and within glial cells as glial cytoplasmic inclusions (GCIs) [4-10]. Interestingly, α-syn may also be present in the amyloid plaques of Alzheimer's disease, where it co-aggregates with amyloid beta [11, 12]. Moreover, co-aggregation of α-syn with other proteins such as tau or TDP-43 has also been reported in synucleinopathies [13, 14]. These observations suggest a common neuropathological mechanism to most neurodegenerative disorders involving the abnormal aggregation and accumulation of a few toxic protein conformers. It is very likely that in the causes and consequences of this pathological protein aggregation, and in the ability of these toxic conformers to propagate within the CNS, lays the foundation of most age-dependent neurodegenerative disorders. Most importantly, targeting these mechanisms is a promising therapeutic alternative for the development of disease-modifying alternatives for those ailments.
MSA as a synucleinopathy model for the development of therapies targeting α-synuclein
While in recent years considerable effort has been devoted at understanding the pathogenesis of PD, less is known about MSA, which is a rapidly progressive and fatal neurodegenerative disease characterized by parkinsonism, dysautonomia [15, 16] and α-syn accumulation within oligodendroglial (GCIs) and neuronal cells (NCIs) [17, 18]. This accumulation is accompanied by neuroinflammation [19, 20], demyelination [21, 22] and neurodegeneration [23, 24]. Parkinsonian features reflecting striato-nigral neurodegeneration predominate in 80% of MSA patients in the United States (MSA-P subtype), while the major motor feature in 20% of patients is cerebellar ataxia due to olivo-pontocerebellar atrophy (MSA-C subtype) [25]. Clinically, the rapid progression, the lack of response to L-DOPA [26], and pathologically the extensive accumulation of α-syn within oligodendrocytes differentiates MSA from other synucleinopathies [27].
However, the mechanisms through which α-syn accumulates within oligodendroglial cells in MSA are not completely understood. One possibility is that α-syn is produced by oligodendroglial cells, which in turn over-express or fail to intrinsically clear α-syn; the other is that α-syn propagates from neurons to oligodendrocytes due to neurons over-expressing and/or displaying defects in the physiological mechanisms of α-syn clearance. While some studies suggest that low levels of α-syn mRNA might be detected in oligodendroglial cells [28], others have not been able to confirm these findings [29-31]. However, given the high levels and widespread distribution of α-syn aggregates in MSA, it is possible that both propagation and oligodendroglial α-syn expression are occurring simultaneously. Supporting the possibility of propagation, several studies have shown that α-syn aggregates can transmit from neuron to neuron [32], neuron to astroglial [33] and oligodendroglial cells [34], and oligodendroglial to astroglial cells [20], leading to neuronal dysfunction [35, 36], apoptosis [32] and neuroinflammation [20, 33]. Furthermore, recent studies have shown that injection of homogenates from MSA brains propagate α-syn pathology in a prion-like fashion in the murine brain [37, 38]. Despite the considerable clinical and pathological overlap between PD and MSA-P in early disease stages, MSA progresses more rapidly and is fatal, making it a good candidate for accelerated drug development. Importantly, the use of MSA as a model in the development of disease-modifying therapeutic strategies targeting common pathophysiological mechanisms for all synucleinopathies (such as α-syn aggregation, prion-like propagation and autophagy deficits) might reduce the number of negative clinical trials and redirect resources towards earlier development stages [39, 40].
Novel therapies targeting alpha-synuclein in MSA and related disorders
Despite sharing other neuropathological features, the accumulation of α-syn is the pathological characteristic that defines synucleinopathies as a group of disorders, and it is probably mechanistically involved in the origin of the disease. It follows that reducing the α-syn pathology would have important disease-modifying consequences for synucleinopathy patients, especially at early stages. Importantly, in MSA, α-syn accumulation is prominent in both neurons and glial cells (astrocytes and oligodendrocytes). While astroglial α-syn accumulation is normally observed in synucleinopathies, oligodendroglial α-syn accumulation is specific to MSA [41]. Although in this review we will focus on α-syn-targeted, disease-modifying approaches for MSA, other review articles have described α-syn-targeted approaches for other synucleinopathies [42-44] or other novel therapeutic approaches for MSA [45].
The molecular events leading to the neuropathological accumulation of α-syn may include increased expression and synthesis of the protein, increased aggregation, defective protein clearance, and cell-to-cell prion-like propagation (Figure 1). Although dysfunction of these molecular mechanisms has been observed in the brain of synucleinopathy patients, it is still unclear which one of these processes is the driving force at the origin of the disease. While it is widely accepted that most over-production of α-syn occurs in neurons, expression of the SNCA mRNA in oligodendrocytes has also been detected [28]. Abnormal aggregation and accumulation of α-syn due to defect in protein clearance mechanisms has been observed in primary oligodendrocytes [46]. Finally, intercellular propagation is believed to be responsible for the accumulation of α-syn in oligodendrocytes and other glial cells, and neuron-to-neuron α-syn propagation is probably concomitant although more difficult to quantify. Moreover, increased α-syn aggregation may lead to defective protein clearance and autophagy deficits [47] and, therefore, to increased release of α-syn to the extracellular environment and consequent spreading. The prion-like characteristics of α-syn aggregates may also lead to abnormal α-syn conformational changes and trigger its accumulation in acceptor cells [37, 38, 48, 49]. Therefore, it follows that α-syn-centric therapies should target one or more of these dysregulated mechanisms in order to effectively reduce α-syn pathology.
Figure 1. Therapies targeting α-syn in MSA.
The pathological behavior of α-syn in synucleinopathies has been associated with the dysregulation of its synthesis, aggregation, clearance and/or cell-to-cell propagation ability. The regulation of such mechanisms is interconnected, leading to a prion-like positive feedback loop and to the progression of the disease. Therefore, current disease-modifying therapeutic strategies targeting α-syn in MSA are primarily focused on reducing α-syn synthesis, aggregation and clearance, and/or blocking the spreading of toxic α-syn conformers.
1. Therapies targeting α-syn expression
An increased dosage of the α-syn gene (SNCA) can cause PD, as observed in patients with SNCA triplication [50]. Moreover, elevated expression levels of SNCA are found in affected regions of the PD brain, supporting the hypothesis that an increase in α-syn expression is associated with the development of sporadic PD [51]. This increased expression of α-syn observed in synucleinopathies could be therapeutically targeted by the delivery of antisense, small interfering RNA (siRNA) or short hairpin RNA (shRNA) constructs directed against the SNCA mRNA. In this sense, in animal and in vitro models of PD with SNCA overexpression, gene therapy with antisense constructs has been reported to have disease-modifying effects [52, 53].
It can be inferred that these therapies might be also applicable to the treatment of other synucleinopathies such as MSA. Although a genetic link between SNCA and MSA has not been found (e.g. SNCA multiplications do not increase the risk of suffering from MSA [54]), and some studies have reported that total SNCA mRNA levels are not significantly elevated in MSA brains [28], single nucleotide polymorphisms at the SNCA locus are significantly associated with an increased risk for MSA [55]. Moreover, the translational repression that could be achieved by the use of antisense sequences in MSA models may prevent the excessive synthesis of this protein. However, further research is still needed in this direction.
2. Therapies targeting α-syn aggregation
Aggregated forms of α-syn are more resistant to degradation than monomeric, non-toxic forms [56], which would explain the prominent α-syn accumulation observed in MSA brains despite the apparent lack of increased SNCA expression. Aggregation of α-syn on the form of dimers, oligomers, proto-fibrils and fibrils is believed to be an early event on the α-syn pathology [57]. Oligomers and proto-fibrils are reportedly the toxic species [58, 59], while fibrillar aggregates have been traditionally associated with reduced toxicity [41]. However, several reports have shown that fibrillar forms can also propagate through the brain and contribute to the neurodegenerative pathology [60-63]. In this sense, therapies targeting this molecular mechanism include anti-aggregation agents and small molecules that act as molecular chaperones. An example of those compounds is Anle138b, an α-syn aggregation inhibitor that strongly blocks oligomer accumulation, neuronal degeneration, and disease progression in animal models of PD [64]. Other small molecules designed to stabilize or reduce the formation of toxic α-syn aggregates are also being developed at the preclinical stage, such as the conformational stabilizer NPT200-11 (Neuropore Therapies).
Degenerative movement disorders such as MSA, PD, Huntington's disease and progressive supranuclear palsy, exhibit increased iron content in brain [65-67]. In the case of MSA, dysregulation of iron metabolism may be relevant to the origin of the pathology, as oligodendrocytes are the predominant iron-containing cells in the brain, and iron stimulates α-syn aggregation [68]. Based on those observations, the use of iron chelation agents has been explored as a potential treatment for MSA. A phase III clinical trial with epigallocatechin gallate (EGCG) as an α-syn anti-aggregation approach for MSA is currently ongoing (NCT02008721). EGCG, a polyphenolic flavonoid extracted from green tea leaves, inhibits the formation of toxic α-syn oligomers in vitro and is able to transform α-syn oligomers into non-toxic species [69]. There is also evidence for a neuroprotective effect of EGCG in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of PD [70]. EGCG acts an antioxidant and iron chelation agent, and this may in part explain its anti-aggregation properties. For these reasons, EGCG has also been evaluated in multiple sclerosis and Huntington's disease [71, 72], as well as in many non-neurological conditions.
3. Therapies targeting α-syn degradation and clearance
Recent evidence supports the notion that failure of protein clearance mechanisms (e.g., autophagy) might play a role in the process of α-syn aggregation [36, 73], release [73] and subsequent accumulation of α-syn pathological species in neurons and oligodendroglial cells in MSA and other synucleinopathies (Figure 2). Moreover, microglial pro-inflammatory activation leading to a M1 phenotype may result in a reduction in the ability of microglia to effectively clear out propagating, prion-like α-syn species, resulting in increased accumulation within oligodendrocytes and other α-syn acceptor cells [74]. Therefore, pharmacological stimulation of α-syn clearance could result in both reduced accumulation and impaired cell-to-cell propagation (Figure 2). Restoring clearance mechanisms in the α-syn donor cells (neurons, oligodendrocytes) would reduce its release to the extracellular medium and therefore its cell-to-cell spreading; and restoring clearance in acceptor cells (oligodendrocytes) would reduce α-syn accumulation after its internalization. Treatments targeting α-syn clearance include stimulation of autophagy mechanisms (e.g. nilotinib [75]), modulation of the phagocytic activity of microglia, and the use of extracellular degrading enzymes. Interestingly, some anti-inflammatory approaches are able to reduce oligodendroglial α-syn accumulation, suggesting that regulating the activation of microglia might have the double effect of reducing inflammation and stimulating α-syn clearance. We have recently reported a reduction in α-syn accumulation by anti-inflammatory agents such as certain antidepressants [20, 76] and immunomodulatory drugs [77] in transgenic (tg) mouse models of synucleinopathy.
Figure 2. Deficits in protein clearance mechanisms might increase α-syn propagation and accumulation in MSA.
In MSA, it is believed that oligodendrocytes accumulate α-syn after a process of propagation from neurons and/or other oligodendroglial cells. The inhibition of clearance mechanisms such as autophagy might increase α-syn propagation by reducing its intracellular degradation in neurons and oligodendrocytes, and therefore potentiating its release to the extracellular medium. Moreover, clearance deficits may also lead to the intracellular accumulation of propagating α-syn within oligodendrocytes. Finally, impairments in microglial phagocytic activity hinder the physiological clearance of extracellular α-syn and further potentiate cell-to-cell propagation and glial pro-inflammatory activation.
Some studies have suggested that myeloperoxidase (MPO) inhibition might represent a novel candidate treatment strategy against MSA-like neurodegeneration, acting through its anti-inflammatory and antioxidant properties. In the 3-nitropropionic acid (3-NP) model of MSA, early-start treatment with MPO inhibitors reduces motor impairment and rescues vulnerable neurons in striatum, substantia nigra pars compacta, and other brain areas. Interestingly, MPO inhibition has been associated with suppression of microglial activation and results in reduced intracellular aggregates of α-synuclein [78]. However, a more recent report has shown that delayed-start MPO inhibition fails to impact on motor impairments and neuronal loss in contrast to the disease-modifying efficacy of early-start therapy in the 3-NP mouse model of MSA [79]. Despite these conflicting results, a Phase II clinical trial to assess the safety and tolerability of the MPO inhibitor AZD3241 in MSA patients is currently ongoing (NCT02388295, AstraZeneca). A similar study recently completed in PD patients (NCT01603069) has shown promising results regarding safety and tolerability.
In our laboratory we have analyzed the effects of the immunomodulatory drug lenalidomide in a tg mouse model of MSA that expresses human α-syn under the control of the oligodendroglial myelin basic protein promoter (MBP-α-syn) (Figure 3). Non-tg and MBP-α-syn tg mice were treated with either vehicle or lenalidomide at 100 mg/kg for 4 weeks, as previously described [77]. Neuropathologically, we observed a reduction in the number of α-syn positive cells in striatum after lenalidomide treatment (Figure 3A, 3B), together with a normalization of the pro-inflammatory microglial phenotype as measured by Iba1 staining in the same brain region (Figure 3A, 3C). The mechanisms that might lead to the reduction in the number of α-syn positive oligodendrocytes in lenalidomide-treated animals are still under investigation; however, our preliminary results suggest that the pro-inflammatory activation of microglia in MBP-α-syn tg animals might impair microglial functioning, and the reduction in this microglial activation induced by lenalidomide could lead to an improvement in the phagocytic activity of these cells towards extracellular α-syn. We have previously observed a similar reduction in pro-inflammatory microglial activation by lenalidomide in a tg mouse model of PD [77]. Interestingly, administration of lenalidomide to non-tg animals may lead to a microgliosis phenotype in striatum that is not accompanied by other neuropathological or behavioral deficits (not shown), suggesting that the multi-target actions of immunomodulatory drugs and other anti-inflammatory compounds might account for their ability to modulate inflammation while simultaneously reducing α-syn accumulation.
Figure 3. Lenalidomide reduces α-syn accumulation and microgliosis in the striatum of MBP-α-syn tg mice.
Mice (n=2-5 per group) were treated via gavage with lenalidomide 100 mg/kg or vehicle (0.5% methocellulose) for 4 weeks, and immunohistochemistry performed as previously described [77]. A, α-synuclein and Iba1 immunostaining of the striatum of MBP1-α-syn tg mice treated with vehicle or lenalidomide. Scale bar, 100 μm. B, Cell counts of α-syn positive cells in striatum. C, Optical density quantification of Iba1 staining in striatum. Statistical comparisons were performed using one-way ANOVA with Tukey's post-hoc test. *, p<0.05; **, p<0.01; ***, p<0.001. All experiments described were approved by the animal subjects committee at the University of California, San Diego (UCSD), and were performed according to NIH guidelines for animal use.
4. Targeting α-syn cell-to-cell propagation
Recent studies have shown that injection of homogenates from MSA brains propagate α-syn pathology in a prion-like fashion in the murine brain [37, 38]. In MSA, neuronal cells (donors) release α-syn aggregates into the extracellular environment in clear vesicles [80] and exosomes [81] (Figure 2), and this extracellular α-syn may be taken up by other neurons, oligodendrocytes and astrocytes (acceptors) via endocytosis [82] (Figure 2). Now believed to be a central event at the origin of the neuropathology, cell-to-cell propagation of α-syn has recently become a prominent pharmacological target. The spreading of α-syn can be inhibited by compounds that recognize extracellular α-syn and stimulate its clearance, directly by degradation or indirectly through microglial mechanisms. These include the use of immunotherapy against α-syn (active and passive), and extracellular α-syn degrading enzymes such as neurosin [83-85], matrix metalloproteinases, and plasmin [86].
Active immunotherapy with small peptides mimicking abnormal α-syn conformations (AFFITOPEs) has yielded positive results in tg models of synucleinopathy [87, 88], leading to a Phase I clinical trial with the AFFITOPEs PD01A and PD03A in patients with early MSA (NCT02270489, Affiris AG). AFFITOPEs mimicking the C-terminus region of α-syn elicit an immune response specific to α-syn oligomers that leads to decreased accumulation of α-syn oligomers, reduced neurodegeneration, and improved motor and memory deficits in α-syn tg models of PD and DLB [88], and to reduced demyelination in a tg model of MSA [87]. Moreover, Phase I passive immunization approaches with the α-syn antibodies PRX002 (NCT02157714, Prothena Biosciences) and BIIB054 (NCT02459886, Biogen) are also currently ongoing. It has been shown that monoclonal antibodies recognizing an epitope in the C-terminus of α-syn are effective at clearing intracellular α-syn aggregates, inhibiting α-syn propagation, and preventing C-terminus cleavage of the protein in tg mouse models of PD [89-92]. Similar reports using antibodies against the N-terminus of α-syn show that passive immunotherapy is able to clear α-syn aggregates, reduce α-syn propagation and diminish motor deficits in diverse in vitro and in vivo models of PD [93, 94]. This evidence further supports the value of immunotherapy against α-syn as a disease-modifying option for MSA and related synucleinopathies. In this sense, recent efforts have been directed towards the development of therapeutic antibodies able to detect individual conformational species of α-syn [95-98]. These antibodies could be used to discriminate among protein conformers [99], potentially including MSA-specific α-syn aggregates [62], for the differential treatment of synucleinopathies or for diagnostic purposes.
Lastly, we have recently observed that a modified, systemically delivered neurosin genetically modified for increased half-life and containing a brain-targeting sequence for delivery into the CNS, is able to reduce α-syn levels in oligodendrocytes and the spreading of α-syn to glial cells in a mouse model of MSA [85]. A similar lentiviral construct is also able to induce neuropathological and behavioral improvements in a tg mouse model of PD [84]. Taken together, these results suggest that the use of gene therapy with brain-targeted α-syn degrading enzymes may warrant further investigation as therapy for MSA and other disorders with α-syn propagation.
Final remarks
In this review we provide a brief overview of the current therapeutic approaches targeting α-syn for the treatment of MSA as a model synucleinopathy. Given the clinical progression and the pathophysiological characteristics of MSA, there is an urgent need for effective disease-modifying therapies for this disorder with potential application to similar neurodegenerative diseases with α-syn accumulation. This is even more relevant in the case of approaches aimed at blocking the cell-to-cell propagation of α-syn, as it is a pathological feature especially relevant in MSA compared to other synucleinopathies. However, it is also important to consider that MSA is a rapidly progressing neurodegenerative disorder often misdiagnosed as PD, therefore identifying early biomarkers and developing accurate diagnostic methods for MSA is a critical step for the success of therapeutic approaches targeting early pathological events such as α-syn accumulation. In this sense, the development of alternative animal models for MSA that fully reproduce all the neuropathological features of MSA is a research priority [100]. Early diagnostic approaches for MSA that have been recently explored include, among others: cerebrospinal fluid biomarkers such as microRNA levels [101, 102], neurofilament light chain levels [103], or the combination of α-syn and the ratio of phospho-tau to total tau [104]; differential pharmacodynamics of subacute levodopa [105]; identification of disease-specific α-syn conformations [62, 98, 99]; progressive thinning of the retinal nerve fiber layer and the macular ganglion cell complex [106]; etc. Moreover, combining the use of two or more biomarkers might be more successful than using single markers to increase sensitivity and specificity for the diagnosis of MSA [107]. Finally, elucidating the molecular mechanisms driving the oligodendroglial accumulation of α-syn in MSA would undoubtedly open the door for the identification of effective disease-modifying alternatives.
ACKNOWLEDGEMENTS
Supported by National Institutes of Health (NIH) grants AG18440, AG022074, NS044233 and MSA Coalition Research Grant.
Footnotes
Conflict of interest: The authors declare no conflict of interest.
REFERENCES
- 1.Savica R, Grossardt BR, Bower JH, Ahlskog JE, Rocca WA. Incidence and Pathology of Synucleinopathies and Tauopathies Related to Parkinsonism. JAMA neurology. 2013:1–7. doi: 10.1001/jamaneurol.2013.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Farrer M, Gwinn-Hardy K, Hutton M, Hardy J. The genetics of disorders with synuclein pathology and parkinsonism. Hum Mol Genet. 1999;8:1901–5. doi: 10.1093/hmg/8.10.1901. [DOI] [PubMed] [Google Scholar]
- 3.Galvin JE, Lee VM, Trojanowski JQ. Synucleinopathies: clinical and pathological implications. Archives of neurology. 2001;58:186–90. doi: 10.1001/archneur.58.2.186. [DOI] [PubMed] [Google Scholar]
- 4.Goedert M, Jakes R, Anthony Crowther R, Grazia Spillantini M. Parkinson's Disease, Dementia with Lewy Bodies, and Multiple System Atrophy as alpha-Synucleinopathies. Methods Mol Med. 2001;62:33–59. doi: 10.1385/1-59259-142-6:33. [DOI] [PubMed] [Google Scholar]
- 5.Spillantini MG. Parkinson's disease, dementia with Lewy bodies and multiple system atrophy are alpha-synucleinopathies. Parkinsonism Relat Disord. 1999;5:157–62. doi: 10.1016/s1353-8020(99)00031-0. [DOI] [PubMed] [Google Scholar]
- 6.Takeda A, Mallory M, Sundsmo M, Honer W, Hansen L, Masliah E. Abnormal accumulation of NACP/alpha-synuclein in neurodegenerative disorders. The American journal of pathology. 1998;152:367–72. [PMC free article] [PubMed] [Google Scholar]
- 7.Wakabayashi K, Hayashi S, Kakita A, Yamada M, Toyoshima Y, Yoshimoto M, Takahashi H. Accumulation of alpha-synuclein/NACP is a cytopathological feature common to Lewy body disease and multiple system atrophy. Acta neuropathologica. 1998;96:445–52. doi: 10.1007/s004010050918. [DOI] [PubMed] [Google Scholar]
- 8.Wakabayashi K, Yoshimoto M, Tsuji S, Takahashi H. Alpha-synuclein immunoreactivity in glial cytoplasmic inclusions in multiple system atrophy. Neurosci Lett. 1998;249:180–2. doi: 10.1016/s0304-3940(98)00407-8. [DOI] [PubMed] [Google Scholar]
- 9.Wakabayashi K, Matsumoto K, Takayama K, Yoshimoto M, Takahashi H. NACP, a presynaptic protein, immunoreactivity in Lewy bodies in Parkinson's disease. Neurosci Lett. 1997;239:45–8. doi: 10.1016/s0304-3940(97)00891-4. [DOI] [PubMed] [Google Scholar]
- 10.Papp MI, Kahn JE, Lantos PL. Glial cytoplasmic inclusions in the CNS of patients with multiple system atrophy (striatonigral degeneration, olivopontocerebellar atrophy and Shy-Drager syndrome). J Neurol Sci. 1989;94:79–100. doi: 10.1016/0022-510x(89)90219-0. [DOI] [PubMed] [Google Scholar]
- 11.Masliah E, Rockenstein E, Veinbergs I, Sagara Y, Mallory M, Hashimoto M, Mucke L. beta-amyloid peptides enhance alpha-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer's disease and Parkinson's disease. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:12245–50. doi: 10.1073/pnas.211412398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Winslow AR, Moussaud S, Zhu L, Post KL, Dickson DW, Berezovska O, McLean PJ. Convergence of pathology in dementia with Lewy bodies and Alzheimer's disease: a role for the novel interaction of alpha-synuclein and presenilin 1 in disease. Brain : a journal of neurology. 2014;137:1958–70. doi: 10.1093/brain/awu119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ishizawa T, Mattila P, Davies P, Wang D, Dickson DW. Colocalization of tau and alpha-synuclein epitopes in Lewy bodies. J Neuropathol Exp Neurol. 2003;62:389–97. doi: 10.1093/jnen/62.4.389. [DOI] [PubMed] [Google Scholar]
- 14.Arai T, Mackenzie IR, Hasegawa M, Nonoka T, Niizato K, Tsuchiya K, Iritani S, Onaya M, Akiyama H. Phosphorylated TDP-43 in Alzheimer's disease and dementia with Lewy bodies. Acta neuropathologica. 2009;117:125–36. doi: 10.1007/s00401-008-0480-1. [DOI] [PubMed] [Google Scholar]
- 15.Dickson DW, Lin W, Liu WK, Yen SH. Multiple system atrophy: a sporadic synucleinopathy. Brain Pathol. 1999;9:721–32. doi: 10.1111/j.1750-3639.1999.tb00553.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wenning GK, Seppi K, Scherfler C, Stefanova N, Puschban Z. Multiple system atrophy. Semin Neurol. 2001;21:33–40. doi: 10.1055/s-2001-13117. [DOI] [PubMed] [Google Scholar]
- 17.Lantos PL, Papp MI. Cellular pathology of multiple system atrophy: a review. J Neurol Neurosurg Psychiatry. 1994;57:129–33. doi: 10.1136/jnnp.57.2.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Jellinger KA. Neuropathology and pathophysiology of multiple system atrophy. Neuropathol Appl Neurobiol. 2012;38:379–80. doi: 10.1111/j.1365-2990.2012.01268.x. author reply 81. [DOI] [PubMed] [Google Scholar]
- 19.Stefanova N, Reindl M, Neumann M, Kahle PJ, Poewe W, Wenning GK. Microglial activation mediates neurodegeneration related to oligodendroglial alpha-synucleinopathy: implications for multiple system atrophy. Mov Disord. 2007;22:2196–203. doi: 10.1002/mds.21671. [DOI] [PubMed] [Google Scholar]
- 20.Valera E, Ubhi K, Mante M, Rockenstein E, Masliah E. Antidepressants reduce neuroinflammatory responses and astroglial alpha-synuclein accumulation in a transgenic mouse model of multiple system atrophy. Glia. 2014;62:317–37. doi: 10.1002/glia.22610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Matsuo A, Akiguchi I, Lee GC, McGeer EG, McGeer PL, Kimura J. Myelin degeneration in multiple system atrophy detected by unique antibodies. The American journal of pathology. 1998;153:735–44. doi: 10.1016/S0002-9440(10)65617-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wenning GK, Stefanova N, Jellinger KA, Poewe W, Schlossmacher MG. Multiple system atrophy: a primary oligodendrogliopathy. Ann Neurol. 2008;64:239–46. doi: 10.1002/ana.21465. [DOI] [PubMed] [Google Scholar]
- 23.Jellinger KA. Neuropathological spectrum of synucleinopathies. Mov Disord. 2003;18(Suppl 6):S2–12. doi: 10.1002/mds.10557. [DOI] [PubMed] [Google Scholar]
- 24.Ubhi K, Low P, Masliah E. Multiple system atrophy: a clinical and neuropathological perspective. Trends Neurosci. 2011;34:581–90. doi: 10.1016/j.tins.2011.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gilman S, Wenning GK, Low PA, Brooks DJ, Mathias CJ, Trojanowski JQ, Wood NW, Colosimo C, Dürr A, Fowler CJ, Kaufmann H, Klockgether T, Lees A, Poewe W, Quinn N, Revesz T, Robertson D, Sandroni P, Seppi K, Vidailhet M. Second consensus statement on the diagnosis of multiple system atrophy. Neurology. 2008;71:670–6. doi: 10.1212/01.wnl.0000324625.00404.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wenning GK, Ben Shlomo Y, Magalhaes M, Daniel SE, Quinn NP. Clinical features and natural history of multiple system atrophy. An analysis of 100 cases. Brain : a journal of neurology. 1994;117(Pt 4):835–45. doi: 10.1093/brain/117.4.835. [DOI] [PubMed] [Google Scholar]
- 27.Dickson DW, Liu W, Hardy J, Farrer M, Mehta N, Uitti R, Mark M, Zimmerman T, Golbe L, Sage J, Sima A, D'Amato C, Albin R, Gilman S, Yen SH. Widespread alterations of alpha-synuclein in multiple system atrophy. The American journal of pathology. 1999;155:1241–51. doi: 10.1016/s0002-9440(10)65226-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Asi YT, Simpson JE, Heath PR, Wharton SB, Lees AJ, Revesz T, Houlden H, Holton JL. Alpha-synuclein mRNA expression in oligodendrocytes in MSA. Glia. 2014;62:964–70. doi: 10.1002/glia.22653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ozawa T, Okuizumi K, Ikeuchi T, Wakabayashi K, Takahashi H, Tsuji S. Analysis of the expression level of alpha-synuclein mRNA using postmortem brain samples from pathologically confirmed cases of multiple system atrophy. Acta neuropathologica. 2001;102:188–90. doi: 10.1007/s004010100367. [DOI] [PubMed] [Google Scholar]
- 30.Miller DW, Johnson JM, Solano SM, Hollingsworth ZR, Standaert DG, Young AB. Absence of alpha-synuclein mRNA expression in normal and multiple system atrophy oligodendroglia. Journal of neural transmission. 2005;112:1613–24. doi: 10.1007/s00702-005-0378-1. [DOI] [PubMed] [Google Scholar]
- 31.Jin H, Ishikawa K, Tsunemi T, Ishiguro T, Amino T, Mizusawa H. Analyses of copy number and mRNA expression level of the alpha-synuclein gene in multiple system atrophy. Journal of medical and dental sciences. 2008;55:145–53. [PubMed] [Google Scholar]
- 32.Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, Spencer B, Masliah E, Lee SJ. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci USA. 2009;106:13010–5. doi: 10.1073/pnas.0903691106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lee HJ, Suk JE, Patrick C, Bae EJ, Cho JH, Rho S, Hwang D, Masliah E, Lee SJ. Direct transfer of alpha-synuclein from neuron to astroglia causes inflammatory responses in synucleinopathies. J Biol Chem. 2010;285:9262–72. doi: 10.1074/jbc.M109.081125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Reyes JF, Rey NL, Bousset L, Melki R, Brundin P, Angot E. Alpha-synuclein transfers from neurons to oligodendrocytes. Glia. 2014:387–98. doi: 10.1002/glia.22611. [DOI] [PubMed] [Google Scholar]
- 35.Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, Meaney DF, Trojanowski JQ, Lee VM. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. 2011;72:57–71. doi: 10.1016/j.neuron.2011.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Klucken J, Poehler AM, Ebrahimi-Fakhari D, Schneider J, Nuber S, Rockenstein E, Schlotzer-Schrehardt U, Hyman BT, McLean PJ, Masliah E, Winkler J. Alpha-synuclein aggregation involves a bafilomycin A 1-sensitive autophagy pathway. Autophagy. 2012;8:754–66. doi: 10.4161/auto.19371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Watts JC, Giles K, Oehler A, Middleton L, Dexter DT, Gentleman SM, DeArmond SJ, Prusiner SB. Transmission of multiple system atrophy prions to transgenic mice. Proc Natl Acad Sci USA. 2013:19555–60. doi: 10.1073/pnas.1318268110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Prusiner SB, Woerman AL, Mordes DA, Watts JC, Rampersaud R, Berry DB, Patel S, Oehler A, Lowe JK, Kravitz SN, Geschwind DH, Glidden DV, Halliday GM, Middleton LT, Gentleman SM, Grinberg LT, Giles K. Evidence for alpha-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E5308–17. doi: 10.1073/pnas.1514475112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fernagut PO, Dehay B, Maillard A, Bezard E, Perez P, Pavy-Le Traon A, Rascol O, Foubert-Samier A, Tison F, Meissner WG. Multiple system atrophy: a prototypical synucleinopathy for disease-modifying therapeutic strategies. Neurobiology of disease. 2014;67:133–9. doi: 10.1016/j.nbd.2014.03.021. [DOI] [PubMed] [Google Scholar]
- 40.Krismer F, Jellinger KA, Scholz SW, Seppi K, Stefanova N, Antonini A, Poewe W, Wenning GK. Multiple system atrophy as emerging template for accelerated drug discovery in alpha-synucleinopathies. Parkinsonism Relat Disord. 2014;20:793–9. doi: 10.1016/j.parkreldis.2014.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lashuel HA, Overk CR, Oueslati A, Masliah E. The many faces of α-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci. 2013;14:38–48. doi: 10.1038/nrn3406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Dehay B, Bourdenx M, Gorry P, Przedborski S, Vila M, Hunot S, Singleton A, Olanow CW, Merchant KM, Bezard E, Petsko GA, Meissner WG. Targeting alpha-synuclein for treatment of Parkinson's disease: mechanistic and therapeutic considerations. Lancet Neurol. 2015;14:855–66. doi: 10.1016/S1474-4422(15)00006-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Valera E, Spencer B, Masliah E. Immunotherapeutic Approaches Targeting Amyloid-beta, alpha-Synuclein, and Tau for the Treatment of Neurodegenerative Disorders. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics. 2015 doi: 10.1007/s13311-015-0397-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lindstrom V, Ihse E, Fagerqvist T, Bergstrom J, Nordstrom E, Moller C, Lannfelt L, Ingelsson M. Immunotherapy targeting alpha-synuclein, with relevance for future treatment of Parkinson's disease and other Lewy body disorders. Immunotherapy. 2014;6:141–53. doi: 10.2217/imt.13.162. [DOI] [PubMed] [Google Scholar]
- 45.Poewe W, Mahlknecht P, Krismer F. Therapeutic advances in multiple system atrophy and progressive supranuclear palsy. Mov Disord. 2015;30:1528–38. doi: 10.1002/mds.26334. [DOI] [PubMed] [Google Scholar]
- 46.Schwarz L, Goldbaum O, Bergmann M, Probst-Cousin S, Richter-Landsberg C. Involvement of macroautophagy in multiple system atrophy and protein aggregate formation in oligodendrocytes. Journal of molecular neuroscience : MN. 2012;47:256–66. doi: 10.1007/s12031-012-9733-5. [DOI] [PubMed] [Google Scholar]
- 47.Winslow AR, Rubinsztein DC. The Parkinson disease protein α-synuclein inhibits autophagy. Autophagy. 2011;7:429–31. doi: 10.4161/auto.7.4.14393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Olanow CW, Brundin P. Parkinson's disease and alpha synuclein: is Parkinson's disease a prion-like disorder? Mov Disord. 2013;28:31–40. doi: 10.1002/mds.25373. [DOI] [PubMed] [Google Scholar]
- 49.Masliah E, Rockenstein E, Inglis C, Adame A, Bett C, Lucero M, Sigurdson CJ. Prion infection promotes extensive accumulation of alpha-synuclein in aged human alpha-synuclein transgenic mice. Prion. 2012;6:184–90. doi: 10.4161/pri.19806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nussbaum R, Lincoln S, Crawley A, Hanson M, Maraganore D, Adler C, Cookson MR, Muenter M, Baptista M, Miller D, Blancato J, Hardy J, Gwinn-Hardy K. alpha-Synuclein locus triplication causes Parkinson's disease. Science. 2003;302:841. doi: 10.1126/science.1090278. [DOI] [PubMed] [Google Scholar]
- 51.Chiba-Falek O, Lopez GJ, Nussbaum RL. Levels of alpha-synuclein mRNA in sporadic Parkinson disease patients. Mov Disord. 2006;21:1703–8. doi: 10.1002/mds.21007. [DOI] [PubMed] [Google Scholar]
- 52.Takahashi M, Suzuki M, Fukuoka M, Fujikake N, Watanabe S, Murata M, Wada K, Nagai Y, Hohjoh H. Normalization of Overexpressed alpha-Synuclein Causing Parkinson's Disease By a Moderate Gene Silencing With RNA Interference. Molecular therapy Nucleic acids. 2015;4:e241. doi: 10.1038/mtna.2015.14. [DOI] [PubMed] [Google Scholar]
- 53.Zharikov AD, Cannon JR, Tapias V, Bai Q, Horowitz MP, Shah V, El Ayadi A, Hastings TG, Greenamyre JT, Burton EA. shRNA targeting alpha-synuclein prevents neurodegeneration in a Parkinson's disease model. The Journal of clinical investigation. 2015;125:2721–35. doi: 10.1172/JCI64502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lincoln SJ, Ross OA, Milkovic NM, Dickson DW, Rajput A, Robinson CA, Papapetropoulos S, Mash DC, Farrer MJ. Quantitative PCR-based screening of alpha-synuclein multiplication in multiple system atrophy. Parkinsonism Relat Disord. 2007;13:340–2. doi: 10.1016/j.parkreldis.2006.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Scholz SW, Houlden H, Schulte C, Sharma M, Li A, Berg D, Melchers A, Paudel R, Gibbs JR, Simon-Sanchez J, Paisan-Ruiz C, Bras J, Ding J, Chen H, Traynor BJ, Arepalli S, Zonozi RR, Revesz T, Holton J, Wood N, Lees A, Oertel W, Wullner U, Goldwurm S, Pellecchia MT, Illig T, Riess O, Fernandez HH, Rodriguez RL, Okun MS, Poewe W, Wenning GK, Hardy JA, Singleton AB, Del Sorbo F, Schneider S, Bhatia KP, Gasser T. SNCA variants are associated with increased risk for multiple system atrophy. Ann Neurol. 2009;65:610–4. doi: 10.1002/ana.21685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Tanik SA, Schultheiss CE, Volpicelli-Daley LA, Brunden KR, Lee VM. Lewy body-like alpha-synuclein aggregates resist degradation and impair macroautophagy. J Biol Chem. 2013;288:15194–210. doi: 10.1074/jbc.M113.457408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging. 2003;24:197–211. doi: 10.1016/s0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
- 58.Winner B, Jappelli R, Maji SK, Desplats PA, Boyer L, Aigner S, Hetzer C, Loher T, Vilar M, Campioni S, Tzitzilonis C, Soragni A, Jessberger S, Mira H, Consiglio A, Pham E, Masliah E, Gage FH, Riek R. In vivo demonstration that alpha-synuclein oligomers are toxic. Proc Natl Acad Sci USA. 2011;108:4194–9. doi: 10.1073/pnas.1100976108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Volles MJ, Lee SJ, Rochet JC, Shtilerman MD, Ding TT, Kessler JC, Lansbury PT., Jr Vesicle permeabilization by protofibrillar alpha-synuclein: implications for the pathogenesis and treatment of Parkinson's disease. Biochemistry. 2001;40:7812–9. doi: 10.1021/bi0102398. [DOI] [PubMed] [Google Scholar]
- 60.El-Agnaf OM, Jakes R, Curran MD, Middleton D, Ingenito R, Bianchi E, Pessi A, Neill D, Wallace A. Aggregates from mutant and wild-type alpha-synuclein proteins and NAC peptide induce apoptotic cell death in human neuroblastoma cells by formation of beta-sheet and amyloid-like filaments. FEBS Lett. 1998;440:71–5. doi: 10.1016/s0014-5793(98)01418-5. [DOI] [PubMed] [Google Scholar]
- 61.Pieri L, Madiona K, Bousset L, Melki R. Fibrillar alpha-synuclein and huntingtin exon 1 assemblies are toxic to the cells. Biophysical journal. 2012;102:2894–905. doi: 10.1016/j.bpj.2012.04.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Peelaerts W, Bousset L, Van der Perren A, Moskalyuk A, Pulizzi R, Giugliano M, Van den Haute C, Melki R, Baekelandt V. alpha-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015;522:340–4. doi: 10.1038/nature14547. [DOI] [PubMed] [Google Scholar]
- 63.Masuda-Suzukake M, Nonaka T, Hosokawa M, Kubo M, Shimozawa A, Akiyama H, Hasegawa M. Pathological alpha-synuclein propagates through neural networks. Acta neuropathologica communications. 2014;2:88. doi: 10.1186/s40478-014-0088-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wagner J, Ryazanov S, Leonov A, Levin J, Shi S, Schmidt F, Prix C, Pan-Montojo F, Bertsch U, Mitteregger-Kretzschmar G, Geissen M, Eiden M, Leidel F, Hirschberger T, Deeg AA, Krauth JJ, Zinth W, Tavan P, Pilger J, Zweckstetter M, Frank T, Bahr M, Weishaupt JH, Uhr M, Urlaub H, Teichmann U, Samwer M, Botzel K, Groschup M, Kretzschmar H, Griesinger C, Giese A. Anle138b: a novel oligomer modulator for disease-modifying therapy of neurodegenerative diseases such as prion and Parkinson's disease. Acta neuropathologica. 2013;125:795–813. doi: 10.1007/s00401-013-1114-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Dusek P, Jankovic J, Le W. Iron dysregulation in movement disorders. Neurobiology of disease. 2012;46:1–18. doi: 10.1016/j.nbd.2011.12.054. [DOI] [PubMed] [Google Scholar]
- 66.Wang Y, Butros SR, Shuai X, Dai Y, Chen C, Liu M, Haacke EM, Hu J, Xu H. Different iron-deposition patterns of multiple system atrophy with predominant parkinsonism and idiopathetic Parkinson diseases demonstrated by phase-corrected susceptibility-weighted imaging. AJNR Am J Neuroradiol. 2012;33:266–73. doi: 10.3174/ajnr.A2765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Visanji NP, Collingwood JF, Finnegan ME, Tandon A, House E, Hazrati L-N. Iron deficiency in parkinsonism: region-specific iron dysregulation in Parkinson's disease and multiple system atrophy. Journal of Parkinson's disease. 2013:523–37. doi: 10.3233/JPD-130197. [DOI] [PubMed] [Google Scholar]
- 68.Li WJ, Jiang H, Song N, Xie JX. Dose- and time-dependent alpha-synuclein aggregation induced by ferric iron in SK-N-SH cells. Neurosci Bull. 2010;26:205–10. doi: 10.1007/s12264-010-1117-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lorenzen N, Nielsen SB, Yoshimura Y, Vad BS, Andersen CB, Betzer C, Kaspersen JD, Christiansen G, Pedersen JS, Jensen PH, Mulder FA, Otzen DE. How epigallocatechin gallate can inhibit alpha-synuclein oligomer toxicity in vitro. J Biol Chem. 2014;289:21299–310. doi: 10.1074/jbc.M114.554667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kim JS, Kim JM, O JJ, Jeon BS. Inhibition of inducible nitric oxide synthase expression and cell death by (-)-epigallocatechin-3-gallate, a green tea catechin, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease. Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia. 2010;17:1165–8. doi: 10.1016/j.jocn.2010.01.042. [DOI] [PubMed] [Google Scholar]
- 71.Mahler A, Steiniger J, Bock M, Klug L, Parreidt N, Lorenz M, Zimmermann BF, Krannich A, Paul F, Boschmann M. Metabolic response to epigallocatechin-3-gallate in relapsing-remitting multiple sclerosis: a randomized clinical trial. The American journal of clinical nutrition. 2015;101:487–95. doi: 10.3945/ajcn.113.075309. [DOI] [PubMed] [Google Scholar]
- 72.Ehrnhoefer DE, Duennwald M, Markovic P, Wacker JL, Engemann S, Roark M, Legleiter J, Marsh JL, Thompson LM, Lindquist S, Muchowski PJ, Wanker EE. Green tea (-)-epigallocatechin-gallate modulates early events in huntingtin misfolding and reduces toxicity in Huntington's disease models. Hum Mol Genet. 2006;15:2743–51. doi: 10.1093/hmg/ddl210. [DOI] [PubMed] [Google Scholar]
- 73.Lee HJ, Cho ED, Lee KW, Kim JH, Cho SG, Lee SJ. Autophagic failure promotes the exocytosis and intercellular transfer of alpha-synuclein. Experimental & molecular medicine. 2013;45:e22. doi: 10.1038/emm.2013.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Tang Y, Le W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol Neurobiol. 2015 doi: 10.1007/s12035-014-9070-5. [DOI] [PubMed] [Google Scholar]
- 75.Hebron ML, Lonskaya I, Moussa CE. Nilotinib reverses loss of dopamine neurons and improves motor behavior via autophagic degradation of alpha-synuclein in Parkinson's disease models. Hum Mol Genet. 2013;22:3315–28. doi: 10.1093/hmg/ddt192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ubhi K, Inglis C, Mante M, Patrick C, Adame A, Spencer B, Rockenstein E, May V, Winkler J, Masliah E. Fluoxetine ameliorates behavioral and neuropathological deficits in a transgenic model mouse of α-synucleinopathy. Exp Neurol. 2012;234:405–16. doi: 10.1016/j.expneurol.2012.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Valera E, Mante M, Anderson S, Rockenstein E, Masliah E. Lenalidomide reduces microglial activation and behavioral deficits in a transgenic model of Parkinson's disease. J Neuroinflammation. 2015;12:93. doi: 10.1186/s12974-015-0320-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Stefanova N, Georgievska B, Eriksson H, Poewe W, Wenning GK. Myeloperoxidase inhibition ameliorates multiple system atrophy-like degeneration in a transgenic mouse model. Neurotox Res. 2012;21:393–404. doi: 10.1007/s12640-011-9294-3. [DOI] [PubMed] [Google Scholar]
- 79.Kaindlstorfer C, Sommer P, Georgievska B, Mather RJ, Kugler AR, Poewe W, Wenning GK, Stefanova N. Failure of Neuroprotection Despite Microglial Suppression by Delayed-Start Myeloperoxidase Inhibition in a Model of Advanced Multiple System Atrophy: Clinical Implications. Neurotox Res. 2015;28:185–94. doi: 10.1007/s12640-015-9547-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lee HJ, Patel S, Lee SJ. Intravesicular localization and exocytosis of alpha-synuclein and its aggregates. J Neurosci. 2005;25:6016–24. doi: 10.1523/JNEUROSCI.0692-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow AR, Zhu L, Vanderburg CR, McLean PJ. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener. 2012;7:42. doi: 10.1186/1750-1326-7-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lee HJ, Suk JE, Bae EJ, Lee JH, Paik SR, Lee SJ. Assembly-dependent endocytosis and clearance of extracellular alpha-synuclein. The international journal of biochemistry & cell biology. 2008;40:1835–49. doi: 10.1016/j.biocel.2008.01.017. [DOI] [PubMed] [Google Scholar]
- 83.Tatebe H, Watanabe Y, Kasai T, Mizuno T, Nakagawa M, Tanaka M, Tokuda T. Extracellular neurosin degrades alpha-synuclein in cultured cells. Neurosci Res. 2010;67:341–6. doi: 10.1016/j.neures.2010.04.008. [DOI] [PubMed] [Google Scholar]
- 84.Spencer B, Michael S, Shen J, Kosberg K, Rockenstein E, Patrick C, Adame A, Masliah E. Lentivirus mediated delivery of neurosin promotes clearance of wild-type alpha-synuclein and reduces the pathology in an alpha-synuclein model of LBD. Mol Ther. 2013;21:31–41. doi: 10.1038/mt.2012.66. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 85.Spencer B, Valera E, Rockenstein E, Trejo-Morales M, Adame A, Masliah E. A brain-targeted, modified neurosin (kallikrein-6) reduces alpha-synuclein accumulation in a mouse model of multiple system atrophy. Mol Neurodegener. 2015;10:48. doi: 10.1186/s13024-015-0043-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Park SM, Kim KS. Proteolytic clearance of extracellular α-synuclein as a new therapeutic approach against Parkinson disease. Prion. 2013:121–6. doi: 10.4161/pri.22850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mandler M, Valera E, Rockenstein E, Mante M, Weninger H, Patrick C, Adame A, Schmidhuber S, Santic R, Schneeberger A, Schmidt W, Mattner F, Masliah E. Active immunization against alpha-synuclein ameliorates the degenerative pathology and prevents demyelination in a model of multiple system atrophy. Mol Neurodegener. 2015;10:10. doi: 10.1186/s13024-015-0008-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Mandler M, Valera E, Rockenstein E, Weninger H, Patrick C, Adame A, Santic R, Meindl S, Vigl B, Smrzka O, Schneeberger A, Mattner F, Masliah E. Next-generation active immunization approach for synucleinopathies: implications for Parkinson's disease clinical trials. Acta neuropathologica. 2014;127:861–79. doi: 10.1007/s00401-014-1256-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Games D, Valera E, Spencer B, Rockenstein E, Mante M, Adame A, Patrick C, Ubhi K, Nuber S, Sacayon P, Zago W, Seubert P, Barbour R, Schenk D, Masliah E. Reducing C-terminal-truncated alpha-synuclein by immunotherapy attenuates neurodegeneration and propagation in Parkinson's disease-like models. J Neurosci. 2014;34:9441–54. doi: 10.1523/JNEUROSCI.5314-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Valera E, Masliah E. Immunotherapy for neurodegenerative diseases: Focus on α-synucleinopathies. Pharmacol Ther. 2013 doi: 10.1016/j.pharmthera.2013.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Masliah E, Rockenstein E, Mante M, Crews L, Spencer B, Adame A, Patrick C, Trejo M, Ubhi K, Rohn TT, Mueller-Steiner S, Seubert P, Barbour R, McConlogue L, Buttini M, Games D, Schenk D. Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease. PloS one. 2011;6:e19338. doi: 10.1371/journal.pone.0019338. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 92.Bae EJ, Lee HJ, Rockenstein E, Ho DH, Park EB, Yang NY, Desplats P, Masliah E, Lee SJ. Antibody-aided clearance of extracellular alpha-synuclein prevents cell-to-cell aggregate transmission. J Neurosci. 2012;32:13454–69. doi: 10.1523/JNEUROSCI.1292-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Tran HT, Chung CH, Iba M, Zhang B, Trojanowski JQ, Luk KC, Lee VM. Alpha-synuclein immunotherapy blocks uptake and templated propagation of misfolded alpha-synuclein and neurodegeneration. Cell reports. 2014;7:2054–65. doi: 10.1016/j.celrep.2014.05.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Shahaduzzaman M, Nash K, Hudson C, Sharif M, Grimmig B, Lin X, Bai G, Liu H, Ugen KE, Cao C, Bickford PC. Anti-human alpha-synuclein N-terminal peptide antibody protects against dopaminergic cell death and ameliorates behavioral deficits in an AAV-alpha-synuclein rat model of Parkinson's disease. PloS one. 2015;10:e0116841. doi: 10.1371/journal.pone.0116841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Spencer B, Emadi S, Desplats P, Eleuteri S, Michael S, Kosberg K, Shen J, Rockenstein E, Patrick C, Adame A, Gonzalez T, Sierks M, Masliah E. ESCRT-mediated uptake and degradation of brain-targeted alpha-synuclein single chain antibody attenuates neuronal degeneration in vivo. Mol Ther. 2014;22:1753–67. doi: 10.1038/mt.2014.129. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 96.Emadi S, Liu R, Yuan B, Schulz P, McAllister C, Lyubchenko Y, Messer A, Sierks MR. Inhibiting aggregation of alpha-synuclein with human single chain antibody fragments. Biochemistry. 2004;43:2871–8. doi: 10.1021/bi036281f. [DOI] [PubMed] [Google Scholar]
- 97.Emadi S, Barkhordarian H, Wang MS, Schulz P, Sierks MR. Isolation of a human single chain antibody fragment against oligomeric alpha-synuclein that inhibits aggregation and prevents alpha-synuclein-induced toxicity. J Mol Biol. 2007;368:1132–44. doi: 10.1016/j.jmb.2007.02.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Vaikath NN, Majbour NK, Paleologou KE, Ardah MT, van Dam E, van de Berg WD, Forrest SL, Parkkinen L, Gai WP, Hattori N, Takanashi M, Lee SJ, Mann DM, Imai Y, Halliday GM, Li JY, El-Agnaf OM. Generation and characterization of novel conformation-specific monoclonal antibodies for alpha-synuclein pathology. Neurobiology of disease. 2015;79:81–99. doi: 10.1016/j.nbd.2015.04.009. [DOI] [PubMed] [Google Scholar]
- 99.Bousset L, Pieri L, Ruiz-Arlandis G, Gath J, Jensen PH, Habenstein B, Madiona K, Olieric V, Bockmann A, Meier BH, Melki R. Structural and functional characterization of two alpha-synuclein strains. Nature communications. 2013;4:2575. doi: 10.1038/ncomms3575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Stefanova N, Tison F, Reindl M, Poewe W, Wenning GK. Animal models of multiple system atrophy. Trends Neurosci. 2005;28:501–6. doi: 10.1016/j.tins.2005.07.002. [DOI] [PubMed] [Google Scholar]
- 101.Vallelunga A, Ragusa M, Di Mauro S, Iannitti T, Pilleri M, Biundo R, Weis L, Di Pietro C, De Iuliis A, Nicoletti A, Zappia M, Purrello M, Antonini A. Identification of circulating microRNAs for the differential diagnosis of Parkinson's disease and Multiple System Atrophy. Frontiers in cellular neuroscience. 2014;8:156. doi: 10.3389/fncel.2014.00156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ubhi K, Rockenstein E, Kragh C, Inglis C, Spencer B, Michael S, Mante M, Adame A, Galasko D, Masliah E. Widespread microRNA dysregulation in multiple system atrophy - disease-related alteration in miR-96. The European journal of neuroscience. 2014;39:1026–41. doi: 10.1111/ejn.12444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Herbert MK, Aerts MB, Beenes M, Norgren N, Esselink RA, Bloem BR, Kuiperij HB, Verbeek MM. CSF Neurofilament Light Chain but not FLT3 Ligand Discriminates Parkinsonian Disorders. Frontiers in neurology. 2015;6:91. doi: 10.3389/fneur.2015.00091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Shi M, Bradner J, Hancock AM, Chung KA, Quinn JF, Peskind ER, Galasko D, Jankovic J, Zabetian CP, Kim HM, Leverenz JB, Montine TJ, Ginghina C, Kang UJ, Cain KC, Wang Y, Aasly J, Goldstein D, Zhang J. Cerebrospinal fluid biomarkers for Parkinson disease diagnosis and progression. Ann Neurol. 2011;69:570–80. doi: 10.1002/ana.22311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Calandra-Buonaura G, Doria A, Lopane G, Guaraldi P, Capellari S, Martinelli P, Cortelli P, Contin M. Pharmacodynamics of a low subacute levodopa dose helps distinguish between multiple system atrophy with predominant Parkinsonism and Parkinson's disease. J Neurol. 2015 doi: 10.1007/s00415-015-7961-7. [DOI] [PubMed] [Google Scholar]
- 106.Mendoza-Santiesteban CE, Palma JA, Martinez J, Norcliffe-Kaufmann L, Hedges TR, 3rd, Kaufmann H. Progressive retinal structure abnormalities in multiple system atrophy. Mov Disord. 2015;30:1944–53. doi: 10.1002/mds.26360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Laurens B, Constantinescu R, Freeman R, Gerhard A, Jellinger K, Jeromin A, Krismer F, Mollenhauer B, Schlossmacher MG, Shaw LM, Verbeek MM, Wenning GK, Winge K, Zhang J, Meissner WG. Fluid biomarkers in multiple system atrophy: A review of the MSA Biomarker Initiative. Neurobiology of disease. 2015;80:29–41. doi: 10.1016/j.nbd.2015.05.004. [DOI] [PubMed] [Google Scholar]