1
|
Priya K, Siddesha JM, Dharini S, Shashanka KP. Interacting Models of Amyloid-β and Tau Proteins: An Approach to Identify Drug Targets in Alzheimer's Disease. J Alzheimers Dis Rep 2021; 5:405-411. [PMID: 34189412 PMCID: PMC8203288 DOI: 10.3233/adr-210018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Alzheimer's disease (AD) is the primary cause of dementia affecting millions each year across the world, though still remains incurable. This might be attributed to the lack of knowledge about the associated proteins, their cellular and molecular mechanisms, and the genesis of the disease. The discovery of drugs that earlier revolved around targeting the amyloid-β cascade has now been reformed with the upgraded knowledge of the cross-seeding ability of tau protein which opens new gateways for therapeutic targets. This article provides a comprehensive review of various direct and indirect connecting pathways between the two main proteins involved in development and progression of AD, enabling us to further expand our repertoire of information regarding the etiology of AD. The current review indicates the need for extensive research in this niche, thus considerable advances can be made in understanding AD which eventually helps to improve the current therapeutics against AD.
Collapse
Affiliation(s)
- Khadgawat Priya
- Department of Genetics, University of Delhi, New Delhi, India
| | - J M Siddesha
- Division of Biochemistry, Faculty of Life Sciences, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, Karnataka, India
| | - Shashank Dharini
- Department of Burns, Plastic and Maxillofacial Surgery, VMMC and Safdarjung Hospital, New Delhi, India
| | - K Prasad Shashanka
- Department of Biotechnology and Bioinformatics, Faculty of Life Sciences, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, Karnataka, India
| |
Collapse
|
2
|
Morphine and HIV-1 Tat interact to cause region-specific hyperphosphorylation of tau in transgenic mice. Neurosci Lett 2020; 741:135502. [PMID: 33202259 DOI: 10.1016/j.neulet.2020.135502] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 12/24/2022]
Abstract
Opiate abuse is prevalent among HIV-infected individuals and may exacerbate HIV-associated age-related neurocognitive disorders. However, the extent to which HIV and opiates converge to accelerate pathological traits indicative of brain aging remains unknown. The pathological phospho-isotypes of tau (pSer396, pSer404, pThr205, pSer202, and pThr181) and the tau kinases GSK3β and CDK5/p35 were explored in the striatum, hippocampus, and prefrontal cortex of inducible male and female HIV-1 Tat-transgenic mice, with some receiving escalating doses of morphine for 2 weeks. In the striatum of male mice, pSer396 was increased by co-exposure to morphine and Tat as compared to all other groups. Striatal pSer404 and pThr205 were increased by Tat alone, while pSer202 and pThr181 were unchanged. A comparison between Tat-transgenic female and male mice revealed disparate outcomes for pThr205. No other sex-related changes to tau phosphorylation were observed. In the hippocampus, Tat increased pSer396, while other phosphorylation sites were unchanged and pSer202 was not detected. In the prefrontal cortex, morphine increased pSer396 levels, which were unaffected by Tat, while other phosphorylation sites were unaffected. Assessment of tau kinases revealed no changes to striatal GSK3β (phosphorylated or total) or the total CDK5 levels. Striatal levels of phosphorylated CDK5 and p35, the activator of CDK5, were increased by Tat and with morphine co-exposure, respectively. P35 levels positively correlated with those of pSer396 with Tat and morphine co-exposure. The results reveal region-specific hyperphosphorylation of tau induced by exposure to morphine, Tat, and unique morphine and Tat interactions.
Collapse
|
3
|
Peyressatre M, Laure A, Pellerano M, Boukhaddaoui H, Soussi I, Morris MC. Fluorescent Biosensor of CDK5 Kinase Activity in Glioblastoma Cell Extracts and Living Cells. Biotechnol J 2020; 15:e1900474. [DOI: 10.1002/biot.201900474] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 04/13/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Marion Peyressatre
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| | - Arthur Laure
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| | - Morgan Pellerano
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| | - Hassan Boukhaddaoui
- Plateau Imagerie Cellulaire MRI‐INM INM‐INSERM U 1051, Hôpital Saint Eloi 80 rue Augustin Fliche Montpellier 34091 France
| | - Ines Soussi
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| | - May C. Morris
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| |
Collapse
|
4
|
Bencze J, Szarka M, Bencs V, Szabó RN, Módis LV, Aarsland D, Hortobágyi T. Lemur Tyrosine Kinase 2 (LMTK2) Level Inversely Correlates with Phospho-Tau in Neuropathological Stages of Alzheimer's Disease. Brain Sci 2020; 10:E68. [PMID: 32012723 PMCID: PMC7071479 DOI: 10.3390/brainsci10020068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/17/2020] [Accepted: 01/25/2020] [Indexed: 12/11/2022] Open
Abstract
: Alzheimer's disease (AD) is the most common neurodegenerative dementia. Mapping the pathomechanism and providing novel therapeutic options have paramount significance. Recent studies have proposed the role of LMTK2 in AD. However, its expression pattern and association with the pathognomonic neurofibrillary tangles (NFTs) in different brain regions and neuropathological stages of AD is not clear. We performed chromogenic (CHR) LMTK2 and fluorescent phospho-tau/LMTK2 double-labelling (FDL) immunohistochemistry (IHC) on 10-10 postmortem middle frontal gyrus (MFG) and anterior hippocampus (aHPC) samples with early and late neuropathological Braak tau stages of AD. MFG in early stage was our 'endogenous control' region as it is not affected by NFTs. Semiquantitative CHR-IHC intensity scoring revealed significantly higher (p < 0.001) LMTK2 values in this group compared to NFT-affected regions. FDL-IHC demonstrated LMTK2 predominance in the endogenous control region, while phospho-tau overburden and decreased LMTK2 immunolabelling were detected in NFT-affected groups (aHPC in early and both regions in late stage). Spearman's correlation coefficient showed strong negative correlation between phospho-tau/LMTK2 signals within each group. According to our results, LMTK2 expression is inversely proportionate to the extent of NFT pathology, and decreased LMTK2 level is not a general feature in AD brain, rather it is characteristic of the NFT-affected regions.
Collapse
Affiliation(s)
- János Bencze
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, 4032 Debrecen, Hungary
| | - Máté Szarka
- Horvath Csaba Memorial Institute of Bioanalytical Research, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Vitrolink Ltd., 4033 Debrecen, Hungary
- Institute for Nuclear Research of the Hungarian Academy of Sciences (ATOMKI), 4026 Debrecen, Hungary
| | - Viktor Bencs
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Renáta Nóra Szabó
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, 4032 Debrecen, Hungary
- Institute of Pathology, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary
| | - László V. Módis
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, 4032 Debrecen, Hungary
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Centre for Age-Related Medicine, SESAM, Stavanger University Hospital, 4011 Stavanger, Norway
| | - Tibor Hortobágyi
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, 4032 Debrecen, Hungary
- Institute of Pathology, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary
- Department of Old Age Psychiatry, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Centre for Age-Related Medicine, SESAM, Stavanger University Hospital, 4011 Stavanger, Norway
| |
Collapse
|
5
|
Lu TT, Wan C, Yang W, Cai Z. Role of Cdk5 in Amyloid-beta Pathology of Alzheimer’s Disease. Curr Alzheimer Res 2020; 16:1206-1215. [PMID: 31820699 DOI: 10.2174/1567205016666191210094435] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/29/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer’s Disease (AD) is a progressive neurodegenerative disease with irreversible cognitive
impairment. So far, successful treatment and prevention for this disease are deficient in spite of delaying
the progression of cognitive impairment and dementia. Cyclin dependent kinase 5 (Cdk5), a
unique member of the cyclin-dependent kinase family, is involved in AD pathogenesis and may be a
pathophysiological mediator that links the major pathological features of AD. Cdk5 dysregulation interferes
with the proteolytic processing of Amyloid-beta Protein Precursor (APP) and modulates amyloidbeta
(Aβ) by affecting three enzymes called α-, β- and γ-secretase, which are critical for the hydrolysis
of APP. Given that the accumulation and deposition of Aβ derived from APP are a common hinge point
in the numerous pathogenic hypotheses of AD, figuring out that influence of specific mechanisms of
Cdk5 on Aβ pathology will deepen our understanding of AD.
Collapse
Affiliation(s)
- Tao-Tao Lu
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, 400013, Chongqing, China
| | - Chengqun Wan
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, 400013, Chongqing, China
| | - Wenming Yang
- Departmentof Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031 Anhui Province, China
| | - Zhiyou Cai
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, 400013, Chongqing, China
| |
Collapse
|
6
|
Brenna A, Olejniczak I, Chavan R, Ripperger JA, Langmesser S, Cameroni E, Hu Z, De Virgilio C, Dengjel J, Albrecht U. Cyclin-dependent kinase 5 (CDK5) regulates the circadian clock. eLife 2019; 8:50925. [PMID: 31687929 PMCID: PMC6890458 DOI: 10.7554/elife.50925] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/03/2019] [Indexed: 12/12/2022] Open
Abstract
Circadian oscillations emerge from transcriptional and post-translational feedback loops. An important step in generating rhythmicity is the translocation of clock components into the nucleus, which is regulated in many cases by kinases. In mammals, the kinase promoting the nuclear import of the key clock component Period 2 (PER2) is unknown. Here, we show that the cyclin-dependent kinase 5 (CDK5) regulates the mammalian circadian clock involving phosphorylation of PER2. Knock-down of Cdk5 in the suprachiasmatic nuclei (SCN), the main coordinator site of the mammalian circadian system, shortened the free-running period in mice. CDK5 phosphorylated PER2 at serine residue 394 (S394) in a diurnal fashion. This phosphorylation facilitated interaction with Cryptochrome 1 (CRY1) and nuclear entry of the PER2-CRY1 complex. Taken together, we found that CDK5 drives nuclear entry of PER2, which is critical for establishing an adequate circadian period of the molecular circadian cycle. Of note is that CDK5 may not exclusively phosphorylate PER2, but in addition may regulate other proteins that are involved in the clock mechanism. Taken together, it appears that CDK5 is critically involved in the regulation of the circadian clock and may represent a link to various diseases affected by a derailed circadian clock. Anyone who has crossed multiple time zones on a long flight will be familiar with jet lag, and that feeling of wanting to sleep at lunchtime and eat in the middle of the night. Many bodily processes, including appetite and wakefulness, roughly follow a 24-hour cycle. These cycles are known as circadian rhythms, from the Latin ‘circa diem’ meaning about a day. An area of the brain called the suprachiasmatic nucleus (SCN) coordinates circadian rhythms. It acts as a master clock by generating a 24-hour signal for the rest of the body to follow. Jet lag occurs when this internal circadian rhythm becomes out of sync with the local day-night cycle. Although jet lag can be uncomfortable, it tends to disappear over the course of a few days. This is because exposure to daylight in our new location resets the SCN master clock, enabling us to adapt to a new time zone. But evidence suggests that long-term disruption of circadian rhythms, for example as a result of shift work, may have lasting harmful effects. These include an increased risk of degenerative brain disorders such as Parkinson's disease and Alzheimer's disease. Brenna et al. now identify a molecular mechanism that could explain this link. A key component of the SCN master clock is a protein called Period2 (PER2). Levels of PER2 rise and fall over each 24-hour period, helping the brain keep track of time. Brenna et al. show that PER2 interacts with CDK5, a protein that helps regulate brain development and that has been implicated in Parkinson's disease and Alzheimer's disease. Reducing CDK5 levels in mice shortened their circadian rhythms by several hours. It also altered the animals’ behavioral patterns over a 24-hour period. Deleting the gene for PER2 had a similar effect, suggesting that CDK5 helps regulate PER2. Future studies should investigate the molecular links between CDK5, circadian rhythms and processes such as neurodegeneration. The results would provide clues to whether manipulating the circadian clock could help prevent or treat neurological disorders.
Collapse
Affiliation(s)
- Andrea Brenna
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Iwona Olejniczak
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Rohit Chavan
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Sonja Langmesser
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Zehan Hu
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Urs Albrecht
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
7
|
Le ST, Guros NB, Bruce RC, Cardone A, Amin ND, Zhang S, Klauda JB, Pant HC, Richter CA, Balijepalli A. Quantum capacitance-limited MoS 2 biosensors enable remote label-free enzyme measurements. NANOSCALE 2019; 11:15622-15632. [PMID: 31407757 PMCID: PMC6792296 DOI: 10.1039/c9nr03171e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
We have demonstrated atomically thin, quantum capacitance-limited, field-effect transistors (FETs) that enable the detection of pH changes with 75-fold higher sensitivity (≈4.4 V per pH) over the Nernst value of 59 mV per pH at room temperature when used as a biosensor. The transistors, which are fabricated from monolayer films of MoS2, use a room temperature ionic liquid (RTIL) in place of a conventional oxide gate dielectric and exhibit very low intrinsic noise resulting in a pH resolution of 92 × 10-6 at 10 Hz. This high device performance, which is a function of the structure of our device, is achieved by remotely connecting the gate to a pH sensing element allowing the FETs to be reused. Because pH measurements are fundamentally important in biotechnology, the increased resolution demonstrated here will benefit numerous applications ranging from pharmaceutical manufacturing to clinical diagnostics. As an example, we experimentally quantified the function of the kinase Cdk5, an enzyme implicated in Alzheimer's disease, at concentrations that are 5-fold lower than physiological values, and with sufficient time-resolution to allow the estimation of both steady-state and kinetic parameters in a single experiment. The high sensitivity, increased resolution, and fast turnaround time of the measurements will allow the development of early diagnostic tools and novel therapeutics to detect and treat neurological conditions years before currently possible.
Collapse
Affiliation(s)
- Son T Le
- Nanoscale Device Characterization Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA and Theiss Research, La Jolla, CA 92037, USA
| | - Nicholas B Guros
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA. and Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA
| | - Robert C Bruce
- Nanoscale Device Characterization Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Antonio Cardone
- Software and Systems Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA and University of Maryland Institute for Advanced Computer Studies, University of Maryland, College Park, MD 20742, USA
| | - Niranjana D Amin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Siyuan Zhang
- Nanoscale Device Characterization Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA and Theiss Research, La Jolla, CA 92037, USA
| | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA
| | - Harish C Pant
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Curt A Richter
- Nanoscale Device Characterization Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Arvind Balijepalli
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| |
Collapse
|
8
|
Selenko P. Quo Vadis Biomolecular NMR Spectroscopy? Int J Mol Sci 2019; 20:ijms20061278. [PMID: 30875725 PMCID: PMC6472163 DOI: 10.3390/ijms20061278] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
In-cell nuclear magnetic resonance (NMR) spectroscopy offers the possibility to study proteins and other biomolecules at atomic resolution directly in cells. As such, it provides compelling means to complement existing tools in cellular structural biology. Given the dominance of electron microscopy (EM)-based methods in current structure determination routines, I share my personal view about the role of biomolecular NMR spectroscopy in the aftermath of the revolution in resolution. Specifically, I focus on spin-off applications that in-cell NMR has helped to develop and how they may provide broader and more generally applicable routes for future NMR investigations. I discuss the use of ‘static’ and time-resolved solution NMR spectroscopy to detect post-translational protein modifications (PTMs) and to investigate structural consequences that occur in their response. I argue that available examples vindicate the need for collective and systematic efforts to determine post-translationally modified protein structures in the future. Furthermore, I explain my reasoning behind a Quinary Structure Assessment (QSA) initiative to interrogate cellular effects on protein dynamics and transient interactions present in physiological environments.
Collapse
Affiliation(s)
- Philipp Selenko
- Weizmann Institute of Science, Department of Biological Regulation, 234 Herzl Street, Rehovot 76100, Israel.
| |
Collapse
|
9
|
Abstract
Tauopathies are neurodegenerative diseases that are characterized by the presence of hyperphosphorylated tau-containing neurofibrillary tangles (NFTs) in the brain and include Alzheimer's disease and frontotemporal dementia, which lack effective disease-modifying treatments. The presence of NFTs is known to correlate with cognition impairment, suggesting that targeting tau hyperphosphorylation may be therapeutically effective. MLC901 is a herbal formulation that is currently used in poststroke recovery and consists of nine herbal components. Previously, several components of MLC901 have been shown to have an effect on tau phosphorylation, but it remains unknown whether MLC901 itself has the same effect. The objective of this study was to assess the effects of MLC901 on ameliorating tau phosphorylation at epitopes associated with NFT formation. A stably transfected cell culture model expressing tau harboring the P301S mutation was generated and treated with various concentrations of MLC901 across different time points. Tau phosphorylation profiles and protein levels of enzymes associated with tau phosphorylation were assessed using western blotting. One-way analysis of variance with Bonferroni post-hoc analysis showed that MLC901 significantly reduced tau phosphorylation at epitopes recognized by the AT8, AT270, and PHF-13 antibodies. MLC901 also induced a significant increase in the s9 phosphorylation of glycogen synthase kinase 3β and a concurrent decrease in the activation of cyclin-dependent kinase 5, as measured by a significant decrease in the levels of p35/cyclin-dependent kinase 5. Our results provide supporting evidence to further study the effects of MLC901 on tau pathology and cognition using mouse models of tauopathy.
Collapse
|
10
|
Bencze J, Mórotz GM, Seo W, Bencs V, Kálmán J, Miller CCJ, Hortobágyi T. Biological function of Lemur tyrosine kinase 2 (LMTK2): implications in neurodegeneration. Mol Brain 2018; 11:20. [PMID: 29631601 PMCID: PMC5891947 DOI: 10.1186/s13041-018-0363-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/26/2018] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders are frequent, incurable diseases characterised by abnormal protein accumulation and progressive neuronal loss. Despite their growing prevalence, the underlying pathomechanism remains unclear. Lemur tyrosine kinase 2 (LMTK2) is a member of a transmembrane serine/threonine-protein kinase family. Although it was described more than a decade ago, our knowledge on LMTK2’s biological functions is still insufficient. Recent evidence has suggested that LMTK2 is implicated in neurodegeneration. After reviewing the literature, we identified three LMTK2-mediated mechanisms which may contribute to neurodegenerative processes: disrupted axonal transport, tau hyperphosphorylation and enhanced apoptosis. Moreover, LMTK2 gene expression is decreased in an Alzheimer’s disease mouse model. According to these features, LMTK2 might be a promising therapeutic target in near future. However, further investigations are required to clarify the exact biological functions of this unique protein.
Collapse
Affiliation(s)
- János Bencze
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., Debrecen, H-4032, Hungary
| | - Gábor Miklós Mórotz
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
| | - Woosung Seo
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., Debrecen, H-4032, Hungary
| | - Viktor Bencs
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., Debrecen, H-4032, Hungary
| | - János Kálmán
- Department of Psychiatry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Christopher Charles John Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
| | - Tibor Hortobágyi
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., Debrecen, H-4032, Hungary. .,MTA-DE Cerebrovascular and Neurodegenerative Research Group, Debrecen, Hungary. .,Department of Pathology, Faculty of Medicine, University of Szeged, Szeged, Hungary. .,Department of Old Age Psychiatry, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
11
|
Wilkaniec A, Gąssowska-Dobrowolska M, Strawski M, Adamczyk A, Czapski GA. Inhibition of cyclin-dependent kinase 5 affects early neuroinflammatory signalling in murine model of amyloid beta toxicity. J Neuroinflammation 2018; 15:1. [PMID: 29301548 PMCID: PMC5753486 DOI: 10.1186/s12974-017-1027-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/07/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cyclin-dependent kinase 5 (Cdk5) belongs to the family of proline-directed serine/threonine kinases and plays a critical role in neuronal differentiation, migration, synaptogenesis, plasticity, neurotransmission and apoptosis. The deregulation of Cdk5 activity was observed in post mortem analysis of brain tissue of Alzheimer's disease (AD) patients, suggesting the involvement of Cdk5 in the pathomechanism of this neurodegenerative disease. However, our recent study demonstrated the important function of Cdk5 in regulating inflammatory reaction. METHODS Since the role of Cdk5 in regulation of inflammatory signalling in AD is unknown, we investigated the involvement of Cdk5 in neuroinflammation induced by single intracerebroventricular (icv) injection of amyloid beta protein (Aβ) oligomers in mouse. The brain tissue was analysed up to 35 days post injection. Roscovitine (intraperitoneal administration) was used as a potent Cdk5 inhibitor. The experiments were also performed on human neuroblastoma SH-SY5Y as well as mouse BV2 cell lines treated with exogenous oligomeric Aβ. RESULTS Our results demonstrated that single injection of Aβ oligomers induces long-lasting activation of microglia and astrocytes in the hippocampus. We observed also profound, early inflammatory response in the mice hippocampus, leading to the significant elevation of pro-inflammatory cytokines expression (e.g. TNF-α, IL-1β, IL-6). Moreover, Aβ oligomers elevated the formation of truncated protein p25 in mouse hippocampus and induced overactivation of Cdk5 in neuronal cells. Importantly, administration of roscovitine reduced the inflammatory processes evoked by Aβ in the hippocampus, leading to the significant decrease of cytokines level. CONCLUSIONS These studies clearly show the involvement of Cdk5 in modulation of brain inflammatory response induced by Aβ and may indicate this kinase as a novel target for pharmacological intervention in AD.
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Marcin Strawski
- Laboratory of Electrochemistry, Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland.
| |
Collapse
|
12
|
Shukla V, Seo J, Binukumar B, Amin ND, Reddy P, Grant P, Kuntz S, Kesavapany S, Steiner J, Mishra SK, Tsai LH, Pant HC. TFP5, a Peptide Inhibitor of Aberrant and Hyperactive Cdk5/p25, Attenuates Pathological Phenotypes and Restores Synaptic Function in CK-p25Tg Mice. J Alzheimers Dis 2017; 56:335-349. [PMID: 28085018 PMCID: PMC10020940 DOI: 10.3233/jad-160916] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It has been reported that cyclin-dependent kinase 5 (cdk5), a critical neuronal kinase, is hyperactivated in Alzheimer's disease (AD) and may be, in part, responsible for the hallmark pathology of amyloid plaques and neurofibrillary tangles (NFTs). It has been proposed by several laboratories that hyperactive cdk5 results from the overexpression of p25 (a truncated fragment of p35, the normal cdk5 regulator), which, when complexed to cdk5, induces hyperactivity, hyperphosphorylated tau/NFTs, amyloid-β plaques, and neuronal death. It has previously been shown that intraperitoneal (i.p.) injections of a modified truncated 24-aa peptide (TFP5), derived from the cdk5 activator p35, penetrated the blood-brain barrier and significantly rescued AD-like pathology in 5XFAD model mice. The principal pathology in the 5XFAD mutant, however, is extensive amyloid plaques; hence, as a proof of concept, we believe it is essential to demonstrate the peptide's efficacy in a mouse model expressing high levels of p25, such as the inducible CK-p25Tg model mouse that overexpresses p25 in CamKII positive neurons. Using a modified TFP5 treatment, here we show that peptide i.p. injections in these mice decrease cdk5 hyperactivity, tau, neurofilament-M/H hyperphosphorylation, and restore synaptic function and behavior (i.e., spatial working memory, motor deficit using Rota-rod). It is noteworthy that TFP5 does not inhibit endogenous cdk5/p35 activity, nor other cdks in vivo suggesting it might have no toxic side effects, and may serve as an excellent therapeutic candidate for neurodegenerative disorders expressing abnormally high brain levels of p25 and hyperactive cdk5.
Collapse
Affiliation(s)
- Varsha Shukla
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jinsoo Seo
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - B.K. Binukumar
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Niranjana D. Amin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Preethi Reddy
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Philip Grant
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Susan Kuntz
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Joseph Steiner
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Santosh K. Mishra
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MA, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Harish C. Pant
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Correspondence to: Dr. Harish C. Pant, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
13
|
Zhang ZH, Chen C, Wu QY, Zheng R, Chen Y, Liu Q, Ni JZ, Song GL. Selenomethionine Ameliorates Neuropathology in the Olfactory Bulb of a Triple Transgenic Mouse Model of Alzheimer's Disease. Int J Mol Sci 2016; 17:ijms17101595. [PMID: 27689994 PMCID: PMC5085628 DOI: 10.3390/ijms17101595] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/23/2016] [Accepted: 09/13/2016] [Indexed: 01/08/2023] Open
Abstract
Olfactory dysfunction is an early and common symptom in Alzheimer's disease (AD) and is reported to be related to several pathologic changes, including the deposition of Aβ and hyperphosphorylated tau protein as well as synaptic impairment. Selenomethionine (Se-Met), the major form of selenium in animals and humans, may be a promising therapeutic option for AD as it decreases the deposition of Aβ and tau hyperphosphorylation in a triple transgenic mouse model of AD (3× Tg-AD). In this study, 4-month-old AD mice were treated with 6 µg/mL Se-Met in drinking water for 12 weeks and the effect of Se-Met on neuropathological deficits in olfactory bulb (OB) of 3× Tg-AD mice was investigated. The administration of Se-Met effectively decreased the production and deposition of Aβ by inhibiting β-site amyloid precursor protein cleaving enzyme 1 (BACE1)-regulated amyloid precursor protein (APP) processing and reduced the level of total tau and phosphorylated tau, which depended on depressing the activity and expression of glycogen synthase kinase-3β (GSK-3β) and cyclin-dependent kinase 5 (CDK5). Meanwhile, Se-Met reduced glial activation, relieved neuroinflammation and attenuated neuronal cell death in the OB of AD mice. So Se-Met could improve pathologic changes of AD in the OB, which further demonstrated the potential therapeutic effect of Se-Met in AD.
Collapse
Affiliation(s)
- Zhong-Hao Zhang
- Changchun Institute of Applied Chemistry, University of Chinese Academy of Sciences, Changchun 130022, China.
| | - Chen Chen
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Qiu-Yan Wu
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Rui Zheng
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Yao Chen
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Jia-Zuan Ni
- Changchun Institute of Applied Chemistry, University of Chinese Academy of Sciences, Changchun 130022, China.
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Guo-Li Song
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
14
|
NMR Meets Tau: Insights into Its Function and Pathology. Biomolecules 2016; 6:biom6020028. [PMID: 27338491 PMCID: PMC4919923 DOI: 10.3390/biom6020028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/02/2016] [Accepted: 05/26/2016] [Indexed: 12/21/2022] Open
Abstract
In this review, we focus on what we have learned from Nuclear Magnetic Resonance (NMR) studies on the neuronal microtubule-associated protein Tau. We consider both the mechanistic details of Tau: the tubulin relationship and its aggregation process. Phosphorylation of Tau is intimately linked to both aspects. NMR spectroscopy has depicted accurate phosphorylation patterns by different kinases, and its non-destructive character has allowed functional assays with the same samples. Finally, we will discuss other post-translational modifications of Tau and its interaction with other cellular factors in relationship to its (dys)function.
Collapse
|
15
|
Curcio M, Salazar IL, Mele M, Canzoniero LMT, Duarte CB. Calpains and neuronal damage in the ischemic brain: The swiss knife in synaptic injury. Prog Neurobiol 2016; 143:1-35. [PMID: 27283248 DOI: 10.1016/j.pneurobio.2016.06.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022]
Abstract
The excessive extracellular accumulation of glutamate in the ischemic brain leads to an overactivation of glutamate receptors with consequent excitotoxic neuronal death. Neuronal demise is largely due to a sustained activation of NMDA receptors for glutamate, with a consequent increase in the intracellular Ca(2+) concentration and activation of calcium- dependent mechanisms. Calpains are a group of Ca(2+)-dependent proteases that truncate specific proteins, and some of the cleavage products remain in the cell, although with a distinct function. Numerous studies have shown pre- and post-synaptic effects of calpains on glutamatergic and GABAergic synapses, targeting membrane- associated proteins as well as intracellular proteins. The resulting changes in the presynaptic proteome alter neurotransmitter release, while the cleavage of postsynaptic proteins affects directly or indirectly the activity of neurotransmitter receptors and downstream mechanisms. These alterations also disturb the balance between excitatory and inhibitory neurotransmission in the brain, with an impact in neuronal demise. In this review we discuss the evidence pointing to a role for calpains in the dysregulation of excitatory and inhibitory synapses in brain ischemia, at the pre- and post-synaptic levels, as well as the functional consequences. Although targeting calpain-dependent mechanisms may constitute a good therapeutic approach for stroke, specific strategies should be developed to avoid non-specific effects given the important regulatory role played by these proteases under normal physiological conditions.
Collapse
Affiliation(s)
- Michele Curcio
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ivan L Salazar
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-789 Coimbra, Portugal
| | - Miranda Mele
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal.
| |
Collapse
|
16
|
Czapski GA, Gąssowska M, Wilkaniec A, Chalimoniuk M, Strosznajder JB, Adamczyk A. The mechanisms regulating cyclin-dependent kinase 5 in hippocampus during systemic inflammatory response: The effect on inflammatory gene expression. Neurochem Int 2016; 93:103-12. [PMID: 26806339 DOI: 10.1016/j.neuint.2016.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 11/19/2022]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is critical for nervous system's development and function, and its aberrant activation contributes to pathomechanism of Alzheimer's disease and other neurodegenerative disorders. It was recently suggested that Cdk5 may participate in regulation of inflammatory signalling. The aim of this study was to analyse the mechanisms involved in regulating Cdk5 activity in the brain during systemic inflammatory response (SIR) as well as the involvement of Cdk5 in controlling the expression of inflammatory genes. Genetic and biochemical alterations in hippocampus were analysed 3 and 12 h after intraperitoneal injection of lipopolysaccharide. We observed an increase in both Cdk5 gene expression and protein level. Moreover, phosphorylation of Cdk5 on Ser159 was significantly enhanced. Also transcription of Cdk5-regulatory protein (p35/Cdk5r1) was augmented, and the level of p25, calpain-dependent cleavage product of p35, was increased. All these results demonstrated rapid activation of Cdk5 in the brain during SIR. Hyperactivity of Cdk5 contributed to enhanced phosphorylation of tau and glycogen synthase kinase 3β. Inhibition of Cdk5 with Roscovitine reduced activation of NF-κB and expression of inflammation-related genes, demonstrating the critical role of Cdk5 in regulation of gene transcription during SIR.
Collapse
Affiliation(s)
- Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, ul. Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Magdalena Gąssowska
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, ul. Pawińskiego 5, 02-106 Warsaw, Poland
| | - Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, ul. Pawińskiego 5, 02-106 Warsaw, Poland
| | - Małgorzata Chalimoniuk
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, ul. Pawińskiego 5, 02-106 Warsaw, Poland
| | - Joanna B Strosznajder
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, ul. Pawińskiego 5, 02-106 Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, ul. Pawińskiego 5, 02-106 Warsaw, Poland
| |
Collapse
|
17
|
Ahuja P, Cantrelle FX, Huvent I, Hanoulle X, Lopez J, Smet C, Wieruszeski JM, Landrieu I, Lippens G. Proline Conformation in a Functional Tau Fragment. J Mol Biol 2015; 428:79-91. [PMID: 26655856 DOI: 10.1016/j.jmb.2015.11.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/05/2015] [Accepted: 11/24/2015] [Indexed: 12/18/2022]
Abstract
The conformational state of distinct prolines can determine the folding of a protein but equally other biological processes when coupled to a conformation-sensitive secondary reaction. For the neuronal tau protein, the importance of proline conformation is underscored by its interaction with different prolyl cis/trans isomerases. The proline conformation would gain even further importance after phosphorylation of the preceding residue by various proline-directed kinases. A number of molecular diseases including Alzheimer's disease and traumatic brain injury were thereby recently qualified as "cistauosis", as they would imply a cis conformation for the pThr231-Pro232 prolyl bond. We here investigate by NMR spectroscopy the conformation of all prolines in a functional Tau fragment, Tau[208-324]. Although we can detect and identify some minor conformers in the cis form, we show that all prolines are for over 90% in the trans conformation. Phosphorylation by CDK2/CycA3, which notably leads to complete modification of the Thr231 residue, does not change this conclusion. Our data hence disagree with the notion that specific prolyl bonds in tau would adopt preferentially the cis conformation.
Collapse
Affiliation(s)
- Puneet Ahuja
- UMR8576 CNRS Lille University, 59658 Villeneuve d'Ascq, France
| | | | - Isabelle Huvent
- UMR8576 CNRS Lille University, 59658 Villeneuve d'Ascq, France
| | - Xavier Hanoulle
- UMR8576 CNRS Lille University, 59658 Villeneuve d'Ascq, France
| | - Juan Lopez
- UMR8576 CNRS Lille University, 59658 Villeneuve d'Ascq, France
| | - Caroline Smet
- UMR8576 CNRS Lille University, 59658 Villeneuve d'Ascq, France
| | | | | | - G Lippens
- UMR8576 CNRS Lille University, 59658 Villeneuve d'Ascq, France.
| |
Collapse
|
18
|
Wilkaniec A, Czapski GA, Adamczyk A. Cdk5 at crossroads of protein oligomerization in neurodegenerative diseases: facts and hypotheses. J Neurochem 2015; 136:222-33. [PMID: 26376455 DOI: 10.1111/jnc.13365] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 09/02/2015] [Accepted: 09/03/2015] [Indexed: 02/06/2023]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is involved in proper neurodevelopment and brain function and serves as a switch between neuronal survival and death. Overactivation of Cdk5 is associated with many neurodegenerative disorders such as Alzheimer's or Parkinson's diseases. It is believed that in those diseases Cdk5 may be an important link between disease-initiating factors and cell death effectors. A common hallmark of neurodegenerative disorders is incorrect folding of specific proteins, thus leading to their intra- and extracellular accumulation in the nervous system. Abnormal Cdk5 signaling contributes to dysfunction of individual proteins and has a substantial role in either direct or indirect interactions of proteins common to, and critical in, different neurodegenerative diseases. While the roles of Cdk5 in α-synuclein (ASN) - tau or β-amyloid peptide (Aβ) - tau interactions are well documented, its contribution to many other pertinent interactions, such as that of ASN with Aβ, or interactions of the Aβ - ASN - tau triad with prion proteins, did not get beyond plausible hypotheses and remains to be proven. Understanding of the exact position of Cdk5 in the deleterious feed-forward loop critical for development and progression of neurodegenerative diseases may help designing successful therapeutic strategies of several fatal neurodegenerative diseases. Cyclin-dependent kinase 5 (Cdk5) is associated with many neurodegenerative disorders such as Alzheimer's or Parkinson's diseases. It is believed that in those diseases Cdk5 may be an important factor involved in protein misfolding, toxicity and interaction. We suggest that Cdk5 may contribute to the vicious circle of neurotoxic events involved in the pathogenesis of different neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
19
|
Zhang S, Lu Z, Mao W, Ahmed AA, Yang H, Zhou J, Jennings N, Rodriguez-Aguayo C, Lopez-Berestein G, Miranda R, Qiao W, Baladandayuthapani V, Li Z, Sood AK, Liu J, Le XF, Bast RC. CDK5 Regulates Paclitaxel Sensitivity in Ovarian Cancer Cells by Modulating AKT Activation, p21Cip1- and p27Kip1-Mediated G1 Cell Cycle Arrest and Apoptosis. PLoS One 2015; 10:e0131833. [PMID: 26146988 PMCID: PMC4492679 DOI: 10.1371/journal.pone.0131833] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/06/2015] [Indexed: 01/12/2023] Open
Abstract
Cyclin-dependent kinase 5 (CDK5) is a cytoplasmic serine/ threonine kinase. Knockdown of CDK5 enhances paclitaxel sensitivity in human ovarian cancer cells. This study explores the mechanisms by which CDK5 regulates paclitaxel sensitivity in human ovarian cancers. Multiple ovarian cancer cell lines and xenografts were treated with CDK5 small interfering RNA (siRNA) with or without paclitaxel to examine the effect on cancer cell viability, cell cycle arrest and tumor growth. CDK5 protein was measured by immunohistochemical staining of an ovarian cancer tissue microarray to correlate CDK5 expression with overall patient survival. Knockdown of CDK5 with siRNAs inhibits activation of AKT which significantly correlates with decreased cell growth and enhanced paclitaxel sensitivity in ovarian cancer cell lines. In addition, CDK5 knockdown alone and in combination with paclitaxel induced G1 cell cycle arrest and caspase 3 dependent apoptotic cell death associated with post-translational upregulation and nuclear translocation of TP53 and p27Kip1 as well as TP53-dependent transcriptional induction of p21Cip1 in wild type TP53 cancer cells. Treatment of HEYA8 and A2780 wild type TP53 xenografts in nu/nu mice with CDK5 siRNA and paclitaxel produced significantly greater growth inhibition than either treatment alone. Increased expression of CDK5 in human ovarian cancers correlates inversely with overall survival. CDK5 modulates paclitaxel sensitivity by regulating AKT activation, the cell cycle and caspase-dependent apoptosis. CDK5 inhibition can potentiate paclitaxel activity in human ovarian cancer cells.
Collapse
Affiliation(s)
- Shu Zhang
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of General Surgery, the Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Zhen Lu
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Weiqun Mao
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ahmed A. Ahmed
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hailing Yang
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jinhua Zhou
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Nicholas Jennings
- Departments of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Cristian Rodriguez-Aguayo
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Gabriel Lopez-Berestein
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Roberto Miranda
- Departments of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, Untied States of America
| | - Wei Qiao
- Bioinformatics Computer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Veera Baladandayuthapani
- Bioinformatics Computer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Zongfang Li
- Department of General Surgery, the Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Anil K. Sood
- Departments of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jinsong Liu
- Departments of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, Untied States of America
| | - Xiao-Feng Le
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (RCB); (XFL)
| | - Robert C. Bast
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (RCB); (XFL)
| |
Collapse
|
20
|
Abstract
Cyclin dependent kinase-5 (Cdk5), a family member of the cyclin-dependent kinases, plays a pivotal role in the central nervous system. During embryogenesis, Cdk5 is indispensable for brain development and, in the adult brain, it is essential for numerous neuronal processes, including higher cognitive functions such as learning and memory formation. However, Cdk5 activity becomes deregulated in several neurological disorders, such as Alzheimer's disease, Parkinson's disease and Huntington's disease, which leads to neurotoxicity. Therefore, precise control over Cdk5 activity is essential for its physiological functions. This Commentary covers the various mechanisms of Cdk5 regulation, including several recently identified protein activators and inhibitors of Cdk5 that control its activity in normal and diseased brains. We also discuss the autoregulatory activity of Cdk5 and its regulation at the transcriptional, post-transcriptional and post-translational levels. We finally highlight physiological and pathological roles of Cdk5 in the brain. Specific modulation of these protein regulators is expected to provide alternative strategies for the development of effective therapeutic interventions that are triggered by deregulation of Cdk5.
Collapse
Affiliation(s)
- Kavita Shah
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Debomoy K Lahiri
- Laboratory of Molecular Neurogenetics, Departments of Psychiatry and of Medical & Molecular Genetics, Indiana University School of Medicine, Institute of Psychiatric Research, Neuroscience Research Building, 320 W. 15th St., Indianapolis, IN 46202, USA
| |
Collapse
|
21
|
Corbel C, Zhang B, Le Parc A, Baratte B, Colas P, Couturier C, Kosik KS, Landrieu I, Le Tilly V, Bach S. Tamoxifen inhibits CDK5 kinase activity by interacting with p35/p25 and modulates the pattern of tau phosphorylation. ACTA ACUST UNITED AC 2015; 22:472-482. [PMID: 25865311 DOI: 10.1016/j.chembiol.2015.03.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/27/2015] [Accepted: 03/06/2015] [Indexed: 12/18/2022]
Abstract
Cyclin-dependent kinase 5 (CDK5) is a multifunctional enzyme that plays numerous roles, notably in brain development. CDK5 is activated through its association with the activators, p35 and p39, rather than by cyclins. Proteolytic procession of the N-terminal part of its activators has been linked to Alzheimer's disease and various other neuropathies. The interaction with the proteolytic product p25 prolongs CDK5 activation and modifies the substrate specificity. In order to discover small-molecule inhibitors of the interaction between CDK5 and p25, we have used a bioluminescence resonance energy transfer (BRET)-based screening assay. Among the 1,760 compounds screened, the generic drug tamoxifen has been identified. The inhibition of the CDK5 activity by tamoxifen was notably validated by monitoring the phosphorylation state of tau protein. The study of the molecular mechanism of inhibition indicates that tamoxifen interacts with p25 to block the CDK5/p25 interaction and pave the way for new treatments of tauopathies.
Collapse
Affiliation(s)
- Caroline Corbel
- USR3151-CNRS/UPMC, Protein Phosphorylation and Disease Laboratory, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, Bretagne, France; EA4250-LIMATB-EG2B, Centre de Recherche et d'Enseignement Yves Coppens, Université de Bretagne Sud, 56017 Vannes, France; Kosik Laboratory, Neuroscience Research Institute, Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Bing Zhang
- School of Renewable Energy, North China Electric Power Electricity, 071003 Beijing, China
| | - Annabelle Le Parc
- EA4250-LIMATB-EG2B, Centre de Recherche et d'Enseignement Yves Coppens, Université de Bretagne Sud, 56017 Vannes, France
| | - Blandine Baratte
- USR3151-CNRS/UPMC, Protein Phosphorylation and Disease Laboratory, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, Bretagne, France
| | - Pierre Colas
- USR3151-CNRS/UPMC, Protein Phosphorylation and Disease Laboratory, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, Bretagne, France
| | - Cyril Couturier
- UMR761-INSERM Lille University, Biostructures and Drug Discovery, 59006 Lille, France
| | - Kenneth S Kosik
- Kosik Laboratory, Neuroscience Research Institute, Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Isabelle Landrieu
- UMR8576 CNRS-Lille North of France University, 59658 Villeneuve d'Ascq, France; Interdisciplinary Research Institute (IRI), 58658 Villeneuve d'Ascq, France
| | - Véronique Le Tilly
- EA4250-LIMATB-EG2B, Centre de Recherche et d'Enseignement Yves Coppens, Université de Bretagne Sud, 56017 Vannes, France
| | - Stéphane Bach
- USR3151-CNRS/UPMC, Protein Phosphorylation and Disease Laboratory, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, Bretagne, France.
| |
Collapse
|
22
|
Peyressatre M, Prével C, Pellerano M, Morris MC. Targeting cyclin-dependent kinases in human cancers: from small molecules to Peptide inhibitors. Cancers (Basel) 2015; 7:179-237. [PMID: 25625291 PMCID: PMC4381256 DOI: 10.3390/cancers7010179] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 01/12/2015] [Indexed: 12/12/2022] Open
Abstract
Cyclin-dependent kinases (CDK/Cyclins) form a family of heterodimeric kinases that play central roles in regulation of cell cycle progression, transcription and other major biological processes including neuronal differentiation and metabolism. Constitutive or deregulated hyperactivity of these kinases due to amplification, overexpression or mutation of cyclins or CDK, contributes to proliferation of cancer cells, and aberrant activity of these kinases has been reported in a wide variety of human cancers. These kinases therefore constitute biomarkers of proliferation and attractive pharmacological targets for development of anticancer therapeutics. The structural features of several of these kinases have been elucidated and their molecular mechanisms of regulation characterized in depth, providing clues for development of drugs and inhibitors to disrupt their function. However, like most other kinases, they constitute a challenging class of therapeutic targets due to their highly conserved structural features and ATP-binding pocket. Notwithstanding, several classes of inhibitors have been discovered from natural sources, and small molecule derivatives have been synthesized through rational, structure-guided approaches or identified in high throughput screens. The larger part of these inhibitors target ATP pockets, but a growing number of peptides targeting protein/protein interfaces are being proposed, and a small number of compounds targeting allosteric sites have been reported.
Collapse
Affiliation(s)
- Marion Peyressatre
- Institut des Biomolécules Max Mousseron, IBMM-CNRS-UMR5247, 15 Av. Charles Flahault, 34093 Montpellier, France.
| | - Camille Prével
- Institut des Biomolécules Max Mousseron, IBMM-CNRS-UMR5247, 15 Av. Charles Flahault, 34093 Montpellier, France.
| | - Morgan Pellerano
- Institut des Biomolécules Max Mousseron, IBMM-CNRS-UMR5247, 15 Av. Charles Flahault, 34093 Montpellier, France.
| | - May C Morris
- Institut des Biomolécules Max Mousseron, IBMM-CNRS-UMR5247, 15 Av. Charles Flahault, 34093 Montpellier, France.
| |
Collapse
|
23
|
Sui D, Xu X, Ye X, Liu M, Mianecki M, Rattanasinchai C, Buehl C, Deng X, Kuo MH. Protein interaction module-assisted function X (PIMAX) approach to producing challenging proteins including hyperphosphorylated tau and active CDK5/p25 kinase complex. Mol Cell Proteomics 2014; 14:251-62. [PMID: 25385071 DOI: 10.1074/mcp.o114.044412] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many biomedically critical proteins are underrepresented in proteomics and biochemical studies because of the difficulty of their production in Escherichia coli. These proteins might possess posttranslational modifications vital to their functions, tend to misfold and be partitioned into bacterial inclusion bodies, or act only in a stoichiometric dimeric complex. Successful production of these proteins requires efficient interaction between these proteins and a specific "facilitator," such as a protein-modifying enzyme, a molecular chaperone, or a natural physical partner within the dimeric complex. Here we report the design and application of a protein interaction module-assisted function X (PIMAX) system that effectively overcomes these hurdles. By fusing two proteins of interest to a pair of well-studied protein-protein interaction modules, we were able to potentiate the association of these two proteins, resulting in successful production of an enzymatically active cyclin-dependent kinase complex and hyperphosphorylated tau protein, which is intimately linked to Alzheimer disease. Furthermore, using tau isoforms quantitatively phosphorylated by GSK-3β and CDK5 kinases via PIMAX, we demonstrated the hyperphosphorylation-stimulated tau oligomerization in vitro, paving the way for new Alzheimer disease drug discoveries. Vectors for PIMAX can be easily modified to meet the needs of different applications. This approach thus provides a convenient and modular suite with broad implications for proteomics and biomedical research.
Collapse
Affiliation(s)
- Dexin Sui
- From the ‡Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Xinjing Xu
- From the ‡Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Xuemei Ye
- From the ‡Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Mengyu Liu
- From the ‡Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Maxwell Mianecki
- From the ‡Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Chotirat Rattanasinchai
- ¶Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan 48824
| | - Christopher Buehl
- ¶Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan 48824
| | - Xiexiong Deng
- From the ‡Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Min-Hao Kuo
- From the ‡Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824;
| |
Collapse
|
24
|
Castro-Alvarez JF, Uribe-Arias SA, Mejía-Raigosa D, Cardona-Gómez GP. Cyclin-dependent kinase 5, a node protein in diminished tauopathy: a systems biology approach. Front Aging Neurosci 2014; 6:232. [PMID: 25225483 PMCID: PMC4150361 DOI: 10.3389/fnagi.2014.00232] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/11/2014] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. One of the main pathological changes that occurs in AD is the intracellular accumulation of hyperphosphorylated Tau protein in neurons. Cyclin-dependent kinase 5 (CDK5) is one of the major kinases involved in Tau phosphorylation, directly phosphorylating various residues and simultaneously regulating various substrates such as kinases and phosphatases that influence Tau phosphorylation in a synergistic and antagonistic way. It remains unknown how the interaction between CDK5 and its substrates promotes Tau phosphorylation, and systemic approaches are needed that allow an analysis of all the proteins involved. In this review, the role of the CDK5 signaling pathway in Tau hyperphosphorylation is described, an in silico model of the CDK5 signaling pathway is presented. The relationship among these theoretical and computational models shows that the regulation of Tau phosphorylation by PP2A and glycogen synthase kinase 3β (GSK3β) is essential under basal conditions and also describes the leading role of CDK5 under excitotoxic conditions, where silencing of CDK5 can generate changes in these enzymes to reverse a pathological condition that simulates AD.
Collapse
Affiliation(s)
- John F Castro-Alvarez
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, University of Antioquia, Sede de Investigación Universitaria Medellin, Colombia
| | - S Alejandro Uribe-Arias
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, University of Antioquia, Sede de Investigación Universitaria Medellin, Colombia
| | - Daniel Mejía-Raigosa
- Group of Biophysics, Faculty of Exact and Natural Sciences, Institute of Physics, University of Antioquia Medellin, Colombia
| | - Gloria P Cardona-Gómez
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, University of Antioquia, Sede de Investigación Universitaria Medellin, Colombia
| |
Collapse
|
25
|
Kimura T, Ishiguro K, Hisanaga SI. Physiological and pathological phosphorylation of tau by Cdk5. Front Mol Neurosci 2014; 7:65. [PMID: 25076872 PMCID: PMC4097945 DOI: 10.3389/fnmol.2014.00065] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 06/26/2014] [Indexed: 11/13/2022] Open
Abstract
Hyperphosphorylation of microtubule-associated protein tau is one of the major pathological events in Alzheimer’s disease (AD) and other related neurodegenerative diseases, including frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17). Mutations in the tau gene MAPT are a cause of FTDP-17, and the mutated tau proteins are hyperphosphorylated in patient brains. Thus, it is important to determine the molecular mechanism of hyperphosphorylation of tau to understand the pathology of these diseases collectively called tauopathy. Tau is phosphorylated at many sites via several protein kinases, and a characteristic is phosphorylation at Ser/Thr residues in Ser/Thr-Pro sequences, which are targeted by proline-directed protein kinases such as ERK, GSK3β, and Cdk5. Among these kinases, Cdk5 is particularly interesting because it could be abnormally activated in AD. Cdk5 is a member of the cyclin-dependent kinases (Cdks), but in contrast to the major Cdks, which promote cell cycle progression in proliferating cells, Cdk5 is activated in post-mitotic neurons via the neuron-specific activator p35. Cdk5-p35 plays a critical role in brain development and physiological synaptic activity. In contrast, in disease brains, Cdk5 is thought to be hyperactivated by p25, which is the N-terminal truncated form of p35 and is generated by cleavage with calpain. Several reports have indicated that tau is hyperphosphorylated by Cdk5-p25. However, normal and abnormal phosphorylation of tau by Cdk5 is still not completely understood. In this article, we summarize the physiological and pathological phosphorylation of tau via Cdk5.
Collapse
Affiliation(s)
- Taeko Kimura
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Tokyo Metropolitan University Hachioji, Japan
| | - Koichi Ishiguro
- Department of Neurology, Graduate School of Medicine, Juntendo University Bunkyo, Japan
| | - Shin-Ichi Hisanaga
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Tokyo Metropolitan University Hachioji, Japan
| |
Collapse
|
26
|
Vázquez-Rosa E, Rodríguez-Cruz EN, Serrano S, Rodríguez-Laureano L, Vega IE. Cdk5 phosphorylation of EFhd2 at S74 affects its calcium binding activity. Protein Sci 2014; 23:1197-207. [PMID: 24917152 DOI: 10.1002/pro.2499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 12/31/2022]
Abstract
EFhd2 is a calcium binding protein, which is highly expressed in the central nervous system and associated with pathological forms of tau proteins in tauopathies. Previous phosphoproteomics studies and bioinformatics analysis suggest that EFhd2 may be phosphorylated. Here, we determine whether Cdk5, a hyperactivated kinase in tauopathies, phosphorylates EFhd2 and influence its known molecular activities. The results indicated that EFhd2 is phosphorylated by brain extract of the transgenic mouse CK-p25, which overexpresses the Cdk5 constitutive activator p25. Consistently, in vitro kinase assays demonstrated that Cdk5, but not GSK3β, directly phosphorylates EFhd2. Biomass, tandem mass spectrometry, and mutagenesis analyses indicated that Cdk5 monophosphorylates EFhd2 at S74, but not the adjacent S76. Furthermore, Cdk5-mediated phosphorylation of EFhd2 affected its calcium binding activity. Finally, a phospho-specific antibody was generated against EFhd2 phosphorylated at S74 and was used to detect this phosphorylation event in postmortem brain tissue from Alzheimer's disease and normal-aging control cases. Results demonstrated that EFhd2 is phosphorylated in vivo at S74. These results imply that EFhd2's physiological and/or pathological function could be regulated by its phosphorylation state.
Collapse
Affiliation(s)
- Edwin Vázquez-Rosa
- Department of Chemistry, College of Natural Sciences, University of Puerto Rico-Río Piedras Campus, San Juan, Puerto Rico, 00931; Protein Mass Spectrometry Core Facility, College of Natural Sciences, University of Puerto Rico - Río Piedras Campus, San Juan, Puerto Rico, 00931
| | | | | | | | | |
Collapse
|
27
|
The Beta-amyloid protein of Alzheimer's disease: communication breakdown by modifying the neuronal cytoskeleton. Int J Alzheimers Dis 2013; 2013:910502. [PMID: 24416616 PMCID: PMC3876695 DOI: 10.1155/2013/910502] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/07/2013] [Indexed: 01/28/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most prevalent severe neurological disorders afflicting our aged population. Cognitive decline, a major symptom exhibited by AD patients, is associated with neuritic dystrophy, a degenerative growth state of neurites. The molecular mechanisms governing neuritic dystrophy remain unclear. Mounting evidence indicates that the AD-causative agent, β-amyloid protein (Aβ), induces neuritic dystrophy. Indeed, neuritic dystrophy is commonly found decorating Aβ-rich amyloid plaques (APs) in the AD brain. Furthermore, disruption and degeneration of the neuronal microtubule system in neurons forming dystrophic neurites may occur as a consequence of Aβ-mediated downstream signaling. This review defines potential molecular pathways, which may be modulated subsequent to Aβ-dependent interactions with the neuronal membrane as a consequence of increasing amyloid burden in the brain.
Collapse
|
28
|
Tang C, Welty DF. A dynamic view to the modulation of phosphorylation and O-GlcNAcylation by inhibition of O-GlcNAcase. Comput Biol Chem 2013; 45:9-16. [DOI: 10.1016/j.compbiolchem.2013.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 12/31/2022]
|
29
|
|
30
|
Yang Y, Lightstone FC, Wong SE. Approaches to efficiently estimate solvation and explicit water energetics in ligand binding: the use of WaterMap. Expert Opin Drug Discov 2013; 8:277-87. [DOI: 10.1517/17460441.2013.749853] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
31
|
Luan K, Rosales JL, Lee KY. Viewpoint: Crosstalks between neurofibrillary tangles and amyloid plaque formation. Ageing Res Rev 2013; 12:174-81. [PMID: 22728532 DOI: 10.1016/j.arr.2012.06.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 05/17/2012] [Accepted: 06/06/2012] [Indexed: 12/29/2022]
Abstract
Since its discovery, the hallmarks of Alzheimer's disease (AD) brain have been recognised as the formation of amyloid plaques and neurofibrillary tangles (NFTs). Mounting evidence has suggested the active interplay between the two pathways. Studies have shown that β-amyloid (Aβ) can be internalized and generated intracellularly, accelerating NFT formation. Conversely, tau elements in NFTs are observed to affect Aβ and amyloid plaque formation. Yet the precise mechanisms which link the pathologies of the two brain lesions remain elusive. In this review, we discuss recent evidence that support five putative mechanisms by which crosstalk occurs between amyloid plaque and NFT formation in AD pathogenesis. Understanding the crosstalks in the formation of AD pathologies could provide new clues for the development of novel therapeutic strategies to delay or halt the progression of AD.
Collapse
Affiliation(s)
- Kailie Luan
- Department of Cell Biology and Anatomy, Southern Alberta Cancer Research and Hotchkiss Brain Institutes, University of Calgary, Alberta, Canada
| | | | | |
Collapse
|
32
|
p10, the N-terminal domain of p35, protects against CDK5/p25-induced neurotoxicity. Proc Natl Acad Sci U S A 2012; 109:20041-6. [PMID: 23151508 DOI: 10.1073/pnas.1212914109] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cyclin-dependent kinase 5(CDK5) in complex with its activator, p35 (protein of 35 kDa), is essential for early neurodevelopment in mammals. However, endogenous cleavage of p35 to p25 is associated with neuron death and neurodegenerative disease. Here we show that a peptide (p10') encoding the N-terminal domain of p35 protects against CDK5/p25-induced toxicity in neurons. p10' also prevented the death of neurons treated with the neurotoxin, 1-methyl-4-phenylpyridinium (MPP(+)), which induces conversion of endogenous p35 to p25, and Parkinson disease (PD)-like symptoms in animals. MPP(+) induces CDK5/p25-dependent phosphorylation of peroxiredoxin 2 (Prx2), resulting in inhibition of its peroxireductase activity and accumulation of reactive oxygen species (ROS). We found that p10' expression inhibited both Prx2 phosphorylation and ROS accumulation in neurons. In addition, p10' inhibited the p25-induced appearance of antigen of the Ki67 antibody (Ki67) and phosphohistone H2AX (γH2AX), classic markers of cell cycle activity and DNA double-strand breakage, respectively, associated with neuron death. Our results suggest that p10 (protein of 10 kDa) is a unique prosurvival domain in p35, essential for normal CDK5/p35 function in neurons. Loss of the p10 domain results in CDK5/p25 toxicity and neurodegeneration in vivo.
Collapse
|
33
|
Modi PK, Komaravelli N, Singh N, Sharma P. Interplay between MEK-ERK signaling, cyclin D1, and cyclin-dependent kinase 5 regulates cell cycle reentry and apoptosis of neurons. Mol Biol Cell 2012; 23:3722-30. [PMID: 22833568 PMCID: PMC3442418 DOI: 10.1091/mbc.e12-02-0125] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In response to neurotoxic signals, postmitotic neurons make attempts to reenter the cell cycle, which results in their death. Although several cell cycle proteins have been implicated in cell cycle-related neuronal apoptosis (CRNA), the molecular mechanisms that underlie this important event are poorly understood. Here, we demonstrate that neurotoxic agents such as β-amyloid peptide cause aberrant activation of mitogen-activated kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) signaling, which promotes the entry of neurons into the cell cycle, resulting in their apoptosis. The MEK-ERK pathway regulates CRNA by elevating the levels of cyclin D1. The increase in cyclin D1 attenuates the activation of cyclin-dependent kinase 5 (cdk5) by its neuronal activator p35. The inhibition of p35-cdk5 activity results in enhanced MEK-ERK signaling, leading to CRNA. These studies highlight how neurotoxic signals reprogram and alter the neuronal signaling machinery to promote their entry into the cell cycle, which eventually leads to neuronal cell death.
Collapse
Affiliation(s)
- Prashant Kumar Modi
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi 110067, India
| | | | | | | |
Collapse
|
34
|
Cheung ZH, Ip NY. Cdk5: a multifaceted kinase in neurodegenerative diseases. Trends Cell Biol 2011; 22:169-75. [PMID: 22189166 DOI: 10.1016/j.tcb.2011.11.003] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 11/09/2011] [Accepted: 11/14/2011] [Indexed: 10/14/2022]
Abstract
Since the identification of cyclin-dependent kinase-5 (Cdk5) as a tau kinase and member of the Cdk family almost 20 years ago, deregulation of Cdk5 activity has been linked to an array of neurodegenerative diseases. As knowledge on the etiopathological mechanisms of these diseases evolved through the years, Cdk5 has also been implicated in additional cellular events that are affected under these pathological conditions. From the role of Cdk5 in the regulation of synaptic functions to its involvement in autophagy deregulation, significant insights have been obtained regarding the role of Cdk5 as a key regulator of neurodegeneration. Here, we summarize recent findings on the involvement of Cdk5 in the pathophysiological mechanisms underlying various neurodegenerative diseases.
Collapse
Affiliation(s)
- Zelda H Cheung
- Division of Life Science, Molecular Neuroscience Center and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | | |
Collapse
|
35
|
López-Tobón A, Castro-Álvarez JF, Piedrahita D, Boudreau RL, Gallego-Gómez JC, Cardona-Gómez GP. Silencing of CDK5 as potential therapy for Alzheimer's disease. Rev Neurosci 2011; 22:143-52. [PMID: 21476938 DOI: 10.1515/rns.2011.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neurodegeneration is one of the greatest public health challenges for the 21st century. Among neurodegenerative diseases, Alzheimer's disease (AD) is the most prevalent and best characterized. Nevertheless, despite the large investment in AD research, currently there is no effective therapeutic option. In the present review, we highlight a novel alternative, which takes advantage of the biotechnological outbreak deployed by the discovery of the RNA interference-based gene silencing mechanism, and its application as a tool for neurodegeneration treatment. Here, we highlight cyclin-dependent kinase 5 (CDK5) as a key candidate target for therapeutic gene silencing. Unlike other members of the cyclin-dependent kinase family, CDK5 does not seem to play a crucial role in cell cycle regulation. By contrast, CDK5 participates in multiple functions during nervous system development and has been established as a key mediator of Tau hyperphosphorylation and neurofibrillary pathology, thus serving as an optimal candidate for targeted therapy in the adult nervous system. We propose that the use of RNA interference for CDK5 silencing presents an attractive and specific therapeutic alternative for AD and perhaps against other tauopathies.
Collapse
Affiliation(s)
- Alejandro López-Tobón
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, Group of Neuroscience of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellin, Colombia
| | | | | | | | | | | |
Collapse
|
36
|
Engmann O, Hortobágyi T, Pidsley R, Troakes C, Bernstein HG, Kreutz MR, Mill J, Nikolic M, Giese KP. Schizophrenia is associated with dysregulation of a Cdk5 activator that regulates synaptic protein expression and cognition. ACTA ACUST UNITED AC 2011; 134:2408-21. [PMID: 21772061 DOI: 10.1093/brain/awr155] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cyclin-dependent kinase 5 is activated by small subunits, of which p35 is the most abundant. The functions of cyclin-dependent kinase 5 signalling in cognition and cognitive disorders remains unclear. Here, we show that in schizophrenia, a disorder associated with impaired cognition, p35 expression is reduced in relevant brain regions. Additionally, the expression of septin 7 and OPA1, proteins downstream of truncated p35, is decreased in schizophrenia. Mimicking a reduction of p35 in heterozygous knockout mice is associated with cognitive endophenotypes. Furthermore, a reduction of p35 in mice results in protein changes similar to schizophrenia post-mortem brain. Hence, heterozygous p35 knockout mice model both cognitive endophenotypes and molecular changes reminiscent of schizophrenia. These changes correlate with reduced acetylation of the histone deacetylase 1 target site H3K18 in mice. This site has previously been shown to be affected by truncated p35. By restoring H3K18 acetylation with the clinically used specific histone deacetylase 1 inhibitor MS-275 both cognitive and molecular endophenotypes of schizophrenia can be rescued in p35 heterozygous knockout mice. In summary, we suggest that reduced p35 expression in schizophrenia has an impact on synaptic protein expression and cognition and that these deficits can be rescued, at least in part, by the inhibition of histone deacetylase 1.
Collapse
Affiliation(s)
- Olivia Engmann
- Department for Neuroscience, King’s College, London SE5 9NU, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Engmann O, Hortobágyi T, Thompson AJ, Guadagno J, Troakes C, Soriano S, Al-Sarraj S, Kim Y, Giese KP. Cyclin-dependent kinase 5 activator p25 is generated during memory formation and is reduced at an early stage in Alzheimer's disease. Biol Psychiatry 2011; 70:159-68. [PMID: 21616478 DOI: 10.1016/j.biopsych.2011.04.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 03/23/2011] [Accepted: 04/06/2011] [Indexed: 01/08/2023]
Abstract
BACKGROUND The cyclin-dependent kinase 5 activator p35 can be cleaved into p25. Formation of p25 has been suggested to contribute to neurodegeneration in Alzheimer's disease (AD). However, overexpression of low levels of p25 in mice enhances memory formation. Therefore, it has been suggested that p25 formation might be an event early in AD to compensate for impairments in synaptic plasticity. Ongoing p25 formation has been hypothesized to contribute to neurodegeneration at the later stages of AD. METHODS Here, we tested the early compensation hypothesis by analyzing the levels of p25 and its precursor p35 in AD postmortem samples from different brain regions at different stages of tau pathology, using quantitative Western blots. Furthermore, we studied p35 and p25 during spatial memory formation. By employing quantitative mass spectrometry, we identified proteins downstream of p25, which were then studied in AD samples. RESULTS We found that p25 is generated during spatial memory formation. Furthermore, we demonstrate that overexpression of p25 in the physiological range increases the expression of two proteins implicated in spine formation, septin 7 and optic atrophy 1. We show that the expression of p35 and p25 is reduced as an early event in AD. Moreover, expression of the p25-regulated protein optic atrophy 1 was reduced in a time course similar to p25 expression. CONCLUSIONS Our findings suggest that p25 generation is a mechanism underlying hippocampal memory formation that is impaired in the early stages of AD. Our findings argue against the previously raised early compensation hypothesis and they propose that p25-mediated neurotoxicity does not occur in AD.
Collapse
Affiliation(s)
- Olivia Engmann
- Department of Neuroscience, Medical Research Council Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mungenast AE, Tsai LH. Addressing the complex etiology of Alzheimer’s disease: the role of p25/Cdk5. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.11.22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder characterized by the progressive loss of forebrain neurons and the deterioration of learning and memory. Therapies for AD have primarily focused upon either the inhibition of amyloid synthesis or its deposition in the brain, but clinical testing to date has not yet found an effective amelioration of cognitive symptoms. Synaptic loss closely correlates with the degree of dementia in AD patients. However, mouse AD models that target the amyloid-β pathway generally do not exhibit a profound loss of synapses, despite extensive synaptic dysfunction. The increased generation of p25, an activator of the cyclin-dependent kinase 5 (Cdk5) has been found in both human patients and mouse models of neurodegeneration. The current work reviews our knowledge, to date, on the role of p25/Cdk5 in Alzheimer’s disease, with a focus upon the interaction of amyloid-β and p25/Cdk5 in synaptic dysfunction and neuronal loss.
Collapse
Affiliation(s)
- Alison E Mungenast
- Picower Institute for Learning & Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Li-Huei Tsai
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
39
|
Landrieu I, Smet-Nocca C, Amniai L, Louis JV, Wieruszeski JM, Goris J, Janssens V, Lippens G. Molecular implication of PP2A and Pin1 in the Alzheimer's disease specific hyperphosphorylation of Tau. PLoS One 2011; 6:e21521. [PMID: 21731772 PMCID: PMC3121875 DOI: 10.1371/journal.pone.0021521] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 05/30/2011] [Indexed: 02/06/2023] Open
Abstract
Background Tau phosphorylation and dephosphorylation regulate in a poorly understood manner its physiological role of microtubule stabilization, and equally its integration in Alzheimer disease (AD) related fibrils. A specific phospho-pattern will result from the balance between kinases and phosphatases. The heterotrimeric Protein Phosphatase type 2A encompassing regulatory subunit PR55/Bα (PP2AT55α) is a major Tau phosphatase in vivo, which contributes to its final phosphorylation state. We use NMR spectroscopy to determine the dephosphorylation rates of phospho-Tau by this major brain phosphatase, and present site-specific and kinetic data for the individual sites including the pS202/pT205 AT8 and pT231 AT180 phospho-epitopes. Methodology/Principal Findings We demonstrate the importance of the PR55/Bα regulatory subunit of PP2A within this enzymatic process, and show that, unexpectedly, phosphorylation at the pT231 AT180 site negatively interferes with the dephosphorylation of the pS202/pT205 AT8 site. This inhibitory effect can be released by the phosphorylation dependent prolyl cis/trans isomerase Pin1. Because the stimulatory effect is lost with the dimeric PP2A core enzyme (PP2AD) or with a phospho-Tau T231A mutant, we propose that Pin1 regulates the interaction between the PR55/Bα subunit and the AT180 phospho-epitope on Tau. Conclusions/Significance Our results show that phosphorylation of T231 (AT180) can negatively influence the dephosphorylation of the pS202/pT205 AT8 epitope, even without an altered PP2A pool. Thus, a priming dephosphorylation of pT231 AT180 is required for efficient PP2AT55α-mediated dephosphorylation of pS202/pT205 AT8. The sophisticated interplay between priming mechanisms reported for certain Tau kinases and the one described here for Tau phosphatase PP2AT55α may contribute to the hyperphosphorylation of Tau observed in AD neurons.
Collapse
Affiliation(s)
- Isabelle Landrieu
- CNRS-UMR 8576, Federative Research Institute IFR 147, University of Lille-North of France, Villeneuve d'Ascq, France.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Barnett DGS, Bibb JA. The role of Cdk5 in cognition and neuropsychiatric and neurological pathology. Brain Res Bull 2011; 85:9-13. [PMID: 21145377 PMCID: PMC3073157 DOI: 10.1016/j.brainresbull.2010.11.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 11/29/2010] [Accepted: 11/30/2010] [Indexed: 01/08/2023]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase that is ubiquitous in the nervous system and interacts with a myriad of substrates. Its modulation of synaptic plasticity and associated mechanisms of learning and memory as well as neurodegeneration and cognitive disease highlights its importance in the human brain. Cdk5 is active throughout the neuron via its kinase activity, protein-protein interactions, and nuclear associations. It regulates functions thought vital to memory and plasticity, including synaptic vesicle recycling, dendritic spine formation, neurotransmitter receptor density, and neuronal excitability. Although conditional knockout of Cdk5 improves learning and plasticity, the associated deleterious effects of increased excitability cast doubts on the therapeutic efficacy of systemic inhibitors. However, through further work on the regulation of Cdk5 and its effectors, this important molecule promises to aid in elucidating key pathways involved in learning and memory and uncover innovative therapeutic targets to treat neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- David G. S. Barnett
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - James A. Bibb
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
41
|
Laha JK, Zhang X, Qiao L, Liu M, Chatterjee S, Robinson S, Kosik KS, Cuny GD. Structure-activity relationship study of 2,4-diaminothiazoles as Cdk5/p25 kinase inhibitors. Bioorg Med Chem Lett 2011; 21:2098-101. [PMID: 21353545 DOI: 10.1016/j.bmcl.2011.01.140] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 01/27/2011] [Accepted: 01/31/2011] [Indexed: 11/30/2022]
Abstract
Cdk5/p25 has emerged as a principle therapeutic target for numerous acute and chronic neurodegenerative diseases, including Alzheimer's disease. A structure-activity relationship study of 2,4-diaminothiazole inhibitors revealed that increased Cdk5/p25 inhibitory activity could be accomplished by incorporating pyridines on the 2-amino group and addition of substituents to the 2- or 3-position of the phenyl ketone moiety. Interpretation of the SAR results for many of the analogs was aided through in silico docking with Cdk5/p25 and calculating protein hydrations sites using WaterMap. Finally, improved in vitro mouse microsomal stability was also achieved.
Collapse
Affiliation(s)
- Joydev K Laha
- Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham & Women's Hospital and Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Xu J, Tsutsumi K, Tokuraku K, Estes KA, Hisanaga SI, Ikezu T. Actin interaction and regulation of cyclin-dependent kinase 5/p35 complex activity. J Neurochem 2011; 116:192-204. [PMID: 20492361 DOI: 10.1111/j.1471-4159.2010.06824.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cyclin-dependent kinase 5 (Cdk5) plays a critical role during neurodevelopment, synaptic plasticity, and neurodegeneration. Cdk5 activity depends on association with neuronal proteins p35 and p25, a proteolytic product of p35. Cdk5 regulates the actin cytoskeletal dynamics that are essential for neuronal migration, neuritic growth, and synaptogenesis. However, little is known about the interaction of actin and Cdk5 and its effect on neuronal Cdk5 activity. In a previous study, we observed that Cdk5/p35 activity is negatively correlated with co-immunoprecipitated F-actin (filamentous actin) amounts in the mouse brain, and suggested that F-actin inhibits the formation of the Cdk5/p35 complex [Journal of Neuroscience (2008) vol. 28, p. 14511]. The experiments reported here were undertaken to elucidate the relationship between actin and the formation of the Cdk5/p35 complex and its activity. Instead of an F-actin-mediated inhibition, we propose that G-actin (globular actin) in the F-actin preparations is responsible for inhibiting Cdk5/p35 and Cdk5/p25 kinase activity. We found that F-actin binds to p35 but not p25 or Cdk5. We have shown that G-actin binds directly to Cdk5 without disrupting the formation of the Cdk5/p35 or Cdk5/p25 complexes. G-actin potently suppressed Cdk5/p35 and Cdk5/p25 activity when either histone H1 or purified human tau protein were used as substrates, indicating a substrate-independent inhibitory effect of G-actin on Cdk5 activity. Finally, G-actin suppressed the activity of Cdk5 immunoprecipitated from wild type and p35-deficient mouse brain, suggesting that G-actin suppresses endogenous Cdk5 activity in a p35-independent manner. Together, these results suggest a novel mechanism of actin cytoskeletal regulation of Cdk5/p35 activity.
Collapse
Affiliation(s)
- Jiqing Xu
- Center for Neurodegenerative Disorders, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | | | | | |
Collapse
|
43
|
Hisanaga SI, Endo R. Regulation and role of cyclin-dependent kinase activity in neuronal survival and death. J Neurochem 2010; 115:1309-21. [PMID: 21044075 DOI: 10.1111/j.1471-4159.2010.07050.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cyclin-dependent kinase (Cdk)5 is a proline-directed Ser/Thr protein kinase that functions mainly in neurons and is activated by binding to a regulatory subunit, p35 or p39. Kinase activity is mainly determined by the amount of p35 available, which is controlled by a balance between synthesis and degradation. Kinase activity is also regulated by Cdk5 phosphorylation, but the activity of phosphorylated Cdk5 is in contrast to that of cycling Cdks. Cdk5 is a versatile protein kinase that regulates multiple neuronal activities including neuronal migration and synaptic signaling. Further, Cdk5 plays a role in both survival and death of neurons. Long-term inactivation of Cdk5 triggers cell death, and the survival activity of Cdk5 is apparent when neurons suffer from stress. In contrast, hyper-activation of Cdk5 by p25 promotes cell death, probably by reactivating cell-cycle machinery in the nucleus. The pro-death activity is suppressed by membrane association of Cdk5 via myristoylation of p35. Appropriate activity, localization, and regulation of Cdk5 may be critical for long-term survival of neurons, which is more than 80 years in the case of humans.
Collapse
Affiliation(s)
- Shin-ichi Hisanaga
- Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan.
| | | |
Collapse
|
44
|
Leroy A, Landrieu I, Huvent I, Legrand D, Codeville B, Wieruszeski JM, Lippens G. Spectroscopic studies of GSK3{beta} phosphorylation of the neuronal tau protein and its interaction with the N-terminal domain of apolipoprotein E. J Biol Chem 2010; 285:33435-33444. [PMID: 20679343 DOI: 10.1074/jbc.m110.149419] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alzheimer disease neurons are characterized by extraneuronal plaques formed by aggregated amyloid-β peptide and by intraneuronal tangles composed of fibrillar aggregates of the microtubule-associated Tau protein. Tau is mostly found in a hyperphosphorylated form in these tangles. Glycogen synthase kinase 3β (GSK3β) is a proline-directed kinase generally considered as one of the major players that (hyper)phosphorylates Tau. The kinase phosphorylates mainly (Ser/Thr)-Pro motifs and is believed to require a priming activity by another kinase. Here, we use an in vitro phosphorylation assay and NMR spectroscopy to characterize in a qualitative and quantitative manner the phosphorylation of Tau by GSK3β. We find that three residues can be phosphorylated (Ser-396, Ser-400, and Ser-404) by GSK3β alone, without priming. Ser-404 is essential in this process, as its mutation to Ala prevents all activity of GSK3β. However, priming enhances the catalytic efficacy of the kinase, as initial phosphorylation of Ser-214 by the cAMP-dependent protein kinase (PKA) leads to the rapid modification by GSK3β of four regularly spaced additional sites. Because the regular incorporation of negative charges by GSK3β leads to a potential parallel between phospho-Tau and heparin, we investigated its interaction with the heparin/low density lipoprotein receptor binding domain of human apolipoprotein E. We indeed observed an interaction between the GSK3β-promoted regular phospho-pattern on Tau and the apolipoprotein E fragment but none in the absence of phosphorylation or the presence of an irregular phosphorylation pattern by the prolonged activity of PKA. Apolipoprotein E is therefore able to discriminate and interact with specific phosphorylation patterns of Tau.
Collapse
Affiliation(s)
- Arnaud Leroy
- From the Structural and Functional Glycobiology Unit, UMR8576 CNRS-University of Sciences and Technologies of Lille, 59655 Villeneuve d'Ascq; Laboratoire de Biochimie Appliquée, Faculté de Pharmacie à Châtenay-Malabry, University of Paris XI, 5 Rue Jean-Baptiste Clément, 92296 Châtenay-Malabry Cedex, France.
| | - Isabelle Landrieu
- From the Structural and Functional Glycobiology Unit, UMR8576 CNRS-University of Sciences and Technologies of Lille, 59655 Villeneuve d'Ascq
| | - Isabelle Huvent
- From the Structural and Functional Glycobiology Unit, UMR8576 CNRS-University of Sciences and Technologies of Lille, 59655 Villeneuve d'Ascq
| | - Dominique Legrand
- From the Structural and Functional Glycobiology Unit, UMR8576 CNRS-University of Sciences and Technologies of Lille, 59655 Villeneuve d'Ascq
| | - Bernadette Codeville
- From the Structural and Functional Glycobiology Unit, UMR8576 CNRS-University of Sciences and Technologies of Lille, 59655 Villeneuve d'Ascq
| | - Jean-Michel Wieruszeski
- From the Structural and Functional Glycobiology Unit, UMR8576 CNRS-University of Sciences and Technologies of Lille, 59655 Villeneuve d'Ascq
| | - Guy Lippens
- From the Structural and Functional Glycobiology Unit, UMR8576 CNRS-University of Sciences and Technologies of Lille, 59655 Villeneuve d'Ascq.
| |
Collapse
|