1
|
Hyde VR, Zhou C, Fernandez JR, Chatterjee K, Ramakrishna P, Lin A, Fisher GW, Çeliker OT, Caldwell J, Bender O, Sauer PJ, Lugo-Martinez J, Bar DZ, D'Aiuto L, Shemesh OA. Anti-herpetic tau preserves neurons via the cGAS-STING-TBK1 pathway in Alzheimer's disease. Cell Rep 2025; 44:115109. [PMID: 39753133 DOI: 10.1016/j.celrep.2024.115109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on the presence of extracellular β-amyloid (Aβ) and intracellular hyperphosphorylated tau (p-tau). Emerging evidence suggests a potential link between AD pathologies and infectious agents, with herpes simplex virus 1 (HSV-1) being a leading candidate. Our investigation, using metagenomics, mass spectrometry, western blotting, and decrowding expansion pathology, detects HSV-1-associated proteins in human brain samples. Expression of the herpesvirus protein ICP27 increases with AD severity and strongly colocalizes with p-tau but not with Aβ. Modeling in human brain organoids shows that HSV-1 infection elevates tau phosphorylation. Notably, p-tau reduces ICP27 expression and markedly decreases post-infection neuronal death from 64% to 7%. This modeling prompts investigation into the cGAS-STING-TBK1 pathway products, nuclear factor (NF)-κB and IRF-3, which colocalizes with ICP27 and p-tau in AD. Furthermore, experimental activation of STING enhances tau phosphorylation, while TBK1 inhibition prevents it. Together, these findings suggest that tau phosphorylation acts as an innate immune response in AD, driven by cGAS-STING.
Collapse
Affiliation(s)
- Vanesa R Hyde
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoming Zhou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Juan R Fernandez
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krishnashis Chatterjee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pururav Ramakrishna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amanda Lin
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Gregory W Fisher
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Orhan Tunç Çeliker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jill Caldwell
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peter Joseph Sauer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Or A Shemesh
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
2
|
Konsta V, Paschou M, Koti N, Vlachou ME, Livanos P, Xilouri M, Papazafiri P. Neurosteroids Alter p-ERK Levels and Tau Distribution, Restraining the Effects of High Extracellular Calcium. Int J Mol Sci 2024; 25:11637. [PMID: 39519194 PMCID: PMC11546054 DOI: 10.3390/ijms252111637] [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: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Neurosteroids are undeniably regarded as neuroprotective mediators, regulating brain function by rapid non-genomic actions involving interference with microtubules. Conversely, hyperphosphorylated Tau is considered responsible for the onset of a plethora of neurodegenerative diseases, as it dissociates from microtubules, leading to their destabilization, thus impairing synaptic vesicle transport and neurotransmission. Consequently, we aimed to investigate the effects of neurosteroids, specifically allopregnanolone (Allo) and dehydroepiandrosterone (DHEA), on the levels of total and phosphorylated at Serine 404 Tau (p-Tau) in C57BL/6 mice brain slices. In total tissue extracts, we found that neurosteroids elevated both total and p-Tau levels without significantly altering the p-Tau/Tau ratio. In addition, the levels of several enzymes implicated in Tau phosphorylation did not display significant differences between conditions, suggesting that neurosteroids influence Tau distribution rather than its phosphorylation. Hence, we subsequently examined the mitochondria-enriched subcellular fraction where, again, both p-Tau and total Tau levels were increased in the presence of neurosteroids. These effects seem actin-dependent, as disrupting actin polymerization by cytochalasin B preserved Tau levels. Furthermore, co-incubation with high [Ca2+] and neurosteroids mitigated the effects of Ca2+ overload, pointing to cytoskeletal remodeling as a potential mechanism underlying neurosteroid-induced neuroprotection.
Collapse
Affiliation(s)
- Vasiliki Konsta
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, University Campus, 15784 Athens, Greece; (V.K.); (M.P.); (N.K.); (M.E.V.)
| | - Maria Paschou
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, University Campus, 15784 Athens, Greece; (V.K.); (M.P.); (N.K.); (M.E.V.)
| | - Nikoleta Koti
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, University Campus, 15784 Athens, Greece; (V.K.); (M.P.); (N.K.); (M.E.V.)
| | - Maria Evangelia Vlachou
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, University Campus, 15784 Athens, Greece; (V.K.); (M.P.); (N.K.); (M.E.V.)
| | - Pantelis Livanos
- Division of Cell Biology, Department of Biology, Friedrich-Alexander University of Erlangen-Nuremberg, Staudtstrasse 5, 91058 Erlangen, Germany;
| | - Maria Xilouri
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 4, Soranou Efesiou Street, 11527 Athens, Greece;
| | - Panagiota Papazafiri
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, University Campus, 15784 Athens, Greece; (V.K.); (M.P.); (N.K.); (M.E.V.)
| |
Collapse
|
3
|
The ubiquitous microtubule-associated protein 4 (MAP4) controls organelle distribution by regulating the activity of the kinesin motor. Proc Natl Acad Sci U S A 2022; 119:e2206677119. [PMID: 36191197 PMCID: PMC9565364 DOI: 10.1073/pnas.2206677119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regulation of organelle transport by molecular motors along the cytoskeletal microtubules is central to maintaining cellular functions. Here, we show that the ubiquitous tau-related microtubule-associated protein 4 (MAP4) can bias the bidirectional transport of organelles toward the microtubule minus-ends. This is concurrent with MAP4 phosphorylation, mediated by the kinase GSK3β. We demonstrate that MAP4 achieves this bias by tethering the cargo to the microtubules, allowing it to impair the force generation of the plus-end motor kinesin-1. Consistent with this mechanism, MAP4 physically interacts with dynein and dynactin and, when phosphorylated, associates with the cargo-motor complex through its projection domain. Its phosphorylation coincides with the perinuclear accumulation of organelles, a phenotype that is rescued by abolishing the cargo-microtubule MAP4 tether or by the pharmacological inhibition of dynein, confirming the ability of kinesin to inch along, albeit inefficiently, in the presence of phosphorylated MAP4. These findings have broad biological significance because of the ubiquity of MAP4 and the involvement of GSK3β in multiple diseases, more specifically in cancer, where the MAP4-dependent redistribution of organelles may be prevalent in cancer cells, as we demonstrate here for mitochondria in lung carcinoma epithelial cells.
Collapse
|
4
|
Nguyen H, Zhu W, Baltan S. Casein Kinase 2 Signaling in White Matter Stroke. Front Mol Biosci 2022; 9:908521. [PMID: 35911974 PMCID: PMC9325966 DOI: 10.3389/fmolb.2022.908521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/21/2022] [Indexed: 11/27/2022] Open
Abstract
The growth of the aging population, together with improved stroke care, has resulted in an increase in stroke survivors and a rise in recurrent events. Axonal injury and white matter (WM) dysfunction are responsible for much of the disability observed after stroke. The mechanisms of WM injury are distinct compared to gray matter and change with age. Therefore, an ideal stroke therapeutic must restore neuronal and axonal function when applied before or after a stroke, and it must also protect across age groups. Casein kinase 2 (CK2), is expressed in the brain, including WM, and is regulated during the development and numerous disease conditions such as cancer and ischemia. CK2 activation in WM mediates ischemic injury by activating the Cdk5 and AKT/GSK3β signaling pathways. Consequently, CK2 inhibition using the small molecule inhibitor CX-4945 (Silmitasertib) correlates with preservation of oligodendrocytes, conservation of axon structure, and axonal mitochondria, leading to improved functional recovery. Remarkably, CK2 inhibition promotes WM function when applied after ischemic injury by specifically regulating the AKT/GSK3β pathways. The blockade of the active conformation of AKT confers post-ischemic protection to young and old WM by preserving mitochondria, implying AKT as a common therapeutic target across age groups. Using a NanoString nCounter miRNA expression profiling, comparative analyses of ischemic WM with or without CX-4945 treatment reveal that miRNAs are expressed at high levels in WM after ischemia, and CX-4945 differentially regulates some of these miRNAs. Therefore, we propose that miRNA regulation may be one of the protective actions of CX-4945 against WM ischemic injury. Silmitasertib is FDA approved and currently in use for cancer and Covid patients; therefore, it is plausible to repurpose CK2 inhibitors for stroke patients.
Collapse
Affiliation(s)
| | | | - Selva Baltan
- Anesthesiology and Peri-Operative Medicine (APOM), Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
5
|
Heo K, Basu H, Gutnick A, Wei W, Shlevkov E, Schwarz TL. Serine/Threonine Protein Phosphatase 2A Regulates the Transport of Axonal Mitochondria. Front Cell Neurosci 2022; 16:852245. [PMID: 35370563 PMCID: PMC8973303 DOI: 10.3389/fncel.2022.852245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Microtubule-based transport provides mitochondria to distant regions of neurons and is essential for neuronal health. To identify compounds that increase mitochondrial motility, we screened 1,641 small-molecules in a high-throughput screening platform. Indirubin and cantharidin increased mitochondrial motility in rat cortical neurons. Cantharidin is known to inhibit protein phosphatase 2A (PP2A). We therefore tested two other inhibitors of PP2A: LB-100 and okadaic acid. LB-100 increased mitochondrial motility, but okadaic acid did not. To resolve this discrepancy, we knocked down expression of the catalytic subunit of PP2A (PP2CA). This long-term inhibition of PP2A more than doubled retrograde transport of axonal mitochondria, confirming the importance of PP2A as a regulator of mitochondrial motility and as the likely mediator of cantharidin's effect.
Collapse
Affiliation(s)
- Keunjung Heo
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Himanish Basu
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Amos Gutnick
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Wei Wei
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Evgeny Shlevkov
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Thomas L Schwarz
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
6
|
Zinsmaier KE. Mitochondrial Miro GTPases coordinate mitochondrial and peroxisomal dynamics. Small GTPases 2021; 12:372-398. [PMID: 33183150 PMCID: PMC8583064 DOI: 10.1080/21541248.2020.1843957] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondria and peroxisomes are highly dynamic, multifunctional organelles. Both perform key roles for cellular physiology and homoeostasis by mediating bioenergetics, biosynthesis, and/or signalling. To support cellular function, they must be properly distributed, of proper size, and be able to interact with other organelles. Accumulating evidence suggests that the small atypical GTPase Miro provides a central signalling node to coordinate mitochondrial as well as peroxisomal dynamics. In this review, I summarize our current understanding of Miro-dependent functions and molecular mechanisms underlying the proper distribution, size and function of mitochondria and peroxisomes.
Collapse
Affiliation(s)
- Konrad E. Zinsmaier
- Departments of Neuroscience and Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
7
|
GSK3 as a Regulator of Cytoskeleton Architecture: Consequences for Health and Disease. Cells 2021; 10:cells10082092. [PMID: 34440861 PMCID: PMC8393567 DOI: 10.3390/cells10082092] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/06/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) was initially isolated as a critical protein in energy metabolism. However, subsequent studies indicate that GSK-3 is a multi-tasking kinase that links numerous signaling pathways in a cell and plays a vital role in the regulation of many aspects of cellular physiology. As a regulator of actin and tubulin cytoskeleton, GSK3 influences processes of cell polarization, interaction with the extracellular matrix, and directional migration of cells and their organelles during the growth and development of an animal organism. In this review, the roles of GSK3–cytoskeleton interactions in brain development and pathology, migration of healthy and cancer cells, and in cellular trafficking of mitochondria will be discussed.
Collapse
|
8
|
Mechanisms and Therapeutic Implications of GSK-3 in Treating Neurodegeneration. Cells 2021; 10:cells10020262. [PMID: 33572709 PMCID: PMC7911291 DOI: 10.3390/cells10020262] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders are spreading worldwide and are one of the greatest threats to public health. There is currently no adequate therapy for these disorders, and therefore there is an urgent need to accelerate the discovery and development of effective treatments. Although neurodegenerative disorders are broad ranging and highly complex, they may share overlapping mechanisms, and thus potentially manifest common targets for therapeutic interventions. Glycogen synthase kinase-3 (GSK-3) is now acknowledged to be a central player in regulating mood behavior, cognitive functions, and neuron viability. Indeed, many targets controlled by GSK-3 are critically involved in progressing neuron deterioration and disease pathogenesis. In this review, we focus on three pathways that represent prominent mechanisms linking GSK-3 with neurodegenerative disorders: cytoskeleton organization, the mammalian target of rapamycin (mTOR)/autophagy axis, and mitochondria. We also consider the challenges and opportunities in the development of GSK-3 inhibitors for treating neurodegeneration.
Collapse
|
9
|
Peng Y, Gao P, Shi L, Chen L, Liu J, Long J. Central and Peripheral Metabolic Defects Contribute to the Pathogenesis of Alzheimer's Disease: Targeting Mitochondria for Diagnosis and Prevention. Antioxid Redox Signal 2020; 32:1188-1236. [PMID: 32050773 PMCID: PMC7196371 DOI: 10.1089/ars.2019.7763] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 02/09/2020] [Accepted: 02/10/2020] [Indexed: 12/20/2022]
Abstract
Significance: Epidemiological studies indicate that metabolic disorders are associated with an increased risk for Alzheimer's disease (AD). Metabolic remodeling occurs in the central nervous system (CNS) and periphery, even in the early stages of AD. Mitochondrial dysfunction has been widely accepted as a molecular mechanism underlying metabolic disorders. Therefore, focusing on early metabolic changes, especially from the perspective of mitochondria, could be of interest for early AD diagnosis and intervention. Recent Advances: We and others have identified that the levels of several metabolites are fluctuated in the periphery before their accumulation in the CNS, which plays an important role in the pathogenesis of AD. Mitochondrial remodeling is likely one of the earliest signs of AD, linking nutritional imbalance to cognitive deficits. Notably, by improving mitochondrial function, mitochondrial nutrients efficiently rescue cellular metabolic dysfunction in the CNS and periphery in individuals with AD. Critical Issues: Peripheral metabolic disorders should be intensively explored and evaluated for the early diagnosis of AD. The circulating metabolites derived from mitochondrial remodeling represent novel potential diagnostic biomarkers for AD that are more readily detected than CNS-oriented biomarkers. Moreover, mitochondrial nutrients provide a promising approach to preventing and delaying AD progression. Future Directions: Abnormal mitochondrial metabolism in the CNS and periphery is involved in AD pathogenesis. More clinical studies provide evidence for the suitability and reliability of circulating metabolites and cytokines for the early diagnosis of AD. Targeting mitochondria to rewire cellular metabolism is a promising approach to preventing AD and ameliorating AD-related metabolic disorders.
Collapse
Affiliation(s)
- Yunhua Peng
- Center for Mitochondrial Biology & Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Peipei Gao
- Center for Mitochondrial Biology & Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Le Shi
- Center for Mitochondrial Biology & Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Lei Chen
- Center for Mitochondrial Biology & Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jiankang Liu
- Center for Mitochondrial Biology & Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jiangang Long
- Center for Mitochondrial Biology & Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
10
|
Chakravorty A, Jetto CT, Manjithaya R. Dysfunctional Mitochondria and Mitophagy as Drivers of Alzheimer's Disease Pathogenesis. Front Aging Neurosci 2019; 11:311. [PMID: 31824296 PMCID: PMC6880761 DOI: 10.3389/fnagi.2019.00311] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022] Open
Abstract
Neurons are highly specialized post-mitotic cells that are inherently dependent on mitochondria owing to their high bioenergetic demand. Mitochondrial dysfunction is therefore associated with various age-related neurodegenerative disorders such as Alzheimer's disease (AD), wherein accumulation of damaged and dysfunctional mitochondria has been reported as an early symptom further contributing to disease progression. In AD, impairment of mitochondrial function causes bioenergetic deficiency, intracellular calcium imbalance and oxidative stress, thereby aggravating the effect of Aβ and tau pathologies, leading to synaptic dysfunction, cognitive impairment and memory loss. Although there are reports suggesting intricate parallelism between mitochondrial dysfunction and AD pathologies such as Aβ aggregation and hyperphosphorylated tau accumulation, the factors that drive the pathogenesis of either are unclear. In addition, emerging evidence suggest that mitochondrial quality control (QC) mechanisms such as mitophagy are impaired in AD. As an important mitochondrial QC mechanism, mitophagy plays a critical role in maintaining neuronal health and function. Studies show that various proteins involved in mitophagy, mitochondrial dynamics, and mitochondrial biogenesis are affected in AD. Compromised mitophagy may also be attributed to impairment in autophagosome-lysosome fusion and defects in lysosomal acidification. Therapeutic interventions aiming to restore mitophagy functions can be used as a strategy for ameliorating AD pathogenesis. Recent evidence implicates the role of microglial activation via mitophagy induction in reducing amyloid plaque load. This review summarizes the current developments in the field of mitophagy and mitochondrial dysfunction in AD.
Collapse
Affiliation(s)
- Anushka Chakravorty
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Cuckoo Teresa Jetto
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| |
Collapse
|
11
|
Amyloid beta-mediated KIF5A deficiency disrupts anterograde axonal mitochondrial movement. Neurobiol Dis 2019; 127:410-418. [PMID: 30923004 DOI: 10.1016/j.nbd.2019.03.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 11/22/2022] Open
Abstract
Mitochondria are crucial organelles for neurophysiology and brain mitochondrial defects constitute a characteristic of Alzheimer's disease (AD). Impaired axonal mitochondrial traffic, especially the anterograde axonal mitochondrial transport is a pronouncing mitochondrial defect that underlies synaptic failure in AD-related conditions. However, the detailed molecular mechanisms of such axonal mitochondrial abnormality have not been fully understood. KIF5A is a key isoform of kinesin-1, which is a key molecular machinery in facilitating anterograde axonal mitochondrial transport. In this study, we have determined a downregulation of KIF5A in postmortem AD temporal lobes. Further experiments on amyloid beta (Aβ)-treated primary neuron culture and 5 × FAD mice suggest a close association of Aβ toxicity and KIF5A loss. Downregulation of KIF5A mimics Aβ-induced axonal mitochondrial transport deficits, indicating a potential role of KIF5A deficiency in AD-relevant axonal mitochondrial traffic abnormalities. Importantly, the restoration of KIF5A corrects Aβ-induced impairments in axonal mitochondrial transport, especially the anterograde traffic, with little or no impact on retrograde axonal mitochondrial motility. Our findings suggest a novel KIF5A-associated mechanism conferring Aβ toxicity to axonal mitochondrial deficits. Furthermore, the results implicate a potential therapeutic avenue by protecting KIF5A function for the treatment of AD.
Collapse
|
12
|
Vergara C, Houben S, Suain V, Yilmaz Z, De Decker R, Vanden Dries V, Boom A, Mansour S, Leroy K, Ando K, Brion JP. Amyloid-β pathology enhances pathological fibrillary tau seeding induced by Alzheimer PHF in vivo. Acta Neuropathol 2019; 137:397-412. [PMID: 30599077 DOI: 10.1007/s00401-018-1953-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022]
Abstract
Neuropathological analysis in Alzheimer's disease (AD) and experimental evidence in transgenic models overexpressing frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17) mutant tau suggest that amyloid-β pathology enhances the development of tau pathology. In this work, we analyzed this interaction independently of the overexpression of an FTDP-17 mutant tau, by analyzing tau pathology in wild-type (WT), 5xFAD, APP-/- and tau-/- mice after stereotaxic injection in the somatosensory cortex of short-length native human AD-PHF. Gallyas and phosphotau-positive tau inclusions developed in WT, 5xFAD, and APP-/- but not in tau-/- mice. Ultrastructural analysis demonstrated their intracellular localization and that they were composed of straight filaments. These seeded tau inclusions were composed only of endogenous murine tau exhibiting a tau antigenic profile similar to tau aggregates in AD. Insoluble tau level was higher and ipsilateral anteroposterior and contralateral cortical spreading of tau inclusions was more important in AD-PHF-injected 5xFAD mice than in WT mice. The formation of large plaque-associated dystrophic neurites positive for oligomeric and phosphotau was observed in 5xFAD mice injected with AD-PHF but never in control-injected or in non-injected 5xFAD mice. An increased level of the p25 activator of CDK5 kinase was found in AD-PHF-injected 5xFAD mice. These data demonstrate in vivo that the presence of Aβ pathology enhances experimentally induced tau seeding of endogenous, wild-type tau expressed at physiological level, and demonstrate the fibrillar nature of heterotopically seeded endogenous tau. These observations further support the hypothesis that Aβ enhances tau pathology development in AD through increased pathological tau spreading.
Collapse
Affiliation(s)
- Cristina Vergara
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Sarah Houben
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Valérie Suain
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Robert De Decker
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Virginie Vanden Dries
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Alain Boom
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Salwa Mansour
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium.
| |
Collapse
|
13
|
Bastian C, Quinn J, Tripathi A, Aquila D, McCray A, Dutta R, Baltan S, Brunet S. CK2 inhibition confers functional protection to young and aging axons against ischemia by differentially regulating the CDK5 and AKT signaling pathways. Neurobiol Dis 2018; 126:47-61. [PMID: 29944965 DOI: 10.1016/j.nbd.2018.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/08/2018] [Accepted: 05/21/2018] [Indexed: 12/25/2022] Open
Abstract
White matter (WM) is injured in most strokes, which contributes to functional deficits during recovery. Casein kinase 2 (CK2) is a protein kinase that is expressed in brain, including WM. To assess the impact of CK2 inhibition on axon recovery following oxygen glucose deprivation (OGD), mouse optic nerves (MONs), which are pure WM tracts, were subjected to OGD with or without the selective CK2 inhibitor CX-4945. CX-4945 application preserved axon function during OGD and promoted axon function recovery when applied before or after OGD. This protective effect of CK2 inhibition correlated with preservation of oligodendrocytes and conservation of axon structure and axonal mitochondria. To investigate the pertinent downstream signaling pathways, siRNA targeting the CK2α subunit identified CDK5 and AKT as downstream molecules. Consequently, MK-2206 and roscovitine, which are selective AKT and CDK5 inhibitors, respectively, protected young and aging WM function only when applied before OGD. However, a novel pan-AKT allosteric inhibitor, ARQ-092, which targets both the inactive and active conformations of AKT, conferred protection to young and aging axons when applied before or after OGD. These results suggest that AKT and CDK5 signaling contribute to the WM functional protection conferred by CK2 inhibition during ischemia, while inhibition of activated AKT signaling plays the primary role in post-ischemic protection conferred by CK2 inhibition in WM independent of age. CK2 inhibitors are currently being used in clinical trials for cancer patients; therefore, our results will provide rationale for repurposing these drugs as therapeutic options for stroke patients by adding novel targets.
Collapse
Affiliation(s)
- Chinthasagar Bastian
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - John Quinn
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Ajai Tripathi
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Danielle Aquila
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Andrew McCray
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Ranjan Dutta
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Selva Baltan
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| | - Sylvain Brunet
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| |
Collapse
|
14
|
Cheng Y, Bai F. The Association of Tau With Mitochondrial Dysfunction in Alzheimer's Disease. Front Neurosci 2018; 12:163. [PMID: 29623026 PMCID: PMC5874499 DOI: 10.3389/fnins.2018.00163] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/28/2018] [Indexed: 01/01/2023] Open
Abstract
Increasing evidence suggests that abnormally hyperphosphorylated tau plays a vital role in the pathogenesis of Alzheimer's disease (AD). Mitochondrial dysfunction also has a recognized role in the pathophysiology of AD. In recent years, mitochondrial dysfunction has been strongly associated with tau pathology in AD. Overexpression of hyperphosphorylated and aggregated tau appears to damage the axonal transport, leading to abnormal mitochondrial distribution. In addition, pathological tau impairs mitochondrial dynamics by regulating mitochondrial fission/fusion proteins, and further causes mitochondrial dysfunction and neuronal damage. Moreover, mitochondrial dysfunction is also involved in promoting tau pathology in AD. In this article, we evaluate the relationship between phosphorylated tau and mitochondrial dysfunction in AD.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Feng Bai
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
15
|
Murphy LC, Millar JK. Regulation of mitochondrial dynamics by DISC1, a putative risk factor for major mental illness. Schizophr Res 2017; 187:55-61. [PMID: 28082141 DOI: 10.1016/j.schres.2016.12.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/22/2022]
Abstract
Mitochondria are dynamic organelles that are essential to power the process of neurotransmission. Neurons must therefore ensure that mitochondria maintain their functional integrity and are efficiently transported along the full extent of the axons and dendrites, from soma to synapses. Mitochondrial dynamics (trafficking, fission and fusion) co-ordinately regulate mitochondrial quality control and function. DISC1 is a component of the mitochondrial transport machinery and regulates mitochondrial dynamics. DISC1's role in this is adversely affected by sequence variants connected to brain structure/function and disease risk, and by mutant truncation. The DISC1 interactors NDE1 and GSK3β are also involved, indicating a convergence of putative risk factors for psychiatric illness upon mitochondrial dynamics.
Collapse
Affiliation(s)
- Laura C Murphy
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetic and Molecular Medicine at the University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - J Kirsty Millar
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetic and Molecular Medicine at the University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
16
|
Seripa D, Solfrizzi V, Imbimbo BP, Daniele A, Santamato A, Lozupone M, Zuliani G, Greco A, Logroscino G, Panza F. Tau-directed approaches for the treatment of Alzheimer's disease: focus on leuco-methylthioninium. Expert Rev Neurother 2016; 16:259-77. [PMID: 26822031 DOI: 10.1586/14737175.2016.1140039] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Small molecular weight compounds able to inhibit formation of tau oligomers and fibrils have already been tested for Alzheimer's disease (AD) treatment. The most advanced tau aggregation inhibitor (TAI) is methylthioninium (MT), a drug existing in equilibrium between a reduced (leuco-methylthioninium) and oxidized form (MT(+)). MT chloride (also known as methylene blue) was investigated in a 24-week Phase II study in 321 mild-to-moderate AD patients at the doses of 69, 138, and 228 mg/day. This trial failed to show significant positive effects of MT in the overall patient population. The dose of 138 mg/day showed potential benefits on cognitive performance of moderately affected patients and cerebral blood flow in mildly affected patients. A follow-up compound (TRx0237) claimed to be more bioavailable and less toxic than MT, is now being developed. Phase III clinical trials on this novel TAI in AD and in the behavioral variant of frontotemporal dementia are underway.
Collapse
Affiliation(s)
- Davide Seripa
- a Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Foggia , Italy
| | - Vincenzo Solfrizzi
- b Geriatric Medicine-Memory Unit and Rare Disease Centre , University of Bari Aldo Moro , Bari , Italy
| | - Bruno P Imbimbo
- c Research & Development Department , Chiesi Farmaceutici , Parma , Italy
| | - Antonio Daniele
- d Institute of Neurology , Catholic University of Sacred Heart , Rome , Italy
| | - Andrea Santamato
- e Physical Medicine and Rehabilitation Section, 'OORR' Hospital , University of Foggia , Foggia , Italy
| | - Madia Lozupone
- f Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs , University of Bari Aldo Moro , Bari , Italy
| | - Giovanni Zuliani
- g Department of Medical Science, Section of Internal and Cardiopulmonary Medicine , University of Ferrara
| | - Antonio Greco
- a Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Foggia , Italy
| | - Giancarlo Logroscino
- f Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs , University of Bari Aldo Moro , Bari , Italy.,h Department of Clinical Research in Neurology , University of Bari Aldo Moro, 'Pia Fondazione Cardinale G. Panico' , Tricase , Lecce , Italy
| | - Francesco Panza
- a Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Foggia , Italy.,f Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs , University of Bari Aldo Moro , Bari , Italy.,h Department of Clinical Research in Neurology , University of Bari Aldo Moro, 'Pia Fondazione Cardinale G. Panico' , Tricase , Lecce , Italy
| |
Collapse
|
17
|
Audouard E, Houben S, Masaracchia C, Yilmaz Z, Suain V, Authelet M, De Decker R, Buée L, Boom A, Leroy K, Ando K, Brion JP. High-Molecular-Weight Paired Helical Filaments from Alzheimer Brain Induces Seeding of Wild-Type Mouse Tau into an Argyrophilic 4R Tau Pathology in Vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2709-22. [PMID: 27497324 DOI: 10.1016/j.ajpath.2016.06.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/14/2016] [Accepted: 06/06/2016] [Indexed: 12/14/2022]
Abstract
In Alzheimer disease, the development of tau pathology follows neuroanatomically connected pathways, suggesting that abnormal tau species might recruit normal tau by passage from cell to cell. Herein, we analyzed the effect of stereotaxic brain injection of human Alzheimer high-molecular-weight paired helical filaments (PHFs) in the dentate gyrus of wild-type and mutant tau THY-Tau22 mice. After 3 months of incubation, wild-type and THY-Tau22 mice developed an atrophy of the dentate gyrus and a tau pathology characterized by Gallyas and tau-positive grain-like inclusions into granule cells that extended in the hippocampal hilus and eventually away into the alveus, and the fimbria. Gallyas-positive neuropil threads and oligodendroglial coiled bodies were also observed. These tau inclusions were composed only of mouse tau, and were immunoreactive with antibodies to 4R tau, phosphotau, misfolded tau, ubiquitin, and p62. Although local hyperphosphorylation of tau was increased in the dentate gyrus in THY-Tau22 mice, the development of neurofibrillary tangles made of mutant human tau was not accelerated in the hippocampus, indicating that wild-type human PHFs were inefficient in seeding tau aggregates made of G272V/P301S mutant human tau. Our results indicate thus that injection of human wild-type Alzheimer PHF seeded aggregation of wild-type murine tau into an argyrophilic 4R tau pathology, and constitutes an interesting model independent of expression of a mutant tau protein.
Collapse
Affiliation(s)
- Emilie Audouard
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Sarah Houben
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Caterina Masaracchia
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Valérie Suain
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Michèle Authelet
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Robert De Decker
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Luc Buée
- INSERM, U837, Université de Lille 2, Lille, France
| | - Alain Boom
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium.
| |
Collapse
|
18
|
Mitochondrial traffic jams in Alzheimer's disease - pinpointing the roadblocks. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1909-17. [PMID: 27460705 DOI: 10.1016/j.bbadis.2016.07.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/12/2016] [Accepted: 07/22/2016] [Indexed: 12/24/2022]
Abstract
The vigorous axonal transport of mitochondria, which serves to distribute these organelles in a dynamic and non-uniform fashion, is crucial to fulfill neuronal energetic requirements allowing the maintenance of neurons structure and function. Particularly, axonal transport of mitochondria and their spatial distribution among the synapses are directly correlated with synaptic activity and integrity. Despite the basis of Alzheimer's disease (AD) remains enigmatic, axonal pathology and synaptic dysfunction occur prior the occurrence of amyloid-β (Aβ) deposition and tau aggregation, the two classical hallmarks of this devastating neurodegenerative disease. Importantly, the early stages of AD are marked by defects on axonal transport of mitochondria as denoted by the abnormal accumulation of mitochondria within large swellings along dystrophic and degenerating neuritis. Within this scenario, this review is devoted to identify the molecular "roadblocks" underlying the abnormal axonal transport of mitochondria and consequent synaptic "starvation" and neuronal degeneration in AD. Understanding the molecular nature of defective mitochondrial transport may provide a new avenue to counteract AD pathology.
Collapse
|
19
|
Abstract
Establishment of cellular networks and calcium homeostasis are essential for embryonic stem cell proliferation and differentiation. We also hypothesized that adult neural progenitor cells form functional cellular networks relevant for their development. We isolated neuronal progenitor cells from the subventricular zone of 5-week-old mice to investigate the role of gap junctions, calcium homeostasis, and cellular networks in cell differentiation and survival. Western blotting and reverse transcription-PCR showed that the cells expressed the gap junction components connexin 26, 36, 43, and 45, and that expression of connexin 43 increased in early (8 days) differentiated cells. Transmission electron microscopy and immunocytochemistry also indicated that gap junctions were present. Scrape-loading experiments showed dye transfer between cells that could be prevented by gapjunction blockers; thus, functional intercellular gap junctions had been established. However, dye transfer was four times stronger in differentiated cultures, correlating with the increased connexin 43 expression. During time-lapse calcium imaging, both differentiated and undifferentiated cultures showed spontaneous calcium activity that was reduced by gap junction blockers. Cross-correlation analysis of the calcium recordings showed that the cells were interconnected through gap junctions and that the early-differentiated cells were organized in small-world networks. Gap junction blockers did not affect proliferation and differentiation, but resulted in twice as many apoptotic cells. mRNAi knockdown of connexin 43 also doubled the number of apoptotic cells. We conclude that adult neural progenitor cells form networks in vitro that are strengthened during early differentiation by increased expression of connexin 43. The networks are functional and improve cell survival.
Collapse
|
20
|
Shi C, Viccaro K, Lee HG, Shah K. Cdk5-Foxo3 axis: initially neuroprotective, eventually neurodegenerative in Alzheimer's disease models. J Cell Sci 2016; 129:1815-1830. [PMID: 28157684 DOI: 10.1242/jcs.185009] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/03/2016] [Indexed: 12/12/2022] Open
Abstract
Deregulated Cdk5 causes neurotoxic amyloid beta peptide (Aβ) processing and cell death, two hallmarks of Alzheimer's disease, through the Foxo3 transcriptional factor in hippocampal cells, primary neurons and an Alzheimer's disease mouse model. Using an innovative chemical genetic screen, we identified Foxo3 as a direct substrate of Cdk5 in brain lysates. Cdk5 directly phosphorylates Foxo3, which increased its levels and nuclear translocation. Nuclear Foxo3 initially rescued cells from ensuing oxidative stress by upregulating MnSOD (also known as SOD2). However, following prolonged exposure, Foxo3 upregulated Bim (also known as BCL2L11) and FasL (also known as FASLG) causing cell death. Active Foxo3 also increased Aβ(1-42) levels in a phosphorylation-dependent manner. These events were completely inhibited either by expressing phosphorylation-resistant Foxo3 or by depleting Cdk5 or Foxo3, highlighting a key role for Cdk5 in regulating Foxo3. These results were confirmed in an Alzheimer's disease mouse model, which exhibited increased levels and nuclear localization of Foxo3 in hippocampal neurons, which preceded neurodegeneration and Aβ plaque formation, indicating this phenomenon is an early event in Alzheimer's disease pathogenesis. Collectively, these results show that Cdk5-mediated phospho-regulation of Foxo3 can activate several genes that promote neuronal death and aberrant Aβ processing, thereby contributing to the progression of neurodegenerative pathologies.
Collapse
Affiliation(s)
- Chun Shi
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Keith Viccaro
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Hyoung-Gon Lee
- Department of Pathology, Case Western Reserve University School of Medicine, Iris S. Bert L. Wolstein Research Building, 2103 Cornell Road, Room 5123, Cleveland, OH 44106, USA
| | - Kavita Shah
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
| |
Collapse
|
21
|
Shah K, Lahiri DK. A Tale of the Good and Bad: Remodeling of the Microtubule Network in the Brain by Cdk5. Mol Neurobiol 2016; 54:2255-2268. [PMID: 26944284 DOI: 10.1007/s12035-016-9792-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/11/2016] [Indexed: 10/22/2022]
Abstract
Cdk5, a cyclin-dependent kinase family member, is a global orchestrator of neuronal cytoskeletal dynamics. During embryogenesis, Cdk5 is indispensable for brain development. In adults, it is essential for numerous neuronal processes, including higher cognitive functions such as learning and memory formation, drug addiction, pain signaling, and long-term behavior changes through long-term potentiation and long-term depression, all of which rely on rapid alterations in the cytoskeleton. Cdk5 activity becomes deregulated in various brain disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, attention-deficit hyperactivity disorder, epilepsy, schizophrenia, and ischemic stroke; these all result in profound remodeling of the neuronal cytoskeleton. This Commentary specifically focuses on the pleiotropic contribution of Cdk5 in regulating neuronal microtubule remodeling. Because the vast majority of the physiological substrates of Cdk5 are associated with the neuronal cytoskeleton, our emphasis is on the Cdk5 substrates, such as CRMP2, stathmin, drebrin, dixdc1, axin, MAP2, MAP1B, doublecortin, kinesin-5, and tau, that have allowed to unravel the molecular mechanisms through which Cdk5 exerts its divergent roles in regulating neuronal microtubule dynamics, both in healthy and disease states.
Collapse
Affiliation(s)
- Kavita Shah
- Department of Chemistry and Purdue University Center for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| | - Debomoy K Lahiri
- Departments of Psychiatry and Medical & Molecular Genetics, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN, 46202-2266, USA
| |
Collapse
|
22
|
Klinman E, Holzbaur ELF. Stress-Induced CDK5 Activation Disrupts Axonal Transport via Lis1/Ndel1/Dynein. Cell Rep 2015; 12:462-73. [PMID: 26166569 DOI: 10.1016/j.celrep.2015.06.032] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 05/06/2015] [Accepted: 06/09/2015] [Indexed: 01/26/2023] Open
Abstract
Axonal transport is essential for neuronal function, and defects in transport are associated with multiple neurodegenerative diseases. Aberrant cyclin-dependent kinase 5 (CDK5) activity, driven by the stress-induced activator p25, also is observed in these diseases. Here we show that elevated CDK5 activity increases the frequency of nonprocessive events for a range of organelles, including lysosomes, autophagosomes, mitochondria, and signaling endosomes. Transport disruption induced by aberrant CDK5 activation depends on the Lis1/Ndel1 complex, which directly regulates dynein activity. CDK5 phosphorylation of Ndel1 favors a high affinity Lis1/Ndel/dynein complex that blocks the ATP-dependent release of dynein from microtubules, inhibiting processive motility of dynein-driven cargo. Similar transport defects observed in neurons from a mouse model of amyotrophic lateral sclerosis are rescued by CDK5 inhibition. Together, these studies identify CDK5 as a Lis1/Ndel1-dependent regulator of transport in stressed neurons, and suggest that dysregulated CDK5 activity contributes to the transport deficits observed during neurodegeneration.
Collapse
Affiliation(s)
- Eva Klinman
- Neuroscience Graduate Group and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - Erika L F Holzbaur
- Neuroscience Graduate Group and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA.
| |
Collapse
|
23
|
Liguori C, Stefani A, Sancesario G, Sancesario GM, Marciani MG, Pierantozzi M. CSF lactate levels, τ proteins, cognitive decline: a dynamic relationship in Alzheimer's disease. J Neurol Neurosurg Psychiatry 2015; 86:655-9. [PMID: 25121572 DOI: 10.1136/jnnp-2014-308577] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/25/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To investigate, in patients with Alzheimer's Disease (AD), the possible interplay linking alteration of neuronal energy metabolism, as measured via cerebrospinal fluid (CSF) lactate concentration, to severity of AD neurodegenerative processes and impairment of cognitive abilities. METHODS In this study we measured and correlated CSF lactate concentrations, AD biomarker levels (τ-proteins and β-amyloid) and Mini-Mental State Examination (MMSE) score in a population of drug-naïve patients with AD ranging from mild (MMSE≥21/30) to moderate-severe (MMSE<21/30) cognitive decline. They were compared to healthy controls and patients with vascular dementia (VaD). RESULTS Patients with AD (n=145) showed a significant increase of CSF lactate concentration compared to controls (n=80) and patients with VaD (n=44), which was higher in mild (n=67) than in patients with moderate-severe AD (n=78). Moreover, we found, in either the whole AD population or both subgroups, a CSF profile in which higher CSF levels of t-τ and p-τ proteins corresponded to lower concentrations of lactate. CONCLUSIONS We verified the occurrence of high CSF lactate levels in patients with AD, which may be ascribed to mitochondria impairment. Hypothesising that τ proteins may exert a detrimental effect on the entire cellular energy metabolism, the negative correlation found between lactate and τ-protein levels may allow speculation that τ toxicity, already demonstrated to have affected mitochondria, could also impair glycolytic metabolism with a less evident increase of lactate levels in more severe AD. Thus, we suggest a dynamic relationship between neuronal energy metabolism, τ proteins and cognitive decline in AD and propose the clinical potential of assessing CSF lactate levels in patients with AD to better define the neuronal brain metabolism damage.
Collapse
Affiliation(s)
- C Liguori
- Department of Systems Medicine, Neurophysiopathology Unit, University of Rome "Tor Vergata", Rome, Italy Department of Systems Medicine, Neurology Unit, University of Rome "Tor Vergata", Rome, Italy
| | - A Stefani
- Department of Systems Medicine, Neurology Unit, University of Rome "Tor Vergata", Rome, Italy Fondazione Santa Lucia IRCCS, Rome, Italy
| | - G Sancesario
- Department of Systems Medicine, Neurology Unit, University of Rome "Tor Vergata", Rome, Italy Fondazione Santa Lucia IRCCS, Rome, Italy
| | - G M Sancesario
- Department of Clinical Biochemistry and Molecular Biology, University of Rome "Tor Vergata", Rome, Italy
| | - M G Marciani
- Department of Systems Medicine, Neurology Unit, University of Rome "Tor Vergata", Rome, Italy Fondazione Santa Lucia IRCCS, Rome, Italy
| | - M Pierantozzi
- Department of Systems Medicine, Neurology Unit, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
24
|
Welte MA. As the fat flies: The dynamic lipid droplets of Drosophila embryos. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1156-85. [PMID: 25882628 DOI: 10.1016/j.bbalip.2015.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 03/31/2015] [Accepted: 04/06/2015] [Indexed: 01/09/2023]
Abstract
Research into lipid droplets is rapidly expanding, and new cellular and organismal roles for these lipid-storage organelles are continually being discovered. The early Drosophila embryo is particularly well suited for addressing certain questions in lipid-droplet biology and combines technical advantages with unique biological phenomena. This review summarizes key features of this experimental system and the techniques available to study it, in order to make it accessible to researchers outside this field. It then describes the two topics most heavily studied in this system, lipid-droplet motility and protein sequestration on droplets, discusses what is known about the molecular players involved, points to open questions, and compares the results from Drosophila embryo studies to what it is known about lipid droplets in other systems.
Collapse
Affiliation(s)
- Michael A Welte
- Department of Biology University of Rochester, RC Box 270211, 317 Hutchison Hall, Rochester, NY 14627, USA.
| |
Collapse
|
25
|
Encalada SE, Goldstein LSB. Biophysical challenges to axonal transport: motor-cargo deficiencies and neurodegeneration. Annu Rev Biophys 2014; 43:141-69. [PMID: 24702007 DOI: 10.1146/annurev-biophys-051013-022746] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Axonal transport is indispensable for the distribution of vesicles, organelles, messenger RNAs (mRNAs), and signaling molecules along the axon. This process is mediated by kinesins and dyneins, molecular motors that bind to cargoes and translocate on microtubule tracks. Tight modulation of motor protein activity is necessary, but little is known about the molecules and mechanisms that regulate transport. Moreover, evidence suggests that transport impairments contribute to the initiation or progression of neurodegenerative diseases, or both, but the mechanisms by which motor activity is affected in disease are unclear. In this review, we discuss some of the physical and biophysical properties that influence motor regulation in healthy neurons. We further discuss the evidence for the role of transport in neurodegeneration, highlighting two pathways that may contribute to transport impairment-dependent disease: genetic mutations or variation, and protein aggregation. Understanding how and when transport parameters change in disease will help delineate molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Sandra E Encalada
- Department of Molecular and Experimental Medicine, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037;
| | | |
Collapse
|
26
|
Barbosa DJ, Serrat R, Mirra S, Quevedo M, Gómez de Barreda E, Avila J, Fernandes E, Bastos MDL, Capela JP, Carvalho F, Soriano E. MDMA impairs mitochondrial neuronal trafficking in a Tau- and Mitofusin2/Drp1-dependent manner. Arch Toxicol 2014; 88:1561-1572. [PMID: 24522274 DOI: 10.1007/s00204-014-1209-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/29/2014] [Indexed: 01/11/2023]
Abstract
Identification of the mechanisms by which drugs of abuse cause neuronal dysfunction is essential for understanding the biological bases of their acute and long-lasting effects in the brain. Here, we performed real-time functional experiments of axonal transport of mitochondria to explore the role of in situ mitochondrial dysfunction in 3,4-methylenedioxymethamphetamine (MDMA; "ecstasy")-related brain actions. We showed that MDMA dramatically reduced mitochondrial trafficking in hippocampal neurons in a Tau-dependent manner, in which glycogen synthase kinase 3β activity was implicated. Furthermore, we found that these trafficking abnormalities were rescued by over-expression of Mitofusin2 and dynamin-related protein 1, but not of Miro1. Given the relevance of mitochondrial targeting for neuronal function and neurotransmission, our data underscore a novel mechanism of action of MDMA that may contribute to our understanding of how this drug of abuse alters neuronal functioning.
Collapse
Affiliation(s)
- Daniel José Barbosa
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Laboratory, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Stygelbout V, Leroy K, Pouillon V, Ando K, D’Amico E, Jia Y, Luo HR, Duyckaerts C, Erneux C, Schurmans S, Brion JP. Inositol trisphosphate 3-kinase B is increased in human Alzheimer brain and exacerbates mouse Alzheimer pathology. Brain 2014; 137:537-52. [DOI: 10.1093/brain/awt344] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
28
|
Kumazawa A, Katoh H, Nonaka D, Watanabe T, Saotome M, Urushida T, Satoh H, Hayashi H. Microtubule Disorganization Affects the Mitochondrial Permeability Transition Pore in Cardiac Myocytes. Circ J 2014; 78:1206-15. [DOI: 10.1253/circj.cj-13-1298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Azumi Kumazawa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Hideki Katoh
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Daishi Nonaka
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Tomoyuki Watanabe
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Masao Saotome
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Tsuyoshi Urushida
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Hiroshi Satoh
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Hideharu Hayashi
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| |
Collapse
|
29
|
Shaw JL, Chang KT. Nebula/DSCR1 upregulation delays neurodegeneration and protects against APP-induced axonal transport defects by restoring calcineurin and GSK-3β signaling. PLoS Genet 2013; 9:e1003792. [PMID: 24086147 PMCID: PMC3784514 DOI: 10.1371/journal.pgen.1003792] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 07/29/2013] [Indexed: 01/06/2023] Open
Abstract
Post-mortem brains from Down syndrome (DS) and Alzheimer's disease (AD) patients show an upregulation of the Down syndrome critical region 1 protein (DSCR1), but its contribution to AD is not known. To gain insights into the role of DSCR1 in AD, we explored the functional interaction between DSCR1 and the amyloid precursor protein (APP), which is known to cause AD when duplicated or upregulated in DS. We find that the Drosophila homolog of DSCR1, Nebula, delays neurodegeneration and ameliorates axonal transport defects caused by APP overexpression. Live-imaging reveals that Nebula facilitates the transport of synaptic proteins and mitochondria affected by APP upregulation. Furthermore, we show that Nebula upregulation protects against axonal transport defects by restoring calcineurin and GSK-3β signaling altered by APP overexpression, thereby preserving cargo-motor interactions. As impaired transport of essential organelles caused by APP perturbation is thought to be an underlying cause of synaptic failure and neurodegeneration in AD, our findings imply that correcting calcineurin and GSK-3β signaling can prevent APP-induced pathologies. Our data further suggest that upregulation of Nebula/DSCR1 is neuroprotective in the presence of APP upregulation and provides evidence for calcineurin inhibition as a novel target for therapeutic intervention in preventing axonal transport impairments associated with AD. Alzheimer's disease (AD) is a debilitating neurodegenerative disease characterized by gradual neuronal cell loss and memory decline. Importantly, Down syndrome (DS) individuals over 40 years of age almost always develop neuropathological features of AD, although most do not develop dementia until at least two decades later. These findings suggest that DS and AD may share common genetic causes and that a neuroprotective mechanism may delay neurodegeneration and cognitive decline. It has been shown that the amyloid precursor protein (APP), which is associated with AD when duplicated and upregulated in DS, is a key gene contributing to AD pathologies and axonal transport abnormalities. Here, using fruit fly as a simple model organism, we examined the role of Down syndrome critical region 1 (DSCR1), another gene located on chromosome 21 and upregulated in both DS and AD, in modulating APP phenotypes. We find that upregulation of DSCR1 (Nebula in flies) is neuroprotective in the presence of APP upregulation. We report that nebula overexpression delays the onset of neurodegeneration and transport blockage in neuronal cells. Our results further suggest that signaling pathways downstream of DSCR1 may be potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Jillian L. Shaw
- Zilkha Neurogenetic Institute and Department of Cell & Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, United States of America
| | - Karen T. Chang
- Zilkha Neurogenetic Institute and Department of Cell & Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
30
|
Eckert A, Nisbet R, Grimm A, Götz J. March separate, strike together--role of phosphorylated TAU in mitochondrial dysfunction in Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2013; 1842:1258-66. [PMID: 24051203 DOI: 10.1016/j.bbadis.2013.08.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/08/2013] [Accepted: 08/12/2013] [Indexed: 12/17/2022]
Abstract
The energy demand and calcium buffering requirements of the brain are met by the high number of mitochondria in neurons and in these, especially at the synapses. Mitochondria are the major producer of reactive oxygen species (ROS); at the same time, they are damaged by ROS that are induced by abnormal protein aggregates that characterize human neurodegenerative diseases such as Alzheimer's disease (AD). Because synaptic mitochondria are long-lived, any damage exerted by these aggregates impacts severely on neuronal function. Here we review how increased TAU, a defining feature of AD and related tauopathies, impairs mitochondrial function by following the principle: 'March separate, strike together!' In the presence of amyloid-β, TAU's toxicity is augmented suggesting synergistic pathomechanisms. In order to restore mitochondrial functions in neurodegeneration as a means of therapeutic intervention it will be important to integrate the various aspects of dysfunction and get a handle on targeting distinct cell types and subcellular compartments.
Collapse
Affiliation(s)
- Anne Eckert
- Neurobiology Laboratory, Psychiatric University Clinics Basel, University of Basel, Switzerland
| | - Rebecca Nisbet
- Centre for Ageing Dementia Research (CADR), Queensland Brain Institute (QBI), The University of Queensland, Australia
| | - Amandine Grimm
- Neurobiology Laboratory, Psychiatric University Clinics Basel, University of Basel, Switzerland
| | - Jürgen Götz
- Centre for Ageing Dementia Research (CADR), Queensland Brain Institute (QBI), The University of Queensland, Australia.
| |
Collapse
|
31
|
Gassman A, Hao LT, Bhoite L, Bradford CL, Chien CB, Beattie CE, Manfredi JP. Small molecule suppressors of Drosophila kinesin deficiency rescue motor axon development in a zebrafish model of spinal muscular atrophy. PLoS One 2013; 8:e74325. [PMID: 24023935 PMCID: PMC3762770 DOI: 10.1371/journal.pone.0074325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 07/31/2013] [Indexed: 12/15/2022] Open
Abstract
Proximal spinal muscular atrophy (SMA) is the most common inherited motor neuropathy and the leading hereditary cause of infant mortality. Currently there is no effective treatment for the disease, reflecting a need for pharmacologic interventions that restore performance of dysfunctional motor neurons or suppress the consequences of their dysfunction. In a series of assays relevant to motor neuron biology, we explored the activities of a collection of tetrahydroindoles that were reported to alter the metabolism of amyloid precursor protein (APP). In Drosophila larvae the compounds suppressed aberrant larval locomotion due to mutations in the Khc and Klc genes, which respectively encode the heavy and light chains of kinesin-1. A representative compound of this class also suppressed the appearance of axonal swellings (alternatively termed axonal spheroids or neuritic beads) in the segmental nerves of the kinesin-deficient Drosophila larvae. Given the importance of kinesin-dependent transport for extension and maintenance of axons and their growth cones, three members of the class were tested for neurotrophic effects on isolated rat spinal motor neurons. Each compound stimulated neurite outgrowth. In addition, consistent with SMA being an axonopathy of motor neurons, the three axonotrophic compounds rescued motor axon development in a zebrafish model of SMA. The results introduce a collection of small molecules as pharmacologic suppressors of SMA-associated phenotypes and nominate specific members of the collection for development as candidate SMA therapeutics. More generally, the results reinforce the perception of SMA as an axonopathy and suggest novel approaches to treating the disease.
Collapse
Affiliation(s)
- Andrew Gassman
- Sera Prognostics, Inc., Salt Lake City, Utah, United States of America
| | - Le T. Hao
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, United States of America
| | - Leena Bhoite
- Technology Commercialization Office, University of Utah, Salt Lake City, Utah, United States of America
| | - Chad L. Bradford
- Sera Prognostics, Inc., Salt Lake City, Utah, United States of America
| | - Chi-Bin Chien
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Christine E. Beattie
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, United States of America
| | - John P. Manfredi
- Sfida BioLogic, Inc., Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
32
|
Weaver C, Leidel C, Szpankowski L, Farley NM, Shubeita GT, Goldstein LSB. Endogenous GSK-3/shaggy regulates bidirectional axonal transport of the amyloid precursor protein. Traffic 2013; 14:295-308. [PMID: 23279138 DOI: 10.1111/tra.12037] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 12/20/2012] [Accepted: 12/27/2012] [Indexed: 12/13/2022]
Abstract
Neurons rely on microtubule (MT) motor proteins such as kinesin-1 and dynein to transport essential cargos between the cell body and axon terminus. Defective axonal transport causes abnormal axonal cargo accumulations and is connected to neurodegenerative diseases, including Alzheimer's disease (AD). Glycogen synthase kinase 3 (GSK-3) has been proposed to be a central player in AD and to regulate axonal transport by the MT motor protein kinesin-1. Using genetic, biochemical and biophysical approaches in Drosophila melanogaster, we find that endogenous GSK-3 is a required negative regulator of both kinesin-1-mediated and dynein-mediated axonal transport of the amyloid precursor protein (APP), a key contributor to AD pathology. GSK-3 also regulates transport of an unrelated cargo, embryonic lipid droplets. By measuring the forces motors generate in vivo, we find that GSK-3 regulates transport by altering the activity of kinesin-1 motors but not their binding to the cargo. These findings reveal a new relationship between GSK-3 and APP, and demonstrate that endogenous GSK-3 is an essential in vivo regulator of bidirectional APP transport in axons and lipid droplets in embryos. Furthermore, they point to a new regulatory mechanism in which GSK-3 controls the number of active motors that are moving a cargo.
Collapse
Affiliation(s)
- Carole Weaver
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
33
|
Pradeep H, Rajanikant GK. A rational approach to selective pharmacophore designing: an innovative strategy for specific recognition of Gsk3β. Mol Divers 2012; 16:553-62. [PMID: 22918724 PMCID: PMC7089308 DOI: 10.1007/s11030-012-9387-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 07/25/2012] [Indexed: 12/21/2022]
Abstract
We propose a novel cheminformatics approach that combines structure and ligand-based design to identify target-specific pharmacophores with well-defined exclusion ability. Our strategy includes the prediction of selective interactions, developing structure, and knowledge-based selective pharmacophore models, followed by database screening and molecular docking. This unique strategy was employed in addressing the off-target toxicity of Gsk3β and CDKs. The connections of Gsk3β in eukaryotic cell apoptosis and the extensive potency of Gsk3β inhibitors to block cell death have made it a potential drug-discovery target for many grievous human disorders. Gsk3β is phylogenetically very closely related to the CDKs, such as CDK1 and CDK2, which are suggested to be the off-target proteins of Gsk3β inhibitors. Here, we have employed novel computational approaches in designing the ligand candidates that are potentially inhibitory against Gsk3β, with well-defined the exclusion ability to CDKs. A structure-ligand -based selective pharmacophore was modeled. This model was used to retrieve molecules from the zinc database. The hits retrieved were further screened by molecular docking and protein–ligand interaction fingerprints. Based on these results, four molecules were predicted as selective Gsk3β antagonists. It is anticipated that this unique approach can be extended to investigate any protein–ligand specificity.
Collapse
Affiliation(s)
- H Pradeep
- Bioinformatics Centre, School of Biotechnology, National Institute of Technology, Calicut 673601, India
| | | |
Collapse
|
34
|
Regulation of mitochondrial transport and inter-microtubule spacing by tau phosphorylation at the sites hyperphosphorylated in Alzheimer's disease. J Neurosci 2012; 32:2430-41. [PMID: 22396417 DOI: 10.1523/jneurosci.5927-11.2012] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The microtubule-associated protein Tau is a major component of the neurofibrillary tangles that serve as a neuropathological hallmark of Alzheimer's disease. Tau is a substrate for protein phosphorylation at multiple sites and occurs in tangles in a hyperphosphorylated state. However, the physiological functions of Tau phosphorylation or how it may contribute mechanistically to Alzheimer's pathophysiology are not completely understood. Here, we examined the function of human Tau phosphorylation at three sites, Ser199, Ser202, and Thr205, which together comprise the AT8 sites that mark abnormal phosphorylation in Alzheimer's disease. Overexpression of wild-type Tau or mutated forms in which these sites had been changed to either unphosphorylatable alanines or phosphomimetic aspartates inhibited mitochondrial movement in the neurite processes of PC12 cells as well as the axons of mouse brain cortical neurons. However, the greatest effects on mitochondrial translocation were induced by phosphomimetic mutations. These mutations also caused expansion of the space between microtubules in cultured cells when membrane tension was reduced by disrupting actin filaments. Thus, Tau phosphorylation at the AT8 sites may have meaningful effects on mitochondrial movement, likely by controlling microtubule spacing. Hyperphosphorylation of the AT8 sites may contribute to axonal degeneration by disrupting mitochondrial transport in Alzheimer's disease.
Collapse
|
35
|
McLinden KA, Trunova S, Giniger E. At the Fulcrum in Health and Disease: Cdk5 and the Balancing Acts of Neuronal Structure and Physiology. ACTA ACUST UNITED AC 2012; 2012:001. [PMID: 25364642 PMCID: PMC4212508 DOI: 10.4172/2168-975x.s1-001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cdk5 has been implicated in a multitude of processes in neuronal development, cell biology and physiology. These influence many neurological disorders, but the very breadth of Cdk5 effects has made it difficult to synthesize a coherent picture of the part played by this protein in health and disease. In this review, we focus on the roles of Cdk5 in neuronal function, particularly synaptic homeostasis, plasticity, neurotransmission, subcellular organization, and trafficking. We then discuss how disruption of these Cdk5 activities may initiate or exacerbate neural disorders. A recurring theme will be the sensitivity of Cdk5 sequelae to the precise biological context under consideration.
Collapse
Affiliation(s)
- Kristina A McLinden
- National Institute of Neurological Disorders and Stroke, USA ; National Human Genome Research Institute, USA
| | - Svetlana Trunova
- National Institute of Neurological Disorders and Stroke, USA ; National Human Genome Research Institute, USA
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, USA ; National Human Genome Research Institute, USA
| |
Collapse
|
36
|
Levels of kinesin light chain and dynein intermediate chain are reduced in the frontal cortex in Alzheimer's disease: implications for axoplasmic transport. Acta Neuropathol 2012; 123:71-84. [PMID: 22094641 DOI: 10.1007/s00401-011-0901-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 10/16/2011] [Accepted: 10/22/2011] [Indexed: 01/08/2023]
Abstract
Fast anterograde and retrograde axoplasmic transports in neurons rely on the activity of molecular motors and are critical for maintenance of neuronal and synaptic functions. Disturbances of axoplasmic transport have been identified in Alzheimer's disease and in animal models of this disease, but their mechanisms are not well understood. In this study we have investigated the distribution and the level of expression of kinesin light chains (KLCs) (responsible for binding of cargos during anterograde transport) and of dynein intermediate chain (DIC) (a component of the dynein complex during retrograde transport) in frontal cortex and cerebellar cortex of control subjects and Alzheimer's disease patients. By immunoblotting, we found a significant decrease in the levels of expression of KLC1 and 2 and DIC in the frontal cortex, but not in the cerebellar cortex, of Alzheimer's disease patients. A significant decrease in the levels of synaptophysin and of tubulin-β3 proteins, two neuronal markers, was also observed. KLC1 and DIC immunoreactivities did not co-localize with neurofibrillary tangles. The mean mRNA levels of KLC1, 2 and DIC were not significantly different between controls and AD patients. In SH-SY5Y neural cells, GSK-3β phosphorylated KLC1, a change associated to decreased association of KLC1 with its cargoes. Increased levels of active GSK-3β and of phosphorylated KLC1 were also observed in AD frontal cortex. We suggest that reduction of KLCs and DIC proteins in AD cortex results from both reduced expression and neuronal loss, and that these reductions and GSK-3β-mediated phosphorylation of KLC1 contribute to disturbances of axoplasmic flows and synaptic integrity in Alzheimer's disease.
Collapse
|
37
|
Llorens-Martín M, López-Doménech G, Soriano E, Avila J. GSK3β is involved in the relief of mitochondria pausing in a Tau-dependent manner. PLoS One 2011; 6:e27686. [PMID: 22110721 PMCID: PMC3215736 DOI: 10.1371/journal.pone.0027686] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 10/21/2011] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial trafficking deficits have been implicated in the pathogenesis of several neurological diseases, including Alzheimer's disease (AD). The Ser/Thre kinase GSK3β is believed to play a fundamental role in AD pathogenesis. Given that GSK3β substrates include Tau protein, here we studied the impact of GSK3β on mitochondrial trafficking and its dependence on Tau protein. Overexpression of GSK3β in neurons resulted in an increase in motile mitochondria, whereas a decrease in the activity of this kinase produced an increase in mitochondria pausing. These effects were dependent on Tau proteins, as Tau (−/−) neurons did not respond to distinct GSK3β levels. Furthermore, differences in GSK3β expression did not affect other parameters like mitochondria velocity or mitochondria run length. We conclude that GSK3B activity regulates mitochondrial axonal trafficking largely in a Tau-dependent manner.
Collapse
Affiliation(s)
- María Llorens-Martín
- Departmento de Neurobiología Molecular, Centro de Biología Molecular Severo Ochoa, Madrid, Spain
| | - Guillermo López-Doménech
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
- Developmental Neurobiology and Regeneration, Institut for Research in Biomedicine, Barcelona, Spain
- Department of Cell Biology, University of Barcelona, Barcelona, Spain
| | - Eduardo Soriano
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
- Developmental Neurobiology and Regeneration, Institut for Research in Biomedicine, Barcelona, Spain
- Department of Cell Biology, University of Barcelona, Barcelona, Spain
| | - Jesús Avila
- Departmento de Neurobiología Molecular, Centro de Biología Molecular Severo Ochoa, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
- * E-mail:
| |
Collapse
|
38
|
Pritchard SM, Dolan PJ, Vitkus A, Johnson GVW. The toxicity of tau in Alzheimer disease: turnover, targets and potential therapeutics. J Cell Mol Med 2011; 15:1621-35. [PMID: 21348938 PMCID: PMC4373356 DOI: 10.1111/j.1582-4934.2011.01273.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 02/08/2011] [Indexed: 11/28/2022] Open
Abstract
It has been almost 25 years since the initial discovery that tau was the primary component of the neurofibrillary tangles (NFTs) in Alzheimer disease (AD) brain. Although AD is defined by both β-amyloid (Aβ) pathology (Aβ plaques) and tau pathology (NFTs), whether or not tau played a critical role in disease pathogenesis was a subject of discussion for many years. However, given the increasing evidence that pathological forms of tau can compromise neuronal function and that tau is likely an important mediator of Aβ toxicity, there is a growing awareness that tau is a central player in AD pathogenesis. In this review we begin with a brief history of tau, then provide an overview of pathological forms of tau, followed by a discussion of the differential degradation of tau by either the proteasome or autophagy and possible mechanisms by which pathological forms of tau may exert their toxicity. We conclude by discussing possible avenues for therapeutic intervention based on these emerging themes of tau's role in AD.
Collapse
Affiliation(s)
- Susanne M Pritchard
- Gail V.W. JOHNSON, Ph.D., Department of Anesthesiology, 601 Elmwood Ave., Box 604, Rm. 4–6314, University of Rochester, Rochester, NY 14642, USA. Tel.: 585-276-3740 Fax: 585-276-2418 E-mail:
| | | | - Alisa Vitkus
- Department of Anesthesiology and the Interdepartmental Graduate Program in Neuroscience, University of RochesterRochester, NY, USA
| | - Gail VW Johnson
- Department of Anesthesiology and the Interdepartmental Graduate Program in Neuroscience, University of RochesterRochester, NY, USA
| |
Collapse
|