1
|
Sultana OF, Bandaru M, Islam MA, Reddy PH. Unraveling the complexity of human brain: Structure, function in healthy and disease states. Ageing Res Rev 2024; 100:102414. [PMID: 39002647 PMCID: PMC11384519 DOI: 10.1016/j.arr.2024.102414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
The human brain stands as an intricate organ, embodying a nexus of structure, function, development, and diversity. This review delves into the multifaceted landscape of the brain, spanning its anatomical intricacies, diverse functional capacities, dynamic developmental trajectories, and inherent variability across individuals. The dynamic process of brain development, from early embryonic stages to adulthood, highlights the nuanced changes that occur throughout the lifespan. The brain, a remarkably complex organ, is composed of various anatomical regions, each contributing uniquely to its overall functionality. Through an exploration of neuroanatomy, neurophysiology, and electrophysiology, this review elucidates how different brain structures interact to support a wide array of cognitive processes, sensory perception, motor control, and emotional regulation. Moreover, it addresses the impact of age, sex, and ethnic background on brain structure and function, and gender differences profoundly influence the onset, progression, and manifestation of brain disorders shaped by genetic, hormonal, environmental, and social factors. Delving into the complexities of the human brain, it investigates how variations in anatomical configuration correspond to diverse functional capacities across individuals. Furthermore, it examines the impact of neurodegenerative diseases on the structural and functional integrity of the brain. Specifically, our article explores the pathological processes underlying neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases, shedding light on the structural alterations and functional impairments that accompany these conditions. We will also explore the current research trends in neurodegenerative diseases and identify the existing gaps in the literature. Overall, this article deepens our understanding of the fundamental principles governing brain structure and function and paves the way for a deeper understanding of individual differences and tailored approaches in neuroscience and clinical practice-additionally, a comprehensive understanding of structural and functional changes that manifest in neurodegenerative diseases.
Collapse
Affiliation(s)
- Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Madhuri Bandaru
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA 5. Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
2
|
Kim AY, Al Jerdi S, MacDonald R, Triggle CR. Alzheimer's disease and its treatment-yesterday, today, and tomorrow. Front Pharmacol 2024; 15:1399121. [PMID: 38868666 PMCID: PMC11167451 DOI: 10.3389/fphar.2024.1399121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/25/2024] [Indexed: 06/14/2024] Open
Abstract
Alois Alzheimer described the first patient with Alzheimer's disease (AD) in 1907 and today AD is the most frequently diagnosed of dementias. AD is a multi-factorial neurodegenerative disorder with familial, life style and comorbidity influences impacting a global population of more than 47 million with a projected escalation by 2050 to exceed 130 million. In the USA the AD demographic encompasses approximately six million individuals, expected to increase to surpass 13 million by 2050, and the antecedent phase of AD, recognized as mild cognitive impairment (MCI), involves nearly 12 million individuals. The economic outlay for the management of AD and AD-related cognitive decline is estimated at approximately 355 billion USD. In addition, the intensifying prevalence of AD cases in countries with modest to intermediate income countries further enhances the urgency for more therapeutically and cost-effective treatments and for improving the quality of life for patients and their families. This narrative review evaluates the pathophysiological basis of AD with an initial focus on the therapeutic efficacy and limitations of the existing drugs that provide symptomatic relief: acetylcholinesterase inhibitors (AChEI) donepezil, galantamine, rivastigmine, and the N-methyl-D-aspartate receptor (NMDA) receptor allosteric modulator, memantine. The hypothesis that amyloid-β (Aβ) and tau are appropriate targets for drugs and have the potential to halt the progress of AD is critically analyzed with a particular focus on clinical trial data with anti-Aβ monoclonal antibodies (MABs), namely, aducanumab, lecanemab and donanemab. This review challenges the dogma that targeting Aβ will benefit the majority of subjects with AD that the anti-Aβ MABs are unlikely to be the "magic bullet". A comparison of the benefits and disadvantages of the different classes of drugs forms the basis for determining new directions for research and alternative drug targets that are undergoing pre-clinical and clinical assessments. In addition, we discuss and stress the importance of the treatment of the co-morbidities, including hypertension, diabetes, obesity and depression that are known to increase the risk of developing AD.
Collapse
Affiliation(s)
- A. Y. Kim
- Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| | | | - R. MacDonald
- Health Sciences Library, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - C. R. Triggle
- Department of Pharmacology and Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| |
Collapse
|
3
|
Seitkazina A, Kim KH, Fagan E, Sung Y, Kim YK, Lim S. The Fate of Tau Aggregates Between Clearance and Transmission. Front Aging Neurosci 2022; 14:932541. [PMID: 35923541 PMCID: PMC9339952 DOI: 10.3389/fnagi.2022.932541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
Neuronal accumulation of mis-folded tau is the pathological hallmark of multiple neurodegenerative disorders, including Alzheimer’s disease. Distinct from amyloid plaques, which appear simultaneously throughout the brain, tau pathology develops first in a specific brain region and then propagates to neuroanatomically connected brain regions, exacerbating the disease. Due to the implication in disease progression, prevention of tau transmission is recognized as an important therapeutic strategy that can halt disease progression in the brain. Recently, accumulating studies have demonstrated diverse cellular mechanisms associated with cell-to-cell transmission of tau. Once transmitted, mis-folded tau species act as a prion-like seed for native tau aggregation in the recipient neuron. In this review, we summarize the diverse cellular mechanisms associated with the secretion and uptake of tau, and highlight tau-trafficking receptors, which mediate tau clearance or cell-to-cell tau transmission.
Collapse
Affiliation(s)
- Assel Seitkazina
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Kyu Hyeon Kim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Erin Fagan
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, United States
| | - Yoonsik Sung
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Yun Kyung Kim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
- *Correspondence: Yun Kyung Kim,
| | - Sungsu Lim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Sungsu Lim,
| |
Collapse
|
4
|
Li DD, Zheng CQ, Zhang F, Shi JS. Potential neuroprotection by Dendrobium nobile Lindl alkaloid in Alzheimer's disease models. Neural Regen Res 2022; 17:972-977. [PMID: 34558510 PMCID: PMC8552836 DOI: 10.4103/1673-5374.324824] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/20/2021] [Accepted: 02/20/2021] [Indexed: 01/05/2023] Open
Abstract
At present, treatments for Alzheimer's disease can temporarily relieve symptoms but cannot prevent the decline of cognitive ability and other neurodegenerative changes. Dendrobium nobile Lindl alkaloid is the main active component of Dendrobium nobile Lindl. Dendrobium nobile Lindl alkaloid has been shown to resist aging, prolong life span, and exhibit immunomodulatory effects in animals. This review summarizes the mechanisms behind the neuroprotective effects reported in Alzheimer's disease animal models. The neuroprotective effects of Dendrobium nobile Lindl alkaloid have not been studied in patients. The mechanisms by which Dendrobium nobile Lindl alkaloid has been reported to improve cognitive dysfunction in Alzheimer's disease animal models may be associated with extracellular amyloid plaque production, regulation of tau protein hyperphosphorylation, inhibition of neuroinflammation and neuronal apoptosis, activation of autophagy, and enhanced synaptic connections.
Collapse
Affiliation(s)
- Dai-Di Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Lab of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang-Qing Zheng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Lab of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Lab of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Jing-Shan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Lab of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
5
|
Feizabadi MS, Castillon VJ. The Effect of Tau and Taxol on Polymerization of MCF7 Microtubules In Vitro. Int J Mol Sci 2022; 23:ijms23020677. [PMID: 35054875 PMCID: PMC8776089 DOI: 10.3390/ijms23020677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/27/2021] [Accepted: 01/06/2022] [Indexed: 02/04/2023] Open
Abstract
Overexpression of Tau protein in breast cancer cells is identified as an indicator for potential resistance to taxane-based therapy. As reported findings have been obtained mostly from clinical studies, the undetermined underlying mechanism of such drug resistance needs to be thoroughly explored through comprehensive in vitro evaluations. Tau and Taxol bind to the beta tubulin site in microtubules’ structure. This is of particular interest in breast cancer, as microtubules of these cancer cells are structurally distinct from some other microtubules, such as neuronal microtubules, due to their unique beta tubulin isotype distribution. The observed changes in the in vitro polymerization of breast cancer microtubules, and the different function of some molecular motors along them, leave open the possibility that the drug resistance mechanism can potentially be associated with different responses of these microtubules to Tau and Taxol. We carried out a series of parallel experiments to allow comparison of the in vitro dual effect of Tau and Taxol on the polymerization of MCF7 microtubules. We observed a concentration-dependent demotion-like alteration in the self-polymerization kinetics of Tau-induced MCF7 microtubules. In contrast, microtubules polymerized under the simultaneous effects of Tau and Taxol showed promoted assembly as compared with those observed in Tau-induced microtubules. The analysis of our data obtained from the length of MCF7 microtubules polymerized under the interaction with Tau and Taxol in vitro suggests that the phenomenon known as drug resistance in microtubule-targeted drugs such as Taxol may not be directly linked to the different responses of microtubules to the drug. The effect of the drug may be mitigated due to the simultaneous interactions with other microtubule-associated proteins such as Tau protein. The observed regulatory effect of Tau and Taxol on the polymerization of breast cancer microtubules in vitro points to additional evidence for the possible role of tubulin isotypes in microtubules’ functions.
Collapse
|
6
|
Gadhavi J, Shah S, Sinha T, Jain A, Gupta S. Charge neutralization of lysine via carbamylation reveals hidden aggregation hot-spots in tau protein flanking regions. FEBS J 2021; 289:2562-2577. [PMID: 34796642 DOI: 10.1111/febs.16284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/19/2021] [Accepted: 11/17/2021] [Indexed: 11/28/2022]
Abstract
Tau protein is found abundantly in neurofibrillary tangles in Alzheimer's disease (AD). The longest human tau isoform (2N4R) has 44 lysine residues. Several lysine-based post-translational modifications (PTMs) such as glycation, acetylation, ubiquitination, and sumoylation have been implicated not only in AD, but also in other tauopathies. Carbamylation is one such lysine neutralizing age-related nonenzymatic PTM which can modulate the aggregation propensity of tau. In this work, we have studied the aggregation potential of lysine-rich regions of tau upon carbamylation which do not aggregate in their native form. Using an array of biophysical and microscopic analyses, such as ThT kinetic assay, fluorescence microscopy, Congo red staining, and scanning electron microscopy, we demonstrate that peptides derived from four of five such regions exhibit robust fibrillar amyloid formation. These regions are found in the N-terminal projection domain that encompasses proline-rich domain (148-153 and 223-230), repeat domain R1 (253-260), as well as fibrillary core region (368-378), and can be described as hidden aggregation hot-spots which become activated upon carbamylation. We have further compared the impact of carbamylation with acetylation on the aggregation propensity of lysine-rich peptide (254 KKVAVV259 ) using biophysical experiments and molecular dynamics simulations and deduced that carbamylation is a much stronger driver of aggregation than acetylation. Our findings may offer more insight into amyloid fibrils' interaction with hidden aggregation-prone nucleating sequences that act as hot-spots for inducing tau fibrillation.
Collapse
Affiliation(s)
- Joshna Gadhavi
- Department of Biological Engineering, Indian Institute of Technology Gandhinagar, India
| | - Sumedha Shah
- Department of Biological Engineering, Indian Institute of Technology Gandhinagar, India
| | - Tulika Sinha
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Ranchi, India
| | - Alok Jain
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Ranchi, India
| | - Sharad Gupta
- Department of Biological Engineering, Indian Institute of Technology Gandhinagar, India
| |
Collapse
|
7
|
Khan MI, Gilpin K, Hasan F, Mahmud KAHA, Adnan A. Effect of Strain Rate on Single Tau, Dimerized Tau and Tau-Microtubule Interface: A Molecular Dynamics Simulation Study. Biomolecules 2021; 11:1308. [PMID: 34572521 PMCID: PMC8472149 DOI: 10.3390/biom11091308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 01/24/2023] Open
Abstract
Microtubule-associated protein (MAP) tau is a cross-linking molecule that provides structural stability to axonal microtubules (MT). It is considered a potential biomarker for Alzheimer's disease (AD), dementia, and other neurological disorders. It is also a signature protein for Traumatic Brain Injury (TBI) assessment. In the case of TBI, extreme dynamic mechanical energies can be felt by the axonal cytoskeletal members. As such, fundamental understandings of the responses of single tau protein, polymerized tau protein, and tau-microtubule interfaces under high-rate mechanical forces are important. This study attempts to determine the high-strain rate mechanical behavior of single tau, dimerized tau, and tau-MT interface using molecular dynamics (MD) simulation. The results show that a single tau protein is a highly stretchable soft polymer. During deformation, first, it significantly unfolds against van der Waals and electrostatic bonds. Then it stretches against strong covalent bonds. We found that tau acts as a viscoelastic material, and its stiffness increases with the strain rate. The unfolding stiffness can be ~50-500 MPa, while pure stretching stiffness can be >2 GPa. The dimerized tau model exhibits similar behavior under similar strain rates, and tau sliding from another tau is not observed until it is stretched to >7 times of original length, depending on the strain rate. The tau-MT interface simulations show that very high strain and strain rates are required to separate tau from MT suggesting Tau-MT bonding is stronger than MT subunit bonding between themselves. The dimerized tau-MT interface simulations suggest that tau-tau bonding is stronger than tau-MT bonding. In summary, this study focuses on the structural response of individual cytoskeletal components, namely microtubule (MT) and tau protein. Furthermore, we consider not only the individual response of a component, but also their interaction with each other (such as tau with tau or tau with MT). This study will eventually pave the way to build a bottom-up multiscale brain model and analyze TBI more comprehensively.
Collapse
Affiliation(s)
- Md Ishak Khan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (M.I.K.); (F.H.); (K.A.H.A.M.)
| | - Kathleen Gilpin
- Academic Partnership and Engagement Experiment (APEX), Wright State Applied Research Corporation, Beavercreek, OH 45431, USA;
| | - Fuad Hasan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (M.I.K.); (F.H.); (K.A.H.A.M.)
| | - Khandakar Abu Hasan Al Mahmud
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (M.I.K.); (F.H.); (K.A.H.A.M.)
| | - Ashfaq Adnan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (M.I.K.); (F.H.); (K.A.H.A.M.)
| |
Collapse
|
8
|
Zhang H, Cao Y, Ma L, Wei Y, Li H. Possible Mechanisms of Tau Spread and Toxicity in Alzheimer's Disease. Front Cell Dev Biol 2021; 9:707268. [PMID: 34395435 PMCID: PMC8355602 DOI: 10.3389/fcell.2021.707268] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/09/2021] [Indexed: 12/31/2022] Open
Abstract
Tau is a protein that associates with microtubules (MTs) and promotes their assembly and stability. The protein loses its ability to bind MTs in tauopathies, and detached tau can misfold and induce the pathological changes that characterize Alzheimer’s disease (AD). A growing body of evidence indicates that tauopathies can spread between cells or connected regions. Pathological tau transmission in the brain of patients with AD and other tauopathies is due to the spread of various tau species along neuroanatomically connected regions in a “prion-like” manner. This complex process involves multiple steps of secretion, cellular uptake, transcellular transfer, and/or seeding, but the precise mechanisms of tau pathology propagation remain unclear. This review summarizes the current evidence on the nature of propagative tau species and the possible steps involved in the process of tau pathology spread, including detachment from MTs, degradations, and secretion, and discusses the different mechanisms underlying the spread of tau pathology.
Collapse
Affiliation(s)
- Huiqin Zhang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yun Wei
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Li
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Khan MI, Hasan F, Hasan Al Mahmud KA, Adnan A. Domain focused and residue focused phosphorylation effect on tau protein: A molecular dynamics simulation study. J Mech Behav Biomed Mater 2020; 113:104149. [PMID: 33125954 DOI: 10.1016/j.jmbbm.2020.104149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 11/19/2022]
Abstract
Phosphorylation has been hypothesized to alter the ability of tau protein to bind with microtubules (MT), and pathological level of phosphorylation can incorporate formation of Paired Helical Filaments (PHF) in affected tau. Study of the effect of phosphorylation on different domains of tau (projection domain, microtubule binding sites and N-terminus tail) is important to obtain insight about tau neuropathology. In an earlier study, we have already obtained the mechanical properties and behavior of single tau and dimerized tau and observed tau-MT interaction for normal level of phosphorylation. This study attempts to obtain insights on the effect of phosphorylation on different domains of tau, using molecular dynamics (MD) simulation with the aid of CHARMM force field under high strain rate. It also determines the effect of residue focused phosphorylation on tau-MT interaction and tau accumulation tendency. The results show that for single tau protein, unfolding stiffness does not differ significantly due to phosphorylation, but stretching stiffness can be much higher than the normally phosphorylated protein. For dimerized tau protein, the stretching required to separate the protein forming the dimer is similar for phosphorylation in individual domains but is significantly less in case of phosphorylation in all domains. For tau-MT interaction simulations, it is found that for normal phosphorylation, the tau separation from MT occurs at higher strain for phosphorylation in projection domain and N-terminus tail, and earlier for phosphorylation in all domains altogether than the normal phosphorylation state. The residue focused phosphorylation study also shows that tau separates earlier from MT and shows stronger accumulation tendency at the phosphorylated state, while preserving the highly stretchable and flexible characteristic of tau. This study provides important insight on mechanochemical phenomena relevant to traumatic brain injury (TBI) scenario, where the result of mechanical loading and posttranslational modification as well as conformation decides the mechanical behavior.
Collapse
Affiliation(s)
- Md Ishak Khan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Fuad Hasan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | | | - Ashfaq Adnan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA.
| |
Collapse
|
10
|
Khan MI, Hasan F, Mahmud KAHA, Adnan A. Recent Computational Approaches on Mechanical Behavior of Axonal Cytoskeletal Components of Neuron: A Brief Review. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42493-020-00043-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
11
|
Hosseini H, Rangchian A, Prins ML, Giza CC, Ruberti JW, Kavehpour HP. Probing Flow-Induced Biomolecular Interactions With Micro-Extensional Rheology: Tau Protein Aggregation. J Biomech Eng 2020; 142:034501. [PMID: 34043752 DOI: 10.1115/1.4046330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Indexed: 07/25/2024]
Abstract
Biomolecules in solutions subjected to extensional strain can form aggregates, which may be important for our understanding of pathologies involving insoluble protein structures where mechanical forces are thought to be causative (e.g., tau fibers in chronic traumatic encephalopathy (CTE)). To examine the behavior of biomolecules in solution under mechanical strains requires applying rheological methods, often to very small sample volumes. There were two primary objectives in this investigation: (1) To probe flow-induced aggregation of proteins in microliter-sized samples and (2) To test the hypothesis that tau protein aggregates under extensional flow. Tau protein (isoform:3R 0 N; 36.7 kDa) was divided into 10 μl droplets and subjected to extensional strain in a modified tensiometer. Sixteen independent tests were performed where one test on a single droplet comprised three extensional events. To assess the rheological performance of the fluid/tau mixture, the diameter of the filament that formed during extension was tracked as function of time and analyzed for signs of aggregation (i.e., increased relaxation time). The results were compared to two molecules of similar and greater size (Polyethylene Oxide: PEO35, 35 kDa and PEO100, 100 kDa). Analysis showed that the tau protein solution and PEO35 are likely to have formed aggregates, albeit at relatively high extensional strain rates (∼10 kHz). The investigation demonstrates an extensional rheological method capable of determining the properties of protein solutions in μl volumes and that tau protein can aggregate when exposed to a single extensional strain with potentially significant biological implications.
Collapse
Affiliation(s)
- H Hosseini
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - A Rangchian
- Mechanical and Aerospace Engineering and Bioengineering, University of California at Los Angeles, Los Angeles, CA 90095
| | - M L Prins
- Departments of Pediatrics, Neurosurgery and Bioengineering, Brain Injury Research Center, University of California at Los Angeles, Los Angeles, CA 90095
| | - C C Giza
- Departments of Pediatrics, Neurosurgery and Bioengineering, Brain Injury Research Center, University of California at Los Angeles, Los Angeles, CA 90095
| | - J W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA 02115
| | - H P Kavehpour
- Mechanical and Aerospace Engineering and Bioengineering, University of California at Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
12
|
Leso A, Bihaqi SW, Masoud A, Chang JK, Lahouel A, Zawia N. Loss in efficacy measures of tolfenamic acid in a tau knock-out model: Relevance to Alzheimer's disease. Exp Biol Med (Maywood) 2019; 244:1062-1069. [PMID: 31450960 DOI: 10.1177/1535370219871249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In the healthy human brain, the protein tau serves the essential function of stabilizing microtubules. However, in a diseased state, tau becomes destabilized and aggregates into a pathogenic form that ultimately creates one of the two major hallmarks of Alzheimer’s disease (AD), tau tangles. Multiple neurodegenerative diseases, termed tauopathies, such as Pick’s disease, and progressive supranuclear palsy, are also linked to mutations in tau. While AD does include a second hallmark in the form of amyloid beta (Aβ) plaques, to date all therapeutics aimed at these hallmark features have failed. The nonsteroidal anti-inflammatory drug tolfenamic acid (TA) has been shown to reduce the levels of multiple neurodegenerative endpoints viz amyloid precursor protein (APP), Aβ, tau, phosphorylated tau (p-tau) and improve cognitive function, in various murine models, via a new mechanism that targets specificity protein 1 ( SP1). Sp1 is a zinc-finger transcription factor essential for the regulation of tau and CDK5 genes (among others). The impact of TA on these neurodegenerative endpoints occurred in animal models and systems in which both the tau and the APP genes were present. The experimental model utilized in this paper tested whether the same beneficial outcomes of TA can take place after the removal of endogenous murine tau. We found that the impact of TA, both molecular and behavioral, was no longer significant in the absence of the tau gene. This ability of TA occurred independently of its action on anti-inflammatory targets. Therefore, these findings suggest the essentiality of tau for the novel mechanism of action of TA. Impact statement The number of people suffering from Alzheimer’s disease (AD) is expected to increase exponentially in the coming decades. It is estimated to cost the economy about $200 billion annually. With the failure of standard therapeutic approaches, there is a need to develop new drugs in order to avoid an “epidemic crisis” in the future. We have discovered that tolfenamic acid (TA) lowers the levels of proteins associated with AD, by targeting common transcriptional mechanisms that regulate genes involved in common pathogenic pathways. Here, we investigated whether TA had effects on both the amyloid and tau pathways, or whether it selectively targets one of these pathways which impacted the other. Behavioral and molecular studies revealed that TA loses its AD therapeutic potential when tau gene is removed. This ability of TA occurred independently of its action on anti-inflammatory targets. These findings suggest that tau is essential for the new action of TA.
Collapse
Affiliation(s)
- Allison Leso
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI 02881, USA
| | - Syed W Bihaqi
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
| | - Anwar Masoud
- Biochemical Technology Program, Faculty of Applied Science, Thamar University, Thamar 87246, Yemen
| | - Joanna K Chang
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - Asma Lahouel
- Department of Molecular and Cellular Biology, Jijel University, Jijel 18000, Algeria
| | - Nasser Zawia
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI 02881, USA.,George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA.,Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
13
|
Yeboah F, Kim TE, Bill A, Dettmer U. Dynamic behaviors of α-synuclein and tau in the cellular context: New mechanistic insights and therapeutic opportunities in neurodegeneration. Neurobiol Dis 2019; 132:104543. [PMID: 31351173 DOI: 10.1016/j.nbd.2019.104543] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 10/26/2022] Open
Abstract
α-Synuclein (αS) and tau have a lot in common. Dyshomeostasis and aggregation of both proteins are central in the pathogenesis of neurodegenerative diseases: Parkinson's disease, dementia with Lewy bodies, multi-system atrophy and other 'synucleinopathies' in the case of αS; Alzheimer's disease, frontotemporal dementia, progressive supranuclear palsy and other 'tauopathies' in the case of tau. The aggregated states of αS and tau are found to be (hyper)phosphorylated, but the relevance of the phosphorylation in health or disease is not well understood. Both tau and αS are typically characterized as 'intrinsically disordered' proteins, while both engage in transient interactions with cellular components, thereby undergoing structural changes and context-specific folding. αS transiently binds to (synaptic) vesicles forming a membrane-induced amphipathic helix; tau transiently interacts with microtubules forming an 'extended structure'. The regulation and exact nature of the interactions are not fully understood. Here we review recent and previous insights into the dynamic, transient nature of αS and tau with regard to the mode of interaction with their targets, the dwell-time while bound, and the cis and trans factors underlying the frequent switching between bound and unbound states. These aspects are intimately linked to hypotheses on how subtle changes in the transient behaviors may trigger the earliest steps in the pathogenesis of the respective brain diseases. Based on a deeper understanding of transient αS and tau conformations in the cellular context, new therapeutic strategies may emerge, and it may become clearer why existing approaches have failed or how they could be optimized.
Collapse
Affiliation(s)
- Fred Yeboah
- Novartis Institute for Biomedical Research, Chemical Biology and Therapeutics, Cambridge, MA 02139, USA
| | - Tae-Eun Kim
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anke Bill
- Novartis Institute for Biomedical Research, Chemical Biology and Therapeutics, Cambridge, MA 02139, USA.
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Naseri NN, Wang H, Guo J, Sharma M, Luo W. The complexity of tau in Alzheimer's disease. Neurosci Lett 2019; 705:183-194. [PMID: 31028844 PMCID: PMC7060758 DOI: 10.1016/j.neulet.2019.04.022] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/14/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is characterized by two major pathological lesions in the brain, amyloid plaques and neurofibrillary tangles (NFTs) composed mainly of amyloid-β (Aβ) peptides and hyperphosphorylated tau, respectively. Although accumulation of toxic Aβ species in the brain has been proposed as one of the important early events in AD, continued lack of success of clinical trials based on Aβ-targeting drugs has triggered the field to seek out alternative disease mechanisms and related therapeutic strategies. One of the new approaches is to uncover novel roles of pathological tau during disease progression. This review will primarily focus on recent advances in understanding the contributions of tau to AD.
Collapse
Affiliation(s)
- Nima N Naseri
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| | - Hong Wang
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, USA
| | - Jennifer Guo
- The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
15
|
Angeloni C, Barbalace MC, Hrelia S. Icariin and Its Metabolites as Potential Protective Phytochemicals Against Alzheimer's Disease. Front Pharmacol 2019; 10:271. [PMID: 30941046 PMCID: PMC6433697 DOI: 10.3389/fphar.2019.00271] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/04/2019] [Indexed: 01/14/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder affecting more than 35 million people worldwide. As the prevalence of AD is dramatically rising, there is an earnest need for the identification of effective therapies. Available drug treatments only target the symptoms and do not halt the progression of this disorder; thus, the use of natural compounds has been proposed as an alternative intervention strategy. Icariin, a prenylated flavonoid, has several therapeutic effects, including osteoporosis prevention, sexual dysfunction amelioration, immune system modulation, and improvement of cardiovascular function. Substantial studies indicate that icariin may be beneficial to AD by reducing the production of extracellular amyloid plaques and intracellular neurofibrillary tangles and inhibiting phosphodiesterase-5 activity. Moreover, increasing evidence has indicated that icariin exerts a protective role in AD also by limiting inflammation, oxidative stress and reducing potential risk factors for AD such as atherosclerosis. This mini-review discusses the multiple potential mechanisms of action of icariin on the pathobiology of AD including explanation regarding its bioavailability, metabolism and pharmacokinetic.
Collapse
Affiliation(s)
| | | | - Silvana Hrelia
- Department for Life Quality Studies, University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Cilento EM, Jin L, Stewart T, Shi M, Sheng L, Zhang J. Mass spectrometry: A platform for biomarker discovery and validation for Alzheimer's and Parkinson's diseases. J Neurochem 2019; 151:397-416. [PMID: 30474862 DOI: 10.1111/jnc.14635] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022]
Abstract
Accurate, reliable, and objective biomarkers for Alzheimer's disease (AD), Parkinson's disease (PD), and related age-associated neurodegenerative disorders are urgently needed to assist in both diagnosis, particularly at early stages, and monitoring of disease progression. Technological advancements in protein detection platforms over the last few decades have resulted in a plethora of reported molecular biomarker candidates for both AD and PD; however, very few of these candidates are developed beyond the discovery phase of the biomarker development pipeline, a reflection of the current bottleneck within the field. In this review, the expanded use of selected reaction monitoring (SRM) targeted mass spectrometry will be discussed in detail as a platform for systematic verification of large panels of protein biomarker candidates prior to costly validation testing. We also advocate for the coupling of discovery-based proteomics with modern targeted MS-based approaches (e.g., SRM) within a single study in future workflows to expedite biomarker development and validation for AD and PD. It is our hope that improving the efficiency within the biomarker development process by use of an SRM pipeline may ultimately hasten the development of biomarkers that both decrease misdiagnosis of AD and PD and ultimately lead to detection at early stages of disease and objective assessment of disease progression. This article is part of the special issue "Proteomics".
Collapse
Affiliation(s)
- Eugene M Cilento
- Department of Pathology, University of Washington, School of Medicine, Seattle, Washington, USA
| | - Lorrain Jin
- Department of Pathology, University of Washington, School of Medicine, Seattle, Washington, USA
| | - Tessandra Stewart
- Department of Pathology, University of Washington, School of Medicine, Seattle, Washington, USA
| | - Min Shi
- Department of Pathology, University of Washington, School of Medicine, Seattle, Washington, USA
| | - Lifu Sheng
- Department of Pathology, University of Washington, School of Medicine, Seattle, Washington, USA
| | - Jing Zhang
- Department of Pathology, University of Washington, School of Medicine, Seattle, Washington, USA.,Department of Pathology, School of Basic Medicine, Peking University Health Science Center, Peking University Third Hospital and Peking Key Laboratory for Early Diagnosis of Neurodegenerative Disorders, Beijing, China
| |
Collapse
|
17
|
Kounakis K, Tavernarakis N. The Cytoskeleton as a Modulator of Aging and Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:227-245. [PMID: 31493230 DOI: 10.1007/978-3-030-25650-0_12] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cytoskeleton consists of filamentous protein polymers that form organized structures, contributing to a multitude of cell life aspects. It includes three types of polymers: the actin microfilaments, the microtubules and the intermediate filaments. Decades of research have implicated the cytoskeleton in processes that regulate cellular and organismal aging, as well as neurodegeneration associated with injury or neurodegenerative disease, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, or Charcot Marie Tooth disease. Here, we provide a brief overview of cytoskeletal structure and function, and discuss experimental evidence linking cytoskeletal function and dynamics with aging and neurodegeneration.
Collapse
Affiliation(s)
- Konstantinos Kounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece.,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece. .,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece.
| |
Collapse
|
18
|
Alavi Naini SM, Soussi-Yanicostas N. Heparan Sulfate as a Therapeutic Target in Tauopathies: Insights From Zebrafish. Front Cell Dev Biol 2018; 6:163. [PMID: 30619849 PMCID: PMC6306439 DOI: 10.3389/fcell.2018.00163] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/15/2018] [Indexed: 12/13/2022] Open
Abstract
Microtubule-associated protein tau (MAPT) hyperphosphorylation and aggregation, are two hallmarks of a family of neurodegenerative disorders collectively referred to as tauopathies. In many tauopathies, including Alzheimer's disease (AD), progressive supranuclear palsy (PSP) and Pick's disease, tau aggregates are found associated with highly sulfated polysaccharides known as heparan sulfates (HSs). In AD, amyloid beta (Aβ) peptide aggregates associated with HS are also characteristic of disease. Heparin, an HS analog, promotes misfolding, hyperphosphorylation and aggregation of tau protein in vitro. HS also provides cell surface receptors for attachment and uptake of tau seeds, enabling their propagation. These findings point to HS-tau interactions as potential therapeutic targets in tauopathies. The zebrafish genome contains genes paralogous to MAPT, genes orthologous to HS biosynthetic and chain modifier enzymes, and other genes implicated in AD. The nervous system in the zebrafish bears anatomical and chemical similarities to that in humans. These homologies, together with numerous technical advantages, make zebrafish a valuable model for investigating basic mechanisms in tauopathies and identifying therapeutic targets. Here, we comprehensively review current knowledge on the role of HSs in tau pathology and HS-targeting therapeutic approaches. We also discuss novel insights from zebrafish suggesting a role for HS 3-O-sulfated motifs in tau pathology and establishing HS antagonists as potential preventive agents or therapies for tauopathies.
Collapse
Affiliation(s)
- Seyedeh Maryam Alavi Naini
- Department of Neuroscience, Institut de Biologie Paris Seine (IBPS), INSERM, CNRS, Sorbonne Université, Paris, France
| | | |
Collapse
|
19
|
Chang HY, Sang TK, Chiang AS. Untangling the Tauopathy for Alzheimer's disease and parkinsonism. J Biomed Sci 2018; 25:54. [PMID: 29991349 PMCID: PMC6038292 DOI: 10.1186/s12929-018-0457-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/04/2018] [Indexed: 12/19/2022] Open
Abstract
Tau is a microtubule-associated protein that mainly localizes to the axon to stabilize axonal microtubule structure and neuronal connectivity. Tau pathology is one of the most common proteinopathies that associates with age-dependent neurodegenerative diseases including Alzheimer's disease (AD), and various Parkinsonism. Tau protein undergoes a plethora of intra-molecular modifications and some altered forms promote the production of toxic oligomeric tau and paired helical filaments, and through which further assemble into neurofibrillary tangles, also known as tauopathy. In this review, we will discuss the recent advances of the tauopathy research, primarily focusing on its association with the early axonal manifestation of axonal transport defect, axonal mitochondrial stress, autophagic vesicle accumulation and the proceeding of axon destruction, and the pathogenic Tau spreading across the synapse. Two alternative strategies either by targeting tau protein itself or by improving the age-related physiological decline are currently racing to find the hopeful treatment for tauopathy. Undoubtedly, more studies are needed to combat this devastating condition that has already affected millions of people in our aging population.
Collapse
Affiliation(s)
- Hui-Yun Chang
- Department of Medical Science, Institute of Systems Neuroscience, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
- Brain Research Center, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| | - Tzu-Kang Sang
- Department of Life Science, Institute of Biotechnology, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
- Brain Research Center, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| | - Ann-Shyn Chiang
- Department of Medical Science, Institute of Systems Neuroscience, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
- Department of Life Science, Institute of Biotechnology, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
- Brain Research Center, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| |
Collapse
|
20
|
CD64-directed microtubule associated protein tau kills leukemic blasts ex vivo. Oncotarget 2018; 7:67166-67174. [PMID: 27564103 PMCID: PMC5341865 DOI: 10.18632/oncotarget.11568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/11/2016] [Indexed: 01/04/2023] Open
Abstract
Fc gamma receptor I (FcγRI, CD64) is a well-known target antigen for passive immunotherapy against acute myeloid leukemia and chronic myelomonocytic leukemia. We recently reported the preclinical immunotherapeutic potential of microtubule associated protein tau (MAP) against a variety of cancer types including breast carcinoma and Hodgkin's lymphoma. Here we demonstrate that the CD64-directed human cytolytic fusion protein H22(scFv)-MAP kills ex vivo 15–50% of CD64+ leukemic blasts derived from seven myeloid leukemia patients. Furthermore, in contrast to the nonspecific cytostatic agent paclitaxel, H22(scFv)-MAP showed no cytotoxicity towards healthy CD64+ PBMC-derived cells and macrophages. The targeted delivery of this microtubule stabilizing agent therefore offers a promising new strategy for specific treatment of CD64+ leukemia.
Collapse
|
21
|
Oliveira J, Costa M, de Almeida MSC, da Cruz e Silva OA, Henriques AG. Protein Phosphorylation is a Key Mechanism in Alzheimer’s Disease. J Alzheimers Dis 2017; 58:953-978. [DOI: 10.3233/jad-170176] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Joana Oliveira
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, iBiMED, University of Aveiro, Aveiro, Portugal
| | - Márcio Costa
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, iBiMED, University of Aveiro, Aveiro, Portugal
| | | | - Odete A.B. da Cruz e Silva
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, iBiMED, University of Aveiro, Aveiro, Portugal
| | - Ana Gabriela Henriques
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
22
|
Bodea L, Eckert A, Ittner LM, Piguet O, Götz J. Tau physiology and pathomechanisms in frontotemporal lobar degeneration. J Neurochem 2016; 138 Suppl 1:71-94. [PMID: 27306859 PMCID: PMC5094566 DOI: 10.1111/jnc.13600] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/31/2016] [Accepted: 02/24/2016] [Indexed: 12/27/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) has been associated with toxic intracellular aggregates of hyperphosphorylated tau (FTLD-tau). Moreover, genetic studies identified mutations in the MAPT gene encoding tau in familial cases of the disease. In this review, we cover a range of aspects of tau function, both in the healthy and diseased brain, discussing several in vitro and in vivo models. Tau structure and function in the healthy brain is presented, accentuating its distinct compartmentalization in neurons and its role in microtubule stabilization and axonal transport. Furthermore, tau-driven pathology is discussed, introducing current concepts and the underlying experimental evidence. Different aspects of pathological tau phosphorylation, the protein's genomic and domain organization as well as its spreading in disease, together with MAPT-associated mutations and their respective models are presented. Dysfunction related to other post-transcriptional modifications and their effect on normal neuronal functions such as cell cycle, epigenetics and synapse dynamics are also discussed, providing a mechanistic explanation for the observations made in FTLD-tau cases, with the possibility for therapeutic intervention. In this review, we cover aspects of tau function, both in the healthy and diseased brain, referring to different in vitro and in vivo models. In healthy neurons, tau is compartmentalized, with higher concentrations found in the distal part of the axon. Cargo molecules are sensitive to this gradient. A disturbed tau distribution, as found in frontotemporal lobar degeneration (FTLD-tau), has severe consequences for cellular physiology: tau accumulates in the neuronal soma and dendrites, leading among others to microtubule depolymerization and impaired axonal transport. Tau forms insoluble aggregates that sequester additional molecules stalling cellular physiology. Neuronal communication is gradually lost as toxic tau accumulates in dendritic spines with subsequent degeneration of synapses and synaptic loss. Thus, by providing a mechanistic explanation for the observations made in FTLD-tau cases, arises a possibility for therapeutic interventions. This article is part of the Frontotemporal Dementia special issue.
Collapse
Affiliation(s)
- Liviu‐Gabriel Bodea
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Anne Eckert
- Neurobiology LaboratoryPsychiatric University Clinics BaselUniversity of BaselBaselSwitzerland
| | - Lars Matthias Ittner
- Dementia Research UnitSchool of Medical SciencesFaculty of MedicineUniversity of New South WalesSydneyNSWAustralia
| | | | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
23
|
Xu L, Zheng J, Margittai M, Nussinov R, Ma B. How Does Hyperphopsphorylation Promote Tau Aggregation and Modulate Filament Structure and Stability? ACS Chem Neurosci 2016; 7:565-75. [PMID: 26854860 PMCID: PMC7831686 DOI: 10.1021/acschemneuro.5b00294] [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: 12/16/2022] Open
Abstract
Tau proteins are hyperphosphorylated at common sites in their N- and C-terminal domains in at least three neurodegenerative diseases, Parkinson, dementia with Lewy bodies, and Alzheimer's, suggesting specific pathology but general mechanism. Full-length human tau filament comprises a rigid core and a two-layered fuzzy coat. Tau is categorized into two groups of isoforms, with either four repeats (R1-R4) or three repeats (R1, R3, and R4); their truncated constructs are respectively called K18 and K19. Using multiscale molecular dynamics simulations, we explored the conformational consequences of hyperhposphorylation on tau's repeats. Our lower conformational energy filament models suggest a rigid filament core with a radius of ∼30 to 40 Å and an outer layer with a thickness of ∼140 Å consisting of a double-layered polyelectrolyte. The presence of the phosphorylated terminal domains alters the relative stabilities in the K18 ensemble, thus shifting the populations of the full-length filaments. However, the structure with the straight repeats in the core region is still the most stable, similar to the truncated K18 peptide species without the N- and C-terminus. Our simulations across different scales of resolution consistently reveal that hyperphosphorylation of the two terminal domains decreases the attractive interactions among the N- and C-terminus and repeat domain. To date, the relationship on the conformational level between phosphorylation and aggregation has not been understood. Our results suggest that the exposure of the repeat domain upon hyperphosphorylation could enhance tau filament aggregation. Thus, we discovered that even though these neurodegenerative diseases vary and their associated tau filaments are phosphorylated to different extents, remarkably, the three pathologies appear to share a common tau aggregation mechanism.
Collapse
Affiliation(s)
- Liang Xu
- School of Chemistry, Dalian University of Technology, Dalian 116024, China
| | - Jie Zheng
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Martin Margittai
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80208, United States
| | - Ruth Nussinov
- Sackler Institute of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Basic Research Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, NCI, Frederick, Maryland 21702, United States
| | - Buyong Ma
- Basic Research Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, NCI, Frederick, Maryland 21702, United States
| |
Collapse
|
24
|
Regulation of Microtubule Assembly by Tau and not by Pin1. J Mol Biol 2016; 428:1742-59. [PMID: 26996940 DOI: 10.1016/j.jmb.2016.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/08/2016] [Accepted: 03/10/2016] [Indexed: 11/21/2022]
Abstract
The molecular mechanism by which the microtubule-associated protein (MAP) tau regulates the formation of microtubules (MTs) is poorly understood. The activity of tau is controlled via phosphorylation at specific Ser/Thr sites. Of those phosphorylation sites, 17 precede a proline, making them potential recognition sites for the peptidyl-prolyl isomerase Pin1. Pin1 binding and catalysis of phosphorylated tau at the AT180 epitope, which was implicated in Alzheimer's disease, has been reported to be crucial for restoring tau's ability to promote MT polymerization in vitro and in vivo [1]. Surprisingly, we discover that Pin1 does not promote phosphorylated tau-induced MT formation in vitro, refuting the commonly accepted model in which Pin1 binding and catalysis on the A180 epitope restores the function of the Alzheimer's associated phosphorylated tau in tubulin assembly [1, 2]. Using turbidity assays, time-resolved small angle X-ray scattering (SAXS), and time-resolved negative stain electron microscopy (EM), we investigate the mechanism of tau-mediated MT assembly and the role of the Thr231 and Ser235 phosphorylation on this process. We discover novel GTP-tubulin ring-shaped species, which are detectable in the earliest stage of tau-induced polymerization and may play a crucial role in the early nucleation phase of MT assembly. Finally, by NMR and SAXS experiments, we show that the tau molecules must be located on the surface of MTs and tubulin rings during the polymerization reaction. The interaction between tau and tubulin is multipartite, with a high affinity interaction of the four tubulin-binding repeats, and a weaker interaction with the proline-rich sequence and the termini of tau.
Collapse
|
25
|
Dutta S, Jho YS. Adsorption of highly charged Gaussian polyelectrolytes onto oppositely charged surfaces. J Chem Phys 2016; 144:094902. [PMID: 26957178 DOI: 10.1063/1.4942023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
In many biological processes highly charged biopolymers are adsorbed onto oppositely charged surfaces of macroions and membranes. They form strongly correlated structures close to the surface which cannot be explained by the conventional Poisson-Boltzmann theory. In this work strong coupling theory is used to study the adsorption of highly charged Gaussian polyelectrolytes. Two cases of adsorptions are considered, when the Gaussian polyelectrolytes are confined (a) by one charged wall, and (b) between two charged walls. The effects of salt and the geometry of the polymers on their adsorption-depletion transitions in the strong coupling regime are discussed.
Collapse
Affiliation(s)
- Sandipan Dutta
- Asia Pacific Center for Theoretical Physics, Pohang, Gyeongbuk 790-784, South Korea
| | - Y S Jho
- Department of Physics, Asia Pacific Center for Theoretical Physics, Pohang University of Science and Technology, Pohang, Gyeongbuk 790-784, South Korea
| |
Collapse
|
26
|
Bougé AL, Parmentier ML. Tau excess impairs mitosis and kinesin-5 function, leading to aneuploidy and cell death. Dis Model Mech 2016; 9:307-19. [PMID: 26822478 PMCID: PMC4833329 DOI: 10.1242/dmm.022558] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 01/21/2016] [Indexed: 12/31/2022] Open
Abstract
In neurodegenerative diseases such as Alzheimer's disease (AD), cell cycle defects and associated aneuploidy have been described. However, the importance of these defects in the physiopathology of AD and the underlying mechanistic processes are largely unknown, in particular with respect to the microtubule (MT)-binding protein Tau, which is found in excess in the brain and cerebrospinal fluid of affected individuals. Although it has long been known that Tau is phosphorylated during mitosis to generate a lower affinity for MTs, there is, to our knowledge, no indication that an excess of this protein could affect mitosis. Here, we studied the effect of an excess of human Tau (hTau) protein on cell mitosis in vivo. Using the Drosophila developing wing disc epithelium as a model, we show that an excess of hTau induces a mitotic arrest, with the presence of monopolar spindles. This mitotic defect leads to aneuploidy and apoptotic cell death. We studied the mechanism of action of hTau and found that the MT-binding domain of hTau is responsible for these defects. We also demonstrate that the effects of hTau occur via the inhibition of the function of the kinesin Klp61F, the Drosophila homologue of kinesin-5 (also called Eg5 or KIF11). We finally show that this deleterious effect of hTau is also found in other Drosophila cell types (neuroblasts) and tissues (the developing eye disc), as well as in human HeLa cells. By demonstrating that MT-bound Tau inhibits the Eg5 kinesin and cell mitosis, our work provides a new framework to consider the role of Tau in neurodegenerative diseases. Drosophila Collection: We show that Tau, a microtubule-binding protein involved in many neurodegenerative diseases, impairs mitosis when in excess. We show that this occurs via the inhibition of the kinesin-5 mitotic motor.
Collapse
Affiliation(s)
- Anne-Laure Bougé
- Department of Neurosciences, Institut de Génomique Fonctionnelle, CNRS-UMR5203, INSERM-U1191, Université Montpellier, 141 Rue de la Cardonille, Montpellier F-34094, Cedex 5, France
| | - Marie-Laure Parmentier
- Department of Neurosciences, Institut de Génomique Fonctionnelle, CNRS-UMR5203, INSERM-U1191, Université Montpellier, 141 Rue de la Cardonille, Montpellier F-34094, Cedex 5, France
| |
Collapse
|
27
|
He E, Yan G, Zhang J, Wang J, Li W. Effects of phosphorylation on the intrinsic propensity of backbone conformations of serine/threonine. J Biol Phys 2016; 42:247-58. [PMID: 26759163 DOI: 10.1007/s10867-015-9405-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/18/2015] [Indexed: 11/28/2022] Open
Abstract
Each amino acid has its intrinsic propensity for certain local backbone conformations, which can be further modulated by the physicochemical environment and post-translational modifications. In this work, we study the effects of phosphorylation on the intrinsic propensity for different local backbone conformations of serine/threonine by molecular dynamics simulations. We showed that phosphorylation has very different effects on the intrinsic propensity for certain local backbone conformations for the serine and threonine. The phosphorylation of serine increases the propensity of forming polyproline II, whereas that of threonine has the opposite effect. Detailed analysis showed that such different responses to phosphorylation mainly arise from their different perturbations to the backbone hydration and the geometrical constraints by forming side-chain-backbone hydrogen bonds due to phosphorylation. Such an effect of phosphorylation on backbone conformations can be crucial for understanding the molecular mechanism of phosphorylation-regulated protein structures/dynamics and functions.
Collapse
Affiliation(s)
- Erbin He
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, People's Republic of China.,Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Guanghui Yan
- Department of Mathematics and Physics, Nanjing Institute of Technology, Nanjing, 211167, People's Republic of China
| | - Jian Zhang
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, People's Republic of China.,Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Jun Wang
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, People's Republic of China. .,Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210093, People's Republic of China.
| | - Wenfei Li
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, People's Republic of China. .,Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
28
|
Chung PJ, Choi MC, Miller HP, Feinstein HE, Raviv U, Li Y, Wilson L, Feinstein SC, Safinya CR. Direct force measurements reveal that protein Tau confers short-range attractions and isoform-dependent steric stabilization to microtubules. Proc Natl Acad Sci U S A 2015; 112:E6416-25. [PMID: 26542680 PMCID: PMC4664379 DOI: 10.1073/pnas.1513172112] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Microtubules (MTs) are hollow cytoskeletal filaments assembled from αβ-tubulin heterodimers. Tau, an unstructured protein found in neuronal axons, binds to MTs and regulates their dynamics. Aberrant Tau behavior is associated with neurodegenerative dementias, including Alzheimer's. Here, we report on a direct force measurement between paclitaxel-stabilized MTs coated with distinct Tau isoforms by synchrotron small-angle X-ray scattering (SAXS) of MT-Tau mixtures under osmotic pressure (P). In going from bare MTs to MTs with Tau coverage near the physiological submonolayer regime (Tau/tubulin-dimer molar ratio; ΦTau = 1/10), isoforms with longer N-terminal tails (NTTs) sterically stabilized MTs, preventing bundling up to PB ∼ 10,000-20,000 Pa, an order of magnitude larger than bare MTs. Tau with short NTTs showed little additional effect in suppressing the bundling pressure (PB ∼ 1,000-2,000 Pa) over the same range. Remarkably, the abrupt increase in PB observed for longer isoforms suggests a mushroom to brush transition occurring at 1/13 < ΦTau < 1/10, which corresponds to MT-bound Tau with NTTs that are considerably more extended than SAXS data for Tau in solution indicate. Modeling of Tau-mediated MT-MT interactions supports the hypothesis that longer NTTs transition to a polyelectrolyte brush at higher coverages. Higher pressures resulted in isoform-independent irreversible bundling because the polyampholytic nature of Tau leads to short-range attractions. These findings suggest an isoform-dependent biological role for regulation by Tau, with longer isoforms conferring MT steric stabilization against aggregation either with other biomacromolecules or into tight bundles, preventing loss of function in the crowded axon environment.
Collapse
Affiliation(s)
- Peter J Chung
- Materials Department, University of California, Santa Barbara, CA 93106; Physics Department, University of California, Santa Barbara, CA 93106; Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| | - Myung Chul Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Republic of Korea
| | - Herbert P Miller
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106; Neuroscience Research Institute, University of California, Santa Barbara, CA 93106
| | - H Eric Feinstein
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106; Neuroscience Research Institute, University of California, Santa Barbara, CA 93106
| | - Uri Raviv
- Institute of Chemistry, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Youli Li
- Materials Research Laboratory, University of California, Santa Barbara, CA 93106
| | - Leslie Wilson
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106; Neuroscience Research Institute, University of California, Santa Barbara, CA 93106
| | - Stuart C Feinstein
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106; Neuroscience Research Institute, University of California, Santa Barbara, CA 93106
| | - Cyrus R Safinya
- Materials Department, University of California, Santa Barbara, CA 93106; Physics Department, University of California, Santa Barbara, CA 93106; Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106;
| |
Collapse
|
29
|
A Stochastic Multiscale Model That Explains the Segregation of Axonal Microtubules and Neurofilaments in Neurological Diseases. PLoS Comput Biol 2015; 11:e1004406. [PMID: 26285012 PMCID: PMC4540448 DOI: 10.1371/journal.pcbi.1004406] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/17/2015] [Indexed: 11/19/2022] Open
Abstract
The organization of the axonal cytoskeleton is a key determinant of the normal function of an axon, which is a long thin projection of a neuron. Under normal conditions two axonal cytoskeletal polymers, microtubules and neurofilaments, align longitudinally in axons and are interspersed in axonal cross-sections. However, in many neurotoxic and neurodegenerative disorders, microtubules and neurofilaments segregate apart from each other, with microtubules and membranous organelles clustered centrally and neurofilaments displaced to the periphery. This striking segregation precedes the abnormal and excessive neurofilament accumulation in these diseases, which in turn leads to focal axonal swellings. While neurofilament accumulation suggests an impairment of neurofilament transport along axons, the underlying mechanism of their segregation from microtubules remains poorly understood for over 30 years. To address this question, we developed a stochastic multiscale model for the cross-sectional distribution of microtubules and neurofilaments in axons. The model describes microtubules, neurofilaments and organelles as interacting particles in a 2D cross-section, and is built upon molecular processes that occur on a time scale of seconds or shorter. It incorporates the longitudinal transport of neurofilaments and organelles through this domain by allowing stochastic arrival and departure of these cargoes, and integrates the dynamic interactions of these cargoes with microtubules mediated by molecular motors. Simulations of the model demonstrate that organelles can pull nearby microtubules together, and in the absence of neurofilament transport, this mechanism gradually segregates microtubules from neurofilaments on a time scale of hours, similar to that observed in toxic neuropathies. This suggests that the microtubule-neurofilament segregation can be a consequence of the selective impairment of neurofilament transport. The model generates the experimentally testable prediction that the rate and extent of segregation will be dependent on the sizes of the moving organelles as well as the density of their traffic. The shape and function of axons is dependent on a dynamic system of microscopic intracellular protein polymers (microtubules, neurofilaments and microfilaments) that comprise the axonal cytoskeleton. Neurofilaments are cargoes of intracellular transport that move along microtubule tracks, and they accumulate abnormally in axons in many neurotoxic and neurodegenerative disorders. Intriguingly, it has been reported that neurofilaments and microtubules, which are normally interspersed in axonal cross-sections, often segregate apart from each other in these disorders, which is something that is never observed in healthy axons. Here we describe a stochastic multiscale computational model that explains the mechanism of this striking segregation and offers insights into the mechanism of neurofilament accumulation in disease.
Collapse
|
30
|
Miller N, Feng Z, Edens BM, Yang B, Shi H, Sze CC, Hong BT, Su SC, Cantu JA, Topczewski J, Crawford TO, Ko CP, Sumner CJ, Ma L, Ma YC. Non-aggregating tau phosphorylation by cyclin-dependent kinase 5 contributes to motor neuron degeneration in spinal muscular atrophy. J Neurosci 2015; 35:6038-50. [PMID: 25878277 PMCID: PMC4397602 DOI: 10.1523/jneurosci.3716-14.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 02/25/2015] [Accepted: 03/03/2015] [Indexed: 01/27/2023] Open
Abstract
Mechanisms underlying motor neuron degeneration in spinal muscular atrophy (SMA), the leading inherited cause of infant mortality, remain largely unknown. Many studies have established the importance of hyperphosphorylation of the microtubule-associated protein tau in various neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. However, tau phosphorylation in SMA pathogenesis has yet to be investigated. Here we show that tau phosphorylation on serine 202 (S202) and threonine 205 (T205) is increased significantly in SMA motor neurons using two SMA mouse models and human SMA patient spinal cord samples. Interestingly, phosphorylated tau does not form aggregates in motor neurons or neuromuscular junctions (NMJs), even at late stages of SMA disease, distinguishing it from other tauopathies. Hyperphosphorylation of tau on S202 and T205 is mediated by cyclin-dependent kinase 5 (Cdk5) in SMA disease condition, because tau phosphorylation at these sites is significantly reduced in Cdk5 knock-out mice; genetic knock-out of Cdk5 activating subunit p35 in an SMA mouse model also leads to reduced tau phosphorylation on S202 and T205 in the SMA;p35(-/-) compound mutant mice. In addition, expression of the phosphorylation-deficient tauS202A,T205A mutant alleviates motor neuron defects in a zebrafish SMA model in vivo and mouse motor neuron degeneration in culture, whereas expression of phosphorylation-mimetic tauS202E,T205E promotes motor neuron defects. More importantly, genetic knock-out of tau in SMA mice rescues synapse stripping on motor neurons, NMJ denervation, and motor neuron degeneration in vivo. Altogether, our findings suggest a novel mechanism for SMA pathogenesis in which hyperphosphorylation of non-aggregating tau by Cdk5 contributes to motor neuron degeneration.
Collapse
Affiliation(s)
- Nimrod Miller
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611
| | - Zhihua Feng
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - Brittany M Edens
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611
| | - Ben Yang
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611
| | - Han Shi
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611
| | - Christie C Sze
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611
| | - Benjamin Taige Hong
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611, State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China
| | - Susan C Su
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Jorge A Cantu
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611
| | - Jacek Topczewski
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611
| | - Thomas O Crawford
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Chien-Ping Ko
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - Charlotte J Sumner
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Long Ma
- State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China
| | - Yong-Chao Ma
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Lurie Children's Hospital of Chicago, Chicago, Illinois 60611,
| |
Collapse
|
31
|
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: 1.9] [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
|
32
|
Tau protein modifications and interactions: their role in function and dysfunction. Int J Mol Sci 2014; 15:4671-713. [PMID: 24646911 PMCID: PMC3975420 DOI: 10.3390/ijms15034671] [Citation(s) in RCA: 234] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 02/11/2014] [Accepted: 03/04/2014] [Indexed: 01/29/2023] Open
Abstract
Tau protein is abundant in the central nervous system and involved in microtubule assembly and stabilization. It is predominantly associated with axonal microtubules and present at lower level in dendrites where it is engaged in signaling functions. Post-translational modifications of tau and its interaction with several proteins play an important regulatory role in the physiology of tau. As a consequence of abnormal modifications and expression, tau is redistributed from neuronal processes to the soma and forms toxic oligomers or aggregated deposits. The accumulation of tau protein is increasingly recognized as the neuropathological hallmark of a number of dementia disorders known as tauopathies. Dysfunction of tau protein may contribute to collapse of cytoskeleton, thereby causing improper anterograde and retrograde movement of motor proteins and their cargos on microtubules. These disturbances in intraneuronal signaling may compromise synaptic transmission as well as trophic support mechanisms in neurons.
Collapse
|
33
|
Microtubule-associated protein tau facilitates the targeted killing of proliferating cancer cells in vitro and in a xenograft mouse tumour model in vivo. Br J Cancer 2013; 109:1570-8. [PMID: 23942071 PMCID: PMC3776980 DOI: 10.1038/bjc.2013.457] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/18/2013] [Accepted: 07/22/2013] [Indexed: 01/01/2023] Open
Abstract
Background: Antibody drug conjugates (ADCs) and immunotoxins (ITs) are promising anticancer immunotherapeutics. Despite their encouraging performance in clinical trials, both ADCs and ITs often suffer from disadvantages such as stoichiometrically undefined chemical linkage of the cytotoxic payload (ADCs) and the potential immunogenicity of toxins derived from bacteria and plants (ITs). Methods: Human microtubule-associated protein tau (MAP) was cloned in-frame with human EGF, expressed in E. coli and purified by standard chromatographic methods. The in vitro activity was confirmed by flow cytometry, cell viability assays and tubulin polymerisation assay. The in vivo efficacy was demonstrated using noninvasive far-red in vivo imaging. Results: The EGF-MAP selectively induced apoptosis in EGFR-overexpressing proliferating cancer cells through stabilisation of microtubules. Nonproliferating cells were not affected, demonstrating superior selectivity of EGF-MAP for cancer cells. The EGF-MAP was well tolerated at high doses in mice compared with the ETA'-based control. The in vivo efficacy of EGF-MAP was demonstrated in a tumour xenograft mouse model. Conclusion: Our data indicate the general mechanism of action for a new class of human immunotherapeutic reagents suitable for the treatment of cancer. This approach combines the binding specificity of targeting ligands with the selective cytotoxicity of MAP towards proliferating cells.
Collapse
|
34
|
Nath A, Sammalkorpi M, DeWitt DC, Trexler AJ, Elbaum-Garfinkle S, O'Hern CS, Rhoades E. The conformational ensembles of α-synuclein and tau: combining single-molecule FRET and simulations. Biophys J 2013. [PMID: 23199922 DOI: 10.1016/j.bpj.2012.09.032] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Intrinsically disordered proteins (IDPs) are increasingly recognized for their important roles in a range of biological contexts, both in normal physiological function and in a variety of devastating human diseases. However, their structural characterization by traditional biophysical methods, for the purposes of understanding their function and dysfunction, has proved challenging. Here, we investigate the model IDPs α-Synuclein (αS) and tau, that are involved in major neurodegenerative conditions including Parkinson's and Alzheimer's diseases, using excluded volume Monte Carlo simulations constrained by pairwise distance distributions from single-molecule fluorescence measurements. Using this, to our knowledge, novel approach we find that a relatively small number of intermolecular distance constraints are sufficient to accurately determine the dimensions and polymer conformational statistics of αS and tau in solution. Moreover, this method can detect local changes in αS and tau conformations that correlate with enhanced aggregation. Constrained Monte Carlo simulations produce ensembles that are in excellent agreement both with experimental measurements on αS and tau and with all-atom, explicit solvent molecular dynamics simulations of αS, with much lower configurational sampling requirements and computational expense.
Collapse
Affiliation(s)
- Abhinav Nath
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The protein tau is the most abundant microtubule associated protein in the central and peripheral nervous system. In the brain, tau plays a role in the assembly and stabilization of microtubules. The function of tau, however, appears to overlap with other microtubule binding proteins. The observation that tau is associated with neurodegenerative diseases has renewed interest in this protein. Various aspects of structure and biochemistry of tau, fibril formation and clinical perspectives, including therapeutic strategies are reviewed in this chapter.
Collapse
|
36
|
Ferrari R, Hardy J, Momeni P. Frontotemporal dementia: from Mendelian genetics towards genome wide association studies. J Mol Neurosci 2011; 45:500-15. [PMID: 21898125 DOI: 10.1007/s12031-011-9635-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 08/17/2011] [Indexed: 12/12/2022]
Abstract
Frontotemporal lobar degeneration is the most common cause of dementia of non-Alzheimer's type worldwide. It manifests, clinically, with behavioural changes and language impairment and is pathologically associated with tau- or ubiquitin-positive inclusions detected in neurons and glial cells of the frontal and temporal lobes in the brain. Genetic variations in the microtubule-associated protein tau and progranulin genes explain almost 50% of familial cases, whilst variations in TAR DNA-binding protein, charged multivescicular body protein 2B, valosin-containing protein and fused in sarcoma genes contribute to <5% of cases. The rapidly developing investigative techniques available to geneticists such as genome-wide association studies, whole-exome sequencing and, soon, whole-genome sequencing promise to contribute to the unravelling of the genetic architecture of this complex disease and, in the future, to the development of more sensitive, accurate and effective diagnostic and treatment measures.
Collapse
Affiliation(s)
- Raffaele Ferrari
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th St. STOP 9410, Lubbock, TX 79430, USA.
| | | | | |
Collapse
|
37
|
Abstract
While the microtubule-binding capacity of the protein tau has been known for many years, new functions of tau in signaling and cytoskeletal organization have recently emerged. In this review, we highlight these functions and the potential roles of tau in neurodegenerative disease. We also discuss the therapeutic potential of drugs targeting various aspects of tau biology.
Collapse
Affiliation(s)
- Meaghan Morris
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|