1
|
Abraham JN, Rawat D, Srikanth P, Sunny LP, Abraham NM. Alpha-synuclein pathology and Parkinson's disease-related olfactory dysfunctions: an update on preclinical models and therapeutic approaches. Mamm Genome 2025:10.1007/s00335-025-10128-w. [PMID: 40293510 DOI: 10.1007/s00335-025-10128-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
Olfactory dysfunction (OD) is considered one of the early signs of Parkinson's disease (PD), affecting over 90% of PD patients. OD often appears several years before the onset of motor symptoms and is therefore considered an early biomarker of PD. Recent studies have shown that COVID-19 infection might lead to worsening of symptoms and acceleration of disease progression in neurodegenerative disorders, where OD is a common symptom to both. Hence, it is essential to accurately monitor olfactory fitness in clinical settings using any of the currently available olfactory function tests. Even after a quarter of a century of the discovery of α-synuclein (α-syn) pathogenesis in PD, many aspects related to the α-syn pathogenesis in OD remain unknown. Currently, there is no definitive cure for PD; the disease management options include dopaminergic medications, deep brain stimulations, stem cells, and immunotherapy. Generating reliable PD animal models is critical for understanding the molecular pathways and neural circuits affected by disease conditions. This might contribute to the development and validation of new therapeutic approaches. This review discusses the known mechanisms of α-syn aggregated forms causing neuronal death, the recent developments in the PD preclinical models with ODs, and the treatment strategies employed.
Collapse
Affiliation(s)
- Jancy Nixon Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India.
- Department of Life Sciences, Centre of Excellence in Epigenetics, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, 201314, India.
| | - Devesh Rawat
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Priyadharshini Srikanth
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Lisni P Sunny
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Nixon M Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India.
| |
Collapse
|
2
|
Pan H, Balbirnie M, Hou K, Sta Maria NS, Sahay S, Denver P, Lepore S, Jones M, Zuo X, Zhu C, Mirbaha H, Shahpasand-Kroner H, Mekkittikul M, Lu J, Hu CJ, Cheng X, Abskharon R, Sawaya MR, Williams CK, Vinters HV, Jacobs RE, Harris NG, Cole GM, Frautschy SA, Eisenberg DS. Liganded magnetic nanoparticles for magnetic resonance imaging of α-synuclein. NPJ Parkinsons Dis 2025; 11:88. [PMID: 40268938 PMCID: PMC12019173 DOI: 10.1038/s41531-025-00918-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/17/2025] [Indexed: 04/25/2025] Open
Abstract
Aggregation of the protein α-synuclein (α-syn) is the histopathological hallmark of neurodegenerative diseases such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), which are collectively known as synucleinopathies. Currently, patients with synucleinopathies are diagnosed by physical examination and medical history, often at advanced stages of disease. Because synucleinopathies are associated with α-syn aggregates, and α-syn aggregation often precedes onset of symptoms, detecting α-syn aggregates would be a valuable early diagnostic for patients with synucleinopathies. Here, we design a liganded magnetic nanoparticle (LMNP) functionalized with an α-syn-targeting peptide to be used as a magnetic resonance imaging (MRI)-based biomarker for α-syn. Our LMNPs bind to aggregates of α-syn in vitro, cross the blood-brain barrier in mice with mannitol adjuvant, and can be used as an MRI contrast agent to distinguish mice with α-synucleinopathy from age-matched, wild-type control mice in vivo. These results provide evidence for the potential of magnetic nanoparticles that target α-syn for diagnosis of synucleinopathies.
Collapse
Affiliation(s)
- Hope Pan
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Melinda Balbirnie
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Ke Hou
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Naomi S Sta Maria
- Department of Research Physiology, Department of Neuroscience, Keck School of Medicine at USC, Los Angeles, CA, USA
| | - Shruti Sahay
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Paul Denver
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, West Los Angeles VA Medical Center, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Stefano Lepore
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Mychica Jones
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, West Los Angeles VA Medical Center, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Xiaohong Zuo
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, West Los Angeles VA Medical Center, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Chunni Zhu
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Brain Research Institute Electron Microscopy Core Facility, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Hilda Mirbaha
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Hedieh Shahpasand-Kroner
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, West Los Angeles VA Medical Center, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Marisa Mekkittikul
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, West Los Angeles VA Medical Center, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jiahui Lu
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Carolyn J Hu
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Xinyi Cheng
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Romany Abskharon
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Michael R Sawaya
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Christopher K Williams
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Harry V Vinters
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Russell E Jacobs
- Department of Research Physiology, Department of Neuroscience, Keck School of Medicine at USC, Los Angeles, CA, USA
| | - Neil G Harris
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Gregory M Cole
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, West Los Angeles VA Medical Center, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sally A Frautschy
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, West Los Angeles VA Medical Center, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - David S Eisenberg
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Putnam GL, Maitta RW. Alpha synuclein and inflammaging. Heliyon 2025; 11:e41981. [PMID: 39897785 PMCID: PMC11786851 DOI: 10.1016/j.heliyon.2025.e41981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
The α-synuclein protein is an established molecule in Lewy body pathology, especially Parkinson's disease (PD). While the pathological role of α-synuclein (α-syn) in PD has been well described, novel evidence may suggest that α-syn interacts with inflammasomes in response to aging. As age is an inevitable physiological state and is also considered the greatest risk factor for PD, this calls for investigation into how α-syn, aging, and PD could be linked. There is a growing amount of data regarding α-syn normal function in the body that includes involvement in cellular transport such as protein complexes assembly, vesicular trafficking, neurotransmitter release, as well as immune cell maturation. Regarding abnormal α-syn, a number of autosomal dominant mutations have been identified as causes of familial PD, however, symptomatology may not become apparent until later in life due to compensatory mechanisms in the dopaminergic response. This potentially links age-related physiological changes not only as a risk factor for PD, but for the concept of "inflammaging ". This is defined as chronic inflammation that accompanies aging observed in many neurodegenerative pathologies, that include α-syn's ability to form oligomers and toxic fibrils seen in PD. This oligomeric α-syn stimulates pro-inflammatory signals, which may worsen PD symptoms and propagate chronic inflammation. Thus, this review will explore a potential link between α-syn's role in the immune system, inflammaging, and PD.
Collapse
Affiliation(s)
| | - Robert W. Maitta
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
4
|
Kim SM, Quagraine Y, Singh M, Kim JH. Rab11 suppresses neuronal stress signaling by localizing dual leucine zipper kinase to axon terminals for protein turnover. eLife 2024; 13:RP96592. [PMID: 39475475 PMCID: PMC11524585 DOI: 10.7554/elife.96592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Dual leucine zipper kinase (DLK) mediates multiple neuronal stress responses, and its expression levels are constantly suppressed to prevent excessive stress signaling. We found that Wallenda (Wnd), the Drosophila ortholog of DLK, is highly enriched in the axon terminals of Drosophila sensory neurons in vivo and that this subcellular localization is necessary for Highwire-mediated Wnd protein turnover under normal conditions. Our structure-function analysis found that Wnd palmitoylation is essential for its axon terminal localization. Palmitoylation-defective Wnd accumulated in neuronal cell bodies, exhibited dramatically increased protein expression levels, and triggered excessive neuronal stress responses. Defective intracellular transport is implicated in neurodegenerative conditions. Comprehensive dominant-negative Rab protein screening identified Rab11 as an essential factor for Wnd localization in axon terminals. Consequently, Rab11 loss-of-function increased the protein levels of Wnd and induced neuronal stress responses. Inhibiting Wnd activity significantly ameliorated neuronal loss and c-Jun N-terminal kinase signaling triggered by Rab11 loss-of-function. Taken together, these suggest that DLK proteins are constantly transported to axon terminals for protein turnover and a failure of such transport can lead to neuronal loss. Our study demonstrates how subcellular protein localization is coupled to protein turnover for neuronal stress signaling.
Collapse
Affiliation(s)
- Seung Mi Kim
- Department of Biology, University of Nevada RenoRenoUnited States
| | - Yaw Quagraine
- Department of Biology, University of Nevada RenoRenoUnited States
| | - Monika Singh
- Department of Biology, University of Nevada RenoRenoUnited States
| | - Jung Hwan Kim
- Department of Biology, University of Nevada RenoRenoUnited States
| |
Collapse
|
5
|
Kim S, Quagraine Y, Singh M, Kim JH. Rab11 suppresses neuronal stress signaling by localizing Dual leucine zipper kinase to axon terminals for protein turnover. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.18.537392. [PMID: 37131782 PMCID: PMC10153120 DOI: 10.1101/2023.04.18.537392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Dual Leucine Zipper Kinase (DLK) mediates multiple neuronal stress responses, and its expression levels are constantly suppressed to prevent excessive stress signaling. We found that Wallenda (Wnd), the Drosophila ortholog of DLK, is highly enriched in the axon terminals of Drosophila sensory neurons in vivo and that this subcellular localization is necessary for Highwire-mediated Wnd protein turnover under normal conditions. Our structure-function analysis found that Wnd palmitoylation is essential for its axon terminal localization. Palmitoylation-defective Wnd accumulated in neuronal cell bodies, exhibited dramatically increased protein expression levels, and triggered excessive neuronal stress responses. Defective intracellular transport is implicated in neurodegenerative conditions. Comprehensive dominant-negative Rab protein screening identified Rab11 as an essential factor for Wnd localization in axon terminals. Consequently, Rab11 loss-of-function increased the protein levels of Wnd and induced neuronal stress responses. Inhibiting Wnd activity significantly ameliorated neuronal loss and c-Jun N-terminal kinase signaling triggered by Rab11 loss-of-function. Taken together, these suggest that DLK proteins are constantly transported to axon terminals for protein turnover and a failure of such transport can lead to neuronal loss. Our study demonstrates how subcellular protein localization is coupled to protein turnover for neuronal stress signaling. Highlights Wnd is highly enriched in axon terminals.Wnd protein turnover by Hiw is restricted in the axon terminals.Protein palmitoylation of Wnd and Rab11 activity is essential for Wnd axonal localization. Rab11 mutations and defective Wnd palmitoylation impair Wnd protein turnover leading to increased Wnd protein levels and neuronal loss. Inhibiting Wnd activity mitigates neuronal stress response caused by Rab11 loss-of-function.
Collapse
|
6
|
Wolf D, Ayon-Olivas M, Sendtner M. BDNF-Regulated Modulation of Striatal Circuits and Implications for Parkinson's Disease and Dystonia. Biomedicines 2024; 12:1761. [PMID: 39200225 PMCID: PMC11351984 DOI: 10.3390/biomedicines12081761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Neurotrophins, particularly brain-derived neurotrophic factor (BDNF), act as key regulators of neuronal development, survival, and plasticity. BDNF is necessary for neuronal and functional maintenance in the striatum and the substantia nigra, both structures involved in the pathogenesis of Parkinson's Disease (PD). Depletion of BDNF leads to striatal degeneration and defects in the dendritic arborization of striatal neurons. Activation of tropomyosin receptor kinase B (TrkB) by BDNF is necessary for the induction of long-term potentiation (LTP), a form of synaptic plasticity, in the hippocampus and striatum. PD is characterized by the degeneration of nigrostriatal neurons and altered striatal plasticity has been implicated in the pathophysiology of PD motor symptoms, leading to imbalances in the basal ganglia motor pathways. Given its essential role in promoting neuronal survival and meditating synaptic plasticity in the motor system, BDNF might have an important impact on the pathophysiology of neurodegenerative diseases, such as PD. In this review, we focus on the role of BDNF in corticostriatal plasticity in movement disorders, including PD and dystonia. We discuss the mechanisms of how dopaminergic input modulates BDNF/TrkB signaling at corticostriatal synapses and the involvement of these mechanisms in neuronal function and synaptic plasticity. Evidence for alterations of BDNF and TrkB in PD patients and animal models are reviewed, and the potential of BDNF to act as a therapeutic agent is highlighted. Advancing our understanding of these mechanisms could pave the way toward innovative therapeutic strategies aiming at restoring neuroplasticity and enhancing motor function in these diseases.
Collapse
Affiliation(s)
| | | | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany (M.A.-O.)
| |
Collapse
|
7
|
Matarazzo M, Pérez-Soriano A, Vafai N, Shahinfard E, Cheng KJC, McKenzie J, Neilson N, Miao Q, Schaffer P, Shinotoh H, Kordower JH, Sossi V, Stoessl AJ. Misfolded protein deposits in Parkinson's disease and Parkinson's disease-related cognitive impairment, a [ 11C]PBB3 study. NPJ Parkinsons Dis 2024; 10:96. [PMID: 38702305 PMCID: PMC11068893 DOI: 10.1038/s41531-024-00708-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/11/2024] [Indexed: 05/06/2024] Open
Abstract
Parkinson's disease (PD) is associated with aggregation of misfolded α-synuclein and other proteins, including tau. We designed a cross-sectional study to quantify the brain binding of [11C]PBB3 (a ligand known to bind to misfolded tau and possibly α-synuclein) as a proxy of misfolded protein aggregation in Parkinson's disease (PD) subjects with and without cognitive impairment and healthy controls (HC). In this cross-sectional study, nineteen cognitively normal PD subjects (CN-PD), thirteen cognitively impaired PD subjects (CI-PD) and ten HC underwent [11C]PBB3 PET. A subset of the PD subjects also underwent PET imaging with [11C](+)DTBZ to assess dopaminergic denervation and [11C]PBR28 to assess neuroinflammation. Compared to HC, PD subjects showed higher [11C]PBB3 binding in the posterior putamen but not the substantia nigra. There was no relationship across subjects between [11C]PBB3 and [11C]PBR28 binding in nigrostriatal regions. [11C]PBB3 binding was increased in the anterior cingulate in CI-PD compared to CN-PD and HC, and there was an inverse correlation between cognitive scores and [11C]PBB3 binding in this region across all PD subjects. Our results support a primary role of abnormal protein deposition localized to the posterior putamen in PD. This suggests that striatal axonal terminals are preferentially involved in the pathophysiology of PD. Furthermore, our findings suggest that anterior cingulate pathology might represent a significant in vivo marker of cognitive impairment in PD, in agreement with previous neuropathological studies.
Collapse
Affiliation(s)
- Michele Matarazzo
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Alexandra Pérez-Soriano
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada
| | - Nasim Vafai
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Elham Shahinfard
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Kevin Ju-Chieh Cheng
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Jessamyn McKenzie
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada
| | - Nicole Neilson
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada
| | | | | | - Hitoshi Shinotoh
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, Chiba, Japan
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - A Jon Stoessl
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada.
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
8
|
Koyanagi Y, Kassai M, Yoneyama H. The Impact of Intestinal Microbiota and Toll-like Receptor 2 Signaling on α-Synuclein Pathology in Nontransgenic Mice Injected with α-Synuclein Preformed Fibrils. Microorganisms 2024; 12:106. [PMID: 38257933 PMCID: PMC10818728 DOI: 10.3390/microorganisms12010106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Intestinal microbiota and Toll-like receptor 2 (TLR2), which can bind lipoteichoic acid produced by microbiota, might contribute to the pathogenesis of Parkinson's disease (PD), which is characterized by α-synuclein accumulation. Although the contribution of intestinal microbiota and TLR2 to PD pathology was validated in genetic PD models, evidence suggests that the effects of TLR2 signaling on proteinopathy might depend on the presence of a genetic etiology. We examined the impact of intestinal microbiota and TLR2 signaling on α-synuclein pathology in a nontransgenic mouse model of sporadic PD. While an α-synuclein preformed fibrils injection successfully reproduced PD pathology by inducing accumulation of α-synuclein aggregates, microglial activation and increased TLR2 expression in the brains of nontransgenic mice, antibiotic-induced reduction in the density of intestinal microbiota and TLR2 knockout had small impact on these changes. These findings, which are in contrast to those reported in transgenic mice harboring transgene encoding α-synuclein, indicate that the contribution of intestinal microbiota and TLR2 signaling to α-synuclein pathogenesis might be influenced by the presence of a genetic etiology. Additionally, these findings suggest that integrating insights from this experimental model and genetic models would further advance our understanding of the molecular mechanisms underlying sporadic PD.
Collapse
Affiliation(s)
- Yukako Koyanagi
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan;
- Sumitomo Pharma Co., Ltd., Osaka 554-0022, Japan
| | - Momoe Kassai
- Sumitomo Pharma Co., Ltd., Osaka 554-0022, Japan
| | - Hiroshi Yoneyama
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan;
| |
Collapse
|
9
|
Abaza A, Jamil A, Gutlapalli SD, Ali M, Oble MJP, Sonia SN, George S, Shahi SR, Ali Z, Khan S. Parkinson's Neuropathology Puzzle: A Systematic Review Uncovering the Pathological Culprits Behind the Neurological Disease. Cureus 2023; 15:e44353. [PMID: 37664277 PMCID: PMC10472082 DOI: 10.7759/cureus.44353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023] Open
Abstract
Being one of the most prevalent progressive neurodegenerative disorders (falling second only to Alzheimer's disease) with a clinical pattern affecting millions of lives all over the world, Parkinson's disease (PD) has never failed to attract a formidable interest from the vast majority of neurologists and researchers worldwide. This review article will analyze the pathophysiology, etiology, genetics, and pathological stages of Parkinson's disease with their corresponding clinical sequels. A review article was conducted using research databases including PubMed, PubMed Central, Springer, and Elsevier. The research articles reviewed using databases were written in English, German, Japanese, and Chinese and published within the preceding 50 years. Based on the article's findings, we concluded that Parkinson's disease is a progressive disorder with a variety of motor and non-motor symptoms that are influenced by a cascade of pathological neuronal abnormalities such as Lewy neurites and Lewy bodies that gradually build up with an eventual consequence of neurodegeneration of dopamine-secreting neurons. Multiple genetic mutations, pathophysiological events, and environmental factors act as the foundation to initiate that cascade.
Collapse
Affiliation(s)
- Abdelrahman Abaza
- Pathology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Aneeque Jamil
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Sai Dheeraj Gutlapalli
- Internal Medicine, Richmond University Medical Center Affiliated With Mount Sinai Health System and Icahn School of Medicine at Mount Sinai, Staten Island, USA
- Internal Medicine Clinical Research, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Marya Ali
- Psychiatry, Nishtar Medical University, Multan, PAK
| | - Mrinal J P Oble
- Medicine, Kempegowda Institute of Medical Sciences and Research Centre, Bengaluru, IND
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Shamsun Nahar Sonia
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | | | - Srushti R Shahi
- Medicine, St. Martinus University Faculty of Medicine (SMUFOM), Willemstad, CUW
| | - Zahra Ali
- Medicine, Bolan Medical College, Quetta, PAK
- Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
10
|
Kee TR, Wehinger JL, Gonzalez PE, Nguyen E, McGill Percy KC, Khan SA, Chaput D, Wang X, Liu T, Kang DE, Woo JAA. Pathological characterization of a novel mouse model expressing the PD-linked CHCHD2-T61I mutation. Hum Mol Genet 2022; 31:3987-4005. [PMID: 35786718 PMCID: PMC9703812 DOI: 10.1093/hmg/ddac083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) is a mitochondrial protein that plays important roles in cristae structure, oxidative phosphorylation and apoptosis. Multiple mutations in CHCHD2 have been associated with Lewy body disorders (LBDs), such as Parkinson's disease (PD) and dementia with Lewy bodies, with the CHCHD2-T61I mutation being the most widely studied. However, at present, only CHCHD2 knockout or CHCHD2/CHCHD10 double knockout mouse models have been investigated. They do not recapitulate the pathology seen in patients with CHCHD2 mutations. We generated the first transgenic mouse model expressing the human PD-linked CHCHD2-T61I mutation driven by the mPrP promoter. We show that CHCHD2-T61I Tg mice exhibit perinuclear mitochondrial aggregates, neuroinflammation, and have impaired long-term synaptic plasticity associated with synaptic dysfunction. Dopaminergic neurodegeneration, a hallmark of PD, is also observed along with α-synuclein pathology. Significant motor dysfunction is seen with no changes in learning and memory at 1 year of age. A minor proportion of the CHCHD2-T61I Tg mice (~10%) show a severe motor phenotype consistent with human Pisa Syndrome, an atypical PD phenotype. Unbiased proteomics analysis reveals surprising increases in many insoluble proteins predominantly originating from mitochondria and perturbing multiple canonical biological pathways as assessed by ingenuity pathway analysis, including neurodegenerative disease-associated proteins such as tau, cofilin, SOD1 and DJ-1. Overall, CHCHD2-T61I Tg mice exhibit pathological and motor changes associated with LBDs, indicating that this model successfully captures phenotypes seen in human LBD patients with CHCHD2 mutations and demonstrates changes in neurodegenerative disease-associated proteins, which delineates relevant pathological pathways for further investigation.
Collapse
Affiliation(s)
- Teresa R Kee
- Department of Pathology, CWRU School of Medicine, Cleveland, OH 44106, USA
- Department of Molecular of Medicine, USF Health College of Medicine, Tampa, FL 33613, USA
| | - Jessica L Wehinger
- Department of Molecular of Medicine, USF Health College of Medicine, Tampa, FL 33613, USA
| | | | - Eric Nguyen
- Department of Molecular of Medicine, USF Health College of Medicine, Tampa, FL 33613, USA
| | | | - Sophia A Khan
- Department of Pathology, CWRU School of Medicine, Cleveland, OH 44106, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Xinming Wang
- Department of Pathology, CWRU School of Medicine, Cleveland, OH 44106, USA
| | - Tian Liu
- Department of Pathology, CWRU School of Medicine, Cleveland, OH 44106, USA
| | - David E Kang
- Department of Pathology, CWRU School of Medicine, Cleveland, OH 44106, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Jung-A A Woo
- Department of Pathology, CWRU School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
11
|
Chen ML, Wu RM. Homozygous mutation of the LRRK2 ROC domain as a novel genetic model of parkinsonism. J Biomed Sci 2022; 29:60. [PMID: 35965315 PMCID: PMC9375908 DOI: 10.1186/s12929-022-00844-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
Background Parkinson’s disease (PD) is one of the most important neurodegenerative disorders in elderly people. Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are found in a large proportion of the patients with sporadic and familial PD. Mutations can occur at different locations in the LRRK2. Patients with LRRK2 ROC-COR mutations face an increased risk of typical motor symptoms of PD, along with cognitive decline. An animal model with a monogenic LRRK2 gene mutation is a suitable model for exploring the pathophysiology of PD and identifying potential drug therapies. However, the effect of homozygous (HOM) LRRK2 in PD pathophysiology is unclear. Methods We established human LRRK2 (hLRRK2) R1441G HOM transgenic (Tg) mice to explore the phenotype and pathological features that are associated with hLRRK2 R1441G Tg mouse models and discuss the potential clinical relevance. The open field test (OFT) was performed to examine motor and nonmotor behaviors. A CatWalk analysis system was used to study gait function. [18F]FDOPA PET was used to investigate functional changes in the nigrostriatal pathway in vivo. Transmission electron microscopy was used to examine the morphological changes in mitochondria and lysosomes in the substantia nigra. Results The R1441G HOM Tg mice demonstrated gait disturbance and exhibited less anxiety-related behavior and exploratory behavior than mice with hLRRK2 at 12 months old. Additionally, [18F]FDOPA PET showed a reduction in FDOPA uptake in the striatum of the HOM Tg mice. Notably, there was significant lysosome and autophagosome accumulation in the cytoplasm of dopaminergic neurons in R1441G hemizygous (HEM) and HOM mice. Moreover, it was observed using transmission electron microscopy (TEM) that the mitochondria of R1441G Tg mice were smaller than those of hLRRK2 mice. Conclusion This animal provides a novel HOM hLRRK2 R1441G Tg mouse model that reproduces some phenotype of Parkinsonism in terms of both motor and behavioral dysfunction. There is an increased level of mitochondrial fission and no change in the fusion process in the group of HOM hLRRK2 R1441G Tg mouse. This mutant animal model of PD might be used to study the mechanisms of mitochondrial dysfunction and explore potential new drug targets. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-022-00844-9.
Collapse
|
12
|
Zamponi E, Meehl JB, Voeltz GK. The ER ladder is a unique morphological feature of developing mammalian axons. Dev Cell 2022; 57:1369-1382.e6. [PMID: 35609616 DOI: 10.1016/j.devcel.2022.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/28/2022] [Accepted: 05/02/2022] [Indexed: 11/25/2022]
Abstract
The endoplasmic reticulum (ER) confronts a challenge to accommodate long, smooth ER tubules into the structural complexity of the axonal compartment. Here, we describe a morphological feature for the axonal ER network in developing neurons we termed the ER ladder. Axonal ER ladders are composed of rungs that wrap tightly around the microtubule bundle and dynamic rails, which slide across microtubules. We found that the ER-shaping protein Reticulon 2 determines the architecture and dynamics of the axonal ER ladder by modulating its interaction with microtubules. Moreover, we show that ER ladder depletion impairs the trafficking of associated vesicular axonal cargoes. Finally, we demonstrate that stromal interaction molecule 1 (Stim1) localizes to ER rungs and translocates to ER-plasma membrane contact sites upon depletion of luminal Ca2+. Our findings uncover fundamental insights into the structural and functional organization of the axonal ER network in developing mammalian neurons.
Collapse
Affiliation(s)
- Emiliano Zamponi
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Boulder, CO 80309, USA
| | - Janet B Meehl
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Boulder, CO 80309, USA
| | - Gia K Voeltz
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Boulder, CO 80309, USA.
| |
Collapse
|
13
|
Kuznetsov IA, Kuznetsov AV. Bidirectional, unlike unidirectional transport, allows transporting axonal cargos against their concentration gradient. J Theor Biol 2022; 546:111161. [DOI: 10.1016/j.jtbi.2022.111161] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/25/2022]
|
14
|
Kuznetsov IA, Kuznetsov AV. Can the lack of fibrillar form of alpha-synuclein in Lewy bodies be explained by its catalytic activity? Math Biosci 2022; 344:108754. [PMID: 34890628 PMCID: PMC8882444 DOI: 10.1016/j.mbs.2021.108754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/03/2023]
Abstract
Finding the causative pathophysiological mechanisms for Parkinson's disease (PD) is important for developing therapeutic interventions. Until recently, it was believed that Lewy bodies (LBs), the hallmark of PD, are mostly composed of alpha-synuclein (α-syn) fibrils. Recent results (Shahmoradian et al. (2019)) demonstrated that the fibrillar form of α-syn is lacking from LBs. Here we propose that this surprising observation can be explained by the catalytic activity of the fibrillar form of α-syn. We assumed that α-syn fibrils catalyze the formation of LBs, but do not become part of them. We developed a mathematical model based on this hypothesis. By using the developed model, we investigated the consequences of this hypothesis. In particular, the model suggests that the long incubation time of PD can be explained by a two-step aggregation process that leads to its development: (i) aggregation of monomeric α-syn into α-syn oligomers and fibrils and (ii) clustering of membrane-bound organelles, which may cause disruption of axonal trafficking and lead to neuron starvation and death. The model shows that decreasing the rate of destruction of α-syn aggregates in somatic lysosomes accelerates the formation of LBs. Another consequence of the model is the prediction that removing α-syn aggregates from the brain after the aggregation of membrane-bound organelles into LBs has started may not stop the progression of PD because LB formation is an autocatalytic process; hence, the formation of LBs will be catalyzed by aggregates of membrane-bound organelles even in the absence of α-syn aggregates. The performed sensitivity study made it possible to establish the hierarchy of model parameters with respect to their effect on the formation of vesicle aggregates in the soma.
Collapse
Affiliation(s)
- Ivan A Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA.
| |
Collapse
|
15
|
Ramirez AE, Fernández-Pérez EJ, Olivos N, Burgos CF, Boopathi S, Armijo-Weingart L, Pacheco CR, González W, Aguayo LG. The Stimulatory Effects of Intracellular α-Synuclein on Synaptic Transmission Are Attenuated by 2-Octahydroisoquinolin-2(1H)-ylethanamine. Int J Mol Sci 2021; 22:ijms222413253. [PMID: 34948050 PMCID: PMC8705949 DOI: 10.3390/ijms222413253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022] Open
Abstract
α-Synuclein (αSyn) species can be detected in synaptic boutons, where they play a crucial role in the pathogenesis of Parkinson's Disease (PD). However, the effects of intracellular αSyn species on synaptic transmission have not been thoroughly studied. Here, using patch-clamp recordings in hippocampal neurons, we report that αSyn oligomers (αSynO), intracellularly delivered through the patch electrode, produced a fast and potent effect on synaptic transmission, causing a substantial increase in the frequency, amplitude and transferred charge of spontaneous synaptic currents. We also found an increase in the frequency of miniature synaptic currents, suggesting an effect located at the presynaptic site of the synapsis. Furthermore, our in silico approximation using docking analysis and molecular dynamics simulations showed an interaction between a previously described small anti-amyloid beta (Aβ) molecule, termed M30 (2-octahydroisoquinolin-2(1H)-ylethanamine), with a central hydrophobic region of αSyn. In line with this finding, our empirical data aimed to obtain oligomerization states with thioflavin T (ThT) and Western blot (WB) indicated that M30 interfered with αSyn aggregation and decreased the formation of higher-molecular-weight species. Furthermore, the effect of αSynO on synaptic physiology was also antagonized by M30, resulting in a decrease in the frequency, amplitude, and charge transferred of synaptic currents. Overall, the present results show an excitatory effect of intracellular αSyn low molecular-weight species, not previously described, that are able to affect synaptic transmission, and the potential of a small neuroactive molecule to interfere with the aggregation process and the synaptic effect of αSyn, suggesting that M30 could be a potential therapeutic strategy for synucleinopathies.
Collapse
Affiliation(s)
- Alejandra E. Ramirez
- Laboratory of Neurophysiology, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, 160-C, Concepción 4030000, Chile; (A.E.R.); (N.O.); (C.F.B.); (L.A.-W.); (C.R.P.)
| | - Eduardo J. Fernández-Pérez
- Laboratory of Neurophysiology, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, 160-C, Concepción 4030000, Chile; (A.E.R.); (N.O.); (C.F.B.); (L.A.-W.); (C.R.P.)
- Correspondence: (E.J.F.-P.); (L.G.A.)
| | - Nicol Olivos
- Laboratory of Neurophysiology, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, 160-C, Concepción 4030000, Chile; (A.E.R.); (N.O.); (C.F.B.); (L.A.-W.); (C.R.P.)
| | - Carlos F. Burgos
- Laboratory of Neurophysiology, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, 160-C, Concepción 4030000, Chile; (A.E.R.); (N.O.); (C.F.B.); (L.A.-W.); (C.R.P.)
| | - Subramanian Boopathi
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico;
- Center for Bioinformatics, Simulations and Modeling, The Center for Bioinformatics and Molecular Simulations (CBSM), University of Talca, Talca 3530000, Chile;
| | - Lorena Armijo-Weingart
- Laboratory of Neurophysiology, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, 160-C, Concepción 4030000, Chile; (A.E.R.); (N.O.); (C.F.B.); (L.A.-W.); (C.R.P.)
| | - Carla R. Pacheco
- Laboratory of Neurophysiology, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, 160-C, Concepción 4030000, Chile; (A.E.R.); (N.O.); (C.F.B.); (L.A.-W.); (C.R.P.)
| | - Wendy González
- Center for Bioinformatics, Simulations and Modeling, The Center for Bioinformatics and Molecular Simulations (CBSM), University of Talca, Talca 3530000, Chile;
- Millennium Nucleus of Ion Channels-Associated Diseases, The Center for Bioinformatics and Molecular Simulations (CBSM), University of Talca, Talca 3530000, Chile
| | - Luis G. Aguayo
- Laboratory of Neurophysiology, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, 160-C, Concepción 4030000, Chile; (A.E.R.); (N.O.); (C.F.B.); (L.A.-W.); (C.R.P.)
- Programa de Neurociencia, Psiquiatría y Salud Mental, Anatomy Building,
Faculty of Medicine, Universidad de Concepción, Concepción 4030000, Chile
- Correspondence: (E.J.F.-P.); (L.G.A.)
| |
Collapse
|
16
|
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
|
17
|
Vernazza S, Oddone F, Tirendi S, Bassi AM. Risk Factors for Retinal Ganglion Cell Distress in Glaucoma and Neuroprotective Potential Intervention. Int J Mol Sci 2021; 22:7994. [PMID: 34360760 PMCID: PMC8346985 DOI: 10.3390/ijms22157994] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a population of neurons of the central nervous system (CNS) extending with their soma to the inner retina and with their axons to the optic nerve. Glaucoma represents a group of neurodegenerative diseases where the slow progressive death of RGCs results in a permanent loss of vision. To date, although Intra Ocular Pressure (IOP) is considered the main therapeutic target, the precise mechanisms by which RGCs die in glaucoma have not yet been clarified. In fact, Primary Open Angle Glaucoma (POAG), which is the most common glaucoma form, also occurs without elevated IOP. This present review provides a summary of some pathological conditions, i.e., axonal transport blockade, glutamate excitotoxicity and changes in pro-inflammatory cytokines along the RGC projection, all involved in the glaucoma cascade. Moreover, neuro-protective therapeutic approaches, which aim to improve RGC degeneration, have also been taken into consideration.
Collapse
Affiliation(s)
- Stefania Vernazza
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
| | | | - Sara Tirendi
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Anna Maria Bassi
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| |
Collapse
|
18
|
Mavroeidi P, Xilouri M. Neurons and Glia Interplay in α-Synucleinopathies. Int J Mol Sci 2021; 22:4994. [PMID: 34066733 PMCID: PMC8125822 DOI: 10.3390/ijms22094994] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of the neuronal presynaptic protein alpha-synuclein within proteinaceous inclusions represents the key histophathological hallmark of a spectrum of neurodegenerative disorders, referred to by the umbrella term a-synucleinopathies. Even though alpha-synuclein is expressed predominantly in neurons, pathological aggregates of the protein are also found in the glial cells of the brain. In Parkinson's disease and dementia with Lewy bodies, alpha-synuclein accumulates mainly in neurons forming the Lewy bodies and Lewy neurites, whereas in multiple system atrophy, the protein aggregates mostly in the glial cytoplasmic inclusions within oligodendrocytes. In addition, astrogliosis and microgliosis are found in the synucleinopathy brains, whereas both astrocytes and microglia internalize alpha-synuclein and contribute to the spread of pathology. The mechanisms underlying the pathological accumulation of alpha-synuclein in glial cells that under physiological conditions express low to non-detectable levels of the protein are an area of intense research. Undoubtedly, the presence of aggregated alpha-synuclein can disrupt glial function in general and can contribute to neurodegeneration through numerous pathways. Herein, we summarize the current knowledge on the role of alpha-synuclein in both neurons and glia, highlighting the contribution of the neuron-glia connectome in the disease initiation and progression, which may represent potential therapeutic target for a-synucleinopathies.
Collapse
Affiliation(s)
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
19
|
Spijkers XM, Pasteuning-Vuhman S, Dorleijn JC, Vulto P, Wevers NR, Pasterkamp RJ. A directional 3D neurite outgrowth model for studying motor axon biology and disease. Sci Rep 2021; 11:2080. [PMID: 33483540 PMCID: PMC7822896 DOI: 10.1038/s41598-021-81335-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/04/2021] [Indexed: 01/30/2023] Open
Abstract
We report a method to generate a 3D motor neuron model with segregated and directed axonal outgrowth. iPSC-derived motor neurons are cultured in extracellular matrix gel in a microfluidic platform. Neurons extend their axons into an adjacent layer of gel, whereas dendrites and soma remain predominantly in the somal compartment, as verified by immunofluorescent staining. Axonal outgrowth could be precisely quantified and was shown to respond to the chemotherapeutic drug vincristine in a highly reproducible dose-dependent manner. The model was shown susceptible to excitotoxicity upon exposure with excess glutamate and showed formation of stress granules upon excess glutamate or sodium arsenite exposure, mimicking processes common in motor neuron diseases. Importantly, outgrowing axons could be attracted and repelled through a gradient of axonal guidance cues, such as semaphorins. The platform comprises 40 chips arranged underneath a microtiter plate providing both throughput and compatibility to standard laboratory equipment. The model will thus prove ideal for studying axonal biology and disease, drug discovery and regenerative medicine.
Collapse
Affiliation(s)
- Xandor M. Spijkers
- grid.474144.6MIMETAS BV, Organ-On-a-Chip Company, 2333 CH Leiden, The Netherlands ,grid.5477.10000000120346234Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Svetlana Pasteuning-Vuhman
- grid.5477.10000000120346234Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Jennifa C. Dorleijn
- grid.474144.6MIMETAS BV, Organ-On-a-Chip Company, 2333 CH Leiden, The Netherlands
| | - Paul Vulto
- grid.474144.6MIMETAS BV, Organ-On-a-Chip Company, 2333 CH Leiden, The Netherlands
| | - Nienke R. Wevers
- grid.474144.6MIMETAS BV, Organ-On-a-Chip Company, 2333 CH Leiden, The Netherlands ,grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - R. Jeroen Pasterkamp
- grid.5477.10000000120346234Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
20
|
Pang C, Zhang N, Falahati M. Acceleration of α-synuclein fibril formation and associated cytotoxicity stimulated by silica nanoparticles as a model of neurodegenerative diseases. Int J Biol Macromol 2020; 169:532-540. [PMID: 33352154 DOI: 10.1016/j.ijbiomac.2020.12.130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
A wide range of biophysical and theoretical analysis were employed to explore the formation of (α-syn) amyloid fibril formation as a model of Parkinson's disease in the presence of silica oxide nanoparticles (SiO2 NPs). Also, different cellular and molecular assays such as MTT, LDH, caspase, ROS, and qPCR were performed to reveal the α-syn amyloid fibrils-associated cytotoxicity against SH-SY5Y cells. Fluorescence measurements showed that SiO2 NPs accelerate the α-syn aggregation and exposure of hydrophobic moieties. Congo red absorbance, circular dichroism (CD), and transmission electron microscopy (TEM) analysis depicted the SiO2 NPs accelerated the formation of α-syn amyloid fibrils. Molecular docking study showed that SiO2 clusters preferably bind to the N-terminal of α-syn as the helix folding site. We also realized that SiO2 NPs increase the cytotoxicity of α-syn amyloid fibrils through a significant decrease in cell viability, increase in membrane leakage, activation of caspase-9 and -3, elevation of ROS, and increase in the ratio of Bax/Bcl2 mRNA. The cellular assay indicated that α-syn amyloid fibrils formed in the presence of SiO2 NPs induce their cytotoxic effects through the mitochondrial-mediated intrinsic apoptosis pathway. We concluded that these data may reveal some adverse effects of NPs on the progression of Parkinson's disease.
Collapse
Affiliation(s)
- Chao Pang
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shengyang 110000, China.
| | - Na Zhang
- Medical Education Research Center, Shenyang Medical College, Shenyang 110000, China
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
21
|
Tran HT, Tsai EHR, Lewis AJ, Moors T, Bol JGJM, Rostami I, Diaz A, Jonker AJ, Guizar-Sicairos M, Raabe J, Stahlberg H, van de Berg WDJ, Holler M, Shahmoradian SH. Alterations in Sub-Axonal Architecture Between Normal Aging and Parkinson's Diseased Human Brains Using Label-Free Cryogenic X-ray Nanotomography. Front Neurosci 2020; 14:570019. [PMID: 33324142 PMCID: PMC7724048 DOI: 10.3389/fnins.2020.570019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/14/2020] [Indexed: 01/25/2023] Open
Abstract
Gaining insight to pathologically relevant processes in continuous volumes of unstained brain tissue is important for a better understanding of neurological diseases. Many pathological processes in neurodegenerative disorders affect myelinated axons, which are a critical part of the neuronal circuitry. Cryo ptychographic X-ray computed tomography in the multi-keV energy range is an emerging technology providing phase contrast at high sensitivity, allowing label-free and non-destructive three dimensional imaging of large continuous volumes of tissue, currently spanning up to 400,000 μm3. This aspect makes the technique especially attractive for imaging complex biological material, especially neuronal tissues, in combination with downstream optical or electron microscopy techniques. A further advantage is that dehydration, additional contrast staining, and destructive sectioning/milling are not required for imaging. We have developed a pipeline for cryo ptychographic X-ray tomography of relatively large, hydrated and unstained biological tissue volumes beyond what is typical for the X-ray imaging, using human brain tissue and combining the technique with complementary methods. We present four imaged volumes of a Parkinson's diseased human brain and five volumes from a non-diseased control human brain using cryo ptychographic X-ray tomography. In both cases, we distinguish neuromelanin-containing neurons, lipid and melanic pigment, blood vessels and red blood cells, and nuclei of other brain cells. In the diseased sample, we observed several swellings containing dense granular material resembling clustered vesicles between the myelin sheaths arising from the cytoplasm of the parent oligodendrocyte, rather than the axoplasm. We further investigated the pathological relevance of such swollen axons in adjacent tissue sections by immunofluorescence microscopy for phosphorylated alpha-synuclein combined with multispectral imaging. Since cryo ptychographic X-ray tomography is non-destructive, the large dataset volumes were used to guide further investigation of such swollen axons by correlative electron microscopy and immunogold labeling post X-ray imaging, a possibility demonstrated for the first time. Interestingly, we find that protein antigenicity and ultrastructure of the tissue are preserved after the X-ray measurement. As many pathological processes in neurodegeneration affect myelinated axons, our work sets an unprecedented foundation for studies addressing axonal integrity and disease-related changes in unstained brain tissues.
Collapse
Affiliation(s)
| | | | - Amanda J. Lewis
- Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, University of Basel, Basel, Switzerland
| | - Tim Moors
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - J. G. J. M. Bol
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Ana Diaz
- Paul Scherrer Institut, Villigen, Switzerland
| | - Allert J. Jonker
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Joerg Raabe
- Paul Scherrer Institut, Villigen, Switzerland
| | - Henning Stahlberg
- Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, University of Basel, Basel, Switzerland
| | - Wilma D. J. van de Berg
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | | |
Collapse
|
22
|
Zorgniotti A, Ditamo Y, Arce CA, Bisig CG. Irreversible incorporation of L-dopa into the C-terminus of α-tubulin inhibits binding of molecular motor KIF5B to microtubules and alters mitochondrial traffic along the axon. Neurobiol Dis 2020; 147:105164. [PMID: 33171229 DOI: 10.1016/j.nbd.2020.105164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
L-dopa is the most effective drug used to date for management of Parkinson's disease symptoms. Unfortunately, long-term administration of L-dopa often results in development of motor disorders, including dyskinesias. Despite extensive research on L-dopa-induced dyskinesia, its pathogenesis remains poorly understood. We demonstrated previously that L-dopa can be post-translationally incorporated into the C-terminus of α-tubulin in living cells. In the present study, we investigated the effect of the presence of L-dopa-tubulin-enriched microtubules on mitochondrial traffic mediated by molecular motor KIF5B. Using biochemical approaches in combination with experiments on neuronal cell lines and mouse hippocampal primary cultures, we demonstrated that L-dopa incorporation into tubulin is irreversible. Transport of mitochondria along the axon was altered after L-dopa treatment of cells. In L-dopa-treated cells, mitochondria had reduced ability to reach the distal segment of the axon, spent more time in pause, and showed reduced velocity of anterograde movement. KIF5B motor, a member of the kinesin family involved in mitochondrial transport in neurons, showed reduced affinity for Dopa-tubulin-containing microtubules. Our findings, taken together, suggest that tyrosination state of tubulin (and microtubules) is altered by L-dopa incorporation into tubulin; the gradual increase in amount of altered microtubules affects microtubule functioning, impairs mitochondrial traffic and distribution, and this could be relevant in Parkinson's disease patients chronically treated with L-dopa.
Collapse
Affiliation(s)
- Agustina Zorgniotti
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina
| | - Yanina Ditamo
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina
| | - Carlos A Arce
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina
| | - C Gaston Bisig
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina.
| |
Collapse
|
23
|
Zhang Y, Burock MA. Diffusion Tensor Imaging in Parkinson's Disease and Parkinsonian Syndrome: A Systematic Review. Front Neurol 2020; 11:531993. [PMID: 33101169 PMCID: PMC7546271 DOI: 10.3389/fneur.2020.531993] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/18/2020] [Indexed: 12/21/2022] Open
Abstract
Diffusion tensor imaging (DTI) allows measuring fractional anisotropy and similar microstructural indices of the brain white matter. Lower than normal fractional anisotropy as well as higher than normal diffusivity is associated with loss of microstructural integrity and neurodegeneration. Previous DTI studies in Parkinson's disease (PD) have demonstrated abnormal fractional anisotropy in multiple white matter regions, particularly in the dopaminergic nuclei and dopaminergic pathways. However, DTI is not considered a diagnostic marker for the earliest Parkinson's disease since anisotropic alterations present a temporally divergent pattern during the earliest Parkinson's course. This article reviews a majority of clinically employed DTI studies in PD, and it aims to prove the utilities of DTI as a marker of diagnosing PD, correlating clinical symptomatology, tracking disease progression, and treatment effects. To address the challenge of DTI being a diagnostic marker for early PD, this article also provides a comparison of the results from a longitudinal, early stage, multicenter clinical cohort of Parkinson's research with previous publications. This review provides evidences of DTI as a promising marker for monitoring PD progression and classifying atypical PD types, and it also interprets the possible pathophysiologic processes under the complex pattern of fractional anisotropic changes in the first few years of PD. Recent technical advantages, limitations, and further research strategies of clinical DTI in PD are additionally discussed.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Psychiatry, War Related Illness and Injury Study Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Marc A Burock
- Department of Psychiatry, Mainline Health, Bryn Mawr Hospital, Bryn Mawr, PA, United States
| |
Collapse
|
24
|
Zampese E, Surmeier DJ. Calcium, Bioenergetics, and Parkinson's Disease. Cells 2020; 9:cells9092045. [PMID: 32911641 PMCID: PMC7564460 DOI: 10.3390/cells9092045] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Degeneration of substantia nigra (SN) dopaminergic (DAergic) neurons is responsible for the core motor deficits of Parkinson’s disease (PD). These neurons are autonomous pacemakers that have large cytosolic Ca2+ oscillations that have been linked to basal mitochondrial oxidant stress and turnover. This review explores the origin of Ca2+ oscillations and their role in the control of mitochondrial respiration, bioenergetics, and mitochondrial oxidant stress.
Collapse
|
25
|
Zhou H, Li S, Li C, Yang X, Li H, Zhong H, Lu JH, Lee SMY. Oxyphylla A Promotes Degradation of α-Synuclein for Neuroprotection via Activation of Immunoproteasome. Aging Dis 2020; 11:559-574. [PMID: 32489702 PMCID: PMC7220298 DOI: 10.14336/ad.2019.0612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/12/2019] [Indexed: 11/23/2022] Open
Abstract
Parkinson’s disease (PD), the second most common neurodegenerative disorder, is neuropathologically characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) and the presence of Lewy bodies in surviving neurons. α-synuclein (α-syn) is the major component of Lewy bodies and its deposition in neurons is critical pathological event in the pathogenesis of PD. Herein, we reported that Oxyphylla A, a novel lead compound from the fruit of Alpinia oxyphylla, significantly promoted α-syn degradation in a cellular PD model. When exploring the molecular pathways, we found that Oxyphylla A promoted α-syn degradation in a ubiquitin proteasome system (UPS)-dependent and autophagy-independent manner. We further confirmed that Oxyphylla A enhanced UPS activity by upregulating 20S subunit PSMB8 expression. A mechanism study revealed that Oxyphylla A activated the PKA/Akt/mTOR pathway to trigger PSMB8 expression and enhance UPS activity. Finally, we illustrated that Oxyphylla A alleviated the accumulation of both Triton-soluble and Triton-insoluble forms of α-syn and protected against α-syn-induced neurotoxicity in A53T α-syn transgenic mice. These findings suggest that the activation of UPS, via small molecular UPS enhancers including Oxyphylla A, may be a therapeutic strategy for intervention against PD and related diseases.
Collapse
Affiliation(s)
- Hefeng Zhou
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Shengnan Li
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Chuwen Li
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xuanjun Yang
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China.,2Department of Biology, South University of Science and Technology, Shenzhen, China
| | - Haitao Li
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hanbing Zhong
- 2Department of Biology, South University of Science and Technology, Shenzhen, China
| | - Jia-Hong Lu
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Simon Ming-Yuen Lee
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
26
|
Salvadores N, Court FA. The necroptosis pathway and its role in age-related neurodegenerative diseases: will it open up new therapeutic avenues in the next decade? Expert Opin Ther Targets 2020; 24:679-693. [PMID: 32310729 DOI: 10.1080/14728222.2020.1758668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Necroptosis is a programmed form of necrotic cell death. Growing evidence demonstrates that necroptosis contributes to cell demise in different pathological conditions including age-dependent neurodegenerative diseases (NDs). These findings open new avenues for understanding the mechanisms of neuronal loss in NDs, which might eventually translate into novel therapeutic interventions. AREAS COVERED We reviewed key aspects of necroptosis, in health and disease, focusing on evidence demonstrating its involvement in the pathogenesis of age-related NDs. We then highlight the activation of this pathway in the mechanism of axonal degeneration. We searched on PubMed the literature regarding necroptosis published between 2008 and 2020 and reviewed all publications were necroptosis was studied in the context of age-related NDs. EXPERT OPINION Axonal loss and neuronal death are the ultimate consequences of NDs that translate into disease phenotypes. Targeting degenerative mechanisms of the neuron appears as a strategy that might cover a wide range of diseases. Thus, the participation of necroptosis as a common mediator of neuronal demise emerges as a promising target for therapeutic intervention. Considering evidence demonstrating that necroptosis mediates axonal degeneration, we propose and discuss the potential of targeting necroptosis-mediated axonal destruction as a strategy to tackle NDs before neuronal loss occurs.
Collapse
Affiliation(s)
- Natalia Salvadores
- Faculty of Sciences, Center for Integrative Biology, Universidad Mayor , Santiago, Chile.,Fondap Geroscience Center for Brain Health and Metabolism , Santiago, Chile
| | - Felipe A Court
- Faculty of Sciences, Center for Integrative Biology, Universidad Mayor , Santiago, Chile.,Fondap Geroscience Center for Brain Health and Metabolism , Santiago, Chile
| |
Collapse
|
27
|
Anderson EN, Hirpa D, Zheng KH, Banerjee R, Gunawardena S. The Non-amyloidal Component Region of α-Synuclein Is Important for α-Synuclein Transport Within Axons. Front Cell Neurosci 2020; 13:540. [PMID: 32038170 PMCID: PMC6984405 DOI: 10.3389/fncel.2019.00540] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/21/2019] [Indexed: 11/13/2022] Open
Abstract
Proper transport of the Parkinson's disease (PD) protein, α-synuclein (α-syn), is thought to be crucial for its localization and function at the synapse. Previous work has shown that defects in long distance transport within narrow caliber axons occur early in PD, but how such defects contribute to PD is unknown. Here we test the hypothesis that the NAC region is involved in facilitating proper transport of α-syn within axons via its association with membranes. Excess α-syn or fPD mutant α-synA53T accumulates within larval axons perturbing the transport of synaptic proteins. These α-syn expressing larvae also show synaptic morphological and larval locomotion defects, which correlate with the extent of α-syn-mediated axonal accumulations. Strikingly, deletion of the NAC region (α-synΔ71-82) prevented α-syn accumulations and axonal blockages, and reduced its synaptic localization due to decreased axonal entry and axonal transport of α-syn, due to less α-syn bound to membranes. Intriguingly, co-expression α-synΔ71-82 with full-length α-syn rescued α-syn accumulations and synaptic morphological defects, and decreased the ratio of the insoluble higher molecular weight (HMW)/soluble low molecular weight (LMW) α-syn, indicating that this region is perhaps important for the dimerization of α-syn on membranes. Together, our observations suggest that under physiological conditions, α-syn associates with membranes via the NAC region, and that too much α-syn perturbs axonal transport via aggregate formation, instigating synaptic and behavioral defects seen in PD.
Collapse
Affiliation(s)
| | | | | | | | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
28
|
Mezias C, Rey N, Brundin P, Raj A. Neural connectivity predicts spreading of alpha-synuclein pathology in fibril-injected mouse models: Involvement of retrograde and anterograde axonal propagation. Neurobiol Dis 2019; 134:104623. [PMID: 31628991 PMCID: PMC7138530 DOI: 10.1016/j.nbd.2019.104623] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/05/2019] [Accepted: 09/23/2019] [Indexed: 12/18/2022] Open
Abstract
In Parkinson’s disease, some of the first alpha-synuclein aggregates appear in the olfactory system and the dorsal motor nucleus of the vagus nerve before spreading to connected brain regions. We previously demonstrated that injection of alpha-synuclein fibrils unilaterally into the olfactory bulb of wild type mice leads to widespread synucleinopathy in brain regions directly and indirectly connected to the injection site, consistently, over the course of periods longer than 6 months. Our previously reported observations support the idea that alpha-synuclein inclusions propagates between brain region through neuronal networks. In the present study, we further defined the pattern of propagation of alpha-synuclein inclusions and developed a mathematical model based on known mouse brain connectivity. Using this model, we first predicted the pattern of alpha-synuclein inclusions propagation following an injection of fibrils into the olfactory bulb. We then analyzed the fitting of these predictions to our published histological data. Our results demonstrate that the pattern of propagation we observed in vivo is consistent with axonal transport of alpha-synuclein aggregate seeds, followed by transsynaptic transmission. By contrast, simple diffusion of alpha-synuclein fits very poorly our in vivo data. We also found that the spread of alpha-synuclein inclusions appeared to primarily follow neural connections retrogradely until 9 months after injection into the olfactory bulb. Thereafter, the pattern of spreading was consistent with anterograde propagation mathematical models. Finally, we applied our mathematical model to a different, previously published, dataset involving alpha-synuclein fibril injections into the striatum, instead of the olfactory bulb. We found that the mathematical model accurately predicts the reported progressive increase in alpha-synuclein neuropathology also in that paradigm. In conclusion, our findings support that the progressive spread of alpha-synuclein inclusions after injection of protein fibrils follows neural networks in the mouse connectome.
Collapse
Affiliation(s)
- Christopher Mezias
- Weill Cornell Medicine of Cornell University, Department of Neuroscience, New York, NY 10065, USA; Weill Cornell Medicine of Cornell University, Department of Radiology, New York, NY 10065, USA.
| | - Nolwen Rey
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ashish Raj
- Weill Cornell Medicine of Cornell University, Department of Radiology, New York, NY 10065, USA; University of California-San Francisco, Department of Biomedical Imaging, USA
| |
Collapse
|
29
|
Hassani OK, Rymar VV, Nguyen KQ, Huo L, Cloutier JF, Miller FD, Sadikot AF. The noradrenergic system is necessary for survival of vulnerable midbrain dopaminergic neurons: implications for development and Parkinson's disease. Neurobiol Aging 2019; 85:22-37. [PMID: 31734438 DOI: 10.1016/j.neurobiolaging.2019.09.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/22/2022]
Abstract
The cause of midbrain dopaminergic (mDA) neuron loss in sporadic Parkinson's disease (PD) is multifactorial, involving cell autonomous factors, cell-cell interactions, and the effects of environmental toxins. Early loss of neurons in the locus coeruleus (LC), the main source of ascending noradrenergic (NA) projections, is an important feature of PD and other neurodegenerative disorders. We hypothesized that NA afferents provide trophic support for vulnerable mDA neurons. We demonstrate that depriving mDA neurons of NA input increases postnatal apoptosis and decreases cell survival in young adult rodents, with relative sparing of calbindin-positive subpopulations known to be resistant to degeneration in PD. As a mechanism, we propose that the neurotrophin brain-derived neurotrophic factor (BDNF) modulates anterograde survival effects of LC inputs to mDA neurons. We demonstrate that the LC is rich in BDNF mRNA in postnatal and young adult brains. Early postnatal NA denervation reduces both BDNF protein and activation of TrkB receptors in the ventral midbrain. Furthermore, overexpression of BDNF in NA afferents in transgenic mice increases mDA neuronal survival. Finally, increasing NA activity in primary cultures of mDA neurons improves survival, an effect that is additive or synergistic in the presence of different concentrations of BDNF. Taken together, our results point to a novel mechanism whereby LC afferents couple BDNF effects and NA activity to provide anterograde trophic support for vulnerable mDA neurons. Early loss of NA activity and anterograde neurotrophin support may contribute to degeneration of vulnerable neurons in PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Oum Kaltoum Hassani
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Vladimir V Rymar
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Khanh Q Nguyen
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Lia Huo
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jean-François Cloutier
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Freda D Miller
- Departments of Medical Genetics, Microbiology & Physiology, The Hospital for Sick Children Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Abbas F Sadikot
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
30
|
Esteves AR, Palma AM, Gomes R, Santos D, Silva DF, Cardoso SM. Acetylation as a major determinant to microtubule-dependent autophagy: Relevance to Alzheimer's and Parkinson disease pathology. Biochim Biophys Acta Mol Basis Dis 2018; 1865:2008-2023. [PMID: 30572013 DOI: 10.1016/j.bbadis.2018.11.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/29/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
Abstract
Protein post-translational modifications (PTMs) that potentiate protein aggregation have been implicated in several neurological disorders, including Alzheimer's (AD) and Parkinson's disease (PD). In fact, Tau and alpha-synuclein (ASYN) undergo several PTMs potentiating their aggregation and neurotoxicity. Recent data posits a role for acetylation in Tau and ASYN aggregation. Herein we aimed to clarify the role of Sirtuin-2 (SIRT2) and HDAC6 tubulin deacetylases as well as p300 acetyltransferase in AD and PD neurodegeneration. We used transmitochondrial cybrids that recapitulate pathogenic alterations observed in sporadic PD and AD patient brains and ASYN and Tau cellular models. We confirmed that Tau protein and ASYN are microtubules (MTs)-associated proteins (MAPs). Moreover, our results suggest that α-tubulin acetylation induced by SIRT2 inhibition is functionally associated with the improvement of MT dynamic determined by decreased Tau phosphorylation and by increased Tau/tubulin and ASYN/tubulin binding. Our data provide a strong evidence for a functional role of tubulin and MAPs acetylation on autophagic vesicular traffic and cargo clearance. Additionally, we showed that an accumulation of ASYN oligomers imbalance mitochondrial dynamics, which further compromise autophagy. We also demonstrated that an increase in Tau acetylation is associated with Tau phosphorylation. We found that p300, HDAC6 and SIRT2 influences Tau phosphorylation and autophagic flux in AD. In addition, we demonstrated that p300 and HDAC6 modulate Tau and Tubulin acetylation. Overall, our data disclose the role of Tau and ASYN modifications through acetylation in AD and PD pathology, respectively. Moreover, this study indicates that MTs can be a promising therapeutic target in the field of neurodegenerative disorders in which intracellular transport is altered.
Collapse
Affiliation(s)
- A R Esteves
- CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Coimbra, Portugal.
| | - A M Palma
- CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Coimbra, Portugal
| | - R Gomes
- CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Coimbra, Portugal
| | - D Santos
- CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Coimbra, Portugal
| | - D F Silva
- CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Coimbra, Portugal
| | - S M Cardoso
- CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Coimbra, Portugal; Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
31
|
Saifetiarova J, Bhat MA. Ablation of cytoskeletal scaffolding proteins, Band 4.1B and Whirlin, leads to cerebellar purkinje axon pathology and motor dysfunction. J Neurosci Res 2018; 97:313-331. [PMID: 30447021 DOI: 10.1002/jnr.24352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/23/2022]
Abstract
The cerebellar cortex receives neural information from other brain regions to allow fine motor coordination and motor learning. The primary output neurons from the cerebellum are the Purkinje neurons that transmit inhibitory responses to deep cerebellar nuclei through their myelinated axons. Altered morphological organization and electrical properties of the Purkinje axons lead to detrimental changes in locomotor activity often leading to cerebellar ataxias. Two cytoskeletal scaffolding proteins Band 4.1B (4.1B) and Whirlin (Whrn) have been previously shown to play independent roles in axonal domain organization and maintenance in myelinated axons in the spinal cord and sciatic nerves. Immunoblot analysis had indicated cerebellar expression for both 4.1B and Whrn; however, their subcellular localization and cerebellum-specific functions have not been characterized. Using 4.1B and Whrn single and double mutant animals, we show that both proteins are expressed in common cellular compartments of the cerebellum and play cooperative roles in preservation of the integrity of Purkinje neuron myelinated axons. We demonstrate that both 4.1B and Whrn are required for the maintenance of axonal ultrastructure and health. Loss of 4.1B and Whrn leads to axonal transport defects manifested by formation of swellings containing cytoskeletal components, membranous organelles, and vesicles. Moreover, ablation of both proteins progressively affects cerebellar function with impairment in locomotor performance detected by altered gait parameters. Together, our data indicate that 4.1B and Whrn are required for maintaining proper axonal cytoskeletal organization and axonal domains, which is necessary for cerebellum-controlled fine motor coordination.
Collapse
Affiliation(s)
- Julia Saifetiarova
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas
| |
Collapse
|
32
|
Shah SH, Goldberg JL. The Role of Axon Transport in Neuroprotection and Regeneration. Dev Neurobiol 2018; 78:998-1010. [PMID: 30027690 DOI: 10.1002/dneu.22630] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Retinal ganglion cells and other central nervous system neurons fail to regenerate after injury. Understanding the obstacles to survival and regeneration, and overcoming them, is key to preserving and restoring function. While comparisons in the cellular changes seen in these non-regenerative cells with those that do have intrinsic regenerative ability has yielded many candidate genes for regenerative therapies, complete visual recovery has not yet been achieved. Insights gained from neurodegenerative diseases, like glaucoma, underscore the importance of axonal transport of organelles, mRNA, and effector proteins in injury and disease. Targeting molecular motor networks, and their cargoes, may be necessary for realizing complete axonal regeneration and vision restoration.
Collapse
Affiliation(s)
- Sahil H Shah
- Byers Eye Institute, Stanford University, Palo Alto, California.,Neurosciences Graduate Program, University of California, San Diego, California.,Medical Scientist Training Program, University of California, San Diego, California
| | | |
Collapse
|
33
|
Liao EH, Gray L, Tsurudome K, El-Mounzer W, Elazzouzi F, Baim C, Farzin S, Calderon MR, Kauwe G, Haghighi AP. Kinesin Khc-73/KIF13B modulates retrograde BMP signaling by influencing endosomal dynamics at the Drosophila neuromuscular junction. PLoS Genet 2018; 14:e1007184. [PMID: 29373576 PMCID: PMC5802963 DOI: 10.1371/journal.pgen.1007184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 02/07/2018] [Accepted: 01/03/2018] [Indexed: 11/18/2022] Open
Abstract
Retrograde signaling is essential for neuronal growth, function and survival; however, we know little about how signaling endosomes might be directed from synaptic terminals onto retrograde axonal pathways. We have identified Khc-73, a plus-end directed microtubule motor protein, as a regulator of sorting of endosomes in Drosophila larval motor neurons. The number of synaptic boutons and the amount of neurotransmitter release at the Khc-73 mutant larval neuromuscular junction (NMJ) are normal, but we find a significant decrease in the number of presynaptic release sites. This defect in Khc-73 mutant larvae can be genetically enhanced by a partial genetic loss of Bone Morphogenic Protein (BMP) signaling or suppressed by activation of BMP signaling in motoneurons. Consistently, activation of BMP signaling that normally enhances the accumulation of phosphorylated form of BMP transcription factor Mad in the nuclei, can be suppressed by genetic removal of Khc-73. Using a number of assays including live imaging in larval motor neurons, we show that loss of Khc-73 curbs the ability of retrograde-bound endosomes to leave the synaptic area and join the retrograde axonal pathway. Our findings identify Khc-73 as a regulator of endosomal traffic at the synapse and modulator of retrograde BMP signaling in motoneurons.
Collapse
Affiliation(s)
- Edward H. Liao
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - Lindsay Gray
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - Kazuya Tsurudome
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | | | - Fatima Elazzouzi
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Christopher Baim
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Sarah Farzin
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Mario R. Calderon
- Buck Institute for Research on Aging, Novato, CA, United States of America
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - A. Pejmun Haghighi
- Buck Institute for Research on Aging, Novato, CA, United States of America
- Department of Physiology, McGill University, Montreal, QC, Canada
- * E-mail:
| |
Collapse
|
34
|
Terron A, Bal-Price A, Paini A, Monnet-Tschudi F, Bennekou SH, Leist M, Schildknecht S. An adverse outcome pathway for parkinsonian motor deficits associated with mitochondrial complex I inhibition. Arch Toxicol 2018; 92:41-82. [PMID: 29209747 PMCID: PMC5773657 DOI: 10.1007/s00204-017-2133-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022]
Abstract
Epidemiological studies have observed an association between pesticide exposure and the development of Parkinson's disease, but have not established causality. The concept of an adverse outcome pathway (AOP) has been developed as a framework for the organization of available information linking the modulation of a molecular target [molecular initiating event (MIE)], via a sequence of essential biological key events (KEs), with an adverse outcome (AO). Here, we present an AOP covering the toxicological pathways that link the binding of an inhibitor to mitochondrial complex I (i.e., the MIE) with the onset of parkinsonian motor deficits (i.e., the AO). This AOP was developed according to the Organisation for Economic Co-operation and Development guidelines and uploaded to the AOP database. The KEs linking complex I inhibition to parkinsonian motor deficits are mitochondrial dysfunction, impaired proteostasis, neuroinflammation, and the degeneration of dopaminergic neurons of the substantia nigra. These KEs, by convention, were linearly organized. However, there was also evidence of additional feed-forward connections and shortcuts between the KEs, possibly depending on the intensity of the insult and the model system applied. The present AOP demonstrates mechanistic plausibility for epidemiological observations on a relationship between pesticide exposure and an elevated risk for Parkinson's disease development.
Collapse
Affiliation(s)
| | | | - Alicia Paini
- European Commission Joint Research Centre, Ispra, Italy
| | | | | | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstr. 10, PO Box M657, 78457, Konstanz, Germany
| | - Stefan Schildknecht
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstr. 10, PO Box M657, 78457, Konstanz, Germany.
| |
Collapse
|
35
|
Lai MC, Bechy AL, Denk F, Collins E, Gavriliouk M, Zaugg JB, Ryan BJ, Wade-Martins R, Caffrey TM. Haplotype-specific MAPT exon 3 expression regulated by common intronic polymorphisms associated with Parkinsonian disorders. Mol Neurodegener 2017; 12:79. [PMID: 29084565 PMCID: PMC5663040 DOI: 10.1186/s13024-017-0224-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/23/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genome wide association studies have identified microtubule associated protein tau (MAPT) H1 haplotype single nucleotide polymorphisms (SNPs) as leading common risk variants for Parkinson's disease, progressive supranuclear palsy and corticobasal degeneration. The MAPT risk variants fall within a large 1.8 Mb region of high linkage disequilibrium, making it difficult to discern the functionally important risk variants. Here, we leverage the strong haplotype-specific expression of MAPT exon 3 to investigate the functionality of SNPs that fall within this H1 haplotype region of linkage disequilibrium. METHODS In this study, we dissect the molecular mechanisms by which haplotype-specific SNPs confer allele-specific effects on the alternative splicing of MAPT exon 3. Firstly, we use haplotype-hybrid whole-locus genomic MAPT vectors studies to identify functional SNPs. Next, we characterise the RNA-protein interactions at two loci by mass spectrometry. Lastly, we knockdown candidate splice factors to determine their effect on MAPT exon 3 using a novel allele-specific qPCR assay. RESULTS Using whole-locus genomic DNA expression vectors to express MAPT haplotype variants, we demonstrate that rs17651213 regulates exon 3 inclusion in a haplotype-specific manner. We further investigated the functionality of this region using RNA-electrophoretic mobility shift assays to show differential RNA-protein complex formation at the H1 and H2 sequence variants of SNP rs17651213 and rs1800547 and subsequently identified candidate trans-acting splicing factors interacting with these functional SNPs sequences by RNA-protein pull-down experiment and mass spectrometry. Finally, gene knockdown of candidate splice factors identified by mass spectrometry demonstrate a role for hnRNP F and hnRNP Q in the haplotype-specific regulation of exon 3 inclusion. CONCLUSIONS We identified common splice factors hnRNP F and hnRNP Q regulating the haplotype-specific splicing of MAPT exon 3 through intronic variants rs1800547 and rs17651213. This work demonstrates an integrated approach to characterise the functionality of risk variants in large regions of linkage disequilibrium.
Collapse
Affiliation(s)
- Mang Ching Lai
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Anne-Laure Bechy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
| | - Franziska Denk
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
| | - Emma Collins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
| | - Maria Gavriliouk
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
| | - Judith B. Zaugg
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Brent J. Ryan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, OX1 3QX UK
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, OX1 3QX UK
| | - Tara M. Caffrey
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX UK
| |
Collapse
|
36
|
Alpha Lipoamide Ameliorates Motor Deficits and Mitochondrial Dynamics in the Parkinson's Disease Model Induced by 6-Hydroxydopamine. Neurotox Res 2017; 33:759-767. [PMID: 29019159 DOI: 10.1007/s12640-017-9819-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 12/18/2022]
Abstract
The precise mechanisms underlying neuronal injury in Parkinson's disease (PD) are not yet fully elucidated; however, evidence from the in vitro and in vivo PD models suggest that mitochondrial dysfunction may play a major role in PD pathogenesis. Alpha lipoamide, a neutral amide derivative of the lipoic acid, is a better cofactor for mitochondrial dehydrogenase with a stronger protective effect on mitochondria than lipoic acid. Identification of these protective effects of alpha lipoamide on mitochondria, together with the evidence that mitochondrial dysfunction plays a critical role in PD, we speculate that alpha lipoamide may exert a protective effect in PD by regulating the mitochondrial function. The present study investigated the neuroprotective effects of alpha lipoamide in an animal model of PD induced by 6-hydroxydopamine (6-OHDA). The results demonstrated that alpha lipoamide could significantly antagonize the 6-OHDA-induced behavioral damages; restore ATP levels in the midbrain; and also improve the fragmentation, vacuolization, and morphology of the mitochondria. The results of Western blot indicated that alpha lipoamide significantly restored the number of dopaminergic neurons in midbrain and substantially recovered the balance between mitochondrial fission, fusion, and transport. In conclusion, the results demonstrated that alpha lipoamide might exert a significant neuroprotective effect in the animal model of PD by regulation of the dynamic properties of mitochondria.
Collapse
|
37
|
Salvadores N, Sanhueza M, Manque P, Court FA. Axonal Degeneration during Aging and Its Functional Role in Neurodegenerative Disorders. Front Neurosci 2017; 11:451. [PMID: 28928628 PMCID: PMC5591337 DOI: 10.3389/fnins.2017.00451] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/25/2017] [Indexed: 12/11/2022] Open
Abstract
Aging constitutes the main risk factor for the development of neurodegenerative diseases. This represents a major health issue worldwide that is only expected to escalate due to the ever-increasing life expectancy of the population. Interestingly, axonal degeneration, which occurs at early stages of neurodegenerative disorders (ND) such as Alzheimer's disease, Amyotrophic lateral sclerosis, and Parkinson's disease, also takes place as a consequence of normal aging. Moreover, the alteration of several cellular processes such as proteostasis, response to cellular stress and mitochondrial homeostasis, which have been described to occur in the aging brain, can also contribute to axonal pathology. Compelling evidence indicate that the degeneration of axons precedes clinical symptoms in NDs and occurs before cell body loss, constituting an early event in the pathological process and providing a potential therapeutic target to treat neurodegeneration before neuronal cell death. Although, normal aging and the development of neurodegeneration are two processes that are closely linked, the molecular basis of the switch that triggers the transition from healthy aging to neurodegeneration remains unrevealed. In this review we discuss the potential role of axonal degeneration in this transition and provide a detailed overview of the literature and current advances in the molecular understanding of the cellular changes that occur during aging that promote axonal degeneration and then discuss this in the context of ND.
Collapse
Affiliation(s)
- Natalia Salvadores
- Center for Integrative Biology, Faculty of Sciences, Universidad MayorSantiago, Chile.,Fondap Geroscience Center for Brain Health and MetabolismSantiago, Chile
| | - Mario Sanhueza
- Center for Integrative Biology, Faculty of Sciences, Universidad MayorSantiago, Chile.,Fondap Geroscience Center for Brain Health and MetabolismSantiago, Chile
| | - Patricio Manque
- Center for Integrative Biology, Faculty of Sciences, Universidad MayorSantiago, Chile
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad MayorSantiago, Chile.,Fondap Geroscience Center for Brain Health and MetabolismSantiago, Chile
| |
Collapse
|
38
|
Pozo Devoto VM, Dimopoulos N, Alloatti M, Pardi MB, Saez TM, Otero MG, Cromberg LE, Marín-Burgin A, Scassa ME, Stokin GB, Schinder AF, Sevlever G, Falzone TL. αSynuclein control of mitochondrial homeostasis in human-derived neurons is disrupted by mutations associated with Parkinson's disease. Sci Rep 2017; 7:5042. [PMID: 28698628 PMCID: PMC5506004 DOI: 10.1038/s41598-017-05334-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/07/2017] [Indexed: 01/10/2023] Open
Abstract
The etiology of Parkinson’s disease (PD) converges on a common pathogenic pathway of mitochondrial defects in which α-Synuclein (αSyn) is thought to play a role. However, the mechanisms by which αSyn and its disease-associated allelic variants cause mitochondrial dysfunction remain unknown. Here, we analyzed mitochondrial axonal transport and morphology in human-derived neurons overexpressing wild-type (WT) αSyn or the mutated variants A30P or A53T, which are known to have differential lipid affinities. A53T αSyn was enriched in mitochondrial fractions, inducing significant mitochondrial transport defects and fragmentation, while milder defects were elicited by WT and A30P. We found that αSyn-mediated mitochondrial fragmentation was linked to expression levels in WT and A53T variants. Targeted delivery of WT and A53T αSyn to the outer mitochondrial membrane further increased fragmentation, whereas A30P did not. Genomic editing to disrupt the N-terminal domain of αSyn, which is important for membrane association, resulted in mitochondrial elongation without changes in fusion-fission protein levels, suggesting that αSyn plays a direct physiological role in mitochondrial size maintenance. Thus, we demonstrate that the association of αSyn with the mitochondria, which is modulated by protein mutation and dosage, influences mitochondrial transport and morphology, highlighting its relevance in a common pathway impaired in PD.
Collapse
Affiliation(s)
- Victorio Martin Pozo Devoto
- Instituto de Biología Celular y Neurociencias, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires, CP1121, Argentina.,International Clinical Research Center (ICRC), St. Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | - Nicolas Dimopoulos
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Montañeses 2325, Buenos Aires, C1428AQK, Argentina
| | - Matías Alloatti
- Instituto de Biología Celular y Neurociencias, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires, CP1121, Argentina
| | - María Belén Pardi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) -CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Trinidad M Saez
- Instituto de Biología Celular y Neurociencias, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires, CP1121, Argentina.,Instituto de Biología y Medicina Experimental, IBYME (CONICET). Vuelta de obligado 2490, Buenos Aires, CP, 1428, Argentina
| | - María Gabriela Otero
- Instituto de Biología Celular y Neurociencias, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires, CP1121, Argentina
| | - Lucas Eneas Cromberg
- Instituto de Biología Celular y Neurociencias, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires, CP1121, Argentina
| | - Antonia Marín-Burgin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) -CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Maria Elida Scassa
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Montañeses 2325, Buenos Aires, C1428AQK, Argentina
| | - Gorazd B Stokin
- International Clinical Research Center (ICRC), St. Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | - Alejandro F Schinder
- Laboratorio de Plasticidad Neuronal, Fundación Instituto Leloir (IIBBA - CONICET), Av. Patricias Argentinas 435, Buenos Aires, CP C1405BWE, Argentina
| | - Gustavo Sevlever
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Montañeses 2325, Buenos Aires, C1428AQK, Argentina
| | - Tomás Luis Falzone
- Instituto de Biología Celular y Neurociencias, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires, CP1121, Argentina. .,Instituto de Biología y Medicina Experimental, IBYME (CONICET). Vuelta de obligado 2490, Buenos Aires, CP, 1428, Argentina.
| |
Collapse
|
39
|
Paillusson S, Gomez-Suaga P, Stoica R, Little D, Gissen P, Devine MJ, Noble W, Hanger DP, Miller CCJ. α-Synuclein binds to the ER-mitochondria tethering protein VAPB to disrupt Ca 2+ homeostasis and mitochondrial ATP production. Acta Neuropathol 2017; 134:129-149. [PMID: 28337542 PMCID: PMC5486644 DOI: 10.1007/s00401-017-1704-z] [Citation(s) in RCA: 288] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 03/17/2017] [Accepted: 03/18/2017] [Indexed: 12/29/2022]
Abstract
α-Synuclein is strongly linked to Parkinson’s disease but the molecular targets for its toxicity are not fully clear. However, many neuronal functions damaged in Parkinson’s disease are regulated by signalling between the endoplasmic reticulum (ER) and mitochondria. This signalling involves close physical associations between the two organelles that are mediated by binding of the integral ER protein vesicle-associated membrane protein-associated protein B (VAPB) to the outer mitochondrial membrane protein, protein tyrosine phosphatase-interacting protein 51 (PTPIP51). VAPB and PTPIP51 thus act as a scaffold to tether the two organelles. Here we show that α-synuclein binds to VAPB and that overexpression of wild-type and familial Parkinson’s disease mutant α-synuclein disrupt the VAPB-PTPIP51 tethers to loosen ER–mitochondria associations. This disruption to the VAPB-PTPIP51 tethers is also seen in neurons derived from induced pluripotent stem cells from familial Parkinson’s disease patients harbouring pathogenic triplication of the α-synuclein gene. We also show that the α-synuclein induced loosening of ER–mitochondria contacts is accompanied by disruption to Ca2+ exchange between the two organelles and mitochondrial ATP production. Such disruptions are likely to be particularly damaging to neurons that are heavily dependent on correct Ca2+ signaling and ATP.
Collapse
Affiliation(s)
- Sébastien Paillusson
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe road, London, SE5 9RX, UK
| | - Patricia Gomez-Suaga
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe road, London, SE5 9RX, UK
| | - Radu Stoica
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe road, London, SE5 9RX, UK
| | - Daniel Little
- MRC Laboratory of Molecular Cell Biology, University College London, London, UK
| | - Paul Gissen
- MRC Laboratory of Molecular Cell Biology, University College London, London, UK
| | - Michael J Devine
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Wendy Noble
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe road, London, SE5 9RX, UK
| | - Diane P Hanger
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe road, London, SE5 9RX, UK
| | - Christopher C J Miller
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe road, London, SE5 9RX, UK.
| |
Collapse
|
40
|
Bal-Price A, Meek MEB. Adverse outcome pathways: Application to enhance mechanistic understanding of neurotoxicity. Pharmacol Ther 2017; 179:84-95. [PMID: 28529068 PMCID: PMC5869951 DOI: 10.1016/j.pharmthera.2017.05.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent developments have prompted the transition of empirically based testing of late stage toxicity in animals for a range of different endpoints including neurotoxicity to more efficient and predictive mechanistically based approaches with greater emphasis on measurable key events early in the progression of disease. The adverse outcome pathway (AOP) has been proposed as a simplified organizational construct to contribute to this transition by linking molecular initiating events and earlier (more predictive) key events at lower levels of biological organization to disease outcomes. As such, AOPs are anticipated to facilitate the compilation of information to increase mechanistic understanding of pathophysiological pathways that are responsible for human disease. In this review, the sequence of key events resulting in adverse outcome (AO) defined as parkinsonian motor impairment and learning and memory deficit in children, triggered by exposure to environmental chemicals has been briefly described using the AOP framework. These AOPs follow convention adopted in an Organization for Economic Cooperation and Development (OECD) AOP development program, publically available, to permit tailored application of AOPs for a range of different purposes. Due to the complexity of disease pathways, including neurodegenerative disorders, a specific symptom of the disease (e.g. parkinsonian motor deficit) is considered as the AO in a developed AOP. Though the description is necessarily limited by the extent of current knowledge, additional characterization of involved pathways through description of related AOPs interlinked into networks for the same disease has potential to contribute to more holistic and mechanistic understanding of the pathophysiological pathways involved, possibly leading to the mechanism-based reclassification of diseases, thus facilitating more personalized treatment.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission Joint Research Centre, Directorate F - Health, Consumers and Reference Materials, Ispra, Italy.
| | - M E Bette Meek
- McLaughlin Centre for Risk Science, University of Ottawa, Ottawa, Canada
| |
Collapse
|
41
|
Tagliaferro P, Burke RE. Retrograde Axonal Degeneration in Parkinson Disease. JOURNAL OF PARKINSONS DISEASE 2017; 6:1-15. [PMID: 27003783 PMCID: PMC4927911 DOI: 10.3233/jpd-150769] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In spite of tremendous research efforts we have not yet achieved two of our principal therapeutic goals in the treatment of Parkinson’s disease (PD), to prevent its onward progression and to provide restoration of systems that have already been damaged by the time of diagnosis. There are many possible reasons for our inability to make progress. One possibility is that our efforts thus far may not have been directed towards the appropriate cellular compartments. Up until now research has been largely focused on the loss of neurons in the disease. Thus, neuroprotection approaches have been largely aimed at blocking mechanisms that lead to destruction of the neuronal cell body. Attempts to provide neurorestoration have been almost entirely focused on replacement of neurons. We herein review the evidence that the axonal component of diseased neuronal systems merit more of our attention. Evidence from imaging studies, from postmortem neurochemical studies, and from genetic animal models suggests that the axons of the dopaminergic system are involved predominantly and early in PD. Since the mechanisms of axonal destruction are distinct from those of neuron cell body degeneration, a focus on axonal neurobiology will offer new opportunities for preventing their degeneration. At present these mechanisms remain largely obscure. However, defining them is likely to offer new opportunities for neuroprotection. In relation to neurorestoration, while it has been classically believed that neurons of the adult central nervous system are incapable of new axon growth, recent evidence shows that this is not true for the dopaminergic projection. In conclusion, the neurobiology of axons is likely to offer many new approaches to protective and restorative therapeutics.
Collapse
Affiliation(s)
| | - Robert E Burke
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.,Departments of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
42
|
Braak H, Del Tredici K. Neuropathological Staging of Brain Pathology in Sporadic Parkinson's disease: Separating the Wheat from the Chaff. JOURNAL OF PARKINSON'S DISEASE 2017; 7:S71-S85. [PMID: 28282810 PMCID: PMC5345633 DOI: 10.3233/jpd-179001] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A relatively small number of especially susceptible nerve cell types within multiple neurotransmitter systems of the human central, peripheral, and enteric nervous systems (CNS, PNS, ENS) become involved in the degenerative process underlying sporadic Parkinson's disease (sPD). The six-stage model we proposed for brain pathology related to sPD (Neurobiol Aging 2003) was a retrospective study of incidental and clinically diagnosed cases performed on unconventionally thick tissue sections (100 μm) from a large number of brain regions.The staging model emphasized what we perceived to be a sequential development of increasing degrees of Lewy pathology in anatomically interconnected regions together with the loss of aminergic projection neurons in, but not limited to, the locus coeruleus and substantia nigra. The same weight was assigned to axonal and somatodendritic Lewy pathology, and the olfactory bulb was included for the first time in a sPD staging system. After years of research, it now appears that the earliest lesions could develop at nonnigral (dopamine agonist nonresponsive) sites, where the surrounding environment is potentially hostile: the olfactory bulb and, possibly, the ENS. The current lack of knowledge regarding the development of Lewy pathology within the peripheral autonomic nervous system, however, means that alternative extra-CNS sites of origin cannot be disregarded as possible candidates. The PD staging system not only caused controversy but contributed a framework for (1) assessing pathology in the spinal cord, ENS, and PNS in relationship to that evolving in the brain, (2) defining prodromal disease and cohorts of at-risk individuals, (3) developing potential prognostic biomarkers for very early disease, (4) testing novel hypotheses and experimental models of α-synuclein propagation and disease progression, and (5) finding causally-oriented therapies that intervene before the substantia nigra becomes involved. The identification of new disease mechanisms at the molecular and cellular levels indicates that physical contacts (transsynaptic) and transneuronal transmission between vulnerable nerve cells are somehow crucial to the pathogenesis of sPD.
Collapse
Affiliation(s)
- Heiko Braak
- Correspondence to: Prof. Heiko Braak, M.D., Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany. Tel.: +49 731 500 63111; Fax: +49 731 500 63133; E-mail:
| | | |
Collapse
|
43
|
Xiong Y, Dawson TM, Dawson VL. Models of LRRK2-Associated Parkinson's Disease. ADVANCES IN NEUROBIOLOGY 2017; 14:163-191. [PMID: 28353284 DOI: 10.1007/978-3-319-49969-7_9] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common genetic causes of Parkinson's disease (PD) and also one of the strongest genetic risk factors in sporadic PD. The LRRK2 protein contains a GTPase and a kinase domain and several protein-protein interaction domains. Both in vitro and in vivo assays in different model systems have provided tremendous insights into the molecular mechanisms underlying LRRK2-induced dopaminergic neurodegeneration. Among all the model systems, animal models are crucial tools to study the pathogenesis of human disease. How do the animal models recapitulate LRRK2-induced dopaminergic neuronal loss in human PD? To answer this question, this review focuses on the discussion of the animal models of LRRK2-associated PD including genetic- and viral-based models.
Collapse
Affiliation(s)
- Yulan Xiong
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Anatomy and Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS, 66506, USA.
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, 70130-2685, USA.
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, 70130-2685, USA. .,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
44
|
Del Tredici K, Braak H. Review: Sporadic Parkinson's disease: development and distribution of α-synuclein pathology. Neuropathol Appl Neurobiol 2016; 42:33-50. [PMID: 26662475 DOI: 10.1111/nan.12298] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 12/04/2015] [Accepted: 12/13/2015] [Indexed: 12/17/2022]
Abstract
The development of α-synuclein immunoreactive aggregates in selectively vulnerable neuronal types of the human central, peripheral, and enteric nervous systems is crucial for the pathogenesis of sporadic Parkinson's disease. The presence of these lesions persists into the end phase of the disease, a process that is not subject to remission. The initial induction of α-synuclein misfolding and subsequent aggregation probably occurs in the olfactory bulb and/or the enteric nervous system. Each of these sites is exposed to potentially hostile environmental factors. Once formed, the aggregates appear to be capable of propagating trans-synaptically from nerve cell to nerve cell in a virtually self-promoting pathological process. A regional distribution pattern of aggregated α-synuclein emerges that entails the involvement of only a few types of susceptible and axonally interconnected projection neurons within the human nervous system. One major route of disease progression may originate in the enteric nervous system and retrogradely reach the dorsal motor nucleus of the vagal nerve in the lower brainstem. From there, the disease process proceeds chiefly in a caudo-rostral direction through visceromotor and somatomotor brainstem centres to the midbrain, forebrain, and cerebral cortex. Spinal cord centres may become involved by means of descending projections from involved lower brainstem nuclei as well as by sympathetic projections connecting the enteric nervous system with postganglionic peripheral ganglia and preganglionic nuclei of the spinal cord. The development of experimental cellular and animal models is helping to explain the mechanisms of how abnormal α-synuclein can undergo aggregation and how transmission along axonal connectivities can occur, thereby encouraging the initiation of potential disease-modifying therapeutic strategies for sporadic Parkinson's disease.
Collapse
Affiliation(s)
- K Del Tredici
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| | - H Braak
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| |
Collapse
|
45
|
Braak H, Del Tredici K. Potential Pathways of Abnormal Tau and α-Synuclein Dissemination in Sporadic Alzheimer's and Parkinson's Diseases. Cold Spring Harb Perspect Biol 2016; 8:a023630. [PMID: 27580631 PMCID: PMC5088528 DOI: 10.1101/cshperspect.a023630] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Experimental data indicate that transneuronal propagation of abnormal protein aggregates in neurodegenerative proteinopathies, such as sporadic Alzheimer's disease (AD) and Parkinson's disease (PD), is capable of a self-propagating process that leads to a progression of neurodegeneration and accumulation of prion-like particles. The mechanisms by which misfolded tau and α-synuclein possibly spread from one involved nerve cell to the next in the neuronal chain to induce abnormal aggregation are still unknown. Based on findings from studies of human autopsy cases, we review potential pathways and mechanisms related to axonal and transneuronal dissemination of tau (sporadic AD) and α-synuclein (sporadic PD) aggregates between anatomically interconnected regions.
Collapse
Affiliation(s)
- Heiko Braak
- Clinical Neuroanatomy Section/Department of Neurology, Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Kelly Del Tredici
- Clinical Neuroanatomy Section/Department of Neurology, Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| |
Collapse
|
46
|
Sidorova YA, Saarma M. Glial cell line-derived neurotrophic factor family ligands and their therapeutic potential. Mol Biol 2016. [DOI: 10.1134/s0026893316040105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
47
|
White JA, Banerjee R, Gunawardena S. Axonal Transport and Neurodegeneration: How Marine Drugs Can Be Used for the Development of Therapeutics. Mar Drugs 2016; 14:E102. [PMID: 27213408 PMCID: PMC4882576 DOI: 10.3390/md14050102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/19/2016] [Accepted: 04/26/2016] [Indexed: 11/23/2022] Open
Abstract
Unlike virtually any other cells in the human body, neurons are tasked with the unique problem of transporting important factors from sites of synthesis at the cell bodies, across enormous distances, along narrow-caliber projections, to distally located nerve terminals in order to maintain cell viability. As a result, axonal transport is a highly regulated process whereby necessary cargoes of all types are packaged and shipped from one end of the neuron to the other. Interruptions in this finely tuned transport have been linked to many neurodegenerative disorders including Alzheimer's (AD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) suggesting that this pathway is likely perturbed early in disease progression. Therefore, developing therapeutics targeted at modifying transport defects could potentially avert disease progression. In this review, we examine a variety of potential compounds identified from marine aquatic species that affect the axonal transport pathway. These compounds have been shown to function in microtubule (MT) assembly and maintenance, motor protein control, and in the regulation of protein degradation pathways, such as the autophagy-lysosome processes, which are defective in many degenerative diseases. Therefore, marine compounds have great potential in developing effective treatment strategies aimed at early defects which, over time, will restore transport and prevent cell death.
Collapse
Affiliation(s)
- Joseph A White
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| | - Rupkatha Banerjee
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| |
Collapse
|
48
|
Kuznetsov IA, Kuznetsov AV. What can trigger the onset of Parkinson's disease - A modeling study based on a compartmental model of α-synuclein transport and aggregation in neurons. Math Biosci 2016; 278:22-9. [PMID: 27211070 DOI: 10.1016/j.mbs.2016.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/29/2016] [Accepted: 05/07/2016] [Indexed: 02/07/2023]
Abstract
The aim of this paper is to develop a minimal model describing events leading to the onset of Parkinson's disease (PD). The model accounts for α-synuclein (α-syn) production in the soma, transport toward the synapse, misfolding, and aggregation. The production and aggregation of polymeric α-syn is simulated using a minimalistic 2-step Finke-Watzky model. We utilized the developed model to analyze what changes in a healthy neuron are likely to lead to the onset of α-syn aggregation. We checked the effects of interruption of α-syn transport toward the synapse, entry of misfolded (infectious) α-syn into the somatic and synaptic compartments, increasing the rate of α-syn synthesis in the soma, and failure of α-syn degradation machinery. Our model suggests that failure of α-syn degradation machinery is probably the most likely cause for the onset of α-syn aggregation leading to PD.
Collapse
Affiliation(s)
- I A Kuznetsov
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218-2694, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA.
| |
Collapse
|
49
|
|
50
|
Hannan SB, Dräger NM, Rasse TM, Voigt A, Jahn TR. Cellular and molecular modifier pathways in tauopathies: the big picture from screening invertebrate models. J Neurochem 2016; 137:12-25. [PMID: 26756400 DOI: 10.1111/jnc.13532] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/14/2015] [Accepted: 01/04/2016] [Indexed: 12/16/2022]
Abstract
Abnormal tau accumulations were observed and documented in post-mortem brains of patients affected by Alzheimer's disease (AD) long before the identification of mutations in the Microtubule-associated protein tau (MAPT) gene, encoding the tau protein, in a different neurodegenerative disease called Frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17). The discovery of mutations in the MAPT gene associated with FTDP-17 highlighted that dysfunctions in tau alone are sufficient to cause neurodegeneration. Invertebrate models have been diligently utilized in investigating tauopathies, contributing to the understanding of cellular and molecular pathways involved in disease etiology. An important discovery came with the demonstration that over-expression of human tau in Drosophila leads to premature mortality and neuronal dysfunction including neurodegeneration, recapitulating some key neuropathological features of the human disease. The simplicity of handling invertebrate models combined with the availability of a diverse range of experimental resources make these models, in particular Drosophila a powerful invertebrate screening tool. Consequently, several large-scale screens have been performed using Drosophila, to identify modifiers of tau toxicity. The screens have revealed not only common cellular and molecular pathways, but in some instances the same modifier has been independently identified in two or more screens suggesting a possible role for these modifiers in regulating tau toxicity. The purpose of this review is to discuss the genetic modifier screens on tauopathies performed in Drosophila and C. elegans models, and to highlight the common cellular and molecular pathways that have emerged from these studies. Here, we summarize results of tau toxicity screens providing mechanistic insights into pathological alterations in tauopathies. Key pathways or modifiers that have been identified are associated with a broad range of processes including, but not limited to, phosphorylation, cytoskeleton organization, axonal transport, regulation of cellular proteostasis, transcription, RNA metabolism, cell cycle regulation, and apoptosis. We discuss the utility and application of invertebrate models in elucidating the cellular and molecular functions of novel and uncharacterized disease modifiers identified in large-scale screens as well as for investigating the function of genes identified as risk factors in genome-wide association studies from human patients in the post-genomic era. In this review, we combined and summarized several large-scale modifier screens performed in invertebrate models to identify modifiers of tau toxicity. A summary of the screens show that diverse cellular processes are implicated in the modification of tau toxicity. Kinases and phosphatases are the most predominant class of modifiers followed by components required for cellular proteostasis and axonal transport and cytoskeleton elements.
Collapse
Affiliation(s)
- Shabab B Hannan
- Schaller Research Group at the University of Heidelberg and DKFZ, Proteostasis in Neurodegenerative Disease (B180), German Cancer Research Center, Heidelberg, Germany.,Graduate School of Cellular and Molecular Neuroscience, Graduate Training Center of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Nina M Dräger
- Schaller Research Group at the University of Heidelberg and DKFZ, Proteostasis in Neurodegenerative Disease (B180), German Cancer Research Center, Heidelberg, Germany
| | - Tobias M Rasse
- Schaller Research Group at the University of Heidelberg and DKFZ, Proteostasis in Neurodegenerative Disease (B180), German Cancer Research Center, Heidelberg, Germany
| | - Aaron Voigt
- Department of Neurology at University Clinic Aachen, RWTH Aachen, Germany
| | - Thomas R Jahn
- Schaller Research Group at the University of Heidelberg and DKFZ, Proteostasis in Neurodegenerative Disease (B180), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|