1
|
Ramalingam M, Jang S, Hwang J, Cho HH, Kim BC, Jeong HS. Neural-induced human adipose tissue-derived stem cell secretome exerts neuroprotection against rotenone-induced Parkinson's disease in rats. Stem Cell Res Ther 2025; 16:193. [PMID: 40254594 PMCID: PMC12010609 DOI: 10.1186/s13287-025-04306-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 04/01/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a multifactorial disease that involves genetic and environmental factors, which play an essential role in the pathogenesis of PD. Mesenchymal stem cells release a set of bioactive molecules called "secretome" that regulates intercellular communication and cargo transfer in signaling pathways for PD treatment. Thus, this study aimed to evaluate the neuroprotective effects of neural-induced human adipose tissue-derived stem cell (NI-hADSC)-conditioned medium (NI-hADSC-CM) and its exosomes (NI-hADSC-Exo) in a rotenone (ROT)-induced model of PD in rats. METHODS The NI-hADSC-CM was collected from NI-hADSC after 14 days of neural differentiation, and its NI-hADSC-Exo were isolated using a tangential flow filtration system. ROT (1 mg/kg) was subcutaneously administered for 28 days to establish a model of PD in rats. The treatment of NI-hADSC-CM or NI-hADSC-Exo was intravenously injected on days 15, 18, 21, 24, and 27. Animal behavioral effects were explored via a rotarod test. After 28 days, histological and western blot analyses were performed to investigate the tyrosine hydroxylase (TH), α-synuclein (α-syn) aggregation, and downstream signaling pathways for experimental validation. RESULTS NI-hADSC-Exo improved the motor balance and coordination skills against ROT toxicity. ROT reproduced the pathological features of PD, such as a decrease in TH-positive dopaminergic neurons and an increase in α-syn aggregation and glial fibrillary acidic protein (GFAP)-positive cells. NI-hADSC-CM and NI-hADSC-Exo improved the TH expression, decreased the Triton X-100 soluble and insoluble oligomeric p-S129 α-syn, and influenced the differential reactivity to astrocytes and microglia. Secretome treatment could reverse the ROT-induced damages in the neuronal structural and functional proteins, mitochondrial apoptosis, and caspase cascade. The treatment of NI-hADSC-CM and NI-hADSC-Exo ameliorated the ROT toxicity-induced serine-threonine protein kinase dysregulation and autophagy impairment to clear the aggregated α-syn. CONCLUSIONS NI-hADSC-CM and NI-hADSC-Exo significantly exerted neuroprotection by decreasing α-syn toxicity, inhibiting neuroinflammation and apoptosis, restoring autophagic flux properties, and promoting the neuronal function in ROT-injected rats; however, the influence of these treatments on signaling pathways differed slightly between the midbrain and striatum regions. Targeting α-syn degradation pathways provides a novel strategy to elucidate the beneficial effects of MSC secretome and future safe cell-free treatments for PD.
Collapse
Affiliation(s)
- Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| |
Collapse
|
2
|
Mazzetti S, Calogero AM, Pezzoli G, Cappelletti G. Cross-talk between α-synuclein and the microtubule cytoskeleton in neurodegeneration. Exp Neurol 2023; 359:114251. [PMID: 36243059 DOI: 10.1016/j.expneurol.2022.114251] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 12/30/2022]
Abstract
Looking at the puzzle that depicts the molecular determinants in neurodegeneration, many pieces are lacking and multiple interconnections among key proteins and intracellular pathways still remain unclear. Here we focus on the concerted action of α-synuclein and the microtubule cytoskeleton, whose interplay, indeed, is emerging but remains largely unexplored in both its physiology and pathology. α-Synuclein is a key protein involved in neurodegeneration, underlying those diseases termed synucleinopathies. Its propensity to interact with other proteins and structures renders the identification of neuronal death trigger extremely difficult. Conversely, the unbalance of microtubule cytoskeleton in terms of structure, dynamics and function is emerging as a point of convergence in neurodegeneration. Interestingly, α-synuclein and microtubules have been shown to interact and mediate cross-talks with other intracellular structures. This is supported by an increasing amount of evidence ranging from their direct interaction to the engagement of in-common partners and culminating with their respective impact on microtubule-dependent neuronal functions. Last, but not least, it is becoming even more clear that α-synuclein and tubulin work synergically towards pathological aggregation, ultimately resulting in neurodegeneration. In this respect, we supply a novel perspective towards the understanding of α-synuclein biology and, most importantly, of the link between α-synuclein with microtubule cytoskeleton and its impact for neurodegeneration and future development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy; Fondazione Grigioni per il Morbo di Parkinson, Milan, Italy
| | | | - Gianni Pezzoli
- Fondazione Grigioni per il Morbo di Parkinson, Milan, Italy
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy; Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
3
|
Przewodowska D, Marzec W, Madetko N. Novel Therapies for Parkinsonian Syndromes-Recent Progress and Future Perspectives. Front Mol Neurosci 2021; 14:720220. [PMID: 34512258 PMCID: PMC8427499 DOI: 10.3389/fnmol.2021.720220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/23/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Atypical parkinsonian syndromes are rare, fatal neurodegenerative diseases associated with abnormal protein accumulation in the brain. Examples of these syndromes include progressive supranuclear palsy, multiple system atrophy, and corticobasal degeneration. A common clinical feature in parkinsonism is a limited improvement with levodopa. So far, there are no disease-modifying treatments to address these conditions, and therapy is only limited to the alleviation of symptoms. Diagnosis is devastating for patients, as prognosis is extremely poor, and the disease tends to progress rapidly. Currently, potential causes and neuropathological mechanisms involved in these diseases are being widely investigated. Objectives: The goal of this review is to summarize recent advances and gather emerging disease-modifying therapies that could slow the progression of atypical parkinsonian syndromes. Methods: PubMed and Google Scholar databases were searched regarding novel perspectives for atypical parkinsonism treatment. The following medical subject headings were used: "atypical parkinsonian syndromes-therapy," "treatment of atypical parkinsonian syndromes," "atypical parkinsonian syndromes-clinical trial," "therapy of tauopathy," "alpha-synucleinopathy treatment," "PSP therapy/treatment," "CBD therapy/treatment," "MSA therapy/treatment," and "atypical parkinsonian syndromes-disease modifying." All search results were manually reviewed prior to inclusion in this review. Results: Neuroinflammation, mitochondrial dysfunction, microglia activation, proteasomal impairment, and oxidative stress play a role in the neurodegenerative process. Ongoing studies and clinical trials target these components in order to suppress toxic protein accumulation. Various approaches such as stem cell therapy, anti-aggregation/anti-phosphorylation agent administration, or usage of active and passive immunization appear to have promising results. Conclusion: Presently, disease-modifying strategies for atypical parkinsonian syndromes are being actively explored, with encouraging preliminary results. This leads to an assumption that developing accurate, safe, and progression-halting treatment is not far off. Nevertheless, the further investigation remains necessary.
Collapse
Affiliation(s)
- Dominika Przewodowska
- Students' Scientific Association of the Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Weronika Marzec
- Students' Scientific Association of the Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Natalia Madetko
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
4
|
Ghosh A, Singh S. Regulation Of Microtubule: Current Concepts And Relevance To Neurodegenerative Diseases. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:656-679. [PMID: 34323203 DOI: 10.2174/1871527320666210728144043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Accepted: 02/23/2021] [Indexed: 11/22/2022]
Abstract
Neurodevelopmental disorders (NDDs) are abnormalities linked to neuronal structure and irregularities associated with the proliferation of cells, transportation, and differentiation. NDD also involves synaptic circuitry and neural network alterations known as synaptopathies. Microtubules (MTs) and MTs-associated proteins help to maintain neuronal health as well as their development. The microtubular dynamic structure plays a crucial role in the division of cells and forms mitotic spindles, thus take part in initiating stages of differentiation and polarization for various types of cells. The MTs also take part in the cellular death but MT-based cellular degenerations are not yet well excavated. In the last few years, studies have provided the protagonist activity of MTs in neuronal degeneration. In this review, we largely engrossed our discussion on the change of MT cytoskeleton structure, describing their organization, dynamics, transportation, and their failure causing NDDs. At end of this review, we are targeting the therapeutic neuroprotective strategies on clinical priority and also try to discuss the clues for the development of new MT-based therapy as a new pharmacological intervention. This will be a new potential site to block not only neurodegeneration but also promotes the regeneration of neurons.
Collapse
Affiliation(s)
- Anirban Ghosh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| | - Shamsher Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| |
Collapse
|
5
|
Ramalingam M, Jang S, Jeong HS. Neural-Induced Human Adipose Tissue-Derived Stem Cells Conditioned Medium Ameliorates Rotenone-Induced Toxicity in SH-SY5Y Cells. Int J Mol Sci 2021; 22:2322. [PMID: 33652595 PMCID: PMC7956615 DOI: 10.3390/ijms22052322] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disease (NDD) characterized by the degenerative loss of dopaminergic neurons in the substantia nigra along with aggregation of α-synuclein (α-syn). Neurogenic differentiation of human adipose-derived stem cells (NI-hADSCs) by supplementary factors for 14 days activates different biological signaling pathways. In this study, we evaluated the therapeutic role of NI-hADSC-conditioned medium (NI-hADSC-CM) in rotenone (ROT)-induced toxicity in SH-SY5Y cells. Increasing concentrations of ROT led to decreased cell survival at 24 and 48 h in a dose- and time-dependent manner. Treatment of NI-hADSC-CM (50% dilution in DMEM) against ROT (0.5 μM) significantly increased the cell survival. ROT toxicity decreased the expression of tyrosine hydroxylase (TH). Western blot analysis of the Triton X-100-soluble fraction revealed that ROT significantly decreased the oligomeric, dimeric, and monomeric phosphorylated Serine129 (p-S129) α-syn, as well as the total monomeric α-syn expression levels. ROT toxicity increased the oligomeric, but decreased the dimeric and monomeric p-S129 α-syn expression levels. Total α-syn expression (in all forms) was increased in the Triton X-100-insoluble fraction, compared to the control. NI-hADSC-CM treatment enhanced the TH expression, stabilized α-syn monomers, reduced the levels of toxic insoluble p-S129 α-syn, improved the expression of neuronal functional proteins, regulated the Bax/Bcl-2 ratio, and upregulated the expression of pro-caspases, along with PARP-1 inactivation. Moreover, hADSC-CM treatment decreased the cell numbers and have no effect against ROT toxicity on SH-SY5Y cells. The therapeutic effects of NI-hADSC-CM was higher than the beneficial effects of hADSC-CM on cellular signaling. From these results, we conclude that NI-hADSC-CM exerts neuroregenerative effects on ROT-induced PD-like impairments in SH-SY5Y cells.
Collapse
Affiliation(s)
- Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Korea
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Korea
| |
Collapse
|
6
|
Ramalingam M, Jang S, Jeong HS. Therapeutic Effects of Conditioned Medium of Neural Differentiated Human Bone Marrow-Derived Stem Cells on Rotenone-Induced Alpha-Synuclein Aggregation and Apoptosis. Stem Cells Int 2021; 2021:6658271. [PMID: 33552161 PMCID: PMC7847328 DOI: 10.1155/2021/6658271] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been used against several diseases. Their potential mainly appears from its secreted biomolecules. Human bone marrow-derived stem cells (hBMSC) displayed neuronal functional characteristics after differentiation by basic fibroblast growth factor (bFGF) and forskolin. PD is a chronic age-related neurodegenerative disease (NDD) characterized by loss of dopaminergic neurons in the substantia nigra (SN) and abnormal accumulation of α-synuclein (α-syn) aggregations. In this present study, we evaluated the therapeutic effects of neural differentiated hBMSC (NI-hBMSC) conditioned medium (NI-hBMSC-CM) to a rotenone- (ROT-) induced Parkinson's disease (PD) model in SH-SY5Y cells. NI-hBMSC-CM treatment (50% diluted) in the last 24 h of 48 h ROT (0.5 μM) toxicity showed a significant increase in cell survival. The decreased tyrosine hydroxylase (TH) expression as a hallmark of PD was increased by NI-hBMSC-CM. The Triton X-100-soluble and Triton X-100-insoluble cell lysate fractions were used in Western blotting. The oligomeric, dimeric, and monomeric phosphorylated serine129 (p-S129) α-syn and total monomeric α-syn were decreased during ROT toxicity in the Triton X-100-soluble fraction. The Triton X-100-insoluble fraction revealed that ROT toxicity significantly increased the oligomeric but decreased the dimeric and monomeric p-S129 α-syn expressions while all forms of total α-syn were increased in SH-SY5Y cells. NI-hBMSC-CM stabilized the physiological α-syn monomers and reduced aggregated insoluble p-S129 α-syn against ROT. The cytoskeletal proteins, neurofilament-H (NF-H), β3-tubulin (Tuj1), neuronal nuclei (NeuN), and synaptophysin (SYP) were significantly decreased during ROT toxicity. In addition, proapoptotic Bax was increased by ROT with decreased antiapoptotic Bcl-2 and Mcl-1 as well as proforms of caspase-9, caspase-3, caspase-7, and PARP-1. NI-hBMSC-CM ameliorated the neurotrophic protein expressions, controlled the Bax/Bcl-2 ratio, upregulated procaspases, and inactivated PARP-1. From our results, we conclude that NI-hBMSC-CM containing released biomolecules during neural differentiation employs regenerative effects on the ROT model of PD in SH-SY5Y cells.
Collapse
Affiliation(s)
- Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Hwasun, Jeollanam-do 58128, Republic of Korea
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun, Jeollanam-do 58128, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun, Jeollanam-do 58128, Republic of Korea
| |
Collapse
|
7
|
Lee HJ, Ricarte D, Ortiz D, Lee SJ. Models of multiple system atrophy. Exp Mol Med 2019; 51:1-10. [PMID: 31740682 PMCID: PMC6861264 DOI: 10.1038/s12276-019-0346-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 11/09/2022] Open
Abstract
Multiple system atrophy (MSA) is a neurodegenerative disease with diverse clinical manifestations, including parkinsonism, cerebellar syndrome, and autonomic failure. Pathologically, MSA is characterized by glial cytoplasmic inclusions in oligodendrocytes, which contain fibrillary forms of α-synuclein. MSA is categorized as one of the α-synucleinopathy, and α-synuclein aggregation is thought to be the culprit of the disease pathogenesis. Studies on MSA pathogenesis are scarce relative to studies on the pathogenesis of other synucleinopathies, such as Parkinson’s disease and dementia with Lewy bodies. However, recent developments in cellular and animal models of MSA, especially α-synuclein transgenic models, have driven advancements in research on this disease. Here, we review the currently available models of MSA, which include toxicant-induced animal models, α-synuclein-overexpressing cellular models, and mouse models that express α-synuclein specifically in oligodendrocytes through cell type-specific promoters. We will also discuss the results of studies in recently developed transmission mouse models, into which MSA brain extracts were intracerebrally injected. By reviewing the findings obtained from these model systems, we will discuss what we have learned about the disease and describe the strengths and limitations of the models, thereby ultimately providing direction for the design of better models and future research. A review of the models available for studying multiple system atrophy (MSA), a Parkinson’s-like disease, may help identify new treatment options. MSA is difficult to diagnose and unresponsive to drugs. Similar to Parkinson’s disease, it involves accumulation of protein aggregates in brain and spinal cord cells, but the causes are poorly understood. He-Jin Lee at Konkuk University, and Seung-Jae Lee at Seoul National University College of Medicine in South Korea and coworkers have reviewed the models available to study the disease, including toxin-induced and transgenic animal models, and recent evidence that transferring the protein aggregates into cells causes MSA symptoms. Each model mimics some aspects of the disease, but none captures the full range of symptoms. This review helps highlight research pathways that may illuminate treatments for this complex and debilitating adult-onset disease.
Collapse
Affiliation(s)
- He-Jin Lee
- Department of Anatomy, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-gu, Seoul, 05029, South Korea. .,Research Institute of Medical Science, Konkuk University, Seoul, 05029, South Korea. .,IBST, Konkuk University, Seoul, 05029, South Korea.
| | - Diadem Ricarte
- Department of Anatomy, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Darlene Ortiz
- Department of Anatomy, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Seung-Jae Lee
- Department of Medicine and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
8
|
Sekaran H, Gan CY, A Latiff A, Harvey TM, Mohd Nazri L, Hanapi NA, Azizi J, Yusof SR. Changes in blood-brain barrier permeability and ultrastructure, and protein expression in a rat model of cerebral hypoperfusion. Brain Res Bull 2019; 152:63-73. [PMID: 31301381 DOI: 10.1016/j.brainresbull.2019.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/16/2019] [Accepted: 07/08/2019] [Indexed: 12/15/2022]
Abstract
Cerebral hypoperfusion involved a reduction in cerebral blood flow, leading to neuronal dysfunction, microglial activation and white matter degeneration. The effects on the blood-brain barrier (BBB) however, have not been well-documented. Here, two-vessel occlusion model was adopted to mimic the condition of cerebral hypoperfusion in Sprague-Dawley rats. The BBB permeability to high and low molecular weight exogenous tracers i.e. Evans blue dye and sodium fluorescein respectively, showed marked extravasation of the Evans blue dye in the frontal cortex, posterior cortex and thalamus-midbrain at day 1 following induction of cerebral hypoperfusion. Transmission electron microscopy revealed brain endothelial cell and astrocyte damages including increased pinocytotic vesicles and formation of membrane invaginations in the endothelial cells, and swelling of the astrocytes' end-feet. Investigation on brain microvessel protein expressions using two-dimensional (2D) gel electrophoresis coupled with LC-MS/MS showed that proteins involved in mitochondrial energy metabolism, transcription regulation, cytoskeleton maintenance and signaling pathways were differently expressed. The expression of aconitate hydratase, heterogeneous nuclear ribonucleoprotein, enoyl Co-A hydratase and beta-synuclein were downregulated, while the opposite observed for calreticulin and enhancer of rudimentary homolog. These findings provide insights into the BBB molecular responses to cerebral hypoperfusion, which may assist development of future therapeutic strategies.
Collapse
Affiliation(s)
- Hema Sekaran
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Chee-Yuen Gan
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Aishah A Latiff
- Toxicology and Multipurpose Lab, Anti-Doping Lab Qatar, Sports City St, 27775, Doha, Qatar
| | - Thomas Michael Harvey
- Toxicology and Multipurpose Lab, Anti-Doping Lab Qatar, Sports City St, 27775, Doha, Qatar
| | - Liyana Mohd Nazri
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Nur Aziah Hanapi
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Juzaili Azizi
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Siti R Yusof
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia.
| |
Collapse
|
9
|
Microtubule Destabilization Paves the Way to Parkinson's Disease. Mol Neurobiol 2016; 54:6762-6774. [PMID: 27757833 DOI: 10.1007/s12035-016-0188-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/30/2016] [Indexed: 01/20/2023]
Abstract
Microtubules are dynamic structures normally associated to the cell division, during which they form the mitotic spindle, as well as to the initial phases of specification and polarization of various cell types, including neurons. Although microtubules could have a role in the death of many cells and tissues, the microtubule-based degenerative mechanisms have been poorly investigated; nevertheless, during the last two decades, many clues have been accumulated suggesting the importance of the microtubule system during neurodegeneration. Thus, the aim of this review is to analyse how the changes of the microtubule cytoskeleton, in terms of organization and dynamics, as well as the failure of the microtubule-dependent neuronal processes, as axonal transport, may play a pivotal role in the chain of events leading to Parkinson's disease. Last but not least, since disease-modifying or neuroprotective strategies are a clinical priority in Parkinson's disease, we will also present the hints about the concrete possibility of a microtubule-targeted therapy, which would have the potentiality to block the running degenerative events and to prompt the regeneration of the lost tissues.
Collapse
|
10
|
Abstract
α-Synuclein is a presynaptic protein associated to Parkinson’s disease, which is unstructured when free in the cytoplasm and adopts α helical conformation when bound to vesicles. After decades of intense studies, α-Synuclein physiology is still difficult to clear up due to its interaction with multiple partners and its involvement in a pletora of neuronal functions. Here, we looked at the remarkably neglected interplay between α-Synuclein and microtubules, which potentially impacts on synaptic functionality. In order to identify the mechanisms underlying these actions, we investigated the interaction between purified α-Synuclein and tubulin. We demonstrated that α-Synuclein binds to microtubules and tubulin α2β2 tetramer; the latter interaction inducing the formation of helical segment(s) in the α-Synuclein polypeptide. This structural change seems to enable α-Synuclein to promote microtubule nucleation and to enhance microtubule growth rate and catastrophe frequency, both in vitro and in cell. We also showed that Parkinson’s disease-linked α-Synuclein variants do not undergo tubulin-induced folding and cause tubulin aggregation rather than polymerization. Our data enable us to propose α-Synuclein as a novel, foldable, microtubule-dynamase, which influences microtubule organisation through its binding to tubulin and its regulating effects on microtubule nucleation and dynamics.
Collapse
|
11
|
Bleasel JM, Halliday GM, Kim WS. Animal modeling an oligodendrogliopathy--multiple system atrophy. Acta Neuropathol Commun 2016; 4:12. [PMID: 26860328 PMCID: PMC4748629 DOI: 10.1186/s40478-016-0279-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/23/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple system atrophy (MSA) is a rare, yet rapidly-progressive neurodegenerative disease that presents clinically with autonomic failure in combination with parkinsonism or cerebellar ataxia. The definitive neuropathology differentiating MSA from Lewy body diseases is the presence of α-synuclein aggregates in oligodendrocytes (called glial cytoplasmic inclusion or GCI) rather than the fibrillar aggregates in neurons (called Lewy bodies). This makes the pathological pathway(s) in MSA unique in that oligodendrocytes are involved rather than predominantly neurons, as is most other neurodegenerative disorders. MSA is therefore regarded as an oligodendrogliopathy. The etiology of MSA is unknown. No definitive risk factors have been identified, although α-synuclein and other genes have been variably linked to MSA risk. Utilization of postmortem brain tissues has greatly advanced our understanding of GCI pathology and the subsequent neurodegeneration. However, extrapolating the early pathogenesis of MSA from such resource has been difficult and limiting. In recent years, cell and animal models developed for MSA have been instrumental in delineating unique MSA pathological pathways, as well as aiding in clinical phenotyping. The purpose of this review is to bring together and discuss various animal models that have been developed for MSA and how they have advanced our understanding of MSA pathogenesis, particularly the dynamics of α-synuclein aggregation. This review will also discuss how animal models have been used to explore potential therapeutic avenues for MSA, and future directions of MSA modeling.
Collapse
|
12
|
Milisav I, Šuput D, Ribarič S. Unfolded Protein Response and Macroautophagy in Alzheimer's, Parkinson's and Prion Diseases. Molecules 2015; 20:22718-56. [PMID: 26694349 PMCID: PMC6332363 DOI: 10.3390/molecules201219865] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 12/13/2022] Open
Abstract
Proteostasis are integrated biological pathways within cells that control synthesis, folding, trafficking and degradation of proteins. The absence of cell division makes brain proteostasis susceptible to age-related changes and neurodegeneration. Two key processes involved in sustaining normal brain proteostasis are the unfolded protein response and autophagy. Alzheimer’s disease (AD), Parkinson’s disease (PD) and prion diseases (PrDs) have different clinical manifestations of neurodegeneration, however, all share an accumulation of misfolded pathological proteins associated with perturbations in unfolded protein response and macroautophagy. While both the unfolded protein response and macroautophagy play an important role in the prevention and attenuation of AD and PD progression, only macroautophagy seems to play an important role in the development of PrDs. Macroautophagy and unfolded protein response can be modulated by pharmacological interventions. However, further research is necessary to better understand the regulatory pathways of both processes in health and neurodegeneration to be able to develop new therapeutic interventions.
Collapse
Affiliation(s)
- Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, Zaloška 4, Ljubljana SI-1000, Slovenia.
- Faculty of Health Sciences, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenija.
| | - Dušan Šuput
- Institute of Pathophysiology, Faculty of Medicine, Zaloška 4, Ljubljana SI-1000, Slovenia.
| | - Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, Zaloška 4, Ljubljana SI-1000, Slovenia.
| |
Collapse
|
13
|
Jin C, Washimi Y, Yoshida K, Hashizume Y, Yazawa I. Characterization of spheroids in hereditary diffuse leukoencephalopathy with axonal spheroids. J Neurol Sci 2015; 352:74-8. [PMID: 25843289 DOI: 10.1016/j.jns.2015.03.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/16/2015] [Accepted: 03/18/2015] [Indexed: 11/28/2022]
Abstract
Hereditary diffuse leukoencephalopathy with axonal spheroids (HDLS) is a neurodegenerative disease clinically characterized by slowly progressive cognitive decline and motor dysfunction. Neuropathology shows diffuse degeneration in the white matter, with prominent presence of widespread axonal spheroids. To investigate the mechanism underlying HDLS neurodegeneration, we characterized spheroids and examined their development in the degenerated white matter. Analysis revealed that the spheroids are an early neuropathological manifestation in the white matter degeneration and involve axonal component proteins and α-synuclein. The development of spheroids facilitates in initiating neurodegeneration in HDLS.
Collapse
Affiliation(s)
- Chenghua Jin
- Laboratory of Research Resources, Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Yukihiko Washimi
- Department of Neurology, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Kunihiro Yoshida
- Division of Neurogenetics, Department of Brain Disease Research, Shinshu University School of Medicine, Nagano 390-8621, Japan
| | - Yoshio Hashizume
- Laboratory of Neuropathology, Fukushimura Hospital, Aichi 441-8124, Japan
| | - Ikuru Yazawa
- Laboratory of Research Resources, Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan.
| |
Collapse
|
14
|
Stefanova N, Wenning GK. Animal models of multiple system atrophy. Clin Auton Res 2015; 25:9-17. [PMID: 25585910 PMCID: PMC4412689 DOI: 10.1007/s10286-014-0266-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/18/2014] [Indexed: 11/27/2022]
Abstract
Since their introduction in 1996, animal models of multiple system atrophy (MSA) have generated important insights into pathogenesis and interventional therapies. Toxin and genetic approaches have been used alone or in combination to replicate progressive motor and non-motor symptoms reflecting human neuropathology. Here, we review these developments and discuss the advantages and limitations of the MSA animal models, as well as their application in preclinical target validation.
Collapse
Affiliation(s)
- Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, Anichstr. 35, 6020, Innsbruck, Austria,
| | | |
Collapse
|
15
|
Chen J, Mills JD, Halliday GM, Janitz M. The role of transcriptional control in multiple system atrophy. Neurobiol Aging 2015; 36:394-400. [DOI: 10.1016/j.neurobiolaging.2014.08.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/29/2014] [Accepted: 08/12/2014] [Indexed: 12/15/2022]
|
16
|
Jellinger KA. Neuropathology of multiple system atrophy: New thoughts about pathogenesis. Mov Disord 2014; 29:1720-41. [DOI: 10.1002/mds.26052] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 08/29/2014] [Accepted: 09/16/2014] [Indexed: 12/14/2022] Open
|
17
|
Suzuki Y, Jin C, Iwase T, Yazawa I. β-III Tubulin fragments inhibit α-synuclein accumulation in models of multiple system atrophy. J Biol Chem 2014; 289:24374-82. [PMID: 25028513 DOI: 10.1074/jbc.m114.557215] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multiple system atrophy (MSA) is a neurodegenerative disease caused by α-synuclein aggregation in oligodendrocytes and neurons. Using a transgenic mouse model overexpressing human α-synuclein in oligodendrocytes, we previously demonstrated that oligodendrocytic α-synuclein inclusions induce neuronal α-synuclein accumulation and progressive neuronal degeneration. α-Synuclein binds to β-III tubulin, leading to the neuronal accumulation of insoluble α-synuclein in an MSA mouse model. The present study demonstrates that α-synuclein co-localizes with β-III tubulin in the brain tissue from patients with MSA and MSA model transgenic mice as well as neurons cultured from these mice. Accumulation of insoluble α-synuclein in MSA mouse neurons was blocked by the peptide fragment β-III tubulin (residues 235-282). We have determined the α-synuclein-binding domain of β-III tubulin and demonstrated that a short fragment containing this domain can suppress α-synuclein accumulation in the primary cultured cells. Administration of a short α-synuclein-binding fragment of β-III tubulin may be a novel therapeutic strategy for MSA.
Collapse
Affiliation(s)
- Yasuyo Suzuki
- From the Laboratory of Research Resources, Research Institute, National Center for Geriatrics and Gerontology, 35 Gengo, Morioka, Obu-shi, Aichi 474-8511, Japan and
| | - Chenghua Jin
- From the Laboratory of Research Resources, Research Institute, National Center for Geriatrics and Gerontology, 35 Gengo, Morioka, Obu-shi, Aichi 474-8511, Japan and
| | - Tamaki Iwase
- the Department of Neurology, Nagoya City Koseiin Medical Welfare Center, Aichi 465-8610, Japan
| | - Ikuru Yazawa
- From the Laboratory of Research Resources, Research Institute, National Center for Geriatrics and Gerontology, 35 Gengo, Morioka, Obu-shi, Aichi 474-8511, Japan and
| |
Collapse
|
18
|
Palma JA, Kaufmann H. Novel therapeutic approaches in multiple system atrophy. Clin Auton Res 2014; 25:37-45. [PMID: 24928797 DOI: 10.1007/s10286-014-0249-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/23/2014] [Indexed: 01/12/2023]
Abstract
Multiple system atrophy (MSA) is a sporadic, adult onset, relentlessly progressive neurodegenerative disease characterized by autonomic abnormalities associated with parkinsonism, cerebellar dysfunction, pyramidal signs, or combinations thereof. Treatments that can halt or reverse the progression of MSA have not yet been identified. MSA is neuropathologically defined by the presence of α-synuclein-containing inclusions, particularly in the cytoplasm of oligodendrocytes (glial cytoplasmic inclusions, GCIs), which are associated with neurodegeneration. The mechanisms by which oligodendrocytic α-synuclein inclusions cause neuronal death in MSA are not completely understood. The MSA neurodegenerative process likely comprises cell-to-cell transmission of α-synuclein in a prion-like manner, α-synuclein aggregation, increased oxidative stress, abnormal expression of tubulin proteins, decreased expression of neurotrophic factors, excitotoxicity and microglial activation, and neuroinflammation. In an attempt to block each of these pathogenic mechanisms, several pharmacologic approaches have been tried and shown to exert neuroprotective effects in transgenic mouse or cellular models of MSA. These include sertraline, paroxetine, and lithium, which hamper arrival of α-synuclein to oligodendroglia; rifampicin, lithium, and non-steroidal anti-inflammatory drugs, which inhibit α-synuclein aggregation in oligodendrocytes; riluzole, rasagiline, fluoxetine and mesenchymal stem cells, which exert neuroprotective actions; and minocycline and intravenous immunoglobulins, which reduce neuroinflammation and microglial activation. These and other potential therapeutic strategies for MSA are summarized in this review.
Collapse
Affiliation(s)
- Jose-Alberto Palma
- Department of Neurology, Dysautonomia Center, New York University School of Medicine, 530 First Av, Suite 9Q, New York, NY, 10016, USA
| | | |
Collapse
|
19
|
Kuzdas-Wood D, Stefanova N, Jellinger KA, Seppi K, Schlossmacher MG, Poewe W, Wenning GK. Towards translational therapies for multiple system atrophy. Prog Neurobiol 2014; 118:19-35. [PMID: 24598411 PMCID: PMC4068324 DOI: 10.1016/j.pneurobio.2014.02.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 02/07/2014] [Accepted: 02/21/2014] [Indexed: 12/28/2022]
Abstract
Multiple system atrophy (MSA) is a fatal adult-onset neurodegenerative disorder of uncertain etiopathogenesis manifesting with autonomic failure, parkinsonism, and ataxia in any combination. The underlying neuropathology affects central autonomic, striatonigral and olivopontocerebellar pathways and it is associated with distinctive glial cytoplasmic inclusions (GCIs, Papp-Lantos bodies) that contain aggregates of α-synuclein. Current treatment options are very limited and mainly focused on symptomatic relief, whereas disease modifying options are lacking. Despite extensive testing, no neuroprotective drug treatment has been identified up to now; however, a neurorestorative approach utilizing autologous mesenchymal stem cells has shown remarkable beneficial effects in the cerebellar variant of MSA. Here, we review the progress made over the last decade in defining pathogenic targets in MSA and summarize insights gained from candidate disease-modifying interventions that have utilized a variety of well-established preclinical MSA models. We also discuss the current limitations that our field faces and suggest solutions for possible approaches in cause-directed therapies of MSA.
Collapse
Affiliation(s)
- Daniela Kuzdas-Wood
- Department of Neurology, Innsbruck Medical University, Anichstraße 35, Innsbruck 6020, Austria
| | - Nadia Stefanova
- Department of Neurology, Innsbruck Medical University, Anichstraße 35, Innsbruck 6020, Austria
| | | | - Klaus Seppi
- Department of Neurology, Innsbruck Medical University, Anichstraße 35, Innsbruck 6020, Austria
| | - Michael G Schlossmacher
- Divisions of Neuroscience and Neurology, The Ottawa Hospital Research Institute, University of Ottawa, 451 Smyth Road, RGH #1412, Ottawa, ON, K1H 8M5, Canada
| | - Werner Poewe
- Department of Neurology, Innsbruck Medical University, Anichstraße 35, Innsbruck 6020, Austria
| | - Gregor K Wenning
- Department of Neurology, Innsbruck Medical University, Anichstraße 35, Innsbruck 6020, Austria.
| |
Collapse
|
20
|
Suzuki Y, Jin C, Yazawa I. Cystatin C triggers neuronal degeneration in a model of multiple system atrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:790-9. [PMID: 24405769 DOI: 10.1016/j.ajpath.2013.11.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 11/04/2013] [Accepted: 11/04/2013] [Indexed: 01/17/2023]
Abstract
Multiple system atrophy is an intractable neurodegenerative disease caused by α-synuclein (α-syn) accumulation in oligodendrocytes and neurons. With the use of a transgenic mouse model overexpressing human α-syn in oligodendrocytes, we demonstrated that oligodendrocytic α-syn inclusions induce neuronal α-syn accumulation, resulting in progressive neuronal degeneration. The mechanism through which oligodendrocytic α-syn inclusions trigger neuronal α-syn accumulation leading to multiple system atrophy is unknown. In this study, we identified cystatin C, an oligodendrocyte-derived secretory protein that triggers α-syn up-regulation and insoluble α-syn accumulation, in neurons of the mouse central nervous system. Cystatin C was released by mouse oligodendrocytes overexpressing human α-syn, and extracellular cystatin C increased the expression of the endogenous α-syn gene in wild-type mouse neurons. These neurons then accumulate insoluble α-syn and may undergo apoptosis. Cystatin C is a potential pathogenic signal triggering neurodegeneration in multiple system atrophy.
Collapse
Affiliation(s)
- Yasuyo Suzuki
- Laboratory of Research Resources, Research Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Chenghua Jin
- Laboratory of Research Resources, Research Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Ikuru Yazawa
- Laboratory of Research Resources, Research Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Aichi, Japan.
| |
Collapse
|
21
|
Abstract
α-Synuclein is a presynaptic neuronal protein that is linked genetically and neuropathologically to Parkinson's disease (PD). α-Synuclein may contribute to PD pathogenesis in a number of ways, but it is generally thought that its aberrant soluble oligomeric conformations, termed protofibrils, are the toxic species that mediate disruption of cellular homeostasis and neuronal death, through effects on various intracellular targets, including synaptic function. Furthermore, secreted α-synuclein may exert deleterious effects on neighboring cells, including seeding of aggregation, thus possibly contributing to disease propagation. Although the extent to which α-synuclein is involved in all cases of PD is not clear, targeting the toxic functions conferred by this protein when it is dysregulated may lead to novel therapeutic strategies not only in PD, but also in other neurodegenerative conditions, termed synucleinopathies.
Collapse
Affiliation(s)
- Leonidas Stefanis
- Laboratory of Neurodegenerative Diseases, Biomedical Research Foundation of the Academy of Athens, and Second Department of Neurology, University of Athens Medical School, Athens 11527, Greece.
| |
Collapse
|
22
|
Prots I, Veber V, Brey S, Campioni S, Buder K, Riek R, Böhm KJ, Winner B. α-Synuclein oligomers impair neuronal microtubule-kinesin interplay. J Biol Chem 2013; 288:21742-54. [PMID: 23744071 DOI: 10.1074/jbc.m113.451815] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Early α-synuclein (α-Syn)-induced alterations are neurite pathologies resulting in Lewy neurites. α-Syn oligomers are a toxic species in synucleinopathies and are suspected to cause neuritic pathology. To investigate how α-Syn oligomers may be linked to aberrant neurite pathology, we modeled different stages of α-Syn aggregation in vitro and investigated the interplay of α-Syn aggregates with proteins involved in axonal transport. The interaction of wild type α-Syn (WTS) and α-Syn variants (E57K, A30P, and aSyn(30-110)) with kinesin, tubulin, and the microtubule (MT)-associated proteins, MAP2 and Tau, is stronger for multimers than for monomers. WTS seeds but not α-Syn oligomers significantly and dose-dependently reduced Tau-promoted MT assembly in vitro. In contrast, MT gliding velocity across kinesin-coated surfaces was significantly decreased in the presence of α-Syn oligomers but not WTS seeds or fibrils (aSyn(30-110) multimers). In a human dopaminergic neuronal cell line, mild overexpression of the oligomerizing E57K α-Syn variant significantly impaired neurite network morphology without causing profound cell death. In accordance with these findings, MT stability, neuritic kinesin, and neuritic kinesin-dependent cargoes were significantly reduced by the presence of α-Syn oligomers. In summary, different α-Syn species act divergently on the axonal transport machinery. These findings provide new insights into α-Syn oligomer-driven neuritic pathology as one of the earliest events in synucleinopathies.
Collapse
Affiliation(s)
- Iryna Prots
- Junior Research Group III, Nikolaus Fiebiger Centre for Molecular Medicine, Universitaetsklinikum Erlangen, Glueckstrasse 6, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Bury A, Pienaar IS. Behavioral testing regimens in genetic-based animal models of Parkinson's disease: cogencies and caveats. Neurosci Biobehav Rev 2013; 37:846-59. [PMID: 23558176 DOI: 10.1016/j.neubiorev.2013.03.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/01/2013] [Accepted: 03/11/2013] [Indexed: 12/20/2022]
Abstract
Although the onset and progression of Parkinson's disease (PD) is fundamentally sporadic, identification of several of the genes implicated in the disease has provided significant insight concerning patho-physiological mechanisms potentially underlying sporadic PD. Moreover, such studies have caused a revolution in the way researchers view the disease. Since single genes responsible for rare familial forms of the disease have only been identified within the past few years, animal models based on these defects have only recently been generated, thereby not leaving a lot of time for their evaluation and subsequent improvement. The current article provides an extensive review of the major motor and non-motor behavioral tests used in genetically-induced Parkinsonian animals. Moreover, we assess the insights concerning the etiopathogenesis of PD generated from use of such tests and how these have improved available treatment strategies for alleviating aspects of sporadic and non-sporadic parkinsonism.
Collapse
Affiliation(s)
- Alexander Bury
- Centre for Neurodegeneration and Neuroinflammation, Division of Brain Sciences, Department of Medicine, Imperial College London, United Kingdom
| | | |
Collapse
|
24
|
Kragh CL, Fillon G, Gysbers A, Hansen HD, Neumann M, Richter-Landsberg C, Haass C, Zalc B, Lubetzki C, Gai WP, Halliday GM, Kahle PJ, Jensen PH. FAS-dependent cell death in α-synuclein transgenic oligodendrocyte models of multiple system atrophy. PLoS One 2013; 8:e55243. [PMID: 23372841 PMCID: PMC3555893 DOI: 10.1371/journal.pone.0055243] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 12/27/2012] [Indexed: 12/26/2022] Open
Abstract
Multiple system atrophy is a parkinsonian neurodegenerative disorder. It is cytopathologically characterized by accumulation of the protein p25α in cell bodies of oligodendrocytes followed by accumulation of aggregated α-synuclein in so-called glial cytoplasmic inclusions. p25α is a stimulator of α-synuclein aggregation, and coexpression of α-synuclein and p25α in the oligodendroglial OLN-t40-AS cell line causes α-synuclein aggregate-dependent toxicity. In this study, we investigated whether the FAS system is involved in α-synuclein aggregate dependent degeneration in oligodendrocytes and may play a role in multiple system atrophy. Using rat oligodendroglial OLN-t40-AS cells we demonstrate that the cytotoxicity caused by coexpressing α-synuclein and p25α relies on stimulation of the death domain receptor FAS and caspase-8 activation. Using primary oligodendrocytes derived from PLP-α-synuclein transgenic mice we demonstrate that they exist in a sensitized state expressing pro-apoptotic FAS receptor, which makes them sensitive to FAS ligand-mediated apoptosis. Immunoblot analysis shows an increase in FAS in brain extracts from multiple system atrophy cases. Immunohistochemical analysis demonstrated enhanced FAS expression in multiple system atrophy brains notably in oligodendrocytes harboring the earliest stages of glial cytoplasmic inclusion formation. Oligodendroglial FAS expression is an early hallmark of oligodendroglial pathology in multiple system atrophy that mechanistically may be coupled to α-synuclein dependent degeneration and thus represent a potential target for protective intervention.
Collapse
Affiliation(s)
| | - Gwenaëlle Fillon
- Laboratory for Alzheimer's and Parkinson's Disease Research, Department of Biochemistry, Ludwig Maximilians University, Munich, Germany
| | - Amanda Gysbers
- Neuroscience Research Australia and University of New South Wales, Sydney, New South Wales, Australia
| | - Hanne D. Hansen
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Manuela Neumann
- Institute for Neuropathology, University of Zürich, Zürich, Switzerland
| | | | - Christian Haass
- Laboratory for Alzheimer's and Parkinson's Disease Research, Department of Biochemistry, Ludwig Maximilians University, Munich, Germany
| | - Bernard Zalc
- Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, Université Pierre et Marie Curie, Paris, France
| | - Catherine Lubetzki
- Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, Université Pierre et Marie Curie, Paris, France
| | - Wei-Ping Gai
- Department of Human Physiology and Centre for Neuroscience, Flinders University School of Medicine, Bedford Park, South Australia, Australia
| | - Glenda M. Halliday
- Neuroscience Research Australia and University of New South Wales, Sydney, New South Wales, Australia
| | - Philipp J. Kahle
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, Tübingen, Germany
- * E-mail: (PHJ); (PJK)
| | - Poul H. Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- * E-mail: (PHJ); (PJK)
| |
Collapse
|
25
|
Abstract
Multiple system atrophy (MSA) is a predominantly sporadic, adult-onset, fatal neurodegenerative disease of unknown etiology. MSA is characterized by autonomic failure, levodopa-unresponsive parkinsonism, cerebellar ataxia and pyramidal signs in any combination. MSA belongs to a group of neurodegenerative disorders termed α-synucleinopathies, which also include Parkinson's disease and dementia with Lewy bodies. Their common pathological feature is the occurrence of abnormal α-synuclein positive inclusions in neurons or glial cells. In MSA, the main cell type presenting aggregates composed of α-synuclein are oligodendroglial cells . This pathological hallmark, also called glial cytoplasmic inclusions (GCIs) , is associated with progressive and profound neuronal loss in various regions of the brain. The development of animal models of MSA is justified by the limited understanding of the mechanisms of neurodegeneration and GCIs formation, which is paralleled by a lack of therapeutic strategies. Two main types of rodent models have been generated to replicate different features of MSA neuropathology. On one hand, neurotoxin-based models have been produced to reproduce neuronal loss in substantia nigra pars compacta and striatum. On the other hand, transgenic mouse models with overexpression of α-synuclein in oligodendroglia have been used to reproduce GCIs-related pathology. This chapter gives an overview of the atypical Parkinson's syndrome MSA and summarizes the currently available MSA animal models and their relevance for pre-clinical testing of disease-modifying therapies.
Collapse
Affiliation(s)
- Lisa Fellner
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria,
| | | | | |
Collapse
|
26
|
α-Synuclein accumulation reduces GABAergic inhibitory transmission in a model of multiple system atrophy. Biochem Biophys Res Commun 2012; 428:348-53. [PMID: 23098910 DOI: 10.1016/j.bbrc.2012.10.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 10/14/2012] [Indexed: 12/22/2022]
Abstract
Multiple system atrophy is a neurodegenerative disease caused by abnormal α-synuclein (α-syn) accumulation in oligodendrocytes and neurons. We previously demonstrated that transgenic (Tg) mice that selectively overexpressed human α-syn in oligodendrocytes exhibited neuronal α-syn accumulation. Microtubule β-III tubulin binds to endogenous neuronal α-syn to form an insoluble complex, leading to progressive neuronal degeneration. α-Syn accumulation is increased in the presynaptic terminals of Tg mice neurons and may reduce neurotransmitter release. To clarify the mechanisms underlying its involvement in neuronal dysfunction, in the present study, we investigated the effects of neuronal α-syn accumulation on synaptic function in Tg mice. Using whole-cell patch-clamp recording, we found that the frequency of miniature inhibitory postsynaptic currents was reduced in Tg mice. Furthermore, a microtubule depolymerizing agent restored normal frequencies of miniature inhibitory postsynaptic currents in Tg mice. These findings suggest that α-syn and β-III tubulin protein complex plays roles for regulation of synaptic vesicle release in GABAergic interneurons, and it causes to reduce GABAergic inhibitory transmission.
Collapse
|
27
|
Nakayama K, Suzuki Y, Yazawa I. Binding of neuronal α-synuclein to β-III tubulin and accumulation in a model of multiple system atrophy. Biochem Biophys Res Commun 2011; 417:1170-5. [PMID: 22227187 DOI: 10.1016/j.bbrc.2011.12.092] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 12/19/2011] [Indexed: 10/14/2022]
Abstract
Multiple system atrophy (MSA) is a neurodegenerative disease caused by α-synuclein (α-syn) accumulation in oligodendrocytes and neurons. We generated a transgenic (Tg) mouse model in which human α-syn was overexpressed in oligodendrocytes. Our previous studies have revealed that oligodendrocytic α-syn inclusions induced neuronal α-syn accumulation, thereby resulting in progressive neuronal degeneration in mice. We also demonstrated that an insoluble complex of α-syn and β-III tubulin in microtubules progressively accumulated in neurons, thereby leading to neuronal degeneration. In the present study, we demonstrated that neuronal accumulation of the insoluble complex was derived from binding of α-syn to β-III tubulin and not from α-syn self-aggregation. Thus, interaction between α-syn and β-III tubulin plays an important role in neuronal α-syn accumulation in an MSA mouse model.
Collapse
Affiliation(s)
- Kimiko Nakayama
- Laboratory of Research Resources, Research Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Obu, Japan
| | | | | |
Collapse
|
28
|
Vekrellis K, Xilouri M, Emmanouilidou E, Rideout HJ, Stefanis L. Pathological roles of α-synuclein in neurological disorders. Lancet Neurol 2011; 10:1015-25. [PMID: 22014436 DOI: 10.1016/s1474-4422(11)70213-7] [Citation(s) in RCA: 286] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Substantial genetic, neuropathological, and biochemical evidence implicates the presynaptic neuronal protein α-synuclein in Parkinson's disease and related Lewy body disorders. How dysregulation of α-synuclein leads to neurodegeneration is, however, unclear. Soluble oligomeric, but not fully fibrillar, α-synuclein is thought to be toxic. The major neuronal target of aberrant α-synuclein might be the synapse. The effects of aberrant α-synuclein might include alteration of calcium homoeostasis or mitochondrial fragmentation and, in turn, mitochondrial dysfunction, which could link α-synuclein dysfunction to recessive and toxin-induced parkinsonism. α-Synuclein also seems to be linked to other genetic forms of Parkinson's disease, such as those linked to mutations in GBA or LRRK2, possibly through common effects on autophagy and lysosomal function. Finally, α-synuclein is physiologically secreted, and this extracellular form could lead to the spread of pathological accumulations and disease progression. Consequently, factors that regulate the levels, post-translational modifications, specific aberrant cellular effects, or secretion of α-synuclein might be targets for therapy.
Collapse
Affiliation(s)
- Kostas Vekrellis
- Biomedical Research Foundation of Academy of Athens, Athens, Greece
| | | | | | | | | |
Collapse
|
29
|
Fleming SM, Mulligan CK, Richter F, Mortazavi F, Lemesre V, Frias C, Zhu C, Stewart A, Gozes I, Morimoto B, Chesselet MF. A pilot trial of the microtubule-interacting peptide (NAP) in mice overexpressing alpha-synuclein shows improvement in motor function and reduction of alpha-synuclein inclusions. Mol Cell Neurosci 2011; 46:597-606. [PMID: 21193046 PMCID: PMC3046337 DOI: 10.1016/j.mcn.2010.12.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 12/01/2010] [Accepted: 12/17/2010] [Indexed: 01/08/2023] Open
Abstract
Abnormal accumulation of α-synuclein is associated with several neurodegenerative disorders (synucleinopathies), including sporadic Parkinson's disease (PD). Genetic mutations and multiplication of α-synuclein cause familial forms of PD and polymorphisms in the α-synuclein gene are associated with PD risk. Overexpression of α-synuclein can impair essential functions within the cell such as microtubule-dependent transport, suggesting that compounds that act on the microtubule system may have therapeutic benefit for synucleinopathies. In this study, mice overexpressing human wildtype α-synuclein under the Thy1 promoter (Thy1-aSyn) and littermate wildtype control mice were administered daily the microtubule-interacting peptide NAPVSIPQ (NAP; also known as davunetide or AL-108) intranasally for 2 months starting at 1 month of age, in a regimen known to produce effective concentrations of the peptide in mouse brain. Motor performance, coordination, and activity were assessed at the end of treatment. Olfactory function, which is altered in PD, was measured 1 month later. Mice were sacrificed at 4.5 months of age, and their brains examined for proteinase K-resistant α-synuclein inclusions in the substantia nigra and olfactory bulb. NAP-treated Thy1-aSyn mice showed a 38% decrease in the number of errors per step in the challenging beam traversal test and a reduction in proteinase K-resistant α-synuclein inclusions in the substantia nigra compared to vehicle treated transgenics. The data indicate a significant behavioral benefit and a long lasting improvement of α-synuclein pathology following administration of a short term (2 months) NAP administration in a mouse model of synucleinopathy.
Collapse
Affiliation(s)
- Sheila M Fleming
- Departments of Neurology and Neurobiology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hasegawa T, Baba T, Kobayashi M, Konno M, Sugeno N, Kikuchi A, Itoyama Y, Takeda A. Role of TPPP/p25 on α-synuclein-mediated oligodendroglial degeneration and the protective effect of SIRT2 inhibition in a cellular model of multiple system atrophy. Neurochem Int 2010; 57:857-66. [PMID: 20849899 DOI: 10.1016/j.neuint.2010.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2010] [Revised: 08/31/2010] [Accepted: 09/02/2010] [Indexed: 10/19/2022]
Abstract
Multiple system atrophy (MSA) is a progressive neurodegenerative disorder presenting variable combinations of parkinsonism, cerebellar ataxia, corticospinal and autonomic dysfunction. Alpha-synuclein (α-SYN)-immunopositive glial cytoplasmic inclusions (GCIs) represent the neuropathological hallmark of MSA, and tubulin polymerization promoting protein (TPPP)/p25 in oligodendroglia has been known as a potent stimulator of α-SYN aggregation. To gain insight into the molecular pathomechanisms of GCI formation and subsequent oligodendroglial degeneration, we ectopically expressed α-SYN and TPPP in HEK293T and oligodendroglial KG1C cell lines. Here we showed that TPPP specifically accelerated α-SYN oligomer formation and co-immunoprecipitation analysis revealed the specific interaction of TPPP and α-SYN. Moreover, phosphorylation of α-SYN at Ser-129 facilitated the TPPP-mediated α-SYN oligomerization. TPPP facilitated α-SYN-positive cytoplasmic perinuclear inclusions mimicking GCI in both cell lines; however, apoptotic cell death was only observed in KG1C cells. This apoptotic cell death was partly rescued by sirtuin 2 (SIRT2) inhibition. Together, our results provide further insight into the molecular pathogenesis of MSA and potential therapeutic approaches.
Collapse
Affiliation(s)
- Takafumi Hasegawa
- Department of Neurology, Tohoku University School of Medicine, 1-1, Seiryomachi, Aobaku, Sendai, Miyagi 980-8574, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Papp-Lantos inclusions and the pathogenesis of multiple system atrophy: an update. Acta Neuropathol 2010; 119:657-67. [PMID: 20309568 DOI: 10.1007/s00401-010-0672-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 03/11/2010] [Accepted: 03/13/2010] [Indexed: 12/20/2022]
Abstract
Multiple systemic atrophy (MSA) is a progressive, adult-onset neurodegenerative disorder of undetermined aetiology characterized by a distinctive oligodendrogliopathy with argyrophilic glial cytoplasmic inclusions (GCIs) and selective neurodegeneration. GCIs or Papp-Lantos inclusions, described more than 20 years ago, are now accepted as the hallmarks for the definite neuropathological diagnosis of MSA and suggested to play a central role in the pathogenesis of this disorder. GCIs are composed of hyperphosphorylated alpha-synuclein (alphaSyn), ubiquitin, LRRK2 (leucin-rich repeat serine/threonine-protein) and many other proteins, suggesting that MSA represents an invariable synucleinopathy of non-neuronal type, a specific form of proteinopathies. The origin of alphaSyn deposition in GCIs is not yet fully understood, but recent findings of dysregulation in the metabolism of myelin basic protein (MBP) and p25alpha, a central nervous system-specific protein, also called TPPP (tubulin polymerization promoting protein), strengthened the working model of MSA as a primary glial disorder and may explain frequent alterations of myelin in MSA. However, it is unknown whether these changes represent an early event or myelin dysregulation occurs further downstream in MSA pathogenesis. The association between polymorphisms at the SNCA gene locus and the risk for developing MSA also points to a primary role of alphaSyn in its pathogenesis, while in a MBP promoter-driven alphaSyn transgenic mouse model gliosis accompanied the neurodegenerative process originating in oligodendrocytes. Because alphaSyn represents a major component in both oligodendroglial and neuronal inclusions in MSA, some authors suggested both a primary oligodendrogliopathy and a neuronal synucleinopathy, but current biomolecular data and animal models support a crucial role of the Papp-Lantos inclusions and of aberrant alphaSyn accumulation as their main constituent, causing oligodendroglial pathology, myelin disruption and, finally, neuronal degeneration in MSA. The relationship between oligodendrocytes involved by Papp-Lantos inclusions and those in degenerating neurons in the course of MSA needs further elucidation.
Collapse
|