51
|
Clemente V, Hoshino A, Meints J, Shetty M, Starr T, Lee M, Bazzaro M. UNC-45A Is Highly Expressed in the Proliferative Cells of the Mouse Genital Tract and in the Microtubule-Rich Areas of the Mouse Nervous System. Cells 2021; 10:1604. [PMID: 34206743 PMCID: PMC8303485 DOI: 10.3390/cells10071604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/16/2022] Open
Abstract
UNC-45A (Protein unc-45 homolog A) is a cytoskeletal-associated protein with a dual and non-mutually exclusive role as a regulator of the actomyosin system and a Microtubule (MT)-destabilizing protein, which is overexpressed in human cancers including in ovarian cancer patients resistant to the MT-stabilizing drug paclitaxel. Mapping of UNC-45A in the mouse upper genital tract and central nervous system reveals its enrichment not only in highly proliferating and prone to remodeling cells, but also in microtubule-rich areas, of the ovaries and the nervous system, respectively. In both apparatuses, UNC-45A is also abundantly expressed in the ciliated epithelium. As regulators of actomyosin contractility and MT stability are essential for the physiopathology of the female reproductive tract and of neuronal development, our findings suggest that UNC-45A may have a role in ovarian cancer initiation and development as well as in neurodegeneration.
Collapse
Affiliation(s)
- Valentino Clemente
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA; (V.C.); (A.H.); (M.S.); (T.S.)
| | - Asumi Hoshino
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA; (V.C.); (A.H.); (M.S.); (T.S.)
| | - Joyce Meints
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (J.M.); (M.L.)
| | - Mihir Shetty
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA; (V.C.); (A.H.); (M.S.); (T.S.)
| | - Tim Starr
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA; (V.C.); (A.H.); (M.S.); (T.S.)
| | - Michael Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (J.M.); (M.L.)
| | - Martina Bazzaro
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA; (V.C.); (A.H.); (M.S.); (T.S.)
| |
Collapse
|
52
|
Fabbretti E, Antognolli G, Tongiorgi E. Amyloid-β Impairs Dendritic Trafficking of Golgi-Like Organelles in the Early Phase Preceding Neurite Atrophy: Rescue by Mirtazapine. Front Mol Neurosci 2021; 14:661728. [PMID: 34149353 PMCID: PMC8209480 DOI: 10.3389/fnmol.2021.661728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/30/2021] [Indexed: 12/20/2022] Open
Abstract
Neurite atrophy with loss of neuronal polarity is a pathological hallmark of Alzheimer's disease (AD) and other neurological disorders. While there is substantial agreement that disruption of intracellular vesicle trafficking is associated with axonal pathology in AD, comparatively less is known regarding its role in dendritic atrophy. This is a significant gap of knowledge because, unlike axons, dendrites are endowed with the complete endomembrane system comprising endoplasmic reticulum (ER), ER-Golgi intermediate compartment (ERGIC), Golgi apparatus, post-Golgi vesicles, and a recycling-degradative route. In this study, using live-imaging of pGOLT-expressing vesicles, indicative of Golgi outposts and satellites, we investigate how amyloid-β (Aβ) oligomers affect the trafficking of Golgi-like organelles in the different dendritic compartments of cultured rat hippocampal neurons. We found that short-term (4 h) treatment with Aβ led to a decrease in anterograde trafficking of Golgi vesicles in dendrites of both resting and stimulated (with 50 mM KCl) neurons. We also characterized the ability of mirtazapine, a noradrenergic and specific serotonergic tetracyclic antidepressant (NaSSA), to rescue Golgi dynamics in dendrites. Mirtazapine treatment (10 μM) increased the number and both anterograde and retrograde motility, reducing the percentage of static Golgi vesicles. Finally, mirtazapine reverted the neurite atrophy induced by 24 h treatment with Aβ oligomers, suggesting that this drug is able to counteract the effects of Aβ by improving the dendritic trafficking of Golgi-related vesicles.
Collapse
Affiliation(s)
- Elsa Fabbretti
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Enrico Tongiorgi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
53
|
Mol MO, Wong TH, Melhem S, Basu S, Viscusi R, Galjart N, Rozemuller AJ, Fallini C, Landers JE, Kaat LD, Seelaar H, van Rooij JG, van Swieten JC. Novel TUBA4A Variant Associated With Familial Frontotemporal Dementia. Neurol Genet 2021; 7:e596. [PMID: 34169147 PMCID: PMC8221227 DOI: 10.1212/nxg.0000000000000596] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 04/06/2021] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Despite the strong genetic component of frontotemporal dementia (FTD), a substantial proportion of patients remain genetically unresolved. We performed an in-depth study of a family with an autosomal dominant form of FTD to investigate the underlying genetic cause. METHODS Following clinical and pathologic characterization of the family, genetic studies included haplotype sharing analysis and exome sequencing. Subsequently, we performed immunohistochemistry, immunoblotting, and a microtubule repolymerization assay to investigate the potential impact of the candidate variant in tubulin alpha 4a (TUBA4A). RESULTS The clinical presentation in this family is heterogeneous, including behavioral changes, parkinsonian features, and uncharacterized dementia. Neuropathologic examination of 2 patients revealed TAR DNA binding protein 43 (TDP-43) pathology with abundant dystrophic neurites and neuronal intranuclear inclusions, consistent with frontotemporal lobar degeneration-TDP type A. We identified a likely pathogenic variant in TUBA4A segregating with disease. TUBA4A encodes for α-tubulin, which is a major component of the microtubule network. Variants in TUBA4A have been suggested as a rare genetic cause of amyotrophic lateral sclerosis (ALS) and have sporadically been reported in patients with FTD without supporting genetic segregation. A decreased trend of TUBA4A protein abundance was observed in patients compared with controls, and a microtubule repolymerization assay demonstrated disrupted α-tubulin function. As opposed to variants found in ALS, TUBA4A variants associated with FTD appear more localized to the N-terminus, indicating different pathogenic mechanisms. CONCLUSIONS Our findings support the role of TUBA4A variants as rare genetic cause of familial FTD.
Collapse
Affiliation(s)
| | | | - Shamiram Melhem
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Sreya Basu
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Riccardo Viscusi
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Niels Galjart
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Annemieke J.M. Rozemuller
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Claudia Fallini
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - John E. Landers
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Laura Donker Kaat
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Harro Seelaar
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jeroen G.J. van Rooij
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - John C. van Swieten
- From the Department of Neurology (M.O.M., T.H.W., S.M., L.D.K., H.S.,
J.G.J.v.R., J.C.v.S.), and Department of Cell Biology (S.B., R.V., N.G.),
Erasmus Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam
University Medical Center, Location VUmc, Amsterdam Neuroscience, the
Netherlands; Department of Cell and Molecular Biology (C.F.), University of
Rhode Island, Kingston; Department of Neurology (J.E.L.), University of
Massachusetts Medical School, Worcester; and Department of Clinical Genetics
(L.D.K.), Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
54
|
Jangampalli Adi P, Reddy PH. Phosphorylated tau targeted small-molecule PROTACs for the treatment of Alzheimer's disease and tauopathies. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166162. [PMID: 33940164 DOI: 10.1016/j.bbadis.2021.166162] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Tau is a microtubule-stabilizing protein that plays an important role in the formation of axonal microtubules in neurons. Phosphorylated tau (p-Tau) has received great attention in the field of Alzheimer's disease (AD) as a potential therapeutic target due to its involvement with synaptic damage and neuronal dysfunction. Mounting evidence suggests that amyloid beta (Aβ)-targeted clinical trials continuously failed; therefore, it is important to consider alternative therapeutic strategies such as p-tau-PROTACs targeted small molecules for AD and other tauopathies. The present article describes the characteristics of tau biology, structure, and function in both healthy and pathological states in AD. It also explains data from studies that have identified the involvement of p-tau in neuronal damage and synaptic and cognitive functions in AD. Current article also covers several aspects, including small molecule inhibitors, and the development of p-tau-PROTACs targeted drug molecules to treat patients with AD and other tauopathies.
Collapse
Affiliation(s)
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
55
|
Shah M, Chacko LA, Joseph JP, Ananthanarayanan V. Mitochondrial dynamics, positioning and function mediated by cytoskeletal interactions. Cell Mol Life Sci 2021; 78:3969-3986. [PMID: 33576841 PMCID: PMC11071877 DOI: 10.1007/s00018-021-03762-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/27/2020] [Accepted: 01/15/2021] [Indexed: 12/22/2022]
Abstract
The ability of a mitochondrion to undergo fission and fusion, and to be transported and localized within a cell are central not just to proper functioning of mitochondria, but also to that of the cell. The cytoskeletal filaments, namely microtubules, F-actin and intermediate filaments, have emerged as prime movers in these dynamic mitochondrial shape and position transitions. In this review, we explore the complex relationship between the cytoskeleton and the mitochondrion, by delving into: (i) how the cytoskeleton helps shape mitochondria via fission and fusion events, (ii) how the cytoskeleton facilitates the translocation and anchoring of mitochondria with the activity of motor proteins, and (iii) how these changes in form and position of mitochondria translate into functioning of the cell.
Collapse
Affiliation(s)
- Mitali Shah
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Leeba Ann Chacko
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Joel P Joseph
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Vaishnavi Ananthanarayanan
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India.
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
56
|
Tapias A, Lázaro D, Yin BK, Rasa SMM, Krepelova A, Kelmer Sacramento E, Grigaravicius P, Koch P, Kirkpatrick J, Ori A, Neri F, Wang ZQ. HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegeneration. eLife 2021; 10:61531. [PMID: 33594975 PMCID: PMC7939550 DOI: 10.7554/elife.61531] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/16/2021] [Indexed: 01/22/2023] Open
Abstract
Brain homeostasis is regulated by the viability and functionality of neurons. HAT (histone acetyltransferase) and HDAC (histone deacetylase) inhibitors have been applied to treat neurological deficits in humans; yet, the epigenetic regulation in neurodegeneration remains elusive. Mutations of HAT cofactor TRRAP (transformation/transcription domain-associated protein) cause human neuropathies, including psychosis, intellectual disability, autism, and epilepsy, with unknown mechanism. Here we show that Trrap deletion in Purkinje neurons results in neurodegeneration of old mice. Integrated transcriptomics, epigenomics, and proteomics reveal that TRRAP via SP1 conducts a conserved transcriptomic program. TRRAP is required for SP1 binding at the promoter proximity of target genes, especially microtubule dynamics. The ectopic expression of Stathmin3/4 ameliorates defects of TRRAP-deficient neurons, indicating that the microtubule dynamics is particularly vulnerable to the action of SP1 activity. This study unravels a network linking three well-known, but up-to-date unconnected, signaling pathways, namely TRRAP, HAT, and SP1 with microtubule dynamics, in neuroprotection.
Collapse
Affiliation(s)
- Alicia Tapias
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - David Lázaro
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Bo-Kun Yin
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | | | - Anna Krepelova
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | | | | | - Philipp Koch
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Joanna Kirkpatrick
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Francesco Neri
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Faculty of Biological Sciences, Friedrich-Schiller-University of Jena, Jena, Germany
| |
Collapse
|
57
|
Marmolejo-Martínez-Artesero S, Casas C, Romeo-Guitart D. Endogenous Mechanisms of Neuroprotection: To Boost or Not to Boost. Cells 2021; 10:cells10020370. [PMID: 33578870 PMCID: PMC7916582 DOI: 10.3390/cells10020370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Postmitotic cells, like neurons, must live through a lifetime. For this reason, organisms/cells have evolved with self-repair mechanisms that allow them to have a long life. The discovery workflow of neuroprotectors during the last years has focused on blocking the pathophysiological mechanisms that lead to neuronal loss in neurodegeneration. Unfortunately, only a few strategies from these studies were able to slow down or prevent neurodegeneration. There is compelling evidence demonstrating that endorsing the self-healing mechanisms that organisms/cells endogenously have, commonly referred to as cellular resilience, can arm neurons and promote their self-healing. Although enhancing these mechanisms has not yet received sufficient attention, these pathways open up new therapeutic avenues to prevent neuronal death and ameliorate neurodegeneration. Here, we highlight the main endogenous mechanisms of protection and describe their role in promoting neuron survival during neurodegeneration.
Collapse
Affiliation(s)
- Sara Marmolejo-Martínez-Artesero
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, 08193 Barcelona, Spain;
| | - Caty Casas
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, 08193 Barcelona, Spain;
| | - David Romeo-Guitart
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, 08193 Barcelona, Spain;
- Laboratory “Hormonal Regulation of Brain Development and Functions”—Team 8, Institut Necker Enfants-Malades (INEM), INSERM U1151, Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France
- Correspondence: ; Tel.: +33-01-40-61-53-57
| |
Collapse
|
58
|
Pokusa M, Hajdúchová D, Menichová V, Evinová A, Hatoková Z, Kráľová-Trančíková A. Vulnerability of subcellular structures to pathogenesis induced by rotenone in SH-SY5Y cells. Physiol Res 2021; 70:89-99. [PMID: 33453717 DOI: 10.33549/physiolres.934477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Numerous pathological changes of subcellular structures are characteristic hallmarks of neurodegeneration. The main research has focused to mitochondria, endoplasmic reticulum, Golgi apparatus, lysosomal networks as well as microtubular system of the cell. The sequence of specific organelle damage during pathogenesis has not been answered yet. Exposition to rotenone is used for simulation of neurodegenerative changes in SH-SY5Y cells, which are widely used for in vitro modelling of Parkinson´s disease pathogenesis. Intracellular effects were investigated in time points from 0 to 24 h by confocal microscopy and biochemical analyses. Analysis of fluorescent images identified the sensitivity of organelles towards rotenone in this order: microtubular cytoskeleton, mitochondrial network, endoplasmic reticulum, Golgi apparatus and lysosomal network. All observed morphological changes of intracellular compartments were identified before alphaS protein accumulation. Therefore, their potential as an early diagnostic marker is of interest. Understanding of subcellular sensitivity in initial stages of neurodegeneration is crucial for designing new approaches and a management of neurodegenerative disorders.
Collapse
Affiliation(s)
- M Pokusa
- Biomedical Center Martin, Martin, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
59
|
Clark J, Zhu Z, Chuckowree J, Dickson T, Blizzard C. Efficacy of epothilones in central nervous system trauma treatment: what has age got to do with it? Neural Regen Res 2021; 16:618-620. [PMID: 33063710 PMCID: PMC8067923 DOI: 10.4103/1673-5374.295312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Central nervous system injury, specifically traumatic brain and spinal cord injury, can have significant long lasting effects. There are no comprehensive treatments to combat the injury and sequalae of events that occurring following a central nervous system trauma. Herein we discuss the potential for the epothilone family of microtubule stabilizing agents to improve outcomes following experimentally induced trauma. These drugs, which are able to cross the blood-brain barrier, may hold great promise for the treatment of central nervous system trauma and the current literature presents the extensive range of beneficial effects these drugs may have following trauma in animal models. Importantly, the effect of the epothilones can vary and our most recent contributions to this field indicate that the efficacy of epothilones following traumatic brain injury is dependent upon the age of the animals. Therefore, we present a case for a greater emphasis to be placed upon age when using an intervention aimed at neural regeneration and highlight the importance of tailoring the therapeutic regime in the clinic to the age of the patient to promote improved patient outcomes.
Collapse
Affiliation(s)
- Jayden Clark
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Zhendan Zhu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Jyoti Chuckowree
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Tracey Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Catherine Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| |
Collapse
|
60
|
Boiarska Z, Passarella D. Microtubule-targeting agents and neurodegeneration. Drug Discov Today 2020; 26:604-615. [PMID: 33279455 DOI: 10.1016/j.drudis.2020.11.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/17/2020] [Accepted: 11/28/2020] [Indexed: 11/25/2022]
Abstract
The association of microtubule (MT) breakdown with neurodegeneration and neurotoxicity has provided an emerging therapeutic approach for neurodegenerative diseases. Tubulin binders are able to modulate MT dynamics and, as a result, are of particular interest both as potential therapeutics and experimental tools used to validate this strategy. Here, we provide a comprehensive overview of current knowledge and recent advancements regarding MT-targeting approaches for neurodegeneration and evaluate the potential application of MT-targeting agents (MTAs) based on available preclinical and clinical data.
Collapse
Affiliation(s)
- Zlata Boiarska
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Daniele Passarella
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy.
| |
Collapse
|
61
|
Cui L, Zheng J, Zhao Q, Chen JR, Liu H, Peng G, Wu Y, Chen C, He Q, Shi H, Yin S, Friedman RA, Chen Y, Guan MX. Mutations of MAP1B encoding a microtubule-associated phosphoprotein cause sensorineural hearing loss. JCI Insight 2020; 5:136046. [PMID: 33268592 PMCID: PMC7714412 DOI: 10.1172/jci.insight.136046] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 10/28/2020] [Indexed: 12/20/2022] Open
Abstract
The pathophysiology underlying spiral ganglion cell defect–induced deafness remains elusive. Using the whole exome sequencing approach, in combination with functional assays and a mouse disease model, we identified the potentially novel deafness-causative MAP1B gene encoding a highly conserved microtubule-associated protein. Three novel heterozygous MAP1B mutations (c.4198A>G, p.1400S>G; c.2768T>C, p.923I>T; c.5512T>C, p.1838F>L) were cosegregated with autosomal dominant inheritance of nonsyndromic sensorineural hearing loss in 3 unrelated Chinese families. Here, we show that MAP1B is highly expressed in the spiral ganglion neurons in the mouse cochlea. Using otic sensory neuron–like cells, generated by pluripotent stem cells from patients carrying the MAP1B mutation and control subject, we demonstrated that the p.1400S>G mutation caused the reduced levels and deficient phosphorylation of MAP1B, which are involved in the microtubule stability and dynamics. Strikingly, otic sensory neuron–like cells exhibited disturbed dynamics of microtubules, axonal elongation, and defects in electrophysiological properties. Dysfunctions of these derived otic sensory neuron–like cells were rescued by genetically correcting MAP1B mutation using CRISPR/Cas9 technology. Involvement of MAP1B in hearing was confirmed by audiometric evaluation of Map1b heterozygous KO mice. These mutant mice displayed late-onset progressive sensorineural hearing loss that was more pronounced in the high frequencies. The spiral ganglion neurons isolated from Map1b mutant mice exhibited the deficient phosphorylation and disturbed dynamics of microtubules. Map1b deficiency yielded defects in the morphology and electrophysiology of spiral ganglion neurons, but it did not affect the morphologies of cochlea in mice. Therefore, our data demonstrate that dysfunctions of spiral ganglion neurons induced by MAP1B deficiency caused hearing loss. Dysfunctions of spiral ganglion neurons caused by Map1b deficiency leads to sensorineural hearing loss.
Collapse
Affiliation(s)
- Limei Cui
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and.,Department of Human Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Zheng
- Division of Medical Genetics and Genomics, The Children's Hospital
| | - Qiong Zhao
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and.,Department of Human Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jia-Rong Chen
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and
| | | | - Guanghua Peng
- Deaprtment of Otorhinolaryngology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yue Wu
- Division of Medical Genetics and Genomics, The Children's Hospital
| | - Chao Chen
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and
| | | | - Haosong Shi
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shankai Yin
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Rick A Friedman
- Division of Otolaryngology, University of California at San Diego School of Medicine, La Jolla California, USA
| | - Ye Chen
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and.,Department of Human Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Min-Xin Guan
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and.,Department of Human Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Division of Otolaryngology, University of California at San Diego School of Medicine, La Jolla California, USA.,Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, Zhejiang, China.,Joint Institute of Genetics and Genomic Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
62
|
Vasudevan A, Koushika SP. Molecular mechanisms governing axonal transport: a C. elegans perspective. J Neurogenet 2020; 34:282-297. [PMID: 33030066 DOI: 10.1080/01677063.2020.1823385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Axonal transport is integral for maintaining neuronal form and function, and defects in axonal transport have been correlated with several neurological diseases, making it a subject of extensive research over the past several years. The anterograde and retrograde transport machineries are crucial for the delivery and distribution of several cytoskeletal elements, growth factors, organelles and other synaptic cargo. Molecular motors and the neuronal cytoskeleton function as effectors for multiple neuronal processes such as axon outgrowth and synapse formation. This review examines the molecular mechanisms governing axonal transport, specifically highlighting the contribution of studies conducted in C. elegans, which has proved to be a tractable model system in which to identify both novel and conserved regulatory mechanisms of axonal transport.
Collapse
Affiliation(s)
- Amruta Vasudevan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
63
|
Sarkar T, Patro N, Patro IK. Neuronal changes and cognitive deficits in a multi-hit rat model following cumulative impact of early life stressors. Biol Open 2020; 9:bio054130. [PMID: 32878878 PMCID: PMC7522020 DOI: 10.1242/bio.054130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/20/2020] [Indexed: 01/06/2023] Open
Abstract
Perinatal protein malnourishment (LP) is a leading cause for mental and physical retardation in children from poor socioeconomic conditions. Such malnourished children are vulnerable to additional stressors that may synergistically act to cause neurological disorders in adulthood. In this study, the above mentioned condition was mimicked via a multi-hit rat model in which pups born to LP mothers were co-injected with polyinosinic:polycytidylic acid (Poly I:C; viral mimetic) at postnatal day (PND) 3 and lipopolysaccharide (LPS; bacterial mimetic) at PND 9. Individual exposure of Poly I:C and LPS was also given to LP pups to correlate chronicity of stress. Similar treatments were also given to control pups. Hippocampal cellular apoptosis, β III tubulin catastrophe, altered neuronal profiling and spatial memory impairments were assessed at PND 180, using specific immunohistochemical markers (active caspase 3, β III tubulin, doublecortin), golgi studies and cognitive mazes (Morris water maze and T maze). Increase in cellular apoptosis, loss of dendritic arborization and spatial memory impairments were higher in the multi-hit group, than the single-hit groups. Such impairments observed due to multi-hit stress mimicked conditions similar to many neurological disorders and hence, it is hypothesized that later life neurological disorders might be an outcome of multiple early life hits.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Tiyasha Sarkar
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India
| | - Nisha Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India
| | - Ishan Kumar Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India
| |
Collapse
|
64
|
Rodrigues-Amorim D, Rivera-Baltanás T, Del Carmen Vallejo-Curto M, Rodriguez-Jamardo C, de Las Heras E, Barreiro-Villar C, Blanco-Formoso M, Fernández-Palleiro P, Álvarez-Ariza M, López M, García-Caballero A, Olivares JM, Spuch C. Plasma β-III tubulin, neurofilament light chain and glial fibrillary acidic protein are associated with neurodegeneration and progression in schizophrenia. Sci Rep 2020; 10:14271. [PMID: 32868793 PMCID: PMC7459108 DOI: 10.1038/s41598-020-71060-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/10/2020] [Indexed: 12/29/2022] Open
Abstract
Schizophrenia is a progressive disorder characterized by multiple psychotic relapses. After every relapse, patients may not fully recover, and this may lead to a progressive loss of functionality. Pharmacological treatment represents a key factor to minimize the biological, psychological and psychosocial impact of the disorder. The number of relapses and the duration of psychotic episodes induce a potential neuronal damage and subsequently, neurodegenerative processes. Thus, a comparative study was performed, including forty healthy controls and forty-two SZ patients divided into first-episode psychosis (FEP) and chronic SZ (CSZ) subgroups, where the CSZ sub group was subdivided by antipsychotic treatment. In order to measure the potential neuronal damage, plasma levels of β-III tubulin, neurofilament light chain (Nf-L), and glial fibrillary acidic protein (GFAP) were performed. The results revealed that the levels of these proteins were increased in the SZ group compared to the control group (P < 0.05). Moreover, multiple comparison analysis showed highly significant levels of β-III tubulin (P = 0.0002), Nf-L (P = 0.0403) and GFAP (P < 0.015) in the subgroup of CSZ clozapine-treated. In conclusion, β-III tubulin, Nf-L and GFAP proteins may be potential biomarkers of neurodegeneration and progression in SZ.
Collapse
Affiliation(s)
- Daniela Rodrigues-Amorim
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María Del Carmen Vallejo-Curto
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Cynthia Rodriguez-Jamardo
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Elena de Las Heras
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Carolina Barreiro-Villar
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María Blanco-Formoso
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María Álvarez-Ariza
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Marta López
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | | | - José Manuel Olivares
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain.
- Hospital Álvaro Cunqueiro, Bloque Técnico, Galicia Sur Health Research Institute - IISGS, Planta 2, Sala de Investigación, Estrada Clara Campoamor, 341, 36212, Vigo, Spain.
| | - Carlos Spuch
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain.
- Hospital Álvaro Cunqueiro, Bloque Técnico, Galicia Sur Health Research Institute - IISGS, Planta 2, Sala de Investigación, Estrada Clara Campoamor, 341, 36212, Vigo, Spain.
| |
Collapse
|
65
|
Ajsuvakova OP, Tinkov AA, Aschner M, Rocha JB, Michalke B, Skalnaya MG, Skalny AV, Butnariu M, Dadar M, Sarac I, Aaseth J, Bjørklund G. Sulfhydryl groups as targets of mercury toxicity. Coord Chem Rev 2020; 417:213343. [PMID: 32905350 PMCID: PMC7470069 DOI: 10.1016/j.ccr.2020.213343] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The present study addresses existing data on the affinity and conjugation of sulfhydryl (thiol; -SH) groups of low- and high-molecular-weight biological ligands with mercury (Hg). The consequences of these interactions with special emphasis on pathways of Hg toxicity are highlighted. Cysteine (Cys) is considered the primary target of Hg, and link its sensitivity with thiol groups and cellular damage. In vivo, Hg complexes play a key role in Hg metabolism. Due to the increased affinity of Hg to SH groups in Cys residues, glutathione (GSH) is reactive. The geometry of Hg(II) glutathionates is less understood than that with Cys. Both Cys and GSH Hg-conjugates are important in Hg transport. The binding of Hg to Cys mediates multiple toxic effects of Hg, especially inhibitory effects on enzymes and other proteins that contain free Cys residues. In blood plasma, albumin is the main Hg-binding (Hg2+, CH3Hg+, C2H5Hg+, C6H5Hg+) protein. At the Cys34 residue, Hg2+ binds to albumin, whereas other metals likely are bound at the N-terminal site and multi-metal binding sites. In addition to albumin, Hg binds to multiple Cys-containing enzymes (including manganese-superoxide dismutase (Mn-SOD), arginase I, sorbitol dehydrogenase, and δ-aminolevulinate dehydratase, etc.) involved in multiple processes. The affinity of Hg for thiol groups may also underlie the pathways of Hg toxicity. In particular, Hg-SH may contribute to apoptosis modulation by interfering with Akt/CREB, Keap1/Nrf2, NF-κB, and mitochondrial pathways. Mercury-induced oxidative stress may ensue from Cys-Hg binding and inhibition of Mn-SOD (Cys196), thioredoxin reductase (TrxR) (Cys497) activity, as well as limiting GSH (GS-HgCH3) and Trx (Cys32, 35, 62, 65, 73) availability. Moreover, Hg-thiol interaction also is crucial in the neurotoxicity of Hg by modulating the cytoskeleton and neuronal receptors, to name a few. However, existing data on the role of Hg-SH binding in the Hg toxicity remains poorly defined. Therefore, more research is needed to understand better the role of Hg-thiol binding in the molecular pathways of Hg toxicology and the critical role of thiols to counteract negative effects of Hg overload.
Collapse
Affiliation(s)
- Olga P. Ajsuvakova
- Yaroslavl State University, Yaroslavl, Russia
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexey A. Tinkov
- Yaroslavl State University, Yaroslavl, Russia
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Michael Aschner
- IM Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - João B.T. Rocha
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | | | - Anatoly V. Skalny
- Yaroslavl State University, Yaroslavl, Russia
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Monica Butnariu
- Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timișoara, Timişoara, Romania
- CONEM Romania Biotechnology and Environmental Sciences Group, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timișoara, Timişoara, Romania
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Ioan Sarac
- Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timișoara, Timişoara, Romania
- CONEM Romania Biotechnology and Environmental Sciences Group, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timișoara, Timişoara, Romania
| | - Jan Aaseth
- IM Sechenov First Moscow State Medical University, Moscow, Russia
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| |
Collapse
|
66
|
Mori T, Kitani Y, Hatakeyama D, Machida K, Goto-Inoue N, Hayakawa S, Yamamoto N, Kashiwagi K, Kashiwagi A. Predation threats for a 24-h period activated the extension of axons in the brains of Xenopus tadpoles. Sci Rep 2020; 10:11737. [PMID: 32678123 PMCID: PMC7367293 DOI: 10.1038/s41598-020-67975-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/24/2020] [Indexed: 11/24/2022] Open
Abstract
The threat of predation is a driving force in the evolution of animals. We have previously reported that Xenopus laevis enhanced their tail muscles and increased their swimming speeds in the presence of Japanese larval salamander predators. Herein, we investigated the induced gene expression changes in the brains of tadpoles under the threat of predation using 3′-tag digital gene expression profiling. We found that many muscle genes were expressed after 24 h of exposure to predation. Ingenuity pathway analysis further showed that after 24 h of a predation threat, various signal transduction genes were stimulated, such as those affecting the actin cytoskeleton and CREB pathways, and that these might increase microtubule dynamics, axonogenesis, cognition, and memory. To verify the increase in microtubule dynamics, DiI was inserted through the tadpole nostrils. Extension of the axons was clearly observed from the nostril to the diencephalon and was significantly increased (P ≤ 0.0001) after 24 h of exposure to predation, compared with that of the control. The dynamic changes in the signal transductions appeared to bring about new connections in the neural networks, as suggested by the microtubule dynamics. These connections may result in improved memory and cognition abilities, and subsequently increase survivability.
Collapse
Affiliation(s)
- Tsukasa Mori
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan.
| | - Yoichiro Kitani
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan.,Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa, Japan
| | - Den Hatakeyama
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan
| | - Kazumasa Machida
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan
| | - Naoko Goto-Inoue
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan
| | - Satoshi Hayakawa
- Department of Pathology and Microbiology, School of Medicine, Nihon University, Tokyo, Japan
| | - Naoyuki Yamamoto
- Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Keiko Kashiwagi
- Amphibian Research Center (Building M), Hiroshima University, Hiroshima, Japan
| | - Akihiko Kashiwagi
- Amphibian Research Center (Building M), Hiroshima University, Hiroshima, Japan
| |
Collapse
|
67
|
Nguyen LD, Ehrlich BE. Cellular mechanisms and treatments for chemobrain: insight from aging and neurodegenerative diseases. EMBO Mol Med 2020; 12:e12075. [PMID: 32346964 PMCID: PMC7278555 DOI: 10.15252/emmm.202012075] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/09/2020] [Accepted: 04/01/2020] [Indexed: 12/22/2022] Open
Abstract
Chemotherapy is a life-saving treatment for cancer patients, but also causes long-term cognitive impairment, or "chemobrain", in survivors. However, several challenges, including imprecise diagnosis criteria, multiple confounding factors, and unclear and heterogeneous molecular mechanisms, impede effective investigation of preventions and treatments for chemobrain. With the rapid increase in the number of cancer survivors, chemobrain is an urgent but unmet clinical need. Here, we leverage the extensive knowledge in various fields of neuroscience to gain insights into the mechanisms for chemobrain. We start by outlining why the post-mitotic adult brain is particularly vulnerable to chemotherapy. Next, through drawing comparisons with normal aging, Alzheimer's disease, and traumatic brain injury, we identify universal cellular mechanisms that may underlie the cognitive deficits in chemobrain. We further identify existing neurological drugs targeting these cellular mechanisms that can be repurposed as treatments for chemobrain, some of which were already shown to be effective in animal models. Finally, we briefly describe future steps to further advance our understanding of chemobrain and facilitate the development of effective preventions and treatments.
Collapse
Affiliation(s)
- Lien D Nguyen
- Department of Pharmacology and Interdepartmental Neuroscience ProgramYale UniversityNew HavenCTUSA
| | - Barbara E Ehrlich
- Department of Pharmacology and Interdepartmental Neuroscience ProgramYale UniversityNew HavenCTUSA
| |
Collapse
|
68
|
Vahsen BF, Ribas VT, Sundermeyer J, Boecker A, Dambeck V, Lenz C, Shomroni O, Caldi Gomes L, Tatenhorst L, Barski E, Roser AE, Michel U, Urlaub H, Salinas G, Bähr M, Koch JC, Lingor P. Inhibition of the autophagic protein ULK1 attenuates axonal degeneration in vitro and in vivo, enhances translation, and modulates splicing. Cell Death Differ 2020; 27:2810-2827. [PMID: 32341448 PMCID: PMC7493890 DOI: 10.1038/s41418-020-0543-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
Axonal degeneration is a key and early pathological feature in traumatic and neurodegenerative disorders of the CNS. Following a focal lesion to axons, extended axonal disintegration by acute axonal degeneration (AAD) occurs within several hours. During AAD, the accumulation of autophagic proteins including Unc-51 like autophagy activating kinase 1 (ULK1) has been demonstrated, but its role is incompletely understood. Here, we study the effect of ULK1 inhibition in different models of lesion-induced axonal degeneration in vitro and in vivo. Overexpression of a dominant negative of ULK1 (ULK1.DN) in primary rat cortical neurons attenuates axotomy-induced AAD in vitro. Both ULK1.DN and the ULK1 inhibitor SBI-0206965 protect against AAD after rat optic nerve crush in vivo. ULK1.DN additionally attenuates long-term axonal degeneration after rat spinal cord injury in vivo. Mechanistically, ULK1.DN decreases autophagy and leads to an mTOR-mediated increase in translational proteins. Consistently, treatment with SBI-0206965 results in enhanced mTOR activation. ULK1.DN additionally modulates the differential splicing of the degeneration-associated genes Kif1b and Ddit3. These findings uncover ULK1 as an important mediator of axonal degeneration in vitro and in vivo, and elucidate its function in splicing, defining it as a putative therapeutic target.
Collapse
Affiliation(s)
- Björn Friedhelm Vahsen
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Vinicius Toledo Ribas
- Department of Morphology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Jonas Sundermeyer
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Alexander Boecker
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 630 Clinical Research Building, 415 Curie Boulevard, Philadelphia, PA, 19104, USA
| | - Vivian Dambeck
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Von-Siebold-Str. 3a, 37075, Göttingen, Germany.,DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Christof Lenz
- Institute of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany
| | - Orr Shomroni
- NGS-Integrative Genomics Core Unit (NIG), Institute of Human Genetics, University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Lucas Caldi Gomes
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Lars Tatenhorst
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Von-Siebold-Str. 3a, 37075, Göttingen, Germany.,DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Elisabeth Barski
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Anna-Elisa Roser
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Uwe Michel
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Henning Urlaub
- Institute of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit (NIG), Institute of Human Genetics, University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Jan Christoph Koch
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Paul Lingor
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany. .,Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Von-Siebold-Str. 3a, 37075, Göttingen, Germany. .,DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany. .,Department of Neurology, Rechts der Isar Hospital, Technical University Munich, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
69
|
Reduced TUBA1A Tubulin Causes Defects in Trafficking and Impaired Adult Motor Behavior. eNeuro 2020; 7:ENEURO.0045-20.2020. [PMID: 32184299 PMCID: PMC7218002 DOI: 10.1523/eneuro.0045-20.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/03/2020] [Indexed: 12/30/2022] Open
Abstract
Newly born neurons express high levels of TUBA1A α-tubulin to assemble microtubules for neurite extension and to provide tracks for intracellular transport. In the adult brain, Tuba1a expression decreases dramatically. A mouse that harbors a loss-of-function mutation in the gene encoding TUBA1A (Tuba1aND/+) allows us to ask whether TUBA1A is important for the function of mature neurons. α-Tubulin levels are about half of wild type in juvenile Tuba1aND/+ brains, but are close to normal in older animals. In postnatal day (P)0 cultured neurons, reduced TUBA1A allows for assembly of less microtubules in axons resulting in more pausing during organelle trafficking. While Tuba1aND/+ mouse behavior is indistinguishable from wild-type siblings at weaning, Tuba1aND/+ mice develop adult-onset ataxia. Neurons important for motor function in Tuba1aND/+ remain indistinguishable from wild-type with respect to morphology and number and display no evidence of axon degeneration. Tuba1aND/+ neuromuscular junction (NMJ) synapses are the same size as wild-type before the onset of ataxia, but are reduced in size in older animals. Together, these data indicate that the TUBA1A-rich microtubule tracks that are assembled during development are essential for mature neuron function and maintenance of synapses over time.
Collapse
|
70
|
Shi Q, Pei F, Silverman GA, Pak SC, Perlmutter DH, Liu B, Bahar I. Mechanisms of Action of Autophagy Modulators Dissected by Quantitative Systems Pharmacology Analysis. Int J Mol Sci 2020; 21:ijms21082855. [PMID: 32325894 PMCID: PMC7215584 DOI: 10.3390/ijms21082855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy plays an essential role in cell survival/death and functioning. Modulation of autophagy has been recognized as a promising therapeutic strategy against diseases/disorders associated with uncontrolled growth or accumulation of biomolecular aggregates, organelles, or cells including those caused by cancer, aging, neurodegeneration, and liver diseases such as α1-antitrypsin deficiency. Numerous pharmacological agents that enhance or suppress autophagy have been discovered. However, their molecular mechanisms of action are far from clear. Here, we collected a set of 225 autophagy modulators and carried out a comprehensive quantitative systems pharmacology (QSP) analysis of their targets using both existing databases and predictions made by our machine learning algorithm. Autophagy modulators include several highly promiscuous drugs (e.g., artenimol and olanzapine acting as activators, fostamatinib as an inhibitor, or melatonin as a dual-modulator) as well as selected drugs that uniquely target specific proteins (~30% of modulators). They are mediated by three layers of regulation: (i) pathways involving core autophagy-related (ATG) proteins such as mTOR, AKT, and AMPK; (ii) upstream signaling events that regulate the activity of ATG pathways such as calcium-, cAMP-, and MAPK-signaling pathways; and (iii) transcription factors regulating the expression of ATG proteins such as TFEB, TFE3, HIF-1, FoxO, and NF-κB. Our results suggest that PKA serves as a linker, bridging various signal transduction events and autophagy. These new insights contribute to a better assessment of the mechanism of action of autophagy modulators as well as their side effects, development of novel polypharmacological strategies, and identification of drug repurposing opportunities.
Collapse
Affiliation(s)
- Qingya Shi
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Fen Pei
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
| | - Gary A. Silverman
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - Stephen C. Pak
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - David H. Perlmutter
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - Bing Liu
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- Correspondence: (B.L.); (I.B.)
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- Correspondence: (B.L.); (I.B.)
| |
Collapse
|
71
|
|
72
|
Kwon Y, Jeon YW, Kwon M, Cho Y, Park D, Shin JE. βPix-d promotes tubulin acetylation and neurite outgrowth through a PAK/Stathmin1 signaling pathway. PLoS One 2020; 15:e0230814. [PMID: 32251425 PMCID: PMC7135283 DOI: 10.1371/journal.pone.0230814] [Citation(s) in RCA: 10] [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: 09/30/2019] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Microtubules are a major cytoskeletal component of neurites, and the regulation of microtubule stability is essential for neurite morphogenesis. βPix (ARHGEF7) is a guanine nucleotide exchange factor for the small GTPases Rac1 and Cdc42, which modulate the organization of actin filaments and microtubules. βPix is expressed as alternatively spliced variants, including the ubiquitous isoform βPix-a and the neuronal isoforms βPix-b and βPix-d, but the function of the neuronal isoforms remains unclear. Here, we reveal the novel role of βPix neuronal isoforms in regulating tubulin acetylation and neurite outgrowth. At DIV4, hippocampal neurons cultured from βPix neuronal isoform knockout (βPix-NIKO) mice exhibit defects in neurite morphology and tubulin acetylation, a type of tubulin modification which often labels stable microtubules. Treating βPix-NIKO neurons with paclitaxel, which stabilizes the microtubules, or reintroducing either neuronal βPix isoform to the KO neurons overcomes the impairment in neurite morphology and tubulin acetylation, suggesting that neuronal βPix isoforms may promote microtubule stabilization during neurite development. βPix-NIKO neurons also exhibit lower phosphorylation levels for Stathmin1, a microtubule-destabilizing protein, at Ser16. Expressing either βPix neuronal isoform in the βPix-NIKO neurons restores Stathmin1 phosphorylation levels, with βPix-d having a greater effect than βPix-b. Furthermore, we find that the recovery of neurite length and Stathmin1 phosphorylation via βPix-d expression requires PAK kinase activity. Taken together, our study demonstrates that βPix-d regulates the phosphorylation of Stathmin1 in a PAK-dependent manner and that neuronal βPix isoforms promote tubulin acetylation and neurite morphogenesis during neuronal development.
Collapse
Affiliation(s)
- Younghee Kwon
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ye Won Jeon
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Minjae Kwon
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Yongcheol Cho
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Dongeun Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jung Eun Shin
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Institute of Life Science and Biotechnology, Korea University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
73
|
Ren L, Li Q, You T, Zhao X, Xu X, Tang C, Zhu L. Humanin analogue, HNG, inhibits platelet activation and thrombus formation by stabilizing platelet microtubules. J Cell Mol Med 2020; 24:4773-4783. [PMID: 32174022 PMCID: PMC7176859 DOI: 10.1111/jcmm.15151] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/13/2020] [Accepted: 02/15/2020] [Indexed: 12/20/2022] Open
Abstract
HNG, a highly potent mutant of the anti-Alzheimer peptide-humanin, has been shown to protect against ischaemia-reperfusion (I/R) injury. However, the underlying mechanism related to platelet activation remains unknown. We proposed that HNG has an effect on platelet function and thrombus formation. In this study, platelet aggregation, granule secretion, clot retraction, integrin activation and adhesion under flow conditions were evaluated. In mice receiving HNG or saline, cremaster arterial thrombus formation induced by laser injury, tail bleeding time and blood loss were recorded. Platelet microtubule depolymerization was evaluated using immunofluorescence staining. Results showed that HNG inhibited platelet aggregation, P-selectin expression, ATP release, and αIIb β3 activation and adhesion under flow conditions. Mice receiving HNG had attenuated cremaster arterial thrombus formation, although the bleeding time was not prolonged. Moreover, HNG significantly inhibited microtubule depolymerization, enhanced tubulin acetylation in platelets stimulated by fibrinogen or microtubule depolymerization reagent, nocodazole, and inhibited AKT and ERK phosphorylation downstream of HDAC6 by collagen stimulation. Therefore, our results identified a novel role of HNG in platelet function and thrombus formation potentially through stabilizing platelet microtubules via tubulin acetylation. These findings suggest a potential benefit of HNG in the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Lijie Ren
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, State Key Laboratory of Radiation Medicine and Protection, Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
| | - Qing Li
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, State Key Laboratory of Radiation Medicine and Protection, Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
| | - Tao You
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, State Key Laboratory of Radiation Medicine and Protection, Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
| | - Xuefei Zhao
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, State Key Laboratory of Radiation Medicine and Protection, Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
| | - Xingshun Xu
- The Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chaojun Tang
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, State Key Laboratory of Radiation Medicine and Protection, Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
| | - Li Zhu
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, State Key Laboratory of Radiation Medicine and Protection, Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, China
| |
Collapse
|
74
|
Affiliation(s)
- Arpan Rai
- Department of Molecular Life Sciences, Ernst Hadorn Chair, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Lucas Pelkmans
- Department of Molecular Life Sciences, Ernst Hadorn Chair, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
75
|
Loehfelm A, Elder MK, Boucsein A, Jones PP, Williams JM, Tups A. Docosahexaenoic acid prevents palmitate-induced insulin-dependent impairments of neuronal health. FASEB J 2020; 34:4635-4652. [PMID: 32030816 DOI: 10.1096/fj.201902517r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/19/2019] [Accepted: 01/20/2020] [Indexed: 02/06/2023]
Abstract
The importance of fatty acids (FAs) for healthy brain development and function has become more evident in the past decades. However, most studies focus on the hypothalamus as an important FA-sensing brain region involved in energy homeostasis. Less work has been done to evaluate the effects of FAs on brain regions such as the hippocampus or cortex, two important centres of learning, memory formation, and cognition. Furthermore, the mechanisms of how FAs modulate the neuronal development and function are incompletely understood. Therefore, this study examined the effects of the saturated FA palmitic acid (PA) and the polyunsaturated FA docosahexaenoic acid (DHA) on primary hippocampal and cortical cultures isolated from P0/P1 Sprague Dawley rat pups. Exposure to PA, but not DHA, resulted in severe morphological changes in primary neurons such as cell body swelling, axonal and dendritic blebbing, and a reduction in synaptic innervation, compromising healthy cell function and excitability. Pharmacological assessment revealed that the PA-mediated alterations were caused by overactivation of neuronal insulin signaling, demonstrated by insulin stimulation and phosphoinositide 3-kinase inhibition. Remarkably, co-exposure to DHA prevented all PA-induced morphological changes. This work provides new insights into how FAs can affect the cytoskeletal rearrangements and neuronal function via modulation of insulin signaling.
Collapse
Affiliation(s)
- Aline Loehfelm
- Department of Physiology, School of Medical Sciences, Centre for Neuroendocrinology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Megan K Elder
- Department of Anatomy, School of Medical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Alisa Boucsein
- Department of Physiology, School of Medical Sciences, Centre for Neuroendocrinology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Peter P Jones
- Department of Physiology and HeartOtago, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Joanna M Williams
- Department of Anatomy, School of Medical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Alexander Tups
- Department of Physiology, School of Medical Sciences, Centre for Neuroendocrinology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
76
|
Dileep Kumar JS, Prabhakaran J, Damuka N, Hines JW, Norman S, Dodda M, John Mann J, Mintz A, Sai KKS. In vivo comparison of N- 11CH 3 vs O- 11CH 3 radiolabeled microtubule targeted PET ligands. Bioorg Med Chem Lett 2020; 30:126785. [PMID: 31753695 PMCID: PMC11659960 DOI: 10.1016/j.bmcl.2019.126785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/17/2019] [Accepted: 10/24/2019] [Indexed: 11/22/2022]
Abstract
Altered dynamics of microtubules (MT) are implicated in the pathophysiology of a number of brain diseases. Therefore, radiolabeled MT targeted ligands that can penetrate the blood brain barrier (BBB) may offer a direct and sensitive approach for diagnosis, and assessing the clinical potential of MT targeted therapeutics using PET imaging. We recently reported two BBB penetrating radioligands, [11C]MPC-6827 and [11C]HD-800 as specific PET ligands for imaging MTs in brain. The major metabolic pathway of the above molecules is anticipated to be via the initial labeling site, O-methyl, compared to the N-methyl group. Herein, we report the radiosynthesis of N-11CH3-MPC-6827 and N-11CH3-HD-800 and a comparison of their in vivo binding with the corresponding O-11CH3 analogues using microPET imaging and biodistribution methods. Both O-11CH3 and N-11CH3 labeled MT tracers exhibit high specific binding and brain. The N-11CH3 labeled PET ligands demonstrated similar in vivo binding characteristics compared with the corresponding O-11CH3 labeled tracers, [11C]MPC-6827 and [11C]HD-800 respectively.
Collapse
Affiliation(s)
- J S Dileep Kumar
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA.
| | - Jaya Prabhakaran
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Medical Center, New York, USA
| | - Naresh Damuka
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA
| | - Justin Wayne Hines
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Skylar Norman
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Meghana Dodda
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - J John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Medical Center, New York, USA; Department of Radiology, Columbia University Medical Center, New York, USA
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, USA
| | | |
Collapse
|
77
|
Chiha W, Bartlett CA, Petratos S, Fitzgerald M, Harvey AR. Intravitreal application of AAV-BDNF or mutant AAV-CRMP2 protects retinal ganglion cells and stabilizes axons and myelin after partial optic nerve injury. Exp Neurol 2020; 326:113167. [PMID: 31904385 DOI: 10.1016/j.expneurol.2019.113167] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/20/2019] [Accepted: 12/31/2019] [Indexed: 12/29/2022]
Abstract
Secondary degeneration following an initial injury to the central nervous system (CNS) results in increased tissue loss and is associated with increasing functional impairment. Unilateral partial dorsal transection of the adult rat optic nerve (ON) has proved to be a useful experimental model in which to study factors that contribute to secondary degenerative events. Using this injury model, we here quantified the protective effects of intravitreally administered bi-cistronic adeno-associated viral (AAV2) vectors encoding either brain derived neurotrophic factor (BDNF) or a mutant, phospho-resistant, version of collapsin response mediator protein 2 (CRMP2T555A) on retinal ganglion cells (RGCs), their axons, and associated myelin. To test for potential synergistic interactions, some animals received combined injections of both vectors. Three months post-injury, all treatments maintained RGC numbers in central retina, but only AAV2-BDNF significantly protected ventrally located RGCs exclusively vulnerable to secondary degeneration. Behaviourally, treatments that involved AAV2-BDNF significantly restored the number of smooth-pursuit phases of optokinetic nystagmus. While all therapeutic regimens preserved axonal density and proportions of typical complexes, including heminodes and single nodes, BDNF treatments were generally more effective in maintaining the length of the node of Ranvier in myelin surrounding ventral ON axons after injury. Both AAV2-BDNF and AAV2-CRMP2T555A prevented injury-induced changes in G-ratio and overall myelin thickness, but only AAV2-BDNF administration protected against large-scale myelin decompaction in ventral ON. In summary, in a model of secondary CNS degeneration, both BDNF and CRMP2T555A vectors were neuroprotective, however different efficacies were observed for these overexpressed proteins in the retina and ON, suggesting disparate cellular and molecular targets driving responses for neural repair. The potential use of these vectors to treat other CNS injuries and pathologies is discussed.
Collapse
Affiliation(s)
- Wissam Chiha
- School of Biological Sciences, The University of Western Australia, WA 6009, Australia; Curtin Health Innovation Research Institute, Curtin University, Belmont, WA 6102, Australia
| | - Carole A Bartlett
- School of Biological Sciences, The University of Western Australia, WA 6009, Australia
| | - Steven Petratos
- Department of Neuroscience, Monash University, VIC 3004, Australia
| | - Melinda Fitzgerald
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia; Curtin Health Innovation Research Institute, Curtin University, Belmont, WA 6102, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, WA 6009, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia.
| |
Collapse
|
78
|
Casagrande FV, Amadeo A, Cartelli D, Calogero AM, Modena D, Costa I, Cantele F, Onelli E, Moscatelli A, Ascagni M, Pezzoli G, Cappelletti G. The imbalance between dynamic and stable microtubules underlies neurodegeneration induced by 2,5-hexanedione. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165581. [DOI: 10.1016/j.bbadis.2019.165581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/17/2019] [Accepted: 10/12/2019] [Indexed: 01/10/2023]
|
79
|
Kovalska M, Hnilicova P, Kalenska D, Tothova B, Adamkov M, Lehotsky J. Effect of Methionine Diet on Metabolic and Histopathological Changes of Rat Hippocampus. Int J Mol Sci 2019; 20:ijms20246234. [PMID: 31835644 PMCID: PMC6941024 DOI: 10.3390/ijms20246234] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022] Open
Abstract
Hyperhomocysteinemia (hHcy) is regarded as an independent and strong risk factor for cerebrovascular diseases, stroke, and dementias. The hippocampus has a crucial role in spatial navigation and memory processes and is being constantly studied for neurodegenerative disorders. We used a moderate methionine (Met) diet at a dose of 2 g/kg of animal weight/day in duration of four weeks to induce mild hHcy in adult male Wistar rats. A novel approach has been used to explore the hippocampal metabolic changes using proton magnetic resonance spectroscopy (1H MRS), involving a 7T MR scanner in combination with histochemical and immunofluorescence analysis. We found alterations in the metabolic profile, as well as remarkable histo-morphological changes such as an increase of hippocampal volume, alterations in number and morphology of astrocytes, neurons, and their processes in the selective vulnerable brain area of animals treated with a Met-enriched diet. Results of both methodologies suggest that the mild hHcy induced by Met-enriched diet alters volume, histo-morphological pattern, and metabolic profile of hippocampal brain area, which might eventually endorse the neurodegenerative processes.
Collapse
Affiliation(s)
- Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.K.); (M.A.)
| | - Petra Hnilicova
- Department of Neuroscience, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Barbara Tothova
- Department of Molecular Medicine, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.K.); (M.A.)
| | - Jan Lehotsky
- Department of Neuroscience, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
- Correspondence: ; Tel.: +421-43-2633-821
| |
Collapse
|
80
|
Amselem S. Remote Controlled Autonomous Microgravity Lab Platforms for Drug Research in Space. Pharm Res 2019; 36:183. [PMID: 31741058 DOI: 10.1007/s11095-019-2703-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/13/2019] [Indexed: 12/23/2022]
Abstract
Research conducted in microgravity conditions has the potential to yield new therapeutics, as advances can be achieved in the absence of phenomena such as sedimentation, hydrostatic pressure and thermally-induced convection. The outcomes of such studies can significantly contribute to many scientific and technological fields, including drug discovery. This article reviews the existing traditional microgravity platforms as well as emerging ideas for enabling microgravity research focusing on SpacePharma's innovative autonomous remote-controlled microgravity labs that can be launched to space aboard nanosatellites to perform drug research in orbit. The scientific literature is reviewed and examples of life science fields that have benefited from studies in microgravity conditions are given. These include the use of microgravity environment for chemical applications (protein crystallization, drug polymorphism, self-assembly of biomolecules), pharmaceutical studies (microencapsulation, drug delivery systems, behavior and stability of colloidal formulations, antibiotic drug resistance), and biological research, including accelerated models for aging, investigation of bacterial virulence , tissue engineering using organ-on-chips in space, enhanced stem cells proliferation and differentiation.
Collapse
Affiliation(s)
- Shimon Amselem
- SpacePharma R&D Israel LTD, 1st Aba Even Av, 4672519, Herzliya Pituach, Israel.
- SpacePharma SA, Rue l'Armeratte 3, 2950, Courgenay, Switzerland.
| |
Collapse
|
81
|
A Synthetic Snake-Venom-Based Tripeptide Protects PC12 Cells from the Neurotoxicity of Acrolein by Improving Axonal Plasticity and Bioenergetics. Neurotox Res 2019; 37:227-237. [DOI: 10.1007/s12640-019-00111-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
|
82
|
Sabharwal V, Koushika SP. Crowd Control: Effects of Physical Crowding on Cargo Movement in Healthy and Diseased Neurons. Front Cell Neurosci 2019; 13:470. [PMID: 31708745 PMCID: PMC6823667 DOI: 10.3389/fncel.2019.00470] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 10/02/2019] [Indexed: 01/22/2023] Open
Abstract
High concentration of cytoskeletal filaments, organelles, and proteins along with the space constraints due to the axon's narrow geometry lead inevitably to intracellular physical crowding along the axon of a neuron. Local cargo movement is essential for maintaining steady cargo transport in the axon, and this may be impeded by physical crowding. Molecular motors that mediate active transport share movement mechanisms that allow them to bypass physical crowding present on microtubule tracks. Many neurodegenerative diseases, irrespective of how they are initiated, show increased physical crowding owing to the greater number of stalled organelles and structural changes associated with the cytoskeleton. Increased physical crowding may be a significant factor in slowing cargo transport to synapses, contributing to disease progression and culminating in the dying back of the neuronal process. This review explores the idea that physical crowding can impede cargo movement along the neuronal process. We examine the sources of physical crowding and strategies used by molecular motors that might enable cargo to circumvent physically crowded locations. Finally, we describe sub-cellular changes in neurodegenerative diseases that may alter physical crowding and discuss the implications of such changes on cargo movement.
Collapse
Affiliation(s)
| | - Sandhya P. Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
83
|
Kim K, Subramaniyam S, Galaleldeen A, Nakazawa H, Umetsu M, Teizer W, Bhattacharyya S. Nanoparticle Assisted Remodeling of Proteotoxic SOD1 Mutants Alters the Biointerface of the Functional Interaction of Microtubules and Kinesin Motors. ACS APPLIED BIO MATERIALS 2019; 2:4121-4128. [PMID: 35021426 DOI: 10.1021/acsabm.9b00501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Transport deficits with motor neuron degeneration have been implicated in amyotrophic lateral sclerosis (ALS). We report a biomimetic system composed of microtubules/kinesin motor that mimics the dysregulated motor dynamics of ALS. Pathogenic ALS mutants A4V SOD1 completely shut off motility. Treatment with 5 nm citrate coated gold nanoparticles recovers the impaired motor stepping by remodeling the A4V SOD1 rather than stabilizing microtubules or protein folding. Instead, gold nanoparticles alter the protein by a mechanism that reforms protein elements of A4V SOD1, in turn fixing the aberrant interaction of kinesin with microtubules. Reinstating kinesin motility holds potential for managing debilitating ALS.
Collapse
Affiliation(s)
| | - Selvaraj Subramaniyam
- Department of Pharmaceutical Science and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Ahmad Galaleldeen
- Department of Biological Science, St Mary's University, San Antonio, Texas 78228, United States
| | | | | | | | - Sanjib Bhattacharyya
- Department of Pharmaceutical Science and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing 400715, China
| |
Collapse
|
84
|
Bernstein AM, Ritch R, Wolosin JM. LOXL1 folding in exfoliation glaucoma. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 118:273-288. [PMID: 31928728 PMCID: PMC7589528 DOI: 10.1016/bs.apcsb.2019.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Exfoliation syndrome (XFS) is an age-related disease defined by the deposition of aggregated fibrous material (XFM) in the peri-cellular space. Principal morbidity occurs in the eye, where XFM accumulates on the anterior ocular tissues. GWAS have found that certain genetic variants of lysyl oxidase-like 1 (LOXL1), a matrix cross-linking enzyme that is required for elastic fiber formation confer risk for the development of XFS, but are not a single causative factor as many genetically affected individuals do not develop XFS or subsequent glaucoma (XFG). We have found that XFG cells display defects in lysosomes, microtubules, autophagy, and mitochondria resembling defects found in cells from age-related syndromes, such as the main neurodegenerative diseases. In the majority of these diseases, the determining cellular factor is a protein containing intrinsically disordered regions (IDRs) and displaying a high propensity for aggregation. We have found that in XFG patient-derived cells, LOXL1 protein is actively subjected to autophagic clearance, suggesting that LOXL1 is undergoing aggregation. In silico analysis demonstrates that LOXL1's first 369 aa constitute an IDR with the highest disorder probability peak centering around the known risk positions. Experimentally, we have found over-expression of either unmodified LOXL1 or fluorescent chimeras preserving the well-structured N-terminus cause copious intracellular aggregation and that aggregation wanes when the high IDR peaks are deleted. Overall, our work suggests that XFS/G results from the aggregation of the LOXL1 protein coupled with a reduction of cellular proteostasis capabilities in aging, resulting in a chronic build-up of LOXL1-containing protein aggregates.
Collapse
Affiliation(s)
- Audrey M. Bernstein
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, Syracuse, NY, United States
| | - Robert Ritch
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY, United States
| | - J. Mario Wolosin
- Eye and Vision Research Institute, Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
85
|
|
86
|
Chafai DE, Sulimenko V, Havelka D, Kubínová L, Dráber P, Cifra M. Reversible and Irreversible Modulation of Tubulin Self-Assembly by Intense Nanosecond Pulsed Electric Fields. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1903636. [PMID: 31408579 DOI: 10.1002/adma.201903636] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/26/2019] [Indexed: 05/21/2023]
Abstract
Tubulin self-assembly into microtubules is a fascinating natural phenomenon. Its importance is not just crucial for functional and structural biological processes, but it also serves as an inspiration for synthetic nanomaterial innovations. The modulation of the tubulin self-assembly process without introducing additional chemical inhibitors/promoters or stabilizers has remained an elusive process. This work reports a versatile and vigorous strategy for controlling tubulin self-assembly by nanosecond electropulses (nsEPs). The polymerization assessed by turbidimetry is dependent on nsEPs dosage. The kinetics of microtubules formation is tightly linked to the nsEPs effects on structural properties of tubulin, and tubulin-solvent interface, assessed by autofluorescence, and the zeta potential. Moreover, the overall size of tubulin assessed by dynamic light scattering is affected as well. Additionally, atomic force microscopy imaging reveals the formation of different assemblies reflecting applied nsEPs. It is suggested that changes in C-terminal modification states alter tubulin polymerization-competent conformations. Although the assembled tubulin preserve their integral structure, they might exhibit a broad range of new properties important for their functions. Thus, these transient conformation changes of tubulin and their collective properties can result in new applications.
Collapse
Affiliation(s)
- Djamel Eddine Chafai
- Bioelectrodynamics Research Team, Institute of Photonics and Electronics of the Czech Academy of Sciences, Chaberská 57, 182 51, Prague, Czech Republic
| | - Vadym Sulimenko
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Daniel Havelka
- Bioelectrodynamics Research Team, Institute of Photonics and Electronics of the Czech Academy of Sciences, Chaberská 57, 182 51, Prague, Czech Republic
| | - Lucie Kubínová
- Department of Biomathematics, Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Pavel Dráber
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Michal Cifra
- Bioelectrodynamics Research Team, Institute of Photonics and Electronics of the Czech Academy of Sciences, Chaberská 57, 182 51, Prague, Czech Republic
| |
Collapse
|
87
|
Neelagandan N, Gonnella G, Dang S, Janiesch PC, Miller KK, Küchler K, Marques RF, Indenbirken D, Alawi M, Grundhoff A, Kurtz S, Duncan KE. TDP-43 enhances translation of specific mRNAs linked to neurodegenerative disease. Nucleic Acids Res 2019; 47:341-361. [PMID: 30357366 PMCID: PMC6326785 DOI: 10.1093/nar/gky972] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
The RNA-binding protein TDP-43 is heavily implicated in neurodegenerative disease. Numerous patient mutations in TARDBP, the gene encoding TDP-43, combined with data from animal and cell-based models, imply that altered RNA regulation by TDP-43 causes Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. However, underlying mechanisms remain unresolved. Increased cytoplasmic TDP-43 levels in diseased neurons suggest a possible role in this cellular compartment. Here, we examined the impact on translation of overexpressing human TDP-43 and the TDP-43A315T patient mutant protein in motor neuron-like cells and primary cultures of cortical neurons. In motor-neuron like cells, TDP-43 associates with ribosomes without significantly affecting global translation. However, ribosome profiling and additional assays revealed enhanced translation and direct binding of Camta1, Mig12, and Dennd4a mRNAs. Overexpressing either wild-type TDP-43 or TDP-43A315T stimulated translation of Camta1 and Mig12 mRNAs via their 5'UTRs and increased CAMTA1 and MIG12 protein levels. In contrast, translational enhancement of Dennd4a mRNA required a specific 3'UTR region and was specifically observed with the TDP-43A315T patient mutant allele. Our data reveal that TDP-43 can function as an mRNA-specific translational enhancer. Moreover, since CAMTA1 and DENND4A are linked to neurodegeneration, they suggest that this function could contribute to disease.
Collapse
Affiliation(s)
- Nagammal Neelagandan
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Giorgio Gonnella
- Universität Hamburg, MIN-Fakultät, ZBH-Center for Bioinformatics, Hamburg 20146, Germany
| | - Stefan Dang
- Universität Hamburg, MIN-Fakultät, ZBH-Center for Bioinformatics, Hamburg 20146, Germany
| | - Philipp C Janiesch
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Katharine K Miller
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Katrin Küchler
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Rita F Marques
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Daniela Indenbirken
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Malik Alawi
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany.,Bioinformatics Core, University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Adam Grundhoff
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Stefan Kurtz
- Universität Hamburg, MIN-Fakultät, ZBH-Center for Bioinformatics, Hamburg 20146, Germany
| | - Kent E Duncan
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| |
Collapse
|
88
|
Berger J, Berger S, Li M, Jacoby AS, Arner A, Bavi N, Stewart AG, Currie PD. In Vivo Function of the Chaperonin TRiC in α-Actin Folding during Sarcomere Assembly. Cell Rep 2019; 22:313-322. [PMID: 29320728 DOI: 10.1016/j.celrep.2017.12.069] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/11/2017] [Accepted: 12/19/2017] [Indexed: 12/14/2022] Open
Abstract
The TCP-1 ring complex (TRiC) is a multi-subunit group II chaperonin that assists nascent or misfolded proteins to attain their native conformation in an ATP-dependent manner. Functional studies in yeast have suggested that TRiC is an essential and generalized component of the protein-folding machinery of eukaryotic cells. However, TRiC's involvement in specific cellular processes within multicellular organisms is largely unknown because little validation of TRiC function exists in animals. Our in vivo analysis reveals a surprisingly specific role of TRiC in the biogenesis of skeletal muscle α-actin during sarcomere assembly in myofibers. TRiC acts at the sarcomere's Z-disk, where it is required for efficient assembly of actin thin filaments. Binding of ATP specifically by the TRiC subunit Cct5 is required for efficient actin folding in vivo. Furthermore, mutant α-actin isoforms that result in nemaline myopathy in patients obtain their pathogenic conformation via this function of TRiC.
Collapse
Affiliation(s)
- Joachim Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Victoria Node, EMBL Australia, Clayton, VIC 3800, Australia.
| | - Silke Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Victoria Node, EMBL Australia, Clayton, VIC 3800, Australia
| | - Mei Li
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Victoria Node, EMBL Australia, Clayton, VIC 3800, Australia; Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Arie S Jacoby
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Victoria Node, EMBL Australia, Clayton, VIC 3800, Australia
| | - Anders Arner
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Navid Bavi
- Department of Physiology, School of Medical Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Alastair G Stewart
- Molecular, Structural and Computational Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; Faculty of Medicine, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Victoria Node, EMBL Australia, Clayton, VIC 3800, Australia.
| |
Collapse
|
89
|
Bercier V, Hubbard JM, Fidelin K, Duroure K, Auer TO, Revenu C, Wyart C, Del Bene F. Dynactin1 depletion leads to neuromuscular synapse instability and functional abnormalities. Mol Neurodegener 2019; 14:27. [PMID: 31291987 PMCID: PMC6617949 DOI: 10.1186/s13024-019-0327-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dynactin subunit 1 is the largest subunit of the dynactin complex, an activator of the molecular motor protein complex dynein. Reduced levels of DCTN1 mRNA and protein have been found in sporadic amyotrophic lateral sclerosis (ALS) patients, and mutations have been associated with disease, but the role of this protein in disease pathogenesis is still unknown. METHODS We characterized a Dynactin1a depletion model in the zebrafish embryo and combined in vivo molecular analysis of primary motor neuron development with live in vivo axonal transport assays in single cells to investigate ALS-related defects. To probe neuromuscular junction (NMJ) function and organization we performed paired motor neuron-muscle electrophysiological recordings and GCaMP calcium imaging in live, intact larvae, and the synapse structure was investigated by electron microscopy. RESULTS Here we show that Dynactin1a depletion is sufficient to induce defects in the development of spinal cord motor neurons and in the function of the NMJ. We observe synapse instability, impaired growth of primary motor neurons, and higher failure rates of action potentials at the NMJ. In addition, the embryos display locomotion defects consistent with NMJ dysfunction. Rescue of the observed phenotype by overexpression of wild-type human DCTN1-GFP indicates a cell-autonomous mechanism. Synaptic accumulation of DCTN1-GFP, as well as ultrastructural analysis of NMJ synapses exhibiting wider synaptic clefts, support a local role for Dynactin1a in synaptic function. Furthermore, live in vivo analysis of axonal transport and cytoskeleton dynamics in primary motor neurons show that the phenotype reported here is independent of modulation of these processes. CONCLUSIONS Our study reveals a novel role for Dynactin1 in ALS pathogenesis, where it acts cell-autonomously to promote motor neuron synapse stability independently of dynein-mediated axonal transport.
Collapse
Affiliation(s)
- Valérie Bercier
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
- Present Address: VIB-KU Leuven, Center for Brain & Disease Research, Leuven, Belgium
| | - Jeffrey M. Hubbard
- Sorbonne Université, Inserm, CNRS, AP-HP, Institut du Cerveau et de la Moelle Épinière, ICM, F-75013 Paris, France
| | - Kevin Fidelin
- Sorbonne Université, Inserm, CNRS, AP-HP, Institut du Cerveau et de la Moelle Épinière, ICM, F-75013 Paris, France
- Present Address: Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA
| | - Karine Duroure
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
| | - Thomas O. Auer
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
- Present Address: Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Céline Revenu
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
| | - Claire Wyart
- Sorbonne Université, Inserm, CNRS, AP-HP, Institut du Cerveau et de la Moelle Épinière, ICM, F-75013 Paris, France
| | - Filippo Del Bene
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
| |
Collapse
|
90
|
Fasterius E, Uhlén M, Al-Khalili Szigyarto C. Single-cell RNA-seq variant analysis for exploration of genetic heterogeneity in cancer. Sci Rep 2019; 9:9524. [PMID: 31267007 PMCID: PMC6606766 DOI: 10.1038/s41598-019-45934-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 06/20/2019] [Indexed: 01/23/2023] Open
Abstract
Inter- and intra-tumour heterogeneity is caused by genetic and non-genetic factors, leading to severe clinical implications. High-throughput sequencing technologies provide unprecedented tools to analyse DNA and RNA in single cells and explore both genetic heterogeneity and phenotypic variation between cells in tissues and tumours. Simultaneous analysis of both DNA and RNA in the same cell is, however, still in its infancy. We have thus developed a method to extract and analyse information regarding genetic heterogeneity that affects cellular biology from single-cell RNA-seq data. The method enables both comparisons and clustering of cells based on genetic variation in single nucleotide variants, revealing cellular subpopulations corroborated by gene expression-based methods. Furthermore, the results show that lymph node metastases have lower levels of genetic heterogeneity compared to their original tumours with respect to variants affecting protein function. The analysis also revealed three previously unknown variants common across cancer cells in glioblastoma patients. These results demonstrate the power and versatility of scRNA-seq variant analysis and highlight it as a useful complement to already existing methods, enabling simultaneous investigations of both gene expression and genetic variation.
Collapse
Affiliation(s)
- Erik Fasterius
- School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.,Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Cristina Al-Khalili Szigyarto
- School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden. .,Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.
| |
Collapse
|
91
|
Charcot-Marie-Tooth 2F (Hsp27 mutations): A review. Neurobiol Dis 2019; 130:104505. [PMID: 31212070 DOI: 10.1016/j.nbd.2019.104505] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
Charcot-Marie-Tooth disease is a commonly inherited form of neuropathy. Although named over 100 years ago, identification of subtypes of Charcot-Marie-Tooth has rapidly expanded in the preceding decades with the advancement of genetic sequencing, including type 2F (CMT2F), due to mutations in heat shock protein 27 (Hsp27). However, despite CMT being one of the most common inherited neurological diseases, definitive mechanistic models of pathology and effective treatments for CMT2F are lacking. This review extensively profiles the published literature on CMT2F and distal hereditary motor neuropathy II (dHMN II), a similar neuropathy with exclusively motor symptoms that is also due to mutations in Hsp27. This includes a review of case reports and sequencing studies detailing disease course. Included are tables listing of all known published mutations of Hsp27 that cause symptoms of CMT2F and dHMN II. Furthermore, pathological mechanisms are assessed. While many groups have established pathologies relating to defective chaperone function, cellular neurofilament and microtubule structure and function, and mitochondrial and metabolic dysfunction, there are still discrepancies in results between different model systems. Moreover, initial mouse models have also produced promising results with similar phenotypes to humans, however discrepancies still exist. Both patient-focused and scientific studies have demonstrated variability in phenotypes even considering specific mutations. Given the clinical heterogeneity in presentation, CMT2F and dHMN II likely result from similar pathological mechanisms of the same general disease process that may present distinctly due to other genetic and environment influences. Determining how these influences exert their effects to produce pathology contributing to the disease phenotype will be a major future challenge ahead in the field.
Collapse
|
92
|
Ali AA, Ahmed HI, Khaleel SA, Abu-Elfotuh K. Vinpocetine mitigates aluminum-induced cognitive impairment in socially isolated rats. Physiol Behav 2019; 208:112571. [PMID: 31175888 DOI: 10.1016/j.physbeh.2019.112571] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 05/11/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022]
Abstract
Several reports have highlighted the role of vinpocetine in Alzheimer's disease (AD). However, the role of vinpocetine in AD under social isolation conditions has not yet been elucidated. Henceforth, this study aimed to investigate the potential neuroprotective effect of vinpocetine in aluminum-induced AD model associated with social isolation. Social isolation increased the escape latency in Morris water maze (MWM) test, elevated the immobility score and decreased swimming score in forced swimming test (FST) in aluminum treated rats. However, vinpocetine enhanced acquisition in MWM test and exerted anti-depressive effect in FST. The histopathological examination showed marked deterioration in the cerebral cortex and hippocampus of AD isolated rats, while vinpocetine revealed overt improvement. In addition, the levels of amyloid-β protein (Aβ), phosphorylated-tau (Ser396), malondialdehyde (MDA), interleukin 1-beta (IL-1β), tumor necrosis alpha (TNFα), p- Glycogen synthase kinase-3β (p-GSK3β) (Tyr216), and β-secretase (BACE1) gene expression were increased in socially isolated aluminum treated rats, yet, vinpocetine treatment reversed these deteriorating effects. Hence, this study provides profound insights into the role of vinpocetine in AD particularly in the conditions of social isolation. The effects of vinpocetine might be attributed not only to its antioxidant and anti-inflammatory properties, but also to its suppressing effect on GSK3β activity and its downstream BACE1.
Collapse
Affiliation(s)
- Azza A Ali
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Hebatalla I Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt; Pharmacology and Toxicology Department, Faculty of Pharmacy, Heliopolis University for sustainable development, Cairo, Egypt
| | - Sahar A Khaleel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt; Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Karema Abu-Elfotuh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
93
|
Afreen S, Ferreira A. Altered Cytoskeletal Composition and Delayed Neurite Elongation in tau 45-230-Expressing Hippocampal Neurons. Neuroscience 2019; 412:1-15. [PMID: 31158440 DOI: 10.1016/j.neuroscience.2019.05.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/15/2022]
Abstract
Calpain-mediated tau cleavage into the neurotoxic tau45-230 fragment plays an important role in Alzheimer's disease (AD). This tau fragment accumulates mainly in the cytoplasm of degenerating neurons. However, subcellular localization studies indicated that a pool of tau45-230 associates with the cytoskeleton in hippocampal neurons. In the present study, we assessed whether such localization could underlie tau45-230 neurotoxic effects. Quantitative Western blot analysis showed decreased levels of full-length tau bound to microtubules in tau45-230-expressing hippocampal neurons when compared to controls. In addition, the presence of this tau fragment induced a transient increase in tyrosinated tubulin, a marker of unstable microtubules, followed by a significant decrease in the levels of this tubulin isoform. The data obtained also showed a significant reduction in actin filaments in tau45-230-expressing neurons. These changes in microtubules and actin filaments correlated with delayed neurite elongation and axonal differentiation in the presence of this tau fragment. Together, these results suggest that tau45-230 could exert its toxic effects, at least in part, by modifying the composition of the neuronal cytoskeleton and impairing neurite elongation in neurons undergoing degeneration.
Collapse
Affiliation(s)
- Sana Afreen
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Adriana Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
94
|
Xu A, Zhang Z, Ko SH, Fisher AL, Liu Z, Chen L. Microtubule regulators act in the nervous system to modulate fat metabolism and longevity through DAF-16 in C. elegans. Aging Cell 2019; 18:e12884. [PMID: 30638295 PMCID: PMC6413656 DOI: 10.1111/acel.12884] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 10/26/2018] [Accepted: 11/03/2018] [Indexed: 12/16/2022] Open
Abstract
Microtubule (MT) regulation is involved in both neuronal function and the maintenance of neuronal structure, and MT dysregulation appears to be a general downstream indicator and effector of age‐related neurodegeneration. But the role of MTs in natural aging is largely unknown. Here, we demonstrate a role of MT regulators in regulating longevity. We find that loss of EFA‐6, a modulator of MT dynamics, can delay both neuronal aging and extend the lifespan of C. elegans. Through the use of genetic mutants affecting other MT‐regulating genes in C. elegans, we find that loss of MT stabilizing genes (including ptrn‐1 and ptl‐1) shortens lifespan, while loss of MT destabilizing gene hdac‐6 extends lifespan. Via the use of tissue‐specific transgenes, we further show that these MT regulators can act in the nervous system to modulate lifespan. Through RNA‐seq analyses, we found that genes involved in lipid metabolism were differentially expressed in MT regulator mutants, and via the use of Nile Red and Oil Red O staining, we show that the MT regulator mutants have altered fat storage. We further find that the increased fat storage and extended lifespan of the long‐lived MT regulator mutants are dependent on the DAF‐16/FOXO transcription factor. Our results suggest that neuronal MT status might affect organismal aging through DAF‐16‐regulated changes in fat metabolism, and therefore, MT‐based therapies might represent a novel intervention to promote healthy aging.
Collapse
Affiliation(s)
- Aiping Xu
- Barshop Institute for Longevity and Aging Studies; San Antonio Texas
- Department of Cell Systems and Anatomy; UTHSCSA; San Antonio Texas
| | - Zhao Zhang
- Department of Molecular Medicine; UTHSCSA; San Antonio Texas
| | - Su-Hyuk Ko
- Barshop Institute for Longevity and Aging Studies; San Antonio Texas
- Department of Cell Systems and Anatomy; UTHSCSA; San Antonio Texas
- Department of Molecular Medicine; UTHSCSA; San Antonio Texas
| | - Alfred L. Fisher
- Center for Healthy Aging; UTHSCSA; San Antonio Texas
- Division of Geriatrics, Gerontology, and Palliative Medicine, Department of Medicine; UTHSCSA; San Antonio Texas
- GRECC, South Texas VA Healthcare System; San Antonio Texas
| | - Zhijie Liu
- Department of Molecular Medicine; UTHSCSA; San Antonio Texas
| | - Lizhen Chen
- Barshop Institute for Longevity and Aging Studies; San Antonio Texas
- Department of Cell Systems and Anatomy; UTHSCSA; San Antonio Texas
- Department of Molecular Medicine; UTHSCSA; San Antonio Texas
| |
Collapse
|
95
|
Martinez Carrera LA, Gabriel E, Donohoe CD, Hölker I, Mariappan A, Storbeck M, Uhlirova M, Gopalakrishnan J, Wirth B. Novel insights into SMALED2: BICD2 mutations increase microtubule stability and cause defects in axonal and NMJ development. Hum Mol Genet 2019. [PMID: 29528393 DOI: 10.1093/hmg/ddy086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Bicaudal D2 (BICD2) encodes a highly conserved motor adaptor protein that regulates the dynein-dynactin complex in different cellular processes. Heterozygous mutations in BICD2 cause autosomal dominant lower extremity-predominant spinal muscular atrophy-2 (SMALED2). Although, various BICD2 mutations have been shown to alter interactions with different binding partners or the integrity of the Golgi apparatus, the specific pathological effects of BICD2 mutations underlying SMALED2 remain elusive. Here, we show that the fibroblasts derived from individuals with SMALED2 exhibit stable microtubules. Importantly, this effect was observed regardless of where the BICD2 mutation is located, which unifies the most likely cellular mechanism affecting microtubules. Significantly, overexpression of SMALED2-causing BICD2 mutations in the disease-relevant cell type, motor neurons, also results in an increased microtubule stability which is accompanied by axonal aberrations such as collateral branching and overgrowth. To study the pathological consequences of BICD2 mutations in vivo, and to address the controversial debate whether two of these mutations are neuron or muscle specific, we generated the first Drosophila model of SMALED2. Strikingly, neuron-specific expression of BICD2 mutants resulted in reduced neuromuscular junction size in larvae and impaired locomotion of adult flies. In contrast, expressing BICD2 mutations in muscles had no obvious effect on motor function, supporting a primarily neurological etiology of the disease. Thus, our findings contribute to the better understanding of SMALED2 pathology by providing evidence for a common pathomechanism of BICD2 mutations that increase microtubule stability in motor neurons leading to increased axonal branching and to impaired neuromuscular junction development.
Collapse
Affiliation(s)
- Lilian A Martinez Carrera
- Institute of Human Genetics, University of Cologne, 50931 Cologne, Germany.,Institute for Genetics, University of Cologne, 50674 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Elke Gabriel
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Colin D Donohoe
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Irmgard Hölker
- Institute of Human Genetics, University of Cologne, 50931 Cologne, Germany.,Institute for Genetics, University of Cologne, 50674 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Aruljothi Mariappan
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Markus Storbeck
- Institute of Human Genetics, University of Cologne, 50931 Cologne, Germany.,Institute for Genetics, University of Cologne, 50674 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Mirka Uhlirova
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Jay Gopalakrishnan
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University of Cologne, 50931 Cologne, Germany.,Institute for Genetics, University of Cologne, 50674 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany.,Center for Rare Diseases Cologne, University Hospital of Cologne, 50931 Cologne, Germany
| |
Collapse
|
96
|
Abstract
Neurons are polarized cells with long branched axons and dendrites. Microtubule generation and organization machineries are crucial to grow and pattern these complex cellular extensions. Microtubule organizing centers (MTOCs) concentrate the molecular machinery for templating microtubules, stabilizing the nascent polymer, and organizing the resultant microtubules into higher-order structures. MTOC formation and function are well described at the centrosome, in the spindle, and at interphase Golgi; we review these studies and then describe recent results about how the machineries acting at these classic MTOCs are repurposed in the postmitotic neuron for axon and dendrite differentiation. We further discuss a constant tug-of-war interplay between different MTOC activities in the cell and how this process can be used as a substrate for transcription factor-mediated diversification of neuron types.
Collapse
Affiliation(s)
- Jason Y Tann
- Laboratory for Neurodiversity, RIKEN Centre for Brain Science, Saitama, Japan
| | - Adrian W Moore
- Laboratory for Neurodiversity, RIKEN Centre for Brain Science, Saitama, Japan.
| |
Collapse
|
97
|
Chernivec E, Cooper J, Naylor K. Exploring the Effect of Rotenone-A Known Inducer of Parkinson's Disease-On Mitochondrial Dynamics in Dictyostelium discoideum. Cells 2018; 7:E201. [PMID: 30413037 PMCID: PMC6262481 DOI: 10.3390/cells7110201] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 02/07/2023] Open
Abstract
Current treatments for Parkinson's disease (PD) only alleviate symptoms doing little to inhibit the onset and progression of the disease, thus we must research the mechanism of Parkinson's. Rotenone is a known inducer of parkinsonian conditions in rats; we use rotenone to induce parkinsonian cellular conditions in Dictyostelium discoideum. In our model we primarily focus on mitochondrial dynamics. We found that rotenone disrupts the actin and microtubule cytoskeleton but mitochondrial morphology remains intact. Rotenone stimulates mitochondrial velocity while inhibiting mitochondrial fusion, increases reactive oxygen species (ROS) but has no effect on ATP levels. Antioxidants have been shown to decrease some PD symptoms thus we added ascorbic acid to our rotenone treated cells. Ascorbic acid administration suggests that rotenone effects may be specific to the disruption of the cytoskeleton rather than the increase in ROS. Our results imply that D. discoideum may be a valid cellular PD model and that the rotenone induced velocity increase and loss of fusion could prevent mitochondria from effectively providing energy and other mitochondrial products in high demand areas. The combination of these defects in mitochondrial dynamics and increased ROS could result in degeneration of neurons in PD.
Collapse
Affiliation(s)
- Ethan Chernivec
- Department of Biology, University of Central Arkansas, Conway, AR 72035, USA.
| | - Jacie Cooper
- Department of Biology, University of Central Arkansas, Conway, AR 72035, USA.
| | - Kari Naylor
- Department of Biology, University of Central Arkansas, Conway, AR 72035, USA.
| |
Collapse
|
98
|
Grønborg S, Risom L, Ek J, Larsen KB, Scheie D, Petkov Y, Larsen VA, Dunø M, Joensen F, Østergaard E. A Faroese founder variant in TBCD causes early onset, progressive encephalopathy with a homogenous clinical course. Eur J Hum Genet 2018; 26:1512-1520. [PMID: 29921875 PMCID: PMC6138752 DOI: 10.1038/s41431-018-0204-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/22/2018] [Accepted: 06/05/2018] [Indexed: 11/08/2022] Open
Abstract
An intact and dynamic microtubule cytoskeleton is crucial for the development, differentiation, and maintenance of the mammalian cortex. Variants in a host of structural microtubulin-associated proteins have been identified to cause a wide spectrum of malformations of cortical development and alterations of microtubule dynamics have been recognized to cause or contribute to progressive neurodegenerative disorders. TBCD is one of the five tubulin-specific chaperones and is required for reversible assembly of the α-/β-tubulin heterodimer. Recently, variants in TBCD, and one other tubulin-specific chaperone, TBCE, have been identified in patients with distinct progressive encephalopathy with a seemingly broad clinical spectrum. Here, we report the clinical, neuroradiological, and neuropathological features in eight patients originating from the Faroe Islands, who presented with an early onset, progressive encephalopathy with features of primary neurodegeneration, and a homogenous clinical course. These patients were homozygous for a TBCD missense variant c.[3099C>G]; p.(Asn1033Lys), which we show has a high carrier frequency in the Faroese population (2.6%). The patients had similar age of onset as the previously reported patients (n = 24), but much shorter survival, which could be caused by either differences in supportive treatment, or alternatively, that shorter survival is intrinsic to the Faroese phenotype. We present a detailed description of the neuropathology and MR imaging characteristics of a subset of these patients, adding insight into the phenotype of TBCD-related encephalopathy. The finding of a Faroese founder variant will allow targeted genetic diagnostics in patients of Faroese descent as well as improved genetic counseling and testing of at-risk couples.
Collapse
Affiliation(s)
- Sabine Grønborg
- Department of Pediatrics, Center for Rare Diseases, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
- Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Lotte Risom
- Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Jakob Ek
- Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Karen Bonde Larsen
- Department of Pathology, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Neuropathology and Ocular Pathology, John Radcliffe Hospital, Oxford University Hospital, Headley Way, Headington, Oxford, OX3 9DU, UK
| | - David Scheie
- Department of Pathology, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Yanko Petkov
- Department of Pediatrics, Esbjerg Hospital, Finsensgade 35, 6700, Esbjerg, Denmark
| | - Vibeke André Larsen
- Department of Radiology, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Morten Dunø
- Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Fróði Joensen
- Department of Pediatrics, National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
| | - Elsebet Østergaard
- Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
99
|
Kim H, Calatayud C, Guha S, Fernández-Carasa I, Berkowitz L, Carballo-Carbajal I, Ezquerra M, Fernández-Santiago R, Kapahi P, Raya Á, Miranda-Vizuete A, Lizcano JM, Vila M, Caldwell KA, Caldwell GA, Consiglio A, Dalfo E. The Small GTPase RAC1/CED-10 Is Essential in Maintaining Dopaminergic Neuron Function and Survival Against α-Synuclein-Induced Toxicity. Mol Neurobiol 2018; 55:7533-7552. [PMID: 29429047 PMCID: PMC6096980 DOI: 10.1007/s12035-018-0881-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/07/2018] [Indexed: 12/22/2022]
Abstract
Parkinson's disease is associated with intracellular α-synuclein accumulation and ventral midbrain dopaminergic neuronal death in the Substantia Nigra of brain patients. The Rho GTPase pathway, mainly linking surface receptors to the organization of the actin and microtubule cytoskeletons, has been suggested to participate to Parkinson's disease pathogenesis. Nevertheless, its exact contribution remains obscure. To unveil the participation of the Rho GTPase family to the molecular pathogenesis of Parkinson's disease, we first used C elegans to demonstrate the role of the small GTPase RAC1 (ced-10 in the worm) in maintaining dopaminergic function and survival in the presence of alpha-synuclein. In addition, ced-10 mutant worms determined an increase of alpha-synuclein inclusions in comparison to control worms as well as an increase in autophagic vesicles. We then used a human neuroblastoma cells (M17) stably over-expressing alpha-synuclein and found that RAC1 function decreased the amount of amyloidogenic alpha-synuclein. Further, by using dopaminergic neurons derived from patients of familial LRRK2-Parkinson's disease we report that human RAC1 activity is essential in the regulation of dopaminergic cell death, alpha-synuclein accumulation, participates in neurite arborization and modulates autophagy. Thus, we determined for the first time that RAC1/ced-10 participates in Parkinson's disease associated pathogenesis and established RAC1/ced-10 as a new candidate for further investigation of Parkinson's disease associated mechanisms, mainly focused on dopaminergic function and survival against α-synuclein-induced toxicity.
Collapse
Affiliation(s)
- Hanna Kim
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Carles Calatayud
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08028, L'Hospitalet de Llobregat, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08908, Spain
- Center of Regenerative Medicine in Barcelona (CMRB), Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Hospital Duran i Reynals, 08908, L'Hospitalet de Llobregat, Spain
| | - Sanjib Guha
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Irene Fernández-Carasa
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08028, L'Hospitalet de Llobregat, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08908, Spain
| | - Laura Berkowitz
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Iria Carballo-Carbajal
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
| | - Mario Ezquerra
- Laboratory of Parkinson Disease and Other Neurodegenerative Movement Disorders, Department of Neurology: Clinical and Experimental Research, IDIBAPS - Hospital Clínic de Barcelona, 08036, Barcelona, Spain
| | - Rubén Fernández-Santiago
- Laboratory of Parkinson Disease and Other Neurodegenerative Movement Disorders, Department of Neurology: Clinical and Experimental Research, IDIBAPS - Hospital Clínic de Barcelona, 08036, Barcelona, Spain
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Ángel Raya
- Center of Regenerative Medicine in Barcelona (CMRB), Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Hospital Duran i Reynals, 08908, L'Hospitalet de Llobregat, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain
| | - Antonio Miranda-Vizuete
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, 41013, Sevilla, Spain
| | - Jose Miguel Lizcano
- Department of Biochemistry and Molecular Biology, Institut de Neurociències, Faculty of Medicine, M2, Universitat Autònoma de Barcelona (UAB), Bellaterra Campus, Cerdanyola del Vallés, Barcelona, Spain
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Institut de Neurociències, Faculty of Medicine, M2, Universitat Autònoma de Barcelona (UAB), Bellaterra Campus, Cerdanyola del Vallés, Barcelona, Spain
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Antonella Consiglio
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08028, L'Hospitalet de Llobregat, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08908, Spain.
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Spain.
| | - Esther Dalfo
- Department of Biochemistry and Molecular Biology, Institut de Neurociències, Faculty of Medicine, M2, Universitat Autònoma de Barcelona (UAB), Bellaterra Campus, Cerdanyola del Vallés, Barcelona, Spain.
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Can Baumann, 08500, Vic, Spain.
| |
Collapse
|
100
|
Capture of Dense Core Vesicles at Synapses by JNK-Dependent Phosphorylation of Synaptotagmin-4. Cell Rep 2018; 21:2118-2133. [PMID: 29166604 PMCID: PMC5714612 DOI: 10.1016/j.celrep.2017.10.084] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 10/05/2017] [Accepted: 10/23/2017] [Indexed: 01/04/2023] Open
Abstract
Delivery of neurotrophins and neuropeptides via long-range trafficking of dense core vesicles (DCVs) from the cell soma to nerve terminals is essential for synapse modulation and circuit function. But the mechanism by which transiting DCVs are captured at specific sites is unknown. Here, we discovered that Synaptotagmin-4 (Syt4) regulates the capture and spatial distribution of DCVs in hippocampal neurons. We found that DCVs are highly mobile and undergo long-range translocation but switch directions only at the distal ends of axons, revealing a circular trafficking pattern. Phosphorylation of serine 135 of Syt4 by JNK steers DCV trafficking by destabilizing Syt4-Kif1A interaction, leading to a transition from microtubule-dependent DCV trafficking to capture at en passant presynaptic boutons by actin. Furthermore, neuronal activity increased DCV capture via JNK-dependent phosphorylation of the S135 site of Syt4. Our data reveal a mechanism that ensures rapid, site-specific delivery of DCVs to synapses. Syt4-bearing dense core vesicles in axons traffic continually in a circular pattern Phosphorylation of S135 of Syt4 by JNK destabilizes Syt4-Kif1A binding Destabilized Syt4-Kif1A binding promotes capture of vesicles at synapses by actin Neuronal activity increases vesicle capture via S135-dependent JNK phosphorylation
Collapse
|