1
|
Uytterhoeven V, Verstreken P, Nachman E. Synaptic sabotage: How Tau and α-Synuclein undermine synaptic health. J Cell Biol 2025; 224:e202409104. [PMID: 39718548 DOI: 10.1083/jcb.202409104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/07/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024] Open
Abstract
Synaptic dysfunction is one of the earliest cellular defects observed in Alzheimer's disease (AD) and Parkinson's disease (PD), occurring before widespread protein aggregation, neuronal loss, and cognitive decline. While the field has focused on the aggregation of Tau and α-Synuclein (α-Syn), emerging evidence suggests that these proteins may drive presynaptic pathology even before their aggregation. Therefore, understanding the mechanisms by which Tau and α-Syn affect presynaptic terminals offers an opportunity for developing innovative therapeutics aimed at preserving synapses and potentially halting neurodegeneration. This review focuses on the molecular defects that converge on presynaptic dysfunction caused by Tau and α-Syn. Both proteins have physiological roles in synapses. However, during disease, they acquire abnormal functions due to aberrant interactions and mislocalization. We provide an overview of current research on different essential presynaptic pathways influenced by Tau and α-Syn. Finally, we highlight promising therapeutic targets aimed at maintaining synaptic function in both tauopathies and synucleinopathies.
Collapse
Affiliation(s)
- Valerie Uytterhoeven
- Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research , Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Patrik Verstreken
- Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research , Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Eliana Nachman
- Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research , Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Gao Y, Zhang J, Tang T, Liu Z. Hypoxia Pathways in Parkinson's Disease: From Pathogenesis to Therapeutic Targets. Int J Mol Sci 2024; 25:10484. [PMID: 39408813 PMCID: PMC11477385 DOI: 10.3390/ijms251910484] [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] [Received: 09/05/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
The human brain is highly dependent on oxygen, utilizing approximately 20% of the body's oxygen at rest. Oxygen deprivation to the brain can lead to loss of consciousness within seconds and death within minutes. Recent studies have identified regions of the brain with spontaneous episodic hypoxia, referred to as "hypoxic pockets". Hypoxia can also result from impaired blood flow due to conditions such as heart disease, blood clots, stroke, or hemorrhage, as well as from reduced oxygen intake or excessive oxygen consumption caused by factors like low ambient oxygen, pulmonary diseases, infections, inflammation, and cancer. Severe hypoxia in the brain can manifest symptoms similar to Parkinson's disease (PD), including cerebral edema, mood disturbances, and cognitive impairments. Additionally, the development of PD appears to be closely associated with hypoxia and hypoxic pathways. This review seeks to investigate the molecular interactions between hypoxia and PD, emphasizing the pathological role of hypoxic pathways in PD and exploring their potential as therapeutic targets.
Collapse
Affiliation(s)
- Yuanyuan Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (Y.G.); (J.Z.)
| | - Jiarui Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (Y.G.); (J.Z.)
| | - Tuoxian Tang
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Zhenjiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (Y.G.); (J.Z.)
| |
Collapse
|
3
|
Bastioli G, Piccirillo S, Graciotti L, Carone M, Sprega G, Taoussi O, Preziuso A, Castaldo P. Calcium Deregulation in Neurodegeneration and Neuroinflammation in Parkinson's Disease: Role of Calcium-Storing Organelles and Sodium-Calcium Exchanger. Cells 2024; 13:1301. [PMID: 39120330 PMCID: PMC11311461 DOI: 10.3390/cells13151301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that lacks effective treatment strategies to halt or delay its progression. The homeostasis of Ca2+ ions is crucial for ensuring optimal cellular functions and survival, especially for neuronal cells. In the context of PD, the systems regulating cellular Ca2+ are compromised, leading to Ca2+-dependent synaptic dysfunction, impaired neuronal plasticity, and ultimately, neuronal loss. Recent research efforts directed toward understanding the pathology of PD have yielded significant insights, particularly highlighting the close relationship between Ca2+ dysregulation, neuroinflammation, and neurodegeneration. However, the precise mechanisms driving the selective loss of dopaminergic neurons in PD remain elusive. The disruption of Ca2+ homeostasis is a key factor, engaging various neurodegenerative and neuroinflammatory pathways and affecting intracellular organelles that store Ca2+. Specifically, impaired functioning of mitochondria, lysosomes, and the endoplasmic reticulum (ER) in Ca2+ metabolism is believed to contribute to the disease's pathophysiology. The Na+-Ca2+ exchanger (NCX) is considered an important key regulator of Ca2+ homeostasis in various cell types, including neurons, astrocytes, and microglia. Alterations in NCX activity are associated with neurodegenerative processes in different models of PD. In this review, we will explore the role of Ca2+ dysregulation and neuroinflammation as primary drivers of PD-related neurodegeneration, with an emphasis on the pivotal role of NCX in the pathology of PD. Consequently, NCXs and their interplay with intracellular organelles may emerge as potentially pivotal players in the mechanisms underlying PD neurodegeneration, providing a promising avenue for therapeutic intervention aimed at halting neurodegeneration.
Collapse
Affiliation(s)
- Guendalina Bastioli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Laura Graciotti
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Marianna Carone
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
- Institute of Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, 8092 Zürich, Switzerland
| | - Giorgia Sprega
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Omayema Taoussi
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Pasqualina Castaldo
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| |
Collapse
|
4
|
Skeens A, Markle JM, Petipas G, Frey SL, Legleiter J. Divalent cations promote huntingtin fibril formation on endoplasmic reticulum derived and model membranes. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184339. [PMID: 38763270 DOI: 10.1016/j.bbamem.2024.184339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
Huntington's Disease (HD) is caused by an abnormal expansion of the polyglutamine (polyQ) domain within the first exon of the huntingtin protein (htt). This expansion promotes disease-related htt aggregation into amyloid fibrils and the formation of proteinaceous inclusion bodies within neurons. Fibril formation is a complex heterogenous process involving an array of aggregate species such as oligomers, protofibrils, and fibrils. In HD, structural abnormalities of membranes of several organelles develop. In particular, the accumulation of htt fibrils near the endoplasmic reticulum (ER) impinges upon the membrane, resulting in ER damage, altered dynamics, and leakage of Ca2+. Here, the aggregation of htt at a bilayer interface assembled from ER-derived liposomes was investigated, and fibril formation directly on these membranes was enhanced. Based on these observations, simplified model systems were used to investigate mechanisms associated with htt aggregation on ER membranes. As the ER-derived liposome fractions contained residual Ca2+, the role of divalent cations was also investigated. In the absence of lipids, divalent cations had minimal impact on htt structure and aggregation. However, the presence of Ca2+ or Mg2+ played a key role in promoting fibril formation on lipid membranes despite reduced htt insertion into and association with lipid interfaces, suggesting that the ability of divalent cations to promote fibril formation on membranes is mediated by induced changes to the lipid membrane physicochemical properties. With enhanced concentrations of intracellular calcium being a hallmark of HD, the ability of divalent cations to influence htt aggregation at lipid membranes may play a role in aggregation events that lead to organelle abnormalities associated with disease.
Collapse
Affiliation(s)
- Adam Skeens
- The C. Eugene Bennett Department of Chemistry, West Virginia University, 217 Clark Hall, Morgantown, WV 26506, USA
| | - Jordyn M Markle
- The Department of Chemistry, Gettysburg College, 300 N. Washington Street, Gettysburg, PA 17325, USA
| | - Gabriella Petipas
- The C. Eugene Bennett Department of Chemistry, West Virginia University, 217 Clark Hall, Morgantown, WV 26506, USA
| | - Shelli L Frey
- The Department of Chemistry, Gettysburg College, 300 N. Washington Street, Gettysburg, PA 17325, USA.
| | - Justin Legleiter
- The C. Eugene Bennett Department of Chemistry, West Virginia University, 217 Clark Hall, Morgantown, WV 26506, USA; Rockefeller Neurosciences Institutes, West Virginia University, 1 Medical Center Dr., P.O. Box 9303, Morgantown, WV 26505, USA; Department of Neuroscience, West Virginia University, 1 Medical Center Dr., P.O. Box 9303, Morgantown, WV 26505, USA.
| |
Collapse
|
5
|
Sandoval A, Duran P, Corzo-López A, Fernández-Gallardo M, Muñoz-Herrera D, Leyva-Leyva M, González-Ramírez R, Felix R. The role of voltage-gated calcium channels in the pathogenesis of Parkinson's disease. Int J Neurosci 2024; 134:452-461. [PMID: 35993158 DOI: 10.1080/00207454.2022.2115905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/07/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Aim: Voltage-gated calcium (CaV) channels play an essential role in maintaining calcium homeostasis and regulating numerous physiological processes in neurons. Therefore, dysregulation of calcium signaling is relevant in many neurological disorders, including Parkinson's disease (PD). This review aims to introduce the role of CaV channels in PD and discuss some novel aspects of channel regulation and its impact on the molecular pathophysiology of the disease. Methods: an exhaustive search of the literature in the field was carried out using the PubMed database of The National Center for Biotechnology Information. Systematic searches were performed from the initial date of publication to May 2022. Results: Although α-synuclein aggregates are the main feature of PD, L-type calcium (CaV1) channels seem to play an essential role in the pathogenesis of PD. Changes in the functional expression of CaV1.3 channels alter Calcium homeostasis and contribute to the degeneration of dopaminergic neurons. Furthermore, recent studies suggest that CaV channel trafficking towards the cell membrane depends on the activity of the ubiquitin-proteasome system (UPS). In PD, there is an increase in the expression of L-type channels associated with a decrease in the expression of Parkin, an E3 enzyme of the UPS. Therefore, a link between Parkin and CaV channels could play a fundamental role in the pathogenesis of PD and, as such, could be a potentially attractive target for therapeutic intervention. Conclusion: The study of alterations in the functional expression of CaV channels will provide a framework to understand better the neurodegenerative processes that occur in PD and a possible path toward identifying new therapeutic targets to treat this condition.
Collapse
Affiliation(s)
- Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of Mexico (UNAM), Tlalnepantla, Mexico
| | - Paz Duran
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Alejandra Corzo-López
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | | | - David Muñoz-Herrera
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Margarita Leyva-Leyva
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Ricardo Felix
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| |
Collapse
|
6
|
Kampmann M. Molecular and cellular mechanisms of selective vulnerability in neurodegenerative diseases. Nat Rev Neurosci 2024; 25:351-371. [PMID: 38575768 DOI: 10.1038/s41583-024-00806-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 04/06/2024]
Abstract
The selective vulnerability of specific neuronal subtypes is a hallmark of neurodegenerative diseases. In this Review, I summarize our current understanding of the brain regions and cell types that are selectively vulnerable in different neurodegenerative diseases and describe the proposed underlying cell-autonomous and non-cell-autonomous mechanisms. I highlight how recent methodological innovations - including single-cell transcriptomics, CRISPR-based screens and human cell-based models of disease - are enabling new breakthroughs in our understanding of selective vulnerability. An understanding of the molecular mechanisms that determine selective vulnerability and resilience would shed light on the key processes that drive neurodegeneration and point to potential therapeutic strategies to protect vulnerable cell populations.
Collapse
Affiliation(s)
- Martin Kampmann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Khamis H, Cohen O. Coupled action potential and calcium dynamics underlie robust spontaneous firing in dopaminergic neurons. Phys Biol 2024; 21:026005. [PMID: 38382117 DOI: 10.1088/1478-3975/ad2bd4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/21/2024] [Indexed: 02/23/2024]
Abstract
Dopaminergic neurons are specialized cells in the substantia nigra, tasked with dopamine secretion. This secretion relies on intracellular calcium signaling coupled to neuronal electrical activity. These neurons are known to display spontaneous calcium oscillationsin-vitroandin-vivo, even in synaptic isolation, controlling the basal dopamine levels. Here we outline a kinetic model for the ion exchange across the neuronal plasma membrane. Crucially, we relax the assumption of constant, cytoplasmic sodium and potassium concentration. We show that sodium-potassium dynamics are strongly coupled to calcium dynamics and are essential for the robustness of spontaneous firing frequency. The model predicts several regimes of electrical activity, including tonic and 'burst' oscillations, and predicts the switch between those in response to perturbations. 'Bursting' correlates with increased calcium amplitudes, while maintaining constant average, allowing for a vast change in the calcium signal responsible for dopamine secretion. All the above traits provide the flexibility to create rich action potential dynamics that are crucial for cellular function.
Collapse
Affiliation(s)
- Hadeel Khamis
- Gateway Institute for Brain Research, Fort Lauderdale, FL 33314, United States of America
| | - Ohad Cohen
- Gateway Institute for Brain Research, Fort Lauderdale, FL 33314, United States of America
| |
Collapse
|
8
|
Al‐kuraishy HM, Al‐Gareeb AI, Albuhadily AK, Elewa YHA, AL‐Farga A, Aqlan F, Zahran MH, Batiha GE. Sleep disorders cause Parkinson's disease or the reverse is true: Good GABA good night. CNS Neurosci Ther 2024; 30:e14521. [PMID: 38491789 PMCID: PMC10943276 DOI: 10.1111/cns.14521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative brain disease due to degeneration of dopaminergic neurons (DNs) presented with motor and non-motor symptoms. PD symptoms are developed in response to the disturbance of diverse neurotransmitters including γ-aminobutyric acid (GABA). GABA has a neuroprotective effect against PD neuropathology by protecting DNs in the substantia nigra pars compacta (SNpc). It has been shown that the degeneration of GABAergic neurons is linked with the degeneration of DNs and the progression of motor and non-motor PD symptoms. GABA neurotransmission is a necessary pathway for normal sleep patterns, thus deregulation of GABAergic neurotransmission in PD could be the potential cause of sleep disorders in PD. AIM Sleep disorders affect GABA neurotransmission leading to memory and cognitive dysfunction in PD. For example, insomnia and short sleep duration are associated with a reduction of brain GABA levels. Moreover, PD-related disorders including rigidity and nocturia influence sleep patterns leading to fragmented sleep which may also affect PD neuropathology. However, the mechanistic role of GABA in PD neuropathology regarding motor and non-motor symptoms is not fully elucidated. Therefore, this narrative review aims to clarify the mechanistic role of GABA in PD neuropathology mainly in sleep disorders, and how good GABA improves PD. In addition, this review of published articles tries to elucidate how sleep disorders such as insomnia and REM sleep behavior disorder (RBD) affect PD neuropathology and severity. The present review has many limitations including the paucity of prospective studies and most findings are taken from observational and preclinical studies. GABA involvement in the pathogenesis of PD has been recently discussed by recent studies. Therefore, future prospective studies regarding the use of GABA agonists in the management of PD are suggested to observe their distinct effects on motor and non-motor symptoms. CONCLUSION There is a bidirectional relationship between the pathogenesis of PD and sleep disorders which might be due to GABA deregulation.
Collapse
Affiliation(s)
- Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary MedicineZagazig UniversityZagazigEgypt
- Faculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Ammar AL‐Farga
- Biochemistry Department, College of SciencesUniversity of JeddahJeddahSaudia Arbia
| | - Faisal Aqlan
- Department of Chemistry, College of SciencesIbb UniversityIbb GovernorateYemen
| | | | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhur UniversityDamanhurEgypt
| |
Collapse
|
9
|
Çakır M, Saçmacı H. The relationship of salusins with Parkinson's Disease, Alzheimer's Disease, and acute ischemic stroke: A preliminary study. Neurosci Lett 2024; 824:137683. [PMID: 38350537 DOI: 10.1016/j.neulet.2024.137683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/15/2024]
Abstract
Neuroinflammation, oxidative stress, and apoptosis play important roles in the pathophysiology of Alzheimer's Disease (AD), Parkinson's Disease (PD), and Acute Ischemic Stroke (AIS). Salusin-α and salusin-β peptides, which were shown to be present in many tissues, including the central nervous system, were also shown to be associated with apoptosis, inflammation, and oxidative stress. In the present study, the relationship between Salusin-α and salusin-β peptides and AD, PD, and AIS were investigated. A total of 179 people were included in the present study, including 46 AD, 44 PD, 42 AIS, and 47 controls. Plasma Salusin-α and salusin-β levels were measured with the ELISA Method. The plasma salusin-β levels of AD, PD, and AIS patients were lower than the control group at significant levels (p < 0.05). It was also found that there were correlations between salusin-α and salusin-β levels and age, triglyceride, LDL-c, total cholesterol, and hemoglobin levels. In this study, we found that salusin- β, an endogenous neuropeptide, was associated with AD, PD and AIS. The low level of salusin-β in these diseases in which neuronal damage occurs may be related to the neuroprotective properties of this endogenous peptide. Further studies are needed to fully understand the relationship between salusin-β and the pathophysiology of these diseases.
Collapse
Affiliation(s)
- Murat Çakır
- Department of Physiology, Faculty of Medicine, University of Yozgat Bozok, Yozgat, 66200, Turkey.
| | - Hikmet Saçmacı
- Department of Neurology, Faculty of Medicine, University of Yozgat Bozok, Yozgat, 66200, Turkey.
| |
Collapse
|
10
|
Lage L, Rodriguez-Perez AI, Villar-Cheda B, Labandeira-Garcia JL, Dominguez-Meijide A. Angiotensin type 1 receptor activation promotes neuronal and glial alpha-synuclein aggregation and transmission. NPJ Parkinsons Dis 2024; 10:37. [PMID: 38368444 PMCID: PMC10874459 DOI: 10.1038/s41531-024-00650-0] [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: 06/28/2023] [Accepted: 02/02/2024] [Indexed: 02/19/2024] Open
Abstract
The brain renin-angiotensin system (RAS) has been related to dopaminergic degeneration, and high expression of the angiotensin II (AngII) type 1 receptor (AT1) gene is a marker of the most vulnerable neurons in humans. However, it is unknown whether AngII/AT1 overactivation affects α-synuclein aggregation and transmission. In vitro, AngII/AT1 activation increased α-synuclein aggregation in dopaminergic neurons and microglial cells, which was related to AngII-induced NADPH-oxidase activation and intracellular calcium raising. In mice, AngII/AT1 activation was involved in MPTP-induced increase in α-synuclein expression and aggregation, as they significantly decreased in mice treated with the AT1 blocker telmisartan and AT1 knockout mice. Cell co-cultures (transwells) revealed strong transmission of α-synuclein from dopaminergic neurons to astrocytes and microglia. AngII induced a higher α-synuclein uptake by microglial cells and an increase in the transfer of α-synuclein among astroglial cells. However, AngII did not increase the release of α-synuclein by neurons. The results further support brain RAS dysregulation as a major mechanism for the progression of Parkinson's disease, and AT1 inhibition and RAS modulation as therapeutic targets.
Collapse
Affiliation(s)
- Lucia Lage
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana I Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Begoña Villar-Cheda
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - Antonio Dominguez-Meijide
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
11
|
O’Day DH. The Complex Interplay between Toxic Hallmark Proteins, Calmodulin-Binding Proteins, Ion Channels, and Receptors Involved in Calcium Dyshomeostasis in Neurodegeneration. Biomolecules 2024; 14:173. [PMID: 38397410 PMCID: PMC10886625 DOI: 10.3390/biom14020173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Calcium dyshomeostasis is an early critical event in neurodegeneration as exemplified by Alzheimer's (AD), Huntington's (HD) and Parkinson's (PD) diseases. Neuronal calcium homeostasis is maintained by a diversity of ion channels, buffers, calcium-binding protein effectors, and intracellular storage in the endoplasmic reticulum, mitochondria, and lysosomes. The function of these components and compartments is impacted by the toxic hallmark proteins of AD (amyloid beta and Tau), HD (huntingtin) and PD (alpha-synuclein) as well as by interactions with downstream calcium-binding proteins, especially calmodulin. Each of the toxic hallmark proteins (amyloid beta, Tau, huntingtin, and alpha-synuclein) binds to calmodulin. Multiple channels and receptors involved in calcium homeostasis and dysregulation also bind to and are regulated by calmodulin. The primary goal of this review is to show the complexity of these interactions and how they can impact research and the search for therapies. A secondary goal is to suggest that therapeutic targets downstream from calcium dyshomeostasis may offer greater opportunities for success.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
12
|
Prymaczok NC, De Francesco PN, Mazzetti S, Humbert-Claude M, Tenenbaum L, Cappelletti G, Masliah E, Perello M, Riek R, Gerez JA. Cell-to-cell transmitted alpha-synuclein recapitulates experimental Parkinson's disease. NPJ Parkinsons Dis 2024; 10:10. [PMID: 38184623 PMCID: PMC10771530 DOI: 10.1038/s41531-023-00618-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/08/2023] [Indexed: 01/08/2024] Open
Abstract
Parkinson's disease is characterized by a progressive accumulation of alpha-Synuclein (αSyn) neuronal inclusions called Lewy bodies in the nervous system. Lewy bodies can arise from the cell-to-cell propagation of αSyn, which can occur via sequential steps of secretion and uptake. Here, by fusing a removable short signal peptide to the N-terminus of αSyn, we developed a novel mouse model with enhanced αSyn secretion and cell-to-cell transmission. Expression of the secreted αSyn in the mouse brain was under the control of a novel hybrid promoter in combination with adeno-associated virus serotype 9 (AAV9). This combination of promoter and viral vector induced a robust expression in neurons but not in the glia of injected mice. Biochemical characterization of the secreted αSyn revealed that, in cultured cells, this protein is released to the extracellular milieu via conventional secretion. The released αSyn is then internalized and processed by acceptor cells via the endosome-lysosome pathway indicating that the secreted αSyn is cell-to-cell transmitted. The secreted αSyn is aggregation-prone and amyloidogenic, and when expressed in the brain of wild-type non-transgenic mice, it induces a Parkinson's disease-like phenotype that includes a robust αSyn pathology in the substantia nigra, neuronal loss, neuroinflammation, and motor deficits, all the key features of experimental animal models of Parkinson's disease. In summary, a novel animal model of Parkinson's disease based on enhanced cell-to-cell transmission of αSyn was developed. The neuron-produced cell-to-cell transmitted αSyn triggers all phenotypic features of experimental Parkinson's disease in mice.
Collapse
Affiliation(s)
- Natalia Cecilia Prymaczok
- Institute of Molecular Physical Science, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Pablo Nicolas De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology (IMBICE), dependent of the Argentine Research Council (CONICET), Scientific Research Commission and University of La Plata Buenos Aires, La Plata, Argentina
| | - Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, Milano, Italy
- Fondazione Grigioni per il Morbo di Parkinson, Milano, Italy
| | - Marie Humbert-Claude
- Laboratory of Neurotherapies and NeuroModulation, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University Hospital, Lausanne, Switzerland
| | - Liliane Tenenbaum
- Laboratory of Neurotherapies and NeuroModulation, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University Hospital, Lausanne, Switzerland
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, Milano, Italy
- Fondazione Grigioni per il Morbo di Parkinson, Milano, Italy
| | - Eliezer Masliah
- Division of Neurosciences, National Institute on Aging/NIH, 7201, Wisconsin Ave, Bethesda, MD, USA
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology (IMBICE), dependent of the Argentine Research Council (CONICET), Scientific Research Commission and University of La Plata Buenos Aires, La Plata, Argentina
| | - Roland Riek
- Institute of Molecular Physical Science, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Juan Atilio Gerez
- Institute of Molecular Physical Science, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Henrich MT, Oertel WH, Surmeier DJ, Geibl FF. Mitochondrial dysfunction in Parkinson's disease - a key disease hallmark with therapeutic potential. Mol Neurodegener 2023; 18:83. [PMID: 37951933 PMCID: PMC10640762 DOI: 10.1186/s13024-023-00676-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/30/2023] [Indexed: 11/14/2023] Open
Abstract
Mitochondrial dysfunction is strongly implicated in the etiology of idiopathic and genetic Parkinson's disease (PD). However, strategies aimed at ameliorating mitochondrial dysfunction, including antioxidants, antidiabetic drugs, and iron chelators, have failed in disease-modification clinical trials. In this review, we summarize the cellular determinants of mitochondrial dysfunction, including impairment of electron transport chain complex 1, increased oxidative stress, disturbed mitochondrial quality control mechanisms, and cellular bioenergetic deficiency. In addition, we outline mitochondrial pathways to neurodegeneration in the current context of PD pathogenesis, and review past and current treatment strategies in an attempt to better understand why translational efforts thus far have been unsuccessful.
Collapse
Affiliation(s)
- Martin T Henrich
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35039, Marburg, Germany
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Wolfgang H Oertel
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Fanni F Geibl
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35039, Marburg, Germany.
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany.
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
14
|
Hastings N, Yu Y, Huang B, Middya S, Inaoka M, Erkamp NA, Mason RJ, Carnicer‐Lombarte A, Rahman S, Knowles TPJ, Bance M, Malliaras GG, Kotter MRN. Electrophysiological In Vitro Study of Long-Range Signal Transmission by Astrocytic Networks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301756. [PMID: 37485646 PMCID: PMC10582426 DOI: 10.1002/advs.202301756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/09/2023] [Indexed: 07/25/2023]
Abstract
Astrocytes are diverse brain cells that form large networks communicating via gap junctions and chemical transmitters. Despite recent advances, the functions of astrocytic networks in information processing in the brain are not fully understood. In culture, brain slices, and in vivo, astrocytes, and neurons grow in tight association, making it challenging to establish whether signals that spread within astrocytic networks communicate with neuronal groups at distant sites, or whether astrocytes solely respond to their local environments. A multi-electrode array (MEA)-based device called AstroMEA is designed to separate neuronal and astrocytic networks, thus allowing to study the transfer of chemical and/or electrical signals transmitted via astrocytic networks capable of changing neuronal electrical behavior. AstroMEA demonstrates that cortical astrocytic networks can induce a significant upregulation in the firing frequency of neurons in response to a theta-burst charge-balanced biphasic current stimulation (5 pulses of 100 Hz × 10 with 200 ms intervals, 2 s total duration) of a separate neuronal-astrocytic group in the absence of direct neuronal contact. This result corroborates the view of astrocytic networks as a parallel mechanism of signal transmission in the brain that is separate from the neuronal connectome. Translationally, it highlights the importance of astrocytic network protection as a treatment target.
Collapse
Affiliation(s)
- Nataly Hastings
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
- Electrical Engineering DivisionDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Yi‐Lin Yu
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Department of Neurological SurgeryTri‐Service General HospitalNational Defence Medical CentreTaipei, Neihu District11490Taiwan
| | - Botian Huang
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | - Sagnik Middya
- Electrical Engineering DivisionDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Misaki Inaoka
- Electrical Engineering DivisionDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Nadia A. Erkamp
- Yusuf Hamied Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Roger J. Mason
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | | | - Saifur Rahman
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Tuomas P. J. Knowles
- Yusuf Hamied Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
- Cavendish LaboratoryDepartment of PhysicsUniversity of CambridgeJ J Thomson AveCambridgeCB3 0HEUK
| | - Manohar Bance
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | - George G. Malliaras
- Electrical Engineering DivisionDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Mark R. N. Kotter
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| |
Collapse
|
15
|
Zhao Q, Tao Y, Zhao K, Ma Y, Xu Q, Liu C, Zhang S, Li D. Structural Insights of Fe 3+ Induced α-synuclein Fibrillation in Parkinson's Disease. J Mol Biol 2023; 435:167680. [PMID: 35690099 DOI: 10.1016/j.jmb.2022.167680] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023]
Abstract
Amyloid aggregation of α-synuclein (α-syn) in Lewy bodies (LBs) is the pathological hallmark of Parkinson's disease (PD). Iron, especially Fe3+, is accumulated in substantia nigra of PD patients and co-deposited with α-syn in LBs. However, how Fe3+ modulates α-syn fibrillation at molecular level remains unclear. In this study, we found that Fe3+ can promote α-syn fibrillation at low concentration while inhibit its fibrillation at high concentration. NMR titration study shows poor interaction between α-syn monomer and Fe3+. Instead, we found that Fe3+ binds to α-syn fibrils. By using cryo-electron microscopy (cryo-EM), we further determined the atomic structure of α-syn fibril in complex with Fe3+ at the resolution of 2.7 Å. Strikingly, two extra electron densities adjacent to His50 and Glu57 were observed as putative binding sites of Fe3+ and water molecules, suggesting that Fe3+ binds to the negative cleft of the fibril and stabilizes the fibril structure for promoting α-syn aggregation. Further mutagenesis study shows mutation of His50 abolishes the Fe3+-facilitated fibrillation of α-syn. Our work illuminates the structural basis of the interaction of Fe3+ and α-syn in both monomeric and fibrillar forms, and sheds light on understanding the pathological role of Fe3+ in α-syn aggregation in PD.
Collapse
Affiliation(s)
- Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China; School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China; School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Kun Zhao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yeyang Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Qianhui Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China; Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
16
|
Naren P, Cholkar A, Kamble S, Khan SS, Srivastava S, Madan J, Mehra N, Tiwari V, Singh SB, Khatri DK. Pathological and Therapeutic Advances in Parkinson's Disease: Mitochondria in the Interplay. J Alzheimers Dis 2023; 94:S399-S428. [PMID: 36093711 PMCID: PMC10473111 DOI: 10.3233/jad-220682] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative illness majorly affecting the population between the ages of 55 to 65 years. Progressive dopaminergic neuronal loss and the collective assemblage of misfolded alpha-synuclein in the substantia nigra, remain notable neuro-pathological hallmarks of the disease. Multitudes of mechanistic pathways have been proposed in attempts to unravel the pathogenesis of PD but still, it remains elusive. The convergence of PD pathology is found in organelle dysfunction where mitochondria remain a major contributor. Mitochondrial processes like bioenergetics, mitochondrial dynamics, and mitophagy are under strict regulation by the mitochondrial genome and nuclear genome. These processes aggravate neurodegenerative activities upon alteration through neuroinflammation, oxidative damage, apoptosis, and proteostatic stress. Therefore, the mitochondria have grabbed a central position in the patho-mechanistic exploration of neurodegenerative diseases like PD. The management of PD remains a challenge to physicians to date, due to the variable therapeutic response of patients and the limitation of conventional chemical agents which only offer symptomatic relief with minimal to no disease-modifying effect. This review describes the patho-mechanistic pathways involved in PD not only limited to protein dyshomeostasis and oxidative stress, but explicit attention has been drawn to exploring mechanisms like organelle dysfunction, primarily mitochondria and mitochondrial genome influence, while delineating the newer exploratory targets such as GBA1, GLP, LRRK2, and miRNAs and therapeutic agents targeting them.
Collapse
Affiliation(s)
- Padmashri Naren
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Anjali Cholkar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Suchita Kamble
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Sabiya Samim Khan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Neelesh Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.) Varanasi (U.P.), India
| | - Shashi Bala Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| |
Collapse
|
17
|
Caulfield ME, Manfredsson FP, Steece-Collier K. The Role of Striatal Cav1.3 Calcium Channels in Therapeutics for Parkinson's Disease. Handb Exp Pharmacol 2023; 279:107-137. [PMID: 36592226 DOI: 10.1007/164_2022_629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Parkinson's disease (PD) is a relentlessly progressive neurodegenerative disorder with typical motor symptoms that include rigidity, tremor, and akinesia/bradykinesia, in addition to a host of non-motor symptoms. Motor symptoms are caused by progressive and selective degeneration of dopamine (DA) neurons in the SN pars compacta (SNpc) and the accompanying loss of striatal DA innervation from these neurons. With the exception of monogenic forms of PD, the etiology of idiopathic PD remains unknown. While there are a number of symptomatic treatment options available to individuals with PD, these therapies do not work uniformly well in all patients, and eventually most are plagued with waning efficacy and significant side-effect liability with disease progression. The incidence of PD increases with aging, and as such the expected burden of this disease will continue to escalate as our aging population increases (Dorsey et al. Neurology 68:384-386, 2007). The daunting personal and socioeconomic burden has pressed scientists and clinicians to find improved symptomatic treatment options devoid side-effect liability and meaningful disease-modifying therapies. Federal and private sources have supported clinical investigations over the past two-plus decades; however, no trial has yet been successful in finding an effective therapy to slow progression of PD, and there is currently just one FDA approved drug to treat the antiparkinsonian side-effect known as levodopa-induced dyskinesia (LID) that impacts approximately 90% of all individuals with PD. In this review, we present biological rationale and experimental evidence on the potential therapeutic role of the L-type voltage-gated Cav1.3 calcium (Ca2+) channels in two distinct brain regions, with two distinct mechanisms of action, in impacting the lives of individuals with PD. Our primary emphasis will be on the role of Cav1.3 channels in the striatum and the compelling evidence of their involvement in LID side-effect liability. We also briefly discuss the role of these same Ca2+ channels in the SNpc and the longstanding interest in Cav1.3 in this brain region in halting or delaying progression of PD.
Collapse
Affiliation(s)
- Margaret E Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
18
|
Heng Y, Li YY, Wen L, Yan JQ, Chen NH, Yuan YH. Gastric Enteric Glial Cells: A New Contributor to the Synucleinopathies in the MPTP-Induced Parkinsonism Mouse. Molecules 2022; 27:7414. [PMID: 36364248 PMCID: PMC9656042 DOI: 10.3390/molecules27217414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 05/19/2024] Open
Abstract
Accumulating evidence has shown that Parkinson's disease (PD) is a systemic disease other than a mere central nervous system (CNS) disorder. One of the most important peripheral symptoms is gastrointestinal dysfunction. The enteric nervous system (ENS) is regarded as an essential gateway to the environment. The discovery of the prion-like behavior of α-synuclein makes it possible for the neurodegenerative process to start in the ENS and spread via the gut-brain axis to the CNS. We first confirmed that synucleinopathies existed in the stomachs of chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid (MPTP/p)-induced PD mice, as indicated by the significant increase in abnormal aggregated and nitrated α-synuclein in the TH-positive neurons and enteric glial cells (EGCs) of the gastric myenteric plexus. Next, we attempted to clarify the mechanisms in single MPTP-injected mice. The stomach naturally possesses high monoamine oxidase-B (MAO-B) activity and low superoxide dismutase (SOD) activity, making the stomach susceptible to MPTP-induced oxidative stress, as indicated by the significant increase in reactive oxygen species (ROS) in the stomach and elevated 4-hydroxynonenal (4-HNE) in the EGCs after MPTP exposure for 3 h. Additionally, stomach synucleinopathies appear before those of the nigrostriatal system, as determined by Western blotting 12 h after MPTP injection. Notably, nitrated α-synuclein was considerably increased in the EGCs after 3 h and 12 h of MPTP exposure. Taken together, our work demonstrated that the EGCs could be new contributors to synucleinopathies in the stomach. The early-initiated synucleinopathies might further influence neighboring neurons in the myenteric plexus and the CNS. Our results offer a new experimental clue for interpreting the etiology of PD.
Collapse
Affiliation(s)
- Yang Heng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yan-Yan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lu Wen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jia-Qing Yan
- Department of Pharmacy, National Cancer Center/National, Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union, Medical College, Beijing 100021, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
19
|
Amorim Neto DP, de Castro Fonseca M. Protocol for rapid obtention and fractionation of anaerobic bacterial conditioned media to study calcium signaling in enteroendocrine cells. STAR Protoc 2022; 3:101486. [PMID: 35755129 PMCID: PMC9214868 DOI: 10.1016/j.xpro.2022.101486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Gut microbiota influences neurodevelopment, behavior and contributes to neurodegenerative disorders. One possible mechanism is the direct modulation of calcium (Ca2+) signaling and protein homeostasis in enteroendocrine cells (EECs), a component of the gut epithelium. Here, we present a protocol to isolate fractions of conditioned media (CM) from the anaerobic bacteria Akkermansia muciniphila and the utilization of this CM to monitor Ca2+ fluctuation in EECs by imaging. This protocol can be adapted and applied to various bacterial cultures and cell types. For complete details on the use and execution of this protocol, please refer to Amorim Neto et al. (2022). Protocol for rapid obtention and fractionation of anaerobic bacterial conditioned media Protocol can be applied to different anaerobic bacterial strains Isolated media can be used to study Ca2+ signaling in several mammalian cells
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
Affiliation(s)
- Dionisio Pedro Amorim Neto
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, Brazil
| | - Matheus de Castro Fonseca
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA, USA.
| |
Collapse
|
20
|
Kulkarni AS, Burns MR, Brundin P, Wesson DW. Linking α-synuclein-induced synaptopathy and neural network dysfunction in early Parkinson's disease. Brain Commun 2022; 4:fcac165. [PMID: 35822101 PMCID: PMC9272065 DOI: 10.1093/braincomms/fcac165] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/11/2022] [Accepted: 06/20/2022] [Indexed: 01/18/2023] Open
Abstract
The prodromal phase of Parkinson's disease is characterized by aggregation of the misfolded pathogenic protein α-synuclein in select neural centres, co-occurring with non-motor symptoms including sensory and cognitive loss, and emotional disturbances. It is unclear whether neuronal loss is significant during the prodrome. Underlying these symptoms are synaptic impairments and aberrant neural network activity. However, the relationships between synaptic defects and network-level perturbations are not established. In experimental models, pathological α-synuclein not only impacts neurotransmission at the synaptic level, but also leads to changes in brain network-level oscillatory dynamics-both of which likely contribute to non-motor deficits observed in Parkinson's disease. Here we draw upon research from both human subjects and experimental models to propose a 'synapse to network prodrome cascade' wherein before overt cell death, pathological α-synuclein induces synaptic loss and contributes to aberrant network activity, which then gives rise to prodromal symptomology. As the disease progresses, abnormal patterns of neural activity ultimately lead to neuronal loss and clinical progression of disease. Finally, we outline goals and research needed to unravel the basis of functional impairments in Parkinson's disease and other α-synucleinopathies.
Collapse
Affiliation(s)
- Aishwarya S Kulkarni
- Department of Pharmacology & Therapeutics, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| | - Matthew R Burns
- Department of Neurology, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
- Norman Fixel Institute for Neurological Disorders, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| | - Patrik Brundin
- Pharma Research and Early Development (pRED), F. Hoffman-La Roche, Little Falls, NJ, USA
| | - Daniel W Wesson
- Department of Pharmacology & Therapeutics, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
- Norman Fixel Institute for Neurological Disorders, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| |
Collapse
|
21
|
Henderson MX, Henrich MT, Geibl FF, Oertel WH, Brundin P, Surmeier DJ. The roles of connectivity and neuronal phenotype in determining the pattern of α-synuclein pathology in Parkinson's disease. Neurobiol Dis 2022; 168:105687. [PMID: 35283326 PMCID: PMC9610381 DOI: 10.1016/j.nbd.2022.105687] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, and motor dysfunction has been attributed to loss of dopaminergic neurons. However, motor dysfunction is only one of many symptoms experienced by patients. A neuropathological hallmark of PD is intraneuronal protein aggregates called Lewy pathology (LP). Neuropathological staging studies have shown that dopaminergic neurons are only one of the many cell types prone to manifest LP. Progressive appearance of LP in multiple brain regions, as well as peripheral nerves, has led to the popular hypothesis that LP and misfolded forms of one of its major components - α-synuclein (aSYN) - can spread through synaptically connected circuits. However, not all brain regions or neurons within connected circuits develop LP, suggesting that cell autonomous factors modulate the development of pathology. Here, we review studies about how LP develops and progressively engages additional brain regions. We focus on how connectivity constrains progression and discuss cell autonomous factors that drive pathology development. We propose a mixed model of cell autonomous factors and trans-synaptic spread as mediators of pathology progression and put forward this model as a framework for future experiments exploring PD pathophysiology.
Collapse
Affiliation(s)
- Michael X Henderson
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America.
| | - Martin T Henrich
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany; Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg 35043, Germany; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| | - Fanni F Geibl
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany; Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg 35043, Germany; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| | - Wolfgang H Oertel
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany
| | - Patrik Brundin
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| |
Collapse
|
22
|
Sugiura A, Kitamura M, Hasegawa Y. Calcium carbonate supplementation causes motor dysfunction. Exp Anim 2022; 71:399-410. [PMID: 35584940 PMCID: PMC9388334 DOI: 10.1538/expanim.22-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
We previously showed that a diet containing calcium carbonate causes impairments in spatial and recognition memory in mice. In this study, we investigated the effects of calcium carbonate
supplementation on motor function. Motor function was determined using different tests that have been used to analyze different aspects of Parkinsonism. A catalepsy test for akinesia; a
muscular strength assessment, pole test, beam-walking test, and gait analysis for motor coordination and balance assessment; and an open-field test for locomotor activity assessment were
performed. The mice were fed diets containing 0.6% or 1.0% calcium carbonate for eight weeks, after which they were evaluated for motor functions. The diets containing calcium carbonate
caused significant motor dysfunction, as revealed by the different tests, although the spontaneous locomotor activity did not change. Calcium carbonate supplementation decreased the dopamine
content in the basal ganglia, including the striatum and substantia nigra, and the number of tyrosine hydroxylase-positive neurons in the substantia nigra. In addition, administration of
L-dopa led to at least a partial recovery of motor dysfunction, suggesting that calcium carbonate supplementation causes motor dysfunction by decreasing the dopamine content in the basal
ganglia. These results suggest that mice with calcium carbonate-induced motor dysfunction may be useful as a new animal model for Parkinson’s disease and Huntington’s disease.
Collapse
Affiliation(s)
- Ami Sugiura
- College of Environmental Technology, Muroran Institute of Technology
| | - Misa Kitamura
- College of Environmental Technology, Muroran Institute of Technology
| | - Yasushi Hasegawa
- College of Environmental Technology, Muroran Institute of Technology
| |
Collapse
|
23
|
Amorim Neto DP, Bosque BP, Pereira de Godoy JV, Rodrigues PV, Meneses DD, Tostes K, Costa Tonoli CC, Faustino de Carvalho H, González-Billault C, de Castro Fonseca M. Akkermansia muciniphila induces mitochondrial calcium overload and α -synuclein aggregation in an enteroendocrine cell line. iScience 2022; 25:103908. [PMID: 35243260 PMCID: PMC8881719 DOI: 10.1016/j.isci.2022.103908] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/06/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota influence neurodevelopment, modulate behavior, and contribute to neurodegenerative disorders. Several studies have consistently reported a greater abundance of Akkermansia muciniphila in Parkinson disease (PD) fecal samples. Therefore, we investigated whether A.muciniphila-conditioned medium (CM) could initiate α-synuclein (αSyn) misfolding in enteroendocrine cells (EEC) — a component of the gut epithelium featuring neuron-like properties. We found that A. muciniphila CM composition is influenced by the ability of the strain to degrade mucin. Our in vitro experiments showed that the protein-enriched fraction of mucin-free CM induces RyR-mediated Ca2+ release and increased mitochondrial Ca2+ uptake leading to ROS generation and αSyn aggregation. Oral administration of A. muciniphila cultivated in the absence of mucin to mice led to αSyn aggregation in cholecystokinin (CCK)-positive EECs but no motor deficits were observed. Noteworthy, buffering mitochondrial Ca2+ reverted the damaging effects observed. These molecular insights offer evidence that bacterial proteins can induce αSyn aggregation in EECs. Gut bacterium Akkermansia muciniphila is increased in patients with Parkinson disease A. muciniphila-conditioned medium induces mitochondrial Ca2+ overload in EECs Mitochondrial Ca2+ overload leads to ROS generation and αSyn aggregation in vitro Buffering mitochondrial Ca2+ inhibits A. muciniphila-induced αSyn aggregation
Collapse
Affiliation(s)
- Dionísio Pedro Amorim Neto
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Beatriz Pelegrini Bosque
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - João Vitor Pereira de Godoy
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Paulla Vieira Rodrigues
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Dario Donoso Meneses
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
| | - Katiane Tostes
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
| | - Celisa Caldana Costa Tonoli
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
| | | | - Christian González-Billault
- Department of Biology, Faculty of Sciences and Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- The Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Matheus de Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
- Corresponding author
| |
Collapse
|
24
|
Ferritinophagy and α-Synuclein: Pharmacological Targeting of Autophagy to Restore Iron Regulation in Parkinson's Disease. Int J Mol Sci 2022; 23:ijms23042378. [PMID: 35216492 PMCID: PMC8878351 DOI: 10.3390/ijms23042378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
A major hallmark of Parkinson’s disease (PD) is the fatal destruction of dopaminergic neurons within the substantia nigra pars compacta. This event is preceded by the formation of Lewy bodies, which are cytoplasmic inclusions composed of α-synuclein protein aggregates. A triad contribution of α-synuclein aggregation, iron accumulation, and mitochondrial dysfunction plague nigral neurons, yet the events underlying iron accumulation are poorly understood. Elevated intracellular iron concentrations up-regulate ferritin expression, an iron storage protein that provides cytoprotection against redox stress. The lysosomal degradation pathway, autophagy, can release iron from ferritin stores to facilitate its trafficking in a process termed ferritinophagy. Aggregated α-synuclein inhibits SNARE protein complexes and destabilizes microtubules to halt vesicular trafficking systems, including that of autophagy effectively. The scope of this review is to describe the physiological and pathological relationship between iron regulation and α-synuclein, providing a detailed understanding of iron metabolism within nigral neurons. The underlying mechanisms of autophagy and ferritinophagy are explored in the context of PD, identifying potential therapeutic targets for future investigation.
Collapse
|
25
|
Nedaei H, Rezaei-Ghaleh N, Giller K, Becker S, Karami L, Moosavi-Movahedi AA, Griesinger C, Saboury AA. The Calcium-free form of Atorvastatin inhibits amyloid-β(1-42) aggregation in vitro. J Biol Chem 2022; 298:101662. [PMID: 35104501 PMCID: PMC8898965 DOI: 10.1016/j.jbc.2022.101662] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease is characterized by the presence of extraneuronal amyloid plaques composed of amyloid-beta (Aβ) fibrillar aggregates in the brains of patients. In mouse models, it has previously been shown that atorvastatin (Ator), a cholesterol-lowering drug, has some reducing effect on the production of cerebral Aβ. A meta-analysis on humans showed moderate effects in the short term but no improvement in the Alzheimer's Disease Assessment Scale—Cognitive Subscale behavioral test. Here, we explore a potential direct effect of Ator on Aβ42 aggregation. Using NMR-based monomer consumption assays and CD spectroscopy, we observed a promoting effect of Ator in its original form (Ator-calcium) on Aβ42 aggregation, as expected because of the presence of calcium ions. The effect was reversed when applying a CaCO3-based calcium ion scavenging method, which was validated by the aforementioned methods as well as thioflavin-T fluorescence assays and transmission electron microscopy. We found that the aggregation was inhibited significantly when the concentration of calcium-free Ator exceeded that of Aβ by at least a factor of 2. The 1H–15N heteronuclear single quantum correlation and saturation-transfer difference NMR data suggest that calcium-free Ator exerts its effect through interaction with the 16KLVF19 binding site on the Aβ peptide via its aromatic rings as well as hydroxyl and methyl groups. On the other hand, molecular dynamics simulations confirmed that the increasing concentration of Ator is necessary for the inhibition of the conformational transition of Aβ from an α-helix-dominant to a β-sheet-dominant structure.
Collapse
Affiliation(s)
- Hadi Nedaei
- Department of Biophysics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Nasrollah Rezaei-Ghaleh
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany; Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karin Giller
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Stefan Becker
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Leila Karami
- Department of Cell and Molecular Biology, Kharazmi University, Tehran, Iran
| | - Ali Akbar Moosavi-Movahedi
- Department of Biophysics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Christian Griesinger
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
| | - Ali Akbar Saboury
- Department of Biophysics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|
26
|
Sawner AS, Ray S, Yadav P, Mukherjee S, Panigrahi R, Poudyal M, Patel K, Ghosh D, Kummerant E, Kumar A, Riek R, Maji SK. Modulating α-Synuclein Liquid-Liquid Phase Separation. Biochemistry 2021; 60:3676-3696. [PMID: 34431665 DOI: 10.1021/acs.biochem.1c00434] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Liquid-liquid phase separation (LLPS) is a crucial phenomenon for the formation of functional membraneless organelles. However, LLPS is also responsible for protein aggregation in various neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease (PD). Recently, several reports, including ours, have shown that α-synuclein (α-Syn) undergoes LLPS and a subsequent liquid-to-solid phase transition, which leads to amyloid fibril formation. However, how the environmental (and experimental) parameters modulate the α-Syn LLPS remains elusive. Here, we show that in vitro α-Syn LLPS is strongly dependent on the presence of salts, which allows charge neutralization at both terminal segments of protein and therefore promotes hydrophobic interactions supportive for LLPS. Using various purification methods and experimental conditions, we showed, depending upon conditions, α-Syn undergoes either spontaneous (instantaneous) or delayed LLPS. Furthermore, we delineate that the kinetics of liquid droplet formation (i.e., the critical concentration and critical time) is relative and can be modulated by the salt/counterion concentration, pH, presence of surface, PD-associated multivalent cations, and N-terminal acetylation, which are all known to regulate α-Syn aggregation in vitro. Together, our observations suggest that α-Syn LLPS and subsequent liquid-to-solid phase transition could be pathological, which can be triggered only under disease-associated conditions (high critical concentration and/or conditions promoting α-Syn self-assembly). This study will significantly improve our understanding of the molecular mechanisms of α-Syn LLPS and the liquid-to-solid transition.
Collapse
Affiliation(s)
- Ajay Singh Sawner
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Soumik Ray
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Preeti Yadav
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Semanti Mukherjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Rajlaxmi Panigrahi
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Manisha Poudyal
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Dhiman Ghosh
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Eric Kummerant
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Roland Riek
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Samir K Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
27
|
Matsumoto S, Tsunematsu T. Association between Sleep, Alzheimer's, and Parkinson's Disease. BIOLOGY 2021; 10:1127. [PMID: 34827122 PMCID: PMC8614785 DOI: 10.3390/biology10111127] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 01/09/2023]
Abstract
The majority of neurodegenerative diseases are pathologically associated with protein misfolding and aggregation. Alzheimer's disease (AD) is a type of dementia that slowly affects memory and cognitive function, and is characterized by the aggregation of the β-amyloid protein and tau neurofibrillary tangles in the brain. Parkinson's disease (PD) is a movement disorder typically resulting in rigidity and tremor, which is pathologically linked to the aggregation of α-synuclein, particularly in dopaminergic neurons in the midbrain. Sleep disorders commonly occur in AD and PD patients, and it can precede the onset of these diseases. For example, cognitively normal older individuals who have highly fragmented sleep had a 1.5-fold increased risk of subsequently developing AD. This suggests that sleep abnormalities may be a potential biomarker of these diseases. In this review, we describe the alterations of sleep in AD and PD, and discuss their potential in the early diagnosis of these diseases. We further discuss whether sleep disturbance could be a target for the treatment of these diseases.
Collapse
Affiliation(s)
- Sumire Matsumoto
- Advanced Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai 980-8578, Japan;
| | - Tomomi Tsunematsu
- Advanced Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai 980-8578, Japan;
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi 332-0012, Japan
| |
Collapse
|
28
|
Habernig L, Broeskamp F, Aufschnaiter A, Diessl J, Peselj C, Urbauer E, Eisenberg T, de Ory A, Büttner S. Ca2+ administration prevents α-synuclein proteotoxicity by stimulating calcineurin-dependent lysosomal proteolysis. PLoS Genet 2021; 17:e1009911. [PMID: 34780474 PMCID: PMC8629384 DOI: 10.1371/journal.pgen.1009911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 11/29/2021] [Accepted: 10/25/2021] [Indexed: 12/02/2022] Open
Abstract
The capacity of a cell to maintain proteostasis progressively declines during aging. Virtually all age-associated neurodegenerative disorders associated with aggregation of neurotoxic proteins are linked to defects in the cellular proteostasis network, including insufficient lysosomal hydrolysis. Here, we report that proteotoxicity in yeast and Drosophila models for Parkinson's disease can be prevented by increasing the bioavailability of Ca2+, which adjusts intracellular Ca2+ handling and boosts lysosomal proteolysis. Heterologous expression of human α-synuclein (αSyn), a protein critically linked to Parkinson's disease, selectively increases total cellular Ca2+ content, while the levels of manganese and iron remain unchanged. Disrupted Ca2+ homeostasis results in inhibition of the lysosomal protease cathepsin D and triggers premature cellular and organismal death. External administration of Ca2+ reduces αSyn oligomerization, stimulates cathepsin D activity and in consequence restores survival, which critically depends on the Ca2+/calmodulin-dependent phosphatase calcineurin. In flies, increasing the availability of Ca2+ discloses a neuroprotective role of αSyn upon manganese overload. In sum, we establish a molecular interplay between cathepsin D and calcineurin that can be activated by Ca2+ administration to counteract αSyn proteotoxicity.
Collapse
Affiliation(s)
- Lukas Habernig
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Filomena Broeskamp
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Andreas Aufschnaiter
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Jutta Diessl
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Carlotta Peselj
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Elisabeth Urbauer
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Field of Excellence BioHealth-University of Graz, Graz, Austria
| | - Ana de Ory
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Sabrina Büttner
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| |
Collapse
|
29
|
Teng X, Sheveleva A, Tuna F, Willison KR, Ying L. Acetylation Rather than H50Q Mutation Impacts the Kinetics of Cu(II) Binding to α-Synuclein. Chemphyschem 2021; 22:2413-2419. [PMID: 34617653 PMCID: PMC9293329 DOI: 10.1002/cphc.202100651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/02/2021] [Indexed: 11/10/2022]
Abstract
The interaction between α‐synuclein (αSyn) and Cu2+ has been suggested to be closely linked to brain copper homeostasis. Disruption of copper levels could induce misfolding and aggregation of αSyn, and thus contribute to the progression of Parkinson's disease (PD). Understanding the molecular mechanism of αSyn‐Cu2+ interaction is important and controversies in Cu2+ coordination geometry with αSyn still exists. Herein, we find that the pathological H50Q mutation has no impact on the kinetics of Cu2+ binding to the high‐affinity site of wild type αSyn (WT‐αSyn), indicating the non‐involvement of His50 in high‐affinity Cu2+ binding to WT‐αSyn. In contrast, the physiological N‐terminally acetylated αSyn (NAc‐αSyn) displays several orders of magnitude weaker Cu2+ binding affinity than WT‐αSyn. Cu2+ coordination mode to NAc‐αSyn has also been proposed based on EPR spectrum. In addition, we find that Cu2+ coordinated WT‐αSyn is reduction‐active in the presence of GSH, but essentially inactive towards ascorbate. Our work provides new insights into αSyn‐Cu2+ interaction, which may help understand the multifaceted normal functions of αSyn as well as pathological consequences of αSyn aggregation.
Collapse
Affiliation(s)
- Xiangyu Teng
- Department of Chemistry, Imperial College London, White City Campus, London, W12 0BZ, UK
| | - Alena Sheveleva
- Department of Chemistry, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Floriana Tuna
- Department of Chemistry, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Keith R Willison
- Department of Chemistry, Imperial College London, White City Campus, London, W12 0BZ, UK
| | - Liming Ying
- National Heart and Lung Institute, Imperial College London, White City Campus, London, W12 0BZ, UK
| |
Collapse
|
30
|
Ramis R, Ortega-Castro J, Vilanova B, Adrover M, Frau J. Cu 2+, Ca 2+, and methionine oxidation expose the hydrophobic α-synuclein NAC domain. Int J Biol Macromol 2020; 169:251-263. [PMID: 33345970 DOI: 10.1016/j.ijbiomac.2020.12.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/04/2020] [Accepted: 12/03/2020] [Indexed: 11/25/2022]
Abstract
α-Synuclein is an intrinsically disordered protein whose aggregation is related to Parkinson's disease and other neurodegenerative disorders. Metal cations are one of the main factors affecting the propensity of α-synuclein to aggregate, either by directly binding to it or by catalyzing the production of reactive oxygen species that oxidize it. His50, Asp121 and several additional C-terminal α-synuclein residues are binding sites for numerous metal cations, while methionine sulfoxidation occurs readily on this protein under oxidative stress conditions. Molecular dynamics simulations are an excellent tool to obtain a microscopic picture of how metal binding or methionine sulfoxidation alter the conformational preferences of α-synuclein and, hence, its aggregation propensity. In this work, we report the first coarse-grained molecular dynamics study comparing the conformational ensembles of the native protein, the protein bound to either Cu2+ or Ca2+ at its main binding sites, and the methionine-sulfoxidized protein. Our results suggest that these events alter the transient α-synuclein intramolecular contacts, inducing a greater solvent exposure of its hydrophobic, aggregation-prone NAC domain, in full agreement with a recent experimental study on Ca2+ binding. Moreover, metal-binding residues directly participate in the long-range contacts that shield this domain and regulate α-synuclein aggregation. These results provide a molecular-level rationalization of the enhanced fibrillation experimentally observed in the presence of Cu2+ or Ca2+ and the oligomerization induced by methionine sulfoxidation.
Collapse
Affiliation(s)
- Rafael Ramis
- Institut Universitari d'Investigació en Cièencies de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), 07020 Palma de Mallorca, Spain
| | - Joaquín Ortega-Castro
- Institut Universitari d'Investigació en Cièencies de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), 07020 Palma de Mallorca, Spain.
| | - Bartolomé Vilanova
- Institut Universitari d'Investigació en Cièencies de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), 07020 Palma de Mallorca, Spain
| | - Miquel Adrover
- Institut Universitari d'Investigació en Cièencies de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), 07020 Palma de Mallorca, Spain
| | - Juan Frau
- Institut Universitari d'Investigació en Cièencies de la Salut (IUNICS), Departament de Química, Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), 07020 Palma de Mallorca, Spain
| |
Collapse
|
31
|
Ulamec SM, Brockwell DJ, Radford SE. Looking Beyond the Core: The Role of Flanking Regions in the Aggregation of Amyloidogenic Peptides and Proteins. Front Neurosci 2020; 14:611285. [PMID: 33335475 PMCID: PMC7736610 DOI: 10.3389/fnins.2020.611285] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
Amyloid proteins are involved in many neurodegenerative disorders such as Alzheimer’s disease [Tau, Amyloid β (Aβ)], Parkinson’s disease [alpha-synuclein (αSyn)], and amyotrophic lateral sclerosis (TDP-43). Driven by the early observation of the presence of ordered structure within amyloid fibrils and the potential to develop inhibitors of their formation, a major goal of the amyloid field has been to elucidate the structure of the amyloid fold at atomic resolution. This has now been achieved for a wide variety of sequences using solid-state NMR, microcrystallography, X-ray fiber diffraction and cryo-electron microscopy. These studies, together with in silico methods able to predict aggregation-prone regions (APRs) in protein sequences, have provided a wealth of information about the ordered fibril cores that comprise the amyloid fold. Structural and kinetic analyses have also shown that amyloidogenic proteins often contain less well-ordered sequences outside of the amyloid core (termed here as flanking regions) that modulate function, toxicity and/or aggregation rates. These flanking regions, which often form a dynamically disordered “fuzzy coat” around the fibril core, have been shown to play key parts in the physiological roles of functional amyloids, including the binding of RNA and in phase separation. They are also the mediators of chaperone binding and membrane binding/disruption in toxic amyloid assemblies. Here, we review the role of flanking regions in different proteins spanning both functional amyloid and amyloid in disease, in the context of their role in aggregation, toxicity and cellular (dys)function. Understanding the properties of these regions could provide new opportunities to target disease-related aggregation without disturbing critical biological functions.
Collapse
Affiliation(s)
- Sabine M Ulamec
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - David J Brockwell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
32
|
Jeon YM, Kwon Y, Jo M, Lee S, Kim S, Kim HJ. The Role of Glial Mitochondria in α-Synuclein Toxicity. Front Cell Dev Biol 2020; 8:548283. [PMID: 33262983 PMCID: PMC7686475 DOI: 10.3389/fcell.2020.548283] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
The abnormal accumulation of alpha-synuclein (α-syn) aggregates in neurons and glial cells is widely known to be associated with many neurodegenerative diseases, including Parkinson's disease (PD), Dementia with Lewy bodies (DLB), and Multiple system atrophy (MSA). Mitochondrial dysfunction in neurons and glia is known as a key feature of α-syn toxicity. Studies aimed at understanding α-syn-induced toxicity and its role in neurodegenerative diseases have primarily focused on neurons. However, a growing body of evidence demonstrates that glial cells such as microglia and astrocytes have been implicated in the initial pathogenesis and the progression of α-Synucleinopathy. Glial cells are important for supporting neuronal survival, synaptic functions, and local immunity. Furthermore, recent studies highlight the role of mitochondrial metabolism in the normal function of glial cells. In this work, we review the complex relationship between glial mitochondria and α-syn-mediated neurodegeneration, which may provide novel insights into the roles of glial cells in α-syn-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu-Mi Jeon
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Younghwi Kwon
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
- Department of Brain and Cognitive Sciences, DGIST, Daegu, South Korea
| | - Myungjin Jo
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Shinrye Lee
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Seyeon Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
- Department of Brain and Cognitive Sciences, DGIST, Daegu, South Korea
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| |
Collapse
|
33
|
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: 44] [Impact Index Per Article: 8.8] [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
|
34
|
Ross A, Xing V, Wang TT, Bureau SC, Link GA, Fortin T, Zhang H, Hayley S, Sun H. Alleviating toxic α-Synuclein accumulation by membrane depolarization: evidence from an in vitro model of Parkinson's disease. Mol Brain 2020; 13:108. [PMID: 32736645 PMCID: PMC7395353 DOI: 10.1186/s13041-020-00648-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/22/2020] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the formation of toxic, fibrillar form alpha-synuclein (α-Syn) protein aggregates in dopaminergic neurons. Accumulating evidence has shown a multifactorial interplay between the intracellular calcium elevation and α-Syn dynamics. However, whether membrane depolarization regulates toxic α-Syn aggregates remains unclear. To understand this better, we used an in vitro α-Syn preformed fibrils (PFF) model of PD in human neural cells. We demonstrated functional membrane depolarization in differentiated SH-SY5Y cells induced by two independent treatments: high extracellular K+ and the GABAA receptor blocker picrotoxin. We then observed that these treatments significantly alleviated toxic α-Syn aggregation in PFF-treated SH-SY5Y cells. Moreover, clinically relevant direct current stimulation (DCS) also remarkably decreased toxic α-Syn aggregation in PFF-treated SH-SY5Y cells. Taken together, our findings suggest that membrane depolarization plays an important role in alleviating PFF-induced toxic α-Syn aggregates, and that it may represent a novel therapeutic mechanism for PD.
Collapse
Affiliation(s)
- Alysia Ross
- Department of Neuroscience, Carleton University, Ottawa, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Viktoria Xing
- Department of Neuroscience, Carleton University, Ottawa, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Ting Ting Wang
- Department of Neuroscience, Carleton University, Ottawa, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Samantha C Bureau
- Department of Neuroscience, Carleton University, Ottawa, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Giovana A Link
- Department of Neuroscience, Carleton University, Ottawa, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Teresa Fortin
- Department of Neuroscience, Carleton University, Ottawa, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Hui Zhang
- Department of Neurology, SUNY Downstate Medical center, Brooklyn, NY, 11226, USA
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.
| | - Hongyu Sun
- Department of Neuroscience, Carleton University, Ottawa, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.
| |
Collapse
|
35
|
Gonzalez-Rodriguez P, Zampese E, Surmeier DJ. Selective neuronal vulnerability in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2020; 252:61-89. [PMID: 32247375 DOI: 10.1016/bs.pbr.2020.02.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, disabling millions worldwide. Despite the imperative PD poses, at present, there is no cure or means of slowing progression. This gap is attributable to our incomplete understanding of the factors driving pathogenesis. Research over the past several decades suggests that both cell-autonomous and non-cell autonomous processes contribute to the neuronal dysfunction underlying PD symptoms. The thesis of this review is that an intersection of these processes governs the pattern of pathology in PD. Studies of substantia nigra pars compacta (SNc) dopaminergic neurons, whose loss is responsible for the core motor symptoms of PD, suggest that they have a combination of traits-a long, highly branched axon, autonomous activity, and elevated mitochondrial oxidant stress-that predispose them to non-cell autonomous drivers of pathogenesis, like misfolded forms of alpha-synuclein (α-SYN) and inflammation. The literature surrounding these issues will be briefly summarized, and the translational implications of an intersectional hypothesis of PD pathogenesis discussed.
Collapse
Affiliation(s)
| | - Enrico Zampese
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| |
Collapse
|
36
|
Palladino P, Torrini F, Scarano S, Minunni M. Colorimetric analysis of the early oxidation of dopamine by hypochlorous acid as preliminary screening tool for chemical determinants of neuronal oxidative stress. J Pharm Biomed Anal 2020; 179:113016. [DOI: 10.1016/j.jpba.2019.113016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
|
37
|
Britzolaki A, Saurine J, Klocke B, Pitychoutis PM. A Role for SERCA Pumps in the Neurobiology of Neuropsychiatric and Neurodegenerative Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:131-161. [PMID: 31646509 DOI: 10.1007/978-3-030-12457-1_6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Calcium (Ca2+) is a fundamental regulator of cell fate and intracellular Ca2+ homeostasis is crucial for proper function of the nerve cells. Given the complexity of neurons, a constellation of mechanisms finely tunes the intracellular Ca2+ signaling. We are focusing on the sarco/endoplasmic reticulum (SR/ER) calcium (Ca2+)-ATPase (SERCA) pump, an integral ER protein. SERCA's well established role is to preserve low cytosolic Ca2+ levels ([Ca2+]cyt), by pumping free Ca2+ ions into the ER lumen, utilizing ATP hydrolysis. The SERCA pumps are encoded by three distinct genes, SERCA1-3, resulting in 12 known protein isoforms, with tissue-dependent expression patterns. Despite the well-established structure and function of the SERCA pumps, their role in the central nervous system is not clear yet. Interestingly, SERCA-mediated Ca2+ dyshomeostasis has been associated with neuropathological conditions, such as bipolar disorder, schizophrenia, Parkinson's disease and Alzheimer's disease. We summarize here current evidence suggesting a role for SERCA in the neurobiology of neuropsychiatric and neurodegenerative disorders, thus highlighting the importance of this pump in brain physiology and pathophysiology.
Collapse
Affiliation(s)
- Aikaterini Britzolaki
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
| | - Joseph Saurine
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
| | - Benjamin Klocke
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
| | - Pothitos M Pitychoutis
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA.
| |
Collapse
|
38
|
Poulson BG, Szczepski K, Lachowicz JI, Jaremko L, Emwas AH, Jaremko M. Aggregation of biologically important peptides and proteins: inhibition or acceleration depending on protein and metal ion concentrations. RSC Adv 2019; 10:215-227. [PMID: 35492549 PMCID: PMC9047971 DOI: 10.1039/c9ra09350h] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 12/14/2019] [Indexed: 01/03/2023] Open
Abstract
The process of aggregation of proteins and peptides is dependent on the concentration of proteins, and the rate of aggregation can be altered by the presence of metal ions, but this dependence is not always a straightforward relationship. In general, aggregation does not occur under normal physiological conditions, yet it can be induced in the presence of certain metal ions. However, the extent of the influence of metal ion interactions on protein aggregation has not yet been fully comprehended. A consensus has thus been difficult to reach because the acceleration/inhibition of the aggregation of proteins in the presence of metal ions depends on several factors such as pH and the concentration of the aggregated proteins involved as well as metal concentration level of metal ions. Metal ions, like Cu2+, Zn2+, Pb2+ etc. may either accelerate or inhibit aggregation simply because the experimental conditions affect the behavior of biomolecules. It is clear that understanding the relationship between metal ion concentration and protein aggregation will prove useful for future scientific applications. This review focuses on the dependence of the aggregation of selected important biomolecules (peptides and proteins) on metal ion concentrations. We review proteins that are prone to aggregation, the result of which can cause serious neurodegenerative disorders. Furthering our understanding of the relationship between metal ion concentration and protein aggregation will prove useful for future scientific applications, such as finding therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Benjamin Gabriel Poulson
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Kacper Szczepski
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Joanna Izabela Lachowicz
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria 09042 Monserrato Italy
| | - Lukasz Jaremko
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Mariusz Jaremko
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| |
Collapse
|
39
|
Tapias V. Editorial: Mitochondrial Dysfunction and Neurodegeneration. Front Neurosci 2019; 13:1372. [PMID: 31920522 PMCID: PMC6930234 DOI: 10.3389/fnins.2019.01372] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/04/2019] [Indexed: 01/17/2023] Open
Affiliation(s)
- Victor Tapias
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
40
|
Fields CR, Bengoa-Vergniory N, Wade-Martins R. Targeting Alpha-Synuclein as a Therapy for Parkinson's Disease. Front Mol Neurosci 2019; 12:299. [PMID: 31866823 PMCID: PMC6906193 DOI: 10.3389/fnmol.2019.00299] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/22/2019] [Indexed: 01/23/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders with a global burden of approximately 6.1 million patients. Alpha-synuclein has been linked to both the sporadic and familial forms of the disease. Moreover, alpha-synuclein is present in Lewy-bodies, the neuropathological hallmark of PD, and the protein and its aggregation have been widely linked to neurotoxic pathways that ultimately lead to neurodegeneration. Such pathways include autophagy/lysosomal dysregulation, synaptic dysfunction, mitochondrial disruption, and endoplasmic reticulum (ER) and oxidative stress. Alpha-synuclein has not only been shown to alter cellular pathways but also to spread between cells, causing aggregation in host cells. Therapeutic approaches will need to address several, if not all, of these angles of alpha-synuclein toxicity. Here we review the current advances in therapeutic efforts for PD that aim to produce a disease-modifying therapy by targeting the spread, production, aggregation, and degradation of alpha-synuclein. These include: receptor blocking strategies whereby putative alpha-synuclein receptors could be blocked inhibiting alpha-synuclein spread, an alpha-synuclein reduction which will decrease the amount alpha-synuclein available for aggregation and pathway disruption, the use of small molecules in order to target alpha-synuclein aggregation, immunotherapy and the increase of alpha-synuclein degradation by increasing autophagy/lysosomal flux. The research discussed here may lead to a disease-modifying therapy that tackles disease onset and progression in the future.
Collapse
Affiliation(s)
| | - Nora Bengoa-Vergniory
- Department of Physiology, Oxford Parkinson's Disease Center, Anatomy and Genetics, Oxford, United Kingdom
| | - Richard Wade-Martins
- Department of Physiology, Oxford Parkinson's Disease Center, Anatomy and Genetics, Oxford, United Kingdom
| |
Collapse
|
41
|
Ginkgolic acid promotes autophagy-dependent clearance of intracellular alpha-synuclein aggregates. Mol Cell Neurosci 2019; 101:103416. [PMID: 31654699 DOI: 10.1016/j.mcn.2019.103416] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022] Open
Abstract
The accumulation of intracytoplasmic inclusion bodies (Lewy bodies) composed of aggregates of the alpha-synuclein (α-syn) protein is the principal pathological characteristic of Parkinson's disease (PD) and may lead to degeneration of dopaminergic neurons. To date there is no medication that can promote the efficient clearance of these pathological aggregates. In this study, the effect on α-syn aggregate clearance of ginkgolic acid (GA), a natural compound extracted from Ginkgo biloba leaves that inhibits SUMOylation amongst other pathways, was assessed in SH-SY5Y neuroblastoma cells and rat primary cortical neurons. Depolarization of SH-SY5Y neuroblastoma cells and rat primary cortical neurons with KCl was used to induce α-syn aggregate formation. Cells pre-treated with either GA or the related compound, anacardic acid, revealed a significant decrease in intracytoplasmic aggregates immunopositive for α-syn and SUMO-1. An increased frequency of autophagosomes was also detected with both compounds. GA post-treatment 24 h after depolarization also significantly diminished α-syn aggregate bearing cells, indicating the clearance of pre-formed aggregates. Autophagy inhibitors blocked GA-dependent clearance of α-syn aggregates, but not increased autophagosome frequency. Western analysis revealed that the reduction in α-syn aggregate frequency obtained with GA pre-treatment was accompanied by little change in the abundance of SUMO conjugates. The current findings show that GA can promote autophagy-dependent clearance of α-syn aggregates and may have potential in disease modifying therapy.
Collapse
|
42
|
Leandrou E, Emmanouilidou E, Vekrellis K. Voltage-Gated Calcium Channels and α-Synuclein: Implications in Parkinson's Disease. Front Mol Neurosci 2019; 12:237. [PMID: 31649506 PMCID: PMC6794345 DOI: 10.3389/fnmol.2019.00237] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/17/2019] [Indexed: 12/15/2022] Open
Abstract
Alpha-synuclein (α-syn) is biochemically and genetically linked to Parkinson's disease (PD) and other synucleinopathies. It is now widely accepted that α-syn can be released in the extracellular space, even though the mechanism of its release is still unclear. In addition, pathology-related aggregated species of α-syn have been shown to propagate between neurons in synaptically connected areas of the brain thereby assisting the spreading of pathology in healthy neighboring neuronal cells. In neurons, calcium channels are key signaling elements that modulate the release of bioactive molecules (hormones, proteins, and neurotransmitters) through calcium sensing. Such calcium sensing activity is determined by the distinct biophysical and pharmacological properties and the ability of calcium channels to interact with other modulatory proteins. Although the function of extracellular α-syn is currently unknown, previous work suggested the presence of a calcium-dependent mechanism for α-syn secretion both in vitro, in neuronal cells in culture, and also in vivo, in the context of a trans-neuronal network in brain. Mechanisms regulating extracellular α-syn levels may be of particular importance as they could represent novel therapeutic targets. We discuss here how calcium channel activity may contribute to α-syn aggregation and secretion as a pathway to disease progression in synucleinopathies.
Collapse
Affiliation(s)
- Emmanouela Leandrou
- Center for Basic Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Evangelia Emmanouilidou
- Center for Basic Research, Biomedical Research Foundation Academy of Athens, Athens, Greece.,Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Vekrellis
- Center for Basic Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| |
Collapse
|
43
|
González N, Arcos-López T, König A, Quintanar L, Menacho Márquez M, Outeiro TF, Fernández CO. Effects of alpha-synuclein post-translational modifications on metal binding. J Neurochem 2019; 150:507-521. [PMID: 31099098 DOI: 10.1111/jnc.14721] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/11/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Abstract
Parkinson's disease is the second most common neurodegenerative disorder worldwide. Neurodegeneration in this pathology is characterized by the loss of dopaminergic neurons in the substantia nigra, coupled with cytoplasmic inclusions known as Lewy bodies containing α-synuclein. The brain is an organ that concentrates metal ions, and there is emerging evidence that a break-down in metal homeostasis may be a critical factor in a variety of neurodegenerative diseases. α-synuclein has emerged as an important metal-binding protein in the brain, whereas these interactions play an important role in its aggregation and might represent a link between protein aggregation, oxidative damage, and neuronal cell loss. Additionally, α-synuclein undergoes several post-translational modifications that regulate its structure and physiological function, and may be linked to the aggregation and/or oligomer formation. This review is focused on the interaction of this protein with physiologically relevant metal ions, highlighting the cases where metal-AS interactions profile as key modulators for its structural, aggregation, and membrane-binding properties. The impact of α-synuclein phosphorylation and N-terminal acetylation in the metal-binding properties of the protein are also discussed, underscoring a potential interplay between PTMs and metal ion binding in regulating α-synuclein physiological functions and its role in pathology. This article is part of the Special Issue "Synuclein".
Collapse
Affiliation(s)
- Nazareno González
- Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC), Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR, UNR-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Trinidad Arcos-López
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Liliana Quintanar
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Mauricio Menacho Márquez
- Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC), Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR, UNR-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| | - Claudio O Fernández
- Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC), Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR, UNR-CONICET), Universidad Nacional de Rosario, Rosario, Argentina.,Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
44
|
Sgobio C, Sun L, Ding J, Herms J, Lovinger DM, Cai H. Unbalanced calcium channel activity underlies selective vulnerability of nigrostriatal dopaminergic terminals in Parkinsonian mice. Sci Rep 2019; 9:4857. [PMID: 30890737 PMCID: PMC6425036 DOI: 10.1038/s41598-019-41091-7] [Citation(s) in RCA: 12] [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: 11/23/2018] [Accepted: 02/18/2019] [Indexed: 11/23/2022] Open
Abstract
Dopamine (DA) release in striatum is functionally segregated across a dorsolateral/ventromedial axis. Interestingly, nigrostriatal DA signaling disruption in Parkinson's disease (PD) preferentially affects the dorsolateral striatum. The relationship between afferent presynaptic calcium transients (PreCaTs) in DA terminals and DA release in dorsolateral (Caudato-Putamen, DLS) and ventromedial (Nucleus Accumbens Shell, VS) striatal subregions was examined by ex vivo real-time dual-recording in conditional transgenic mice expressing the calcium indicator protein GCaMP3. In DLS, minimal increases in cytosolic calcium trigger steep DA release while PreCaTs and DA release in VS both were proportional to the number of pulses in burst stimulation. Co-expressing α-synuclein with the Parkinson's disease (PD)-associated A53T mutation and GCaMP3 in midbrain DA neurons revealed augmented cytosolic steady state and activity-dependent intra-terminal calcium levels preferentially in DLS, as well as hyperactivation and enhanced expression of N-type calcium channels. Thus, unbalanced calcium channel activity is a presynaptic mechanism to consider in the multifaceted pathogenic pathways of progressive neurodegeneration.
Collapse
Affiliation(s)
- Carmelo Sgobio
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Translational research division, German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jochen Herms
- Translational research division, German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, Munich, 81377, Germany
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, Section on Synaptic Pharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA.
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
45
|
Rane P, Sarmah D, Bhute S, Kaur H, Goswami A, Kalia K, Borah A, Dave KR, Sharma N, Bhattacharya P. Novel Targets for Parkinson's Disease: Addressing Different Therapeutic Paradigms and Conundrums. ACS Chem Neurosci 2019; 10:44-57. [PMID: 29957921 DOI: 10.1021/acschemneuro.8b00180] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease that is pathologically characterized by degeneration of dopamine neurons in the substantia nigra pars compacta (SNpc). PD leads to clinical motor features that include rigidity, tremor, and bradykinesia. Despite multiple available therapies for PD, the clinical features continue to progress, and patients suffer progressive disability. Many advances have been made in PD therapy which directly target the cause of the disease rather than providing symptomatic relief. A neuroprotective or disease modifying strategy that can slow or cease clinical progression and worsening disability remains as a major unmet medical need for PD management. The present review discusses potential novel therapies for PD that include recent interventions in the form of immunomodulatory techniques and stem cell therapy. Further, an introspective approach to identify numerous other novel targets that can alleviate PD pathogenesis and enable physicians to practice multitargeted therapy and that may provide a ray of hope to PD patients in the future are discussed.
Collapse
Affiliation(s)
- Pallavi Rane
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Shashikala Bhute
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Avirag Goswami
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| | - Anupom Borah
- Department of Life Science and Bioinformatics, Assam University, Silchar, Assam 788011, India
| | - Kunjan R. Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Nutan Sharma
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat-382355, India
| |
Collapse
|
46
|
The Cellular Environment Affects Monomeric α-Synuclein Structure. Trends Biochem Sci 2018; 44:453-466. [PMID: 30527975 DOI: 10.1016/j.tibs.2018.11.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022]
Abstract
The presynaptic protein α-synuclein (aSyn) is an 'intrinsically disordered protein' that is highly dynamic in conformation. Transient intramolecular interactions between its charged N and C termini, and between its hydrophobic region and the C terminus, prevent self-association. These interactions inhibit the formation of insoluble inclusions, which are the pathological hallmark of Parkinson's disease and many other synucleinopathies. This review discusses how these intramolecular interactions are influenced by the specific environment aSyn is in. We discuss how charge, pH, calcium, and salt affect the physiological structure of monomeric aSyn, and how they may favour the formation of toxic structures. The more we understand the dynamic conformations of aSyn, the better we can design desperately needed therapeutics to prevent disease progression.
Collapse
|
47
|
Rowlands J, Heng J, Newsholme P, Carlessi R. Pleiotropic Effects of GLP-1 and Analogs on Cell Signaling, Metabolism, and Function. Front Endocrinol (Lausanne) 2018; 9:672. [PMID: 30532733 PMCID: PMC6266510 DOI: 10.3389/fendo.2018.00672] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
The incretin hormone Glucagon-Like Peptide-1 (GLP-1) is best known for its "incretin effect" in restoring glucose homeostasis in diabetics, however, it is now apparent that it has a broader range of physiological effects in the body. Both in vitro and in vivo studies have demonstrated that GLP-1 mimetics alleviate endoplasmic reticulum stress, regulate autophagy, promote metabolic reprogramming, stimulate anti-inflammatory signaling, alter gene expression, and influence neuroprotective pathways. A substantial body of evidence has accumulated with respect to how GLP-1 and its analogs act to restore and maintain normal cellular functions. These findings have prompted several clinical trials which have reported GLP-1 analogs improve cardiac function, restore lung function and reduce mortality in patients with obstructive lung disease, influence blood pressure and lipid storage, and even prevent synaptic loss and neurodegeneration. Mechanistically, GLP-1 elicits its effects via acute elevation in cAMP levels, and subsequent protein kinase(s) activation, pathways well-defined in pancreatic β-cells which stimulate insulin secretion in conjunction with elevated Ca2+ and ATP. More recently, new studies have shed light on additional downstream pathways stimulated by chronic GLP-1 exposure, findings which have direct relevance to our understanding of the potential therapeutic effects of longer lasting analogs recently developed for clinical use. In this review, we provide a comprehensive description of the diverse roles for GLP-1 across multiple tissues, describe downstream pathways stimulated by acute and chronic exposure, and discuss novel pleiotropic applications of GLP-1 mimetics in the treatment of human disease.
Collapse
Affiliation(s)
| | | | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, WA, Australia
| | - Rodrigo Carlessi
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, WA, Australia
| |
Collapse
|
48
|
Melo TQ, Copray SJCVM, Ferrari MFR. Alpha-Synuclein Toxicity on Protein Quality Control, Mitochondria and Endoplasmic Reticulum. Neurochem Res 2018; 43:2212-2223. [DOI: 10.1007/s11064-018-2673-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/14/2018] [Accepted: 10/25/2018] [Indexed: 12/16/2022]
|
49
|
Camacho R, Täuber D, Hansen C, Shi J, Bousset L, Melki R, Li JY, Scheblykin IG. 2D polarization imaging as a low-cost fluorescence method to detect α-synuclein aggregation ex vivo in models of Parkinson's disease. Commun Biol 2018; 1:157. [PMID: 30302401 PMCID: PMC6168587 DOI: 10.1038/s42003-018-0156-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 08/28/2018] [Indexed: 12/15/2022] Open
Abstract
A hallmark of Parkinson's disease is the formation of large protein-rich aggregates in neurons, where α-synuclein is the most abundant protein. A standard approach to visualize aggregation is to fluorescently label the proteins of interest. Then, highly fluorescent regions are assumed to contain aggregated proteins. However, fluorescence brightness alone cannot discriminate micrometer-sized regions with high expression of non-aggregated proteins from regions where the proteins are aggregated on the molecular scale. Here, we demonstrate that 2-dimensional polarization imaging can discriminate between preformed non-aggregated and aggregated forms of α-synuclein, and detect increased aggregation in brain tissues of transgenic mice. This imaging method assesses homo-FRET between labels by measuring fluorescence polarization in excitation and emission simultaneously, which translates into higher contrast than fluorescence anisotropy imaging. Exploring earlier aggregation states of α-synuclein using such technically simple imaging method could lead to crucial improvements in our understanding of α-synuclein-mediated pathology in Parkinson's Disease.
Collapse
Affiliation(s)
- Rafael Camacho
- Chemical Physics and NanoLund, Lund University, P.O. Box 124,, 22100, Lund, Sweden
- Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001, Leuven, Belgium
| | - Daniela Täuber
- Chemical Physics and NanoLund, Lund University, P.O. Box 124,, 22100, Lund, Sweden
- Biopolarisation, Leibniz Institute of Photonic Technology, Albert-Einstein-Str. 9, 07745, Jena, Germany
- Institute of Solid State Physics, FSU Jena, Helmholtzweg 3, 07743, Jena, Germany
| | - Christian Hansen
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC A10, 22184, Lund, Sweden
- Molecular Neurobiology, Department of Experimental Medical Science, BMC B11, 221 84, Lund, Sweden
| | - Juanzi Shi
- Chemical Physics and NanoLund, Lund University, P.O. Box 124,, 22100, Lund, Sweden
| | - Luc Bousset
- Institut Fancois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, 18 Route du Panorama, 92265, Fontenay-Aux-Roses cedex, France
| | - Ronald Melki
- Institut Fancois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, 18 Route du Panorama, 92265, Fontenay-Aux-Roses cedex, France
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC A10, 22184, Lund, Sweden.
- Institute of Health Sciences, China Medical University, 110122, Shenyang, People's Republic of China.
| | - Ivan G Scheblykin
- Chemical Physics and NanoLund, Lund University, P.O. Box 124,, 22100, Lund, Sweden.
| |
Collapse
|
50
|
Choi ML, Gandhi S. Crucial role of protein oligomerization in the pathogenesis of Alzheimer's and Parkinson's diseases. FEBS J 2018; 285:3631-3644. [PMID: 29924502 DOI: 10.1111/febs.14587] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/11/2018] [Accepted: 06/14/2018] [Indexed: 12/16/2022]
Abstract
Misfolding and aggregation of the proteins amyloid-β, tau and alpha-synuclein is the predominant pathology underlying the neurodegenerative disorders, Alzheimer's and Parkinson's disease. While end stage insoluble products of aggregation have been well characterised in human and animal models of disease, accumulating evidence from biophysical, cellular and in vivo studies has shown that soluble intermediates of aggregation, or oligomers, may be the key species that mediate toxicity and underlie seeding and spreading in disease. Here, we review the process of protein misfolding, and the intrinsic and extrinsic processes that cause the native states of the key aggregating proteins to undergo conformational change to form oligomers and ultimately fibrils. We discuss the structural features of the key toxic intermediate, and describe the putative mechanisms by which oligomers may cause cell toxicity. Finally, we explore the potential therapeutic approaches raised by the oligomer hypothesis in neurodegenerative disease.
Collapse
Affiliation(s)
- Minee L Choi
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK.,The Francis Crick Institute, London, UK
| | - Sonia Gandhi
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK.,The Francis Crick Institute, London, UK
| |
Collapse
|