1
|
Fischer I, Connors T, Bouyer J, Jin Y. The unique properties of Big tau in the visual system. Cytoskeleton (Hoboken) 2024; 81:488-499. [PMID: 38761116 DOI: 10.1002/cm.21875] [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: 02/06/2024] [Revised: 03/29/2024] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
Tau is a microtubule associated protein that plays important roles in regulating the properties of microtubules and axonal transport, as well as tauopathies associated with toxic aggregates leading to neurodegenerative diseases. It is encoded by the MAPT gene forming multiple isoforms (45-60 kDa) by alternative splicing which are developmentally regulated. The high molecular weight (MW) tau isoform of 105 kDa, termed Big tau, was originally discovered in the peripheral nervous system (PNS) but later found in selective CNS areas. It contains an additional large exon 4a generating a long projecting domain of about 250 amino acids. Here we investigated the properties of Big tau in the visual system of rats, its distribution in retinal ganglion cells and the optic nerve as well as its developmental regulation using biochemical, molecular and histological analyses. We discovered that Big tau is expresses as a 95 kDa protein (termed middle MW) containing exons 4a, 6 as well as exon 10 which defines a 4 microtubule-binding repeats (4R). It lacks exons 2/3 but shares the extensive phosphorylation characteristic of other tau isoforms. Importantly, early in development the visual system expresses only the low MW isoform (3R) switching to both the low and middle MW isoforms (4R) in adult retinal ganglion neurons and their corresponding axons. This is a unique structure and expression pattern of Big tau, which we hypothesize is associated with the specific properties of the visual system different from what has been previously described in the PNS and other areas of the nervous system.
Collapse
Affiliation(s)
- Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Theresa Connors
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Julien Bouyer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Ying Jin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Neven J, Issayama LK, Dewachter I, Wilson DM. Genomic stress and impaired DNA repair in Alzheimer disease. DNA Repair (Amst) 2024; 139:103678. [PMID: 38669748 DOI: 10.1016/j.dnarep.2024.103678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024]
Abstract
Alzheimer disease (AD) is the most prominent form of dementia and has received considerable attention due to its growing burden on economic, healthcare and basic societal infrastructures. The two major neuropathological hallmarks of AD, i.e., extracellular amyloid beta (Aβ) peptide plaques and intracellular hyperphosphorylated Tau neurofibrillary tangles, have been the focus of much research, with an eye on understanding underlying disease mechanisms and identifying novel therapeutic avenues. One often overlooked aspect of AD is how Aβ and Tau may, through indirect and direct mechanisms, affect genome integrity. Herein, we review evidence that Aβ and Tau abnormalities induce excessive genomic stress and impair genome maintenance mechanisms, events that can promote DNA damage-induced neuronal cell loss and associated brain atrophy.
Collapse
Affiliation(s)
- Jolien Neven
- Hasselt University, Biomedical Research Institute, BIOMED, Hasselt 3500, Belgium
| | - Luidy Kazuo Issayama
- Hasselt University, Biomedical Research Institute, BIOMED, Hasselt 3500, Belgium
| | - Ilse Dewachter
- Hasselt University, Biomedical Research Institute, BIOMED, Hasselt 3500, Belgium
| | - David M Wilson
- Hasselt University, Biomedical Research Institute, BIOMED, Hasselt 3500, Belgium.
| |
Collapse
|
3
|
Wang B, Pan X, Teng IT, Li X, Kobeissy F, Wu ZY, Zhu J, Cai G, Yan H, Yan X, Liang M, Yu F, Lu J, Yang Z, Biondi E, Haskins W, Cao YC, Benner SA, Tan W, Wang KK. Functional Selection of Tau Oligomerization-Inhibiting Aptamers. Angew Chem Int Ed Engl 2024; 63:e202402007. [PMID: 38407551 DOI: 10.1002/anie.202402007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
Pathological hyperphosphorylation and aggregation of microtubule-associated Tau protein contribute to Alzheimer's Disease (AD) and other related tauopathies. Currently, no cure exists for Alzheimer's Disease. Aptamers offer significant potential as next-generation therapeutics in biotechnology and the treatment of neurological disorders. Traditional aptamer selection methods for Tau protein focus on binding affinity rather than interference with pathological Tau. In this study, we developed a new selection strategy to enrich DNA aptamers that bind to surviving monomeric Tau protein under conditions that would typically promote Tau aggregation. Employing this approach, we identified a set of aptamer candidates. Notably, BW1c demonstrates a high binding affinity (Kd=6.6 nM) to Tau protein and effectively inhibits arachidonic acid (AA)-induced Tau protein oligomerization and aggregation. Additionally, it inhibits GSK3β-mediated Tau hyperphosphorylation in cell-free systems and okadaic acid-mediated Tau hyperphosphorylation in cellular milieu. Lastly, retro-orbital injection of BW1c tau aptamer shows the ability to cross the blood brain barrier and gain access to neuronal cell body. Through further refinement and development, these Tau aptamers may pave the way for a first-in-class neurotherapeutic to mitigate tauopathy-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Bang Wang
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
- Foundation for Applied Molecular Evolution, Firebird Biomolecular Sciences LLC, 13709 Progress Boulevard, No. 7, Alachua, FL 32615, USA
| | - Xiaoshu Pan
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
| | - I-Ting Teng
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
| | - Xiaowei Li
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
| | - Firas Kobeissy
- Center for Neurotrauma, Multiomics & Biomarkers, Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310-1458, (USA). Department of Emergency Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Zo-Yu Wu
- Center for Neurotrauma, Multiomics & Biomarkers, Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310-1458, (USA). Department of Emergency Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Jiepei Zhu
- Center for Neurotrauma, Multiomics & Biomarkers, Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310-1458, (USA). Department of Emergency Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Guangzheng Cai
- Center for Neurotrauma, Multiomics & Biomarkers, Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310-1458, (USA). Department of Emergency Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - He Yan
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
| | - Xin Yan
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
| | - Mingwei Liang
- Department of Biochemistry and Molecular Biology, UF Health Cancer Center, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Fahong Yu
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
| | - Jianrong Lu
- Department of Biochemistry and Molecular Biology, UF Health Cancer Center, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Zunyi Yang
- Foundation for Applied Molecular Evolution, Firebird Biomolecular Sciences LLC, 13709 Progress Boulevard, No. 7, Alachua, FL 32615, USA
| | - Elisa Biondi
- Foundation for Applied Molecular Evolution, Firebird Biomolecular Sciences LLC, 13709 Progress Boulevard, No. 7, Alachua, FL 32615, USA
| | - William Haskins
- Gryphon Bio, Inc., 611 Gateway Blvd. Suite 120 #253, South San Francisco, CA 94080-7066, USA
| | - Y Charles Cao
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
| | - Steven A Benner
- Foundation for Applied Molecular Evolution, Firebird Biomolecular Sciences LLC, 13709 Progress Boulevard, No. 7, Alachua, FL 32615, USA
| | - Weihong Tan
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32611, USA
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Kevin K Wang
- Center for Neurotrauma, Multiomics & Biomarkers, Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310-1458, (USA). Department of Emergency Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
- Gryphon Bio, Inc., 611 Gateway Blvd. Suite 120 #253, South San Francisco, CA 94080-7066, USA
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608, USA
| |
Collapse
|
4
|
James LM, Strickland Z, Lopez N, Whited JL, Maden M, Lewis J. Identification and Analysis of Axolotl Homologs for Proteins Implicated in Human Neurodegenerative Proteinopathies. Genes (Basel) 2024; 15:310. [PMID: 38540368 PMCID: PMC10969905 DOI: 10.3390/genes15030310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 06/14/2024] Open
Abstract
Neurodegenerative proteinopathies such as Alzheimer's Disease are characterized by abnormal protein aggregation and neurodegeneration. Neuroresilience or regenerative strategies to prevent neurodegeneration, preserve function, or restore lost neurons may have the potential to combat human proteinopathies; however, the adult human brain possesses a limited capacity to replace lost neurons. In contrast, axolotls (Ambystoma mexicanum) show robust brain regeneration. To determine whether axolotls may help identify potential neuroresilience or regenerative strategies in humans, we first interrogated whether axolotls express putative proteins homologous to human proteins associated with neurodegenerative diseases. We compared the homology between human and axolotl proteins implicated in human proteinopathies and found that axolotls encode proteins highly similar to human microtubule-binding protein tau (tau), amyloid precursor protein (APP), and β-secretase 1 (BACE1), which are critically involved in human proteinopathies like Alzheimer's Disease. We then tested monoclonal Tau and BACE1 antibodies previously used in human and rodent neurodegenerative disease studies using immunohistochemistry and western blotting to validate the homology for these proteins. These studies suggest that axolotls may prove useful in studying the role of these proteins in disease within the context of neuroresilience and repair.
Collapse
Affiliation(s)
- Lucas M. James
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; (L.M.J.); (Z.S.)
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Zachary Strickland
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; (L.M.J.); (Z.S.)
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Noah Lopez
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- The Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jessica L. Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- The Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Malcolm Maden
- Department of Biology and UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Jada Lewis
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; (L.M.J.); (Z.S.)
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
5
|
Grandizoli Saletti P, Casillas-Espinosa PM, Panagiotis Lisgaras C, Bi Mowrey W, Li Q, Liu W, Brady RD, Ali I, Silva J, Yamakawa G, Hudson M, Li C, Braine EL, Coles L, Cloyd JC, Jones NC, Shultz SR, Moshé SL, O'Brien TJ, Galanopoulou AS. Tau Phosphorylation Patterns in the Rat Cerebral Cortex After Traumatic Brain Injury and Sodium Selenate Effects: An Epibios4rx Project 2 Study. J Neurotrauma 2024; 41:222-243. [PMID: 36950806 PMCID: PMC11079442 DOI: 10.1089/neu.2022.0219] [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] [Indexed: 03/24/2023] Open
Abstract
Sodium selenate (SS) activates protein phosphatase 2 (PP2A) and reduces phosphorylated tau (pTAU) and late post-traumatic seizures after lateral fluid percussion injury (LFPI). In EpiBioS4Rx Project 2, a multi-center international study for post-traumatic targets, biomarkers, and treatments, we tested the target relevance and modification by SS of pTAU forms and PP2A and in the LFPI model, at two sites: Einstein and Melbourne. In Experiment 1, adult male rats were assigned to LFPI and sham (both sites) and naïve controls (Einstein). Motor function was monitored by neuroscores. Brains were studied with immunohistochemistry (IHC), Western blots (WBs), or PP2A activity assay, from 2 days to 8 weeks post-operatively. In Experiment 2, LFPI rats received SS for 7 days (SS0.33: 0.33 mg/kg/day; SS1: 1 mg/kg/day, subcutaneously) or vehicle (Veh) post-LFPI and pTAU, PR55 expression, or PP2A activity were studied at 2 days and 1 week (on treatment), or 2 weeks (1 week off treatment). Plasma selenium and SS levels were measured. In Experiment 1 IHC, LFPI rats had higher cortical pTAU-Ser202/Thr205-immunoreactivity (AT8-ir) and pTAU-Ser199/202-ir at 2 days, and pTAU-Thr231-ir (AT180-ir) at 2 days, 2 weeks, and 8 weeks, ipsilaterally to LFPI, than controls. LFPI-2d rats also had higher AT8/total-TAU5-ir in cortical extracts ipsilateral to the lesion (WB). PP2A (PR55-ir) showed time- and region-dependent changes in IHC, but not in WB. PP2A activity was lower in LFPI-1wk than in sham rats. In Experiment 2, SS did not affect neuroscores or cellular AT8-ir, AT180-ir, or PR55-ir in IHC. In WB, total cortical AT8/total-TAU-ir was lower in SS0.33 and SS1 LFPI rats than in Veh rats (2 days, 1 week); total cortical PR55-ir (WB) and PP2A activity were higher in SS1 than Veh rats (2 days). SS dose dependently increased plasma selenium and SS levels. Concordant across-sites data confirm time and pTAU form-specific cortical increases ipsilateral to LFPI. The discordant SS effects may either suggest SS-induced reduction in the numbers of cells with increased pTAU-ir, need for longer treatment, or the involvement of other mechanisms of action.
Collapse
Affiliation(s)
- Patricia Grandizoli Saletti
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Pablo M. Casillas-Espinosa
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Christos Panagiotis Lisgaras
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Wenzhu Bi Mowrey
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx New York, USA
| | - Qianyun Li
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Wei Liu
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Rhys D. Brady
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Idrish Ali
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Juliana Silva
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Glenn Yamakawa
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Matt Hudson
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Crystal Li
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Emma L. Braine
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Lisa Coles
- University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - James C. Cloyd
- University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx New York, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx New York, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx New York, USA
| | - Terence J. O'Brien
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx New York, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx New York, USA
| |
Collapse
|
6
|
Vidal-Palencia L, Font C, Rebollada-Merino A, Santpere G, Andrés-Benito P, Ferrer I, Pumarola M. Primary Feline Tauopathy: Clinical, Morphological, Immunohistochemical, and Genetic Studies. Animals (Basel) 2023; 13:2985. [PMID: 37760385 PMCID: PMC10525166 DOI: 10.3390/ani13182985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Tauopathies are a group of neurodegenerative diseases characterized by the pathological aggregation of hyperphosphorylated tau in neurons and glia. Primary tauopathies are not uncommon in humans but exceptional in other species. We evaluate the clinical, neuropathological, and genetic alterations related to tau pathology in 16 cats aged from 1 to 21 years with different clinical backgrounds. Interestingly, a 10-year-old female cat presented a six-year progressive history of mental status and gait abnormalities. The imaging study revealed generalized cortical atrophy. Due to the poor prognosis, the cat was euthanatized at the age of ten. Neuropathological lesions were characterized by massive neuronal loss with marked spongiosis and associated moderate reactive gliosis in the parietal cortex, being less severe in other areas of the cerebral cortex, and the loss of Purkinje cells of the cerebellum. Immunohistochemical methods revealed a 4R-tauopathy with granular pre-tangles in neurons and coiled bodies in oligodendrocytes. Deposits were recognized with several phospho-site antibodies (4Rtau, tau5, AT8, PFH, tau-P Thr181, tau-P-Ser 262, tau-P Ser 422) and associated with increased granular expression of active tau kinases (p38-P Thr180/Tyr182 and SAPK/JNK-P Thr138/Thr185). The genetic study revealed well-preserved coding regions of MAPT. No similar alterations related to tau pathology were found in the other 15 cats processed in parallel. To our knowledge, this is the first case reporting a primary 4R-tauopathy with severe cerebral and Purkinje cell degeneration in an adult cat with neurological signs starting at a young age.
Collapse
Affiliation(s)
- Laura Vidal-Palencia
- Hospital del Mar Research Institute, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain;
- Unitat de Patologia Murina i Comparada, Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Campus UAB, Travessera dels Turons s/n, 08193 Barcelona, Spain;
| | - Cristina Font
- Hospital Veterinari Canis, Can Pau Birol, 38, 17006 Girona, Spain;
| | - Agustín Rebollada-Merino
- VISAVET Health Surveillance Centre, Complutense University of Madrid, 28040 Madrid, Spain;
- Department of Internal Medicine and Animal Surgery, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Gabriel Santpere
- Hospital del Mar Research Institute, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain;
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Pol Andrés-Benito
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain; (P.A.-B.); (I.F.)
| | - Isidro Ferrer
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain; (P.A.-B.); (I.F.)
- Department of Pathology and Experimental Therapeutics, University of Barcelona, 08007 Barcelona, Spain
| | - Martí Pumarola
- Unitat de Patologia Murina i Comparada, Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Campus UAB, Travessera dels Turons s/n, 08193 Barcelona, Spain;
| |
Collapse
|
7
|
Gambardella JC, Schoephoerster W, Bondarenko V, Yandell BS, Emborg ME. Expression of tau and phosphorylated tau in the brain of normal and hemiparkinsonian rhesus macaques. J Comp Neurol 2023; 531:1198-1216. [PMID: 37098996 PMCID: PMC10247506 DOI: 10.1002/cne.25490] [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: 11/19/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/27/2023]
Abstract
Tau is a neuronal protein involved in microtubule stabilization and intracellular vesicle transport in axons. In neurodegenerative disorders termed "tauopathies," like Alzheimer's and Parkinson's disease, tau becomes hyperphosphorylated and forms intracellular inclusions. Rhesus macaques are widely used for studying ageing processes and modeling neurodegenerative disorders, yet little is known about endogenous tau expression in their brains. In this study, immunohistochemical methods were used to map and characterize total tau, 3R- and 4R-tau isoforms, and phosphorylated tau (pThr231-tau and pSer202/Thr205-tau/AT8) expression bilaterally in 16 brain regions of normal and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced hemiparkinsonian adult rhesus macaques. Tau-immunoreactivity (-ir), including both 3R and 4R isoforms, was observed throughout the brain, with varying regional intensities. The anterior cingulate cortex, entorhinal cortex, and hippocampus displayed the most robust tau-ir, while the subthalamic nucleus and white matter regions had minimal expression. Tau was present in neurons of gray matter regions; it was preferentially observed in fibers of the globus pallidus and substantia nigra and in cell bodies of the thalamus and subthalamic nucleus. In white matter regions, tau was abundantly present in oligodendrocytes. Additionally, neuronal pThr231-tau-ir was abundant in all brain regions, but not AT8-ir. Differences in regional and intracellular protein expression were not detected between control subjects and both brain hemispheres of MPTP-treated animals. Specifically, tau-ir in the substantia nigra of all subjects colocalized with GABAergic neurons. Overall, this report provides an in-depth characterization of tau expression in the rhesus macaque brain to facilitate future investigations for understanding and modeling tau pathology in this species.
Collapse
Affiliation(s)
- Julia C. Gambardella
- Preclinical Parkinson’s Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison
| | - Wyatt Schoephoerster
- Preclinical Parkinson’s Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison
| | - Viktoriya Bondarenko
- Preclinical Parkinson’s Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison
| | | | - Marina E. Emborg
- Preclinical Parkinson’s Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison
- Department of Medical Physics, University of Wisconsin-Madison
| |
Collapse
|
8
|
Zhang S, Dong H, Bian J, Li D, Liu C. Targeting amyloid proteins for clinical diagnosis of neurodegenerative diseases. FUNDAMENTAL RESEARCH 2023; 3:505-519. [PMID: 38933553 PMCID: PMC11197785 DOI: 10.1016/j.fmre.2022.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Abnormal aggregation and accumulation of pathological amyloid proteins such as amyloid-β, Tau, and α-synuclein play key pathological roles and serve as histological hallmarks in different neurodegenerative diseases (NDs) such as Alzheimer's disease (AD) and Parkinson's disease (PD). In addition, various post-translational modifications (PTMs) have been identified on pathological amyloid proteins and are subjected to change during disease progression. Given the central role of amyloid proteins in NDs, tremendous efforts have been made to develop amyloid-targeting strategies for clinical diagnosis and molecular classification of NDs. In this review, we summarize two major strategies for targeting amyloid aggregates, with a focus on the trials in AD diagnosis. The first strategy is a positron emission tomography (PET) scan of protein aggregation in the brain. We mainly focus on introducing the development of small-molecule PET tracers for specifically recognizing pathological amyloid fibrils. The second strategy is the detection of PTM biomarkers on amyloid proteins in cerebrospinal fluid and plasma. We discuss the pathological roles of different PTMs in diseases and how we can use the PTM profile of amyloid proteins for clinical diagnosis. Finally, we point out the potential technical challenges of these two strategies, and outline other potential strategies, as well as a combination of multiple strategies, for molecular diagnosis of NDs.
Collapse
Affiliation(s)
- Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hui Dong
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang Bian
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
9
|
Li P, Chen J, Wang X, Su Z, Gao M, Huang Y. Liquid - liquid phase separation of tau: Driving forces, regulation, and biological implications. Neurobiol Dis 2023; 183:106167. [PMID: 37230179 DOI: 10.1016/j.nbd.2023.106167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/16/2023] [Accepted: 05/21/2023] [Indexed: 05/27/2023] Open
Abstract
The past 15 years have witnessed an explosion in the studies of biomolecular condensates that are implicated in numerous biological processes and play vital roles in human health and diseases. Recent findings demonstrate that the microtubule-associated protein tau forms liquid condensates through liquid-liquid phase separation (LLPS) in in vitro experiments using purified recombinant proteins and cell-based experiments. Although in vivo studies are lacking, liquid condensates have emerged as an important assembly state of physiological and pathological tau and LLPS can regulate the function of microtubules, mediate stress granule formation, and accelerate tau amyloid aggregation. In this review, we summarize recent advances in tau LLPS, aiming to unveiling the delicate interactions driving tau LLPS. We further discuss the association of tau LLPS with physiology and disease in the context of the sophisticated regulation of tau LLPS. Deciphering the mechanisms underlying tau LLPS and the liquid-to-solid transition enables rational design of molecules that inhibit or delay the formation of tau solid species, thus providing novel targeted therapeutic strategies for tauopathies.
Collapse
Affiliation(s)
- Ping Li
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Jingxin Chen
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Xi Wang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Zhengding Su
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Meng Gao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China.
| | - Yongqi Huang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China.
| |
Collapse
|
10
|
Rahman MM, Islam MR, Alam Tumpa MA, Shohag S, Shakil Khan Shuvo, Ferdous J, Kajol SA, Aljohani ASM, Al Abdulmonem W, Rauf A, Thiruvengadam M. Insights into the promising prospect of medicinal chemistry studies against neurodegenerative disorders. Chem Biol Interact 2023; 373:110375. [PMID: 36739931 DOI: 10.1016/j.cbi.2023.110375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/06/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Medicinal chemistry is an interdisciplinary field that incorporates organic chemistry, biochemistry, physical chemistry, pharmacology, informatics, molecular biology, structural biology, cell biology, and other disciplines. Additionally, it considers molecular factors such as the mode of action of the drugs, their chemical structure-activity relationship (SAR), and pharmacokinetic aspects like absorption, distribution, metabolism, elimination, and toxicity. Neurodegenerative disorders (NDs), which are defined by the breakdown of neurons over time, are affecting an increasing number of people. Oxidative stress, particularly the increased production of Reactive Oxygen Species (ROS), plays a crucial role in the growth of various disorders, as indicated by the identification of protein, lipid, and Deoxyribonucleic acid (DNA) oxidation products in vivo. Because of their inherent nature, most biological molecules are vulnerable to ROS, even if they play a role in metabolic parameters and cell signaling. Due to their high polyunsaturated fatty acid content, low antioxidant barrier, and high oxygen uptake, neurons are particularly vulnerable to oxidation by nature. As a result, excessive ROS generation in neurons looks especially harmful, and the mechanisms associated with biomolecule oxidative destruction are several and complex. This review focuses on the formation and management of ROS, as well as their chemical characteristics (both thermodynamic and kinetic), interactions, and implications in NDs.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Mst Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Sheikh Shohag
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University Buraydah, 52571, Saudi Arabia
| | - Shakil Khan Shuvo
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Jannatul Ferdous
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Saima Akter Kajol
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University Buraydah, 52571, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine Qassim University, Buraydah, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, 23430, Khyber Pakhtunkhwa (KP), Pakistan.
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Sciences, Konkuk University, Seoul, 05029, South Korea; Department of Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| |
Collapse
|
11
|
Nicastro N, Nencha U, Burkhard PR, Garibotto V. Dopaminergic imaging in degenerative parkinsonisms, an established clinical diagnostic tool. J Neurochem 2023; 164:346-363. [PMID: 34935143 DOI: 10.1111/jnc.15561] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) and other neurodegenerative parkinsonisms are characterised by loss of striatal dopaminergic neurons. Dopamine functional deficits can be measured in vivo using positron emission tomography (PET) and single-photon emission computed tomography (SPECT) ligands assessing either presynaptic (e.g. dopamine synthesis and storage, transporter density) or postsynaptic terminals (i.e. D2 receptors availability). Nuclear medicine imaging thus helps the clinician to separate degenerative forms of parkinsonism with other neurological conditions, e.g. essential tremor or drug-induced parkinsonism. With the present study, we aimed at summarizing the current evidence about dopaminergic molecular imaging in the diagnostic evaluation of PD, atypical parkinsonian syndromes and dementia with Lewy bodies (DLB), as well as its potential to distinguish these conditions and to estimate disease progression. In fact, PET/SPECT methods are clinically validated and have been increasingly integrated into diagnostic guidelines (e.g. for PD and DLB). In addition, there is novel evidence on the classification properties of extrastriatal signal. Finally, dopamine imaging has an outstanding potential to detect neurodegeneration at the premotor stage, including REM-sleep behavior disorder and olfactory loss. Therefore, inclusion of subjects at an early stage for clinical trials can largely benefit from a validated in vivo biomarker such as presynaptic dopamine pathways PET/SPECT assessment.
Collapse
Affiliation(s)
- Nicolas Nicastro
- Division of Neurorehabilitation, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Umberto Nencha
- Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre R Burkhard
- Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Valentina Garibotto
- Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
12
|
Jin Y, Connors T, Bouyer J, Fischer I. Regulation of Tau Expression in Superior Cervical Ganglion (SCG) Neurons In Vivo and In Vitro. Cells 2023; 12:cells12020226. [PMID: 36672160 PMCID: PMC9856632 DOI: 10.3390/cells12020226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
The superior cervical ganglion (SCG) is part of the autonomic nervous system providing sympathetic innervation to the head and neck, and has been regularly used to prepare postnatal neuronal cultures for cell biological studies. We found that during development these neurons change tau expression from the low molecular weight (LMW) isoforms to Big tau, with the potential to affect functions associated with tau such as microtubule dynamic and axonal transport. Big tau contains the large 4a exon that transforms tau from LMW isoforms of 45-60 kDa to 110 kDa. We describe tau expression during postnatal development reporting that the transition from LMW tau to Big tau which started at late embryonic stages is completed by about 4-5 weeks postnatally. We confirmed the presence of Big tau in dissociated postnatal SCG neurons making them an ideal system to study the function of Big tau in neurons. We used SCG explants to examine the response of SCG neurons to lesion and found that Big tau expression returned gradually along the regrowing neurites suggesting that it does not drives regeneration, but facilitates the structure/function of mature SCG neurons. The structural/functional roles of Big tau remain unknown, but it is intriguing that neurons that express Big tau appear less vulnerable to tauopathies.
Collapse
|
13
|
Abstract
Tauopathies are a clinically and neuropathologically heterogeneous group of neurodegenerative disorders, characterized by abnormal tau aggregates. Tau, a microtubule-associated protein, is important for cytoskeletal structure and intracellular transport. Aberrant posttranslational modification of tau results in abnormal tau aggregates causing neurodegeneration. Tauopathies may be primary, or secondary, where a second protein, such as Aß, is necessary for pathology, for example, in Alzheimer's disease, the most common tauopathy. Primary tauopathies are classified based on tau isoform and cell types where pathology predominates. Primary tauopathies include Pick disease, corticobasal degeneration, progressive supranuclear palsy, and argyrophilic grain disease. Environmental tauopathies include chronic traumatic encephalopathy and geographically isolated tauopathies such as the Guam-Parkinsonian-dementia complex. The clinical presentation of tauopathies varies based on the brain areas affected, generally presenting with a combination of cognitive and motor symptoms either earlier or later in the disease course. As symptoms overlap and tauopathies such as Alzheimer's disease and argyrophilic grain disease often coexist, accurate clinical diagnosis is challenging when biomarkers are unavailable. Available treatments target cognitive, motor, and behavioral symptoms. Disease-modifying therapies have been the focus of drug development, particularly agents targeting Aß and tau pathology in Alzheimer's disease, although most of these trials have failed.
Collapse
Affiliation(s)
- Gayatri Devi
- Department of Neurology and Psychiatry, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.
| |
Collapse
|
14
|
Szénási T, Turu G, Hunyady L. Interactions between β-arrestin proteins and the cytoskeletal system, and their relevance to neurodegenerative disorders. Front Endocrinol (Lausanne) 2023; 14:957981. [PMID: 36843600 PMCID: PMC9947276 DOI: 10.3389/fendo.2023.957981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/04/2023] [Indexed: 02/11/2023] Open
Abstract
β-arrestins, which have multiple cellular functions, were initially described as proteins that desensitize rhodopsin and other G protein-coupled receptors. The cytoskeletal system plays a role in various cellular processes, including intracellular transport, cell division, organization of organelles, and cell cycle. The interactome of β-arrestins includes the major proteins of the three main cytoskeletal systems: tubulins for microtubules, actins for the actin filaments, and vimentin for intermediate filaments. β-arrestins bind to microtubules and regulate their activity by recruiting signaling proteins and interacting with assembly proteins that regulate the actin cytoskeleton and the intermediate filaments. Altered regulation of the cytoskeletal system plays an essential role in the development of Alzheimer's, Parkinson's and other neurodegenerative diseases. Thus, β-arrestins, which interact with the cytoskeleton, were implicated in the pathogenesis progression of these diseases and are potential targets for the treatment of neurodegenerative disorders in the future.
Collapse
Affiliation(s)
- Tibor Szénási
- Institute of Enzymology, Research Center for Natural Sciences, Centre of Excellence of the Hungarian Academy of Sciences, Budapest, Hungary
| | - Gábor Turu
- Institute of Enzymology, Research Center for Natural Sciences, Centre of Excellence of the Hungarian Academy of Sciences, Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - László Hunyady
- Institute of Enzymology, Research Center for Natural Sciences, Centre of Excellence of the Hungarian Academy of Sciences, Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- *Correspondence: László Hunyady,
| |
Collapse
|
15
|
Driscoll I, Ma Y, Lose SR, Gallagher CL, Johnson SC, Asthana S, Hermann BP, Sager MA, Blennow K, Zetterberg H, Carlsson CM, Engelman CD, Dubal DB, Okonkwo OC. AD-associated CSF biomolecular changes are attenuated in KL-VS heterozygotes. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12383. [PMID: 36505396 PMCID: PMC9728548 DOI: 10.1002/dad2.12383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022]
Abstract
Introduction Dementia as an inevitable aging consequence has been challenged and underscores the need for investigations of the factors that confer resilience. We examine whether the functionally advantageous KL-VS variant of the putative aging suppressor KLOTHO gene attenuates age-related cognitive decline and deleterious biomolecular changes. Methods Trajectories of change in memory and executive function (N = 360; 2-12 visits) and cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers-amyloid beta (Aβ)42, total tau (t-tau), phosphorylated tau (p-tau) (N = 112; 2-4 samplings)-were compared between KL-VS non-carriers and heterozygotes in middle-aged and older adults from the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center studies. Results Memory and executive function declined (p's≤ 0.001) and CSF t-tau, p-tau, t-tau/Aβ42, and p-tau/Aβ42 levels increased (all p's≤ 0.004) with age. The rate of p-tau accumulation was attenuated for KL-VS heterozygotes (p = 0.03). Discussion KL-VS heterozygosity may confer resilience to AD-associated biomolecular changes.
Collapse
Affiliation(s)
- Ira Driscoll
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Department of PsychologyUniversity of Wisconsin‐MilwaukeeMilwaukeeWisconsinUSA
| | - Yue Ma
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
| | - Sarah R. Lose
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
| | - Catherine L. Gallagher
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | - Bruce P. Hermann
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Mark A. Sager
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGöteborgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGöteborgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | - Corinne D. Engelman
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Departments of Population Health SciencesUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Dena B. Dubal
- Department of Neurology and Weill Institute for NeurosciencesUniversity of CaliforniaCaliforniaSan FranciscoUSA
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| |
Collapse
|
16
|
Fischer I. Evolutionary perspective of Big tau structure: 4a exon variants of MAPT. Front Mol Neurosci 2022; 15:1019999. [PMID: 36533137 PMCID: PMC9755724 DOI: 10.3389/fnmol.2022.1019999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/17/2022] [Indexed: 08/15/2023] Open
Abstract
The MAPT gene encoding the microtubule-associated protein tau can generate multiple isoforms by alternative splicing giving rise to proteins which are differentially expressed in specific areas of the nervous system and at different developmental stages. Tau plays important roles in modulating microtubule dynamics, axonal transport, synaptic plasticity, and DNA repair, and has also been associated with neurodegenerative diseases (tauopathies) including Alzheimer's disease and frontotemporal dementia. A unique high-molecular-weight isoform of tau, originally found to be expressed in the peripheral nervous system and projecting neurons, has been termed Big tau and has been shown to uniquely contain the large exon 4a that significantly increases the size and 3D structure of tau. With little progress since the original discovery of Big tau, more than 25 years ago, we have now completed a comprehensive comparative study to analyze the structure of the MAPT gene against available databases with respect to the composition of the tau exons as they evolved from early vertebrates to primates and human. We focused the analysis on the evolution of the 4a exon variants and their homology relative to humans. We discovered that the 4a exon defining Big tau appears to be present early in vertebrate evolution as a large insert that dramatically changed the size of the tau protein with low sequence conservation despite a stable size range of about 250aa, and in some species a larger 4a-L exon of 355aa. We suggest that 4a exon variants evolved independently in different species by an exonization process using new alternative splicing to address the growing complexities of the evolving nervous systems. Thus, the appearance of a significantly larger isoform of tau independently repeated itself multiple times during evolution, accentuating the need across vertebrate species for an elongated domain that likely endows Big tau with novel physiological functions as well as properties related to neurodegeneration.
Collapse
Affiliation(s)
- Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
17
|
Karimi N, Bayram Çatak F, Arslan E, Saghazadeh A, Rezaei N. Tau immunotherapy in Alzheimer’s disease and progressive supranuclear palsy. Int Immunopharmacol 2022; 113:109445. [DOI: 10.1016/j.intimp.2022.109445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
|
18
|
Luzzi S, Cherubini V, Falsetti L, Viticchi G, Silvestrini M, Toraldo A. Homocysteine, Cognitive Functions, and Degenerative Dementias: State of the Art. Biomedicines 2022; 10:2741. [PMID: 36359260 PMCID: PMC9687733 DOI: 10.3390/biomedicines10112741] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 07/30/2023] Open
Abstract
There is strong evidence that homocysteine is a risk factor not only for cerebrovascular diseases but also for degenerative dementias. A recent consensus statement renewed the importance and the role of high levels of homocysteine in cognitive decline in several forms of degenerative dementia, such as Alzheimer's disease. Although the molecular mechanisms by which homocysteine causes cell dysfunction are known, both the impact of homocysteine on specific cognitive functions and the relationship between homocysteine level and non-Alzheimer dementias have been poorly investigated. Most of the studies addressing the impact of hyperhomocysteinemia on dementias have not examined the profile of performance across different cognitive domains, and have only relied on screening tests, which provide a very general and coarse-grained picture of the cognitive status of the patients. Yet, trying to understand whether hyperhomocysteinemia is associated with the impairment of specific cognitive functions would be crucial, as it would be, in parallel, learning whether some brain circuits are particularly susceptible to the damage caused by hyperhomocysteinemia. These steps would allow one to (i) understand the actual role of homocysteine in the pathogenesis of cognitive decline and (ii) improve the diagnostic accuracy, differential diagnosis and prognostic implications. This review is aimed at exploring and revising the state of the art of these two strictly related domains. Suggestions for future research are provided.
Collapse
Affiliation(s)
- Simona Luzzi
- Neurology Unit, Department of Experimental and Clinical Medicine, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Veronica Cherubini
- Neurology Unit, Department of Experimental and Clinical Medicine, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Lorenzo Falsetti
- Internal and Subintensive Medicine Department, Azienda Ospedaliero-Universitaria “Ospedali Riuniti” di Ancona, 60126 Ancona, Italy
| | - Giovanna Viticchi
- Neurology Unit, Azienda Ospedaliero-Universitaria “Ospedali Riuniti” di Ancona, 60126 Ancona, Italy
| | - Mauro Silvestrini
- Neurology Unit, Department of Experimental and Clinical Medicine, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Alessio Toraldo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
- Milan Center for Neuroscience (NeuroMI), 20126 Milan, Italy
| |
Collapse
|
19
|
Shilian M, Even A, Gast H, Nguyen L, Weil M. Elongator promotes neuritogenesis via regulation of tau stability through acly activity. Front Cell Dev Biol 2022; 10:1015125. [PMID: 36393857 PMCID: PMC9644021 DOI: 10.3389/fcell.2022.1015125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/05/2022] [Indexed: 11/23/2022] Open
Abstract
The six subunits (Elp1 to Elp6) Elongator complex promotes specific uridine modifications in tRNA’s wobble site. Moreover, this complex has been indirectly involved in the regulation of α-tubulin acetylation in microtubules (MTs) via the stabilization of ATP-Citrate Lyase (Acly), the main cytosolic source of acetyl-CoA production in cells, a key substrate used for global protein acetylation. Here, we report additional evidence that Elongator activity is important for proper cytoskeleton remodeling as cells lacking expression of Elp1 show morphology impairment; including distinct neurite process formation and disorganization and instability of MTs. Here, we show that loss of Elongator results in a reduction of expression of the microtubule associated protein Tau (MAPT). Tau, is a well-known key MT regulator in neurons whose lysines can be competitively acetylated or ubiquitylated. Therefore, we tested whether Tau is an indirect acetylation target of Elongator. We found that a reduction of Elongator activity leads to a decrease of lysine acetylation on Tau that favors its proteasomal degradation. This phenotype was prevented by using selective deacetylase or proteasomal inhibitors. Moreover, our data demonstrate that Acly’s activity regulates the mechanism underlying Tau mediated neurite morphology defects found in Elp1 KD since both Tau levels and neurites morphology are restored due to Acly overexpression. This suggests a possible involvement of both Tau and Acly dysfunction in Familial Dysautonomia (FD), which is an autosomal recessive peripheral neuropathy caused by mutation in the ELP1 gene that severely affects Elp1 expression levels in the nervous system in FD patients in a similar way as found previously in Elp1 KD neuroblastoma cells.
Collapse
Affiliation(s)
- Michal Shilian
- Laboratory for Neurodegenerative Diseases and Personalized Medicine, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Aviel Even
- Laboratory for Neurodegenerative Diseases and Personalized Medicine, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Hila Gast
- Laboratory for Neurodegenerative Diseases and Personalized Medicine, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Laurent Nguyen
- GIGA-Stem Cells and GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGAR), University of Liège, C.H.U. Sart Tilman, Belgium, BIOMED Research Institute, Hasselt, Belgium
| | - Miguel Weil
- Laboratory for Neurodegenerative Diseases and Personalized Medicine, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Miguel Weil,
| |
Collapse
|
20
|
Natale C, Barzago MM, Colnaghi L, De Luigi A, Orsini F, Fioriti L, Diomede L. A Combined Cell-Worm Approach to Search for Compounds Counteracting the Toxicity of Tau Oligomers In Vivo. Int J Mol Sci 2022; 23:11277. [PMID: 36232578 PMCID: PMC9569484 DOI: 10.3390/ijms231911277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
A clear relationship between the tau assemblies and toxicity has still to be established. To correlate the tau conformation with its proteotoxic effect in vivo, we developed an innovative cell-worm-based approach. HEK293 cells expressing tau P301L under a tetracycline-inducible system (HEK T-Rex) were employed to produce different tau assemblies whose proteotoxic potential was evaluated using C. elegans. Lysates from cells induced for five days significantly reduced the worm's locomotor activity. This toxic effect was not related to the total amount of tau produced by cells or to its phosphorylation state but was related to the formation of multimeric tau assemblies, particularly tetrameric ones. We investigated the applicability of this approach for testing compounds acting against oligomeric tau toxicity, using doxycycline (Doxy) as a prototype drug. Doxy affected tau solubility and promoted the disassembly of already formed toxic aggregates in lysates of cells induced for five days. These effects translated into a dose-dependent protective action in C. elegans. These findings confirm the validity of the combined HEK T-Rex cells and the C. elegans-based approach as a platform for pharmacological screening.
Collapse
Affiliation(s)
- Carmina Natale
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Maria Monica Barzago
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luca Colnaghi
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Ada De Luigi
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Franca Orsini
- Dulbecco Telethon Institute and Department of Neuroscience, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luana Fioriti
- Dulbecco Telethon Institute and Department of Neuroscience, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| |
Collapse
|
21
|
Yousef MH, Salama M, El-Fawal HAN, Abdelnaser A. Selective GSK3β Inhibition Mediates an Nrf2-Independent Anti-inflammatory Microglial Response. Mol Neurobiol 2022; 59:5591-5611. [PMID: 35739410 PMCID: PMC9395457 DOI: 10.1007/s12035-022-02923-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/10/2022] [Indexed: 12/15/2022]
Abstract
Glycogen synthase kinase 3 (GSK3) is associated with the proinflammatory phenotype of microglia and has been shown to act in concert with nuclear factor kappa B (NF-κB). GSK3 is also a suppressor of nuclear factor erythroid 2-related factor 2 (Nrf2), the principal regulator of redox homeostasis. Agreeing with the oxidative paradigm of aging, Nrf2 is often deregulated in parainflammatory and neurodegenerative diseases. In this study, we aimed to explore a multimodal disease-modifying utility of GSK3 inhibition, beyond neuronal proteopathologies. Furthermore, we aimed to underscore the difference in therapeutic value between the two GSK3 paralogs by isoform-selective chemical inhibition. The anti-inflammatory effects of paralog-selective GSK3 inhibitors were evaluated as a function of the reductive capacity of each to mitigate LPS-induced activation of SIM-A9 microglia. The Griess method was employed to detect the nitrate-lowering capacity of selective GSK3 inhibition. Real-time PCR was used to assess post-treatment expression levels of pro-inflammatory markers and antioxidant genes; pro-inflammatory cytokines were assayed by ELISA. Nuclear lysates of treated cells were examined for Nrf2 and NF-κB accumulation by immunoblotting. Finally, to infer whether the counter-inflammatory activity of GSK3 inhibition was Nrf2-dependent, DsiRNA-mediated knockdown of Nrf2 was attempted. Results from our experiments reveal a superior anti-inflammatory and anti-oxidative efficacy for GSK3β-selective inhibition, compared to GSK3α-selective and non-selective pan-inhibition; hence, use of selective GSK3β inhibitors is likely to be more propitious than non-selective dual inhibitors administered at comparable doses. Moreover, our results suggest that the anti-inflammatory effects of GSK3 inhibition are not Nrf2 dependent.
Collapse
Affiliation(s)
- Mohamed H Yousef
- School of Sciences and Engineering, Biotechnology Graduate Program, The American University in Cairo, P.O. Box: 74, Cairo, Egypt
| | - Mohamed Salama
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, Cairo, Egypt
| | - Hassan A N El-Fawal
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, Cairo, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, Cairo, Egypt.
| |
Collapse
|
22
|
Sonuç Karaboğa MN, Sezgintürk M. A practical approach for the detection of protein tau with a portable potentiostat. ELECTROANAL 2022. [DOI: 10.1002/elan.202200072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
23
|
Martinez B, Peplow PV. MicroRNA biomarkers in frontotemporal dementia and to distinguish from Alzheimer's disease and amyotrophic lateral sclerosis. Neural Regen Res 2022; 17:1412-1422. [PMID: 34916411 PMCID: PMC8771095 DOI: 10.4103/1673-5374.330591] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 11/04/2022] Open
Abstract
Frontotemporal lobar degeneration describes a group of progressive brain disorders that primarily are associated with atrophy of the prefrontal and anterior temporal lobes. Frontotemporal lobar degeneration is considered to be equivalent to frontotemporal dementia. Frontotemporal dementia is characterized by progressive impairments in behavior, executive function, and language. There are two main clinical subtypes: behavioral-variant frontotemporal dementia and primary progressive aphasia. The early diagnosis of frontotemporal dementia is critical for developing management strategies and interventions for these patients. Without validated biomarkers, the clinical diagnosis depends on recognizing all the core or necessary neuropsychiatric features, but misdiagnosis often occurs due to overlap with a range of neurologic and psychiatric disorders. In the studies reviewed a very large number of microRNAs were found to be dysregulated but with limited overlap between individual studies. Measurement of specific miRNAs singly or in combination, or as miRNA pairs (as a ratio) in blood plasma, serum, or cerebrospinal fluid enabled frontotemporal dementia to be discriminated from healthy controls, Alzheimer's disease, and amyotrophic lateral sclerosis. Furthermore, upregulation of miR-223-3p and downregulation of miR-15a-5p, which occurred both in blood serum and cerebrospinal fluid, distinguished behavioral-variant frontotemporal dementia from healthy controls. Downregulation of miR-132-3p in frontal and temporal cortical tissue distinguished frontotemporal lobar degeneration and frontotemporal dementia, respectively, from healthy controls. Possible strong miRNA biofluid biomarker contenders for behavioral-variant frontotemporal dementia are miR-223-3p, miR-15a-5p, miR-22-3p in blood serum and cerebrospinal fluid, and miR-124 in cerebrospinal fluid. No miRNAs were identified able to distinguish between behavioral-variant frontotemporal dementia and primary progressive aphasia subtypes. Further studies are warranted on investigating miRNA expression in biofluids and frontal/temporal cortical tissue to validate and extend these findings.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Medicine, St. Georges University School of Medicine, Grenada
| | - Philip V. Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
24
|
You Y, Hersh SW, Aslebagh R, Shaffer SA, Ikezu S, Mez J, Lunetta KL, Logue MW, Farrer LA, Ikezu T. Alzheimer's disease associated AKAP9 I2558M mutation alters posttranslational modification and interactome of tau and cellular functions in CRISPR-edited human neuronal cells. Aging Cell 2022; 21:e13617. [PMID: 35567427 PMCID: PMC9197405 DOI: 10.1111/acel.13617] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 12/01/2022] Open
Abstract
Alzheimer's disease (AD) is a pervasive neurodegeneration disease with high heritability. In this study, we employed CRISPR-Cas9-engineered technology to investigate the effects of a rare mutation (rs144662445) in the A kinase anchoring protein 9 (AKAP9) gene, which is associated with AD in African Americans (AA), on tau pathology and the tau interactome in SH-SY5Y P301L neuron-like cells. The mutation significantly increased the level of phosphorylated tau, specifically at the site Ser396/Ser404. Moreover, analyses of the tau interactome measured by affinity purification-mass spectrometry revealed that differentially expressed tau-interacting proteins in AKAP9 mutant cells were associated with RNA translation, RNA localization and oxidative activity, recapitulating the tau interactome signature previously reported with human AD brain samples. Importantly, these results were further validated by functional studies showing a significant reduction in protein synthesis activity and excessive oxidative stress in AKAP9 mutant compared with wild type cells in a tau-dependent manner, which are mirrored with pathological phenotype frequently seen in AD. Our results demonstrated specific effects of rs14462445 on mis-processing of tau and suggest a potential role of AKAP9 in AD pathogenesis.
Collapse
Affiliation(s)
- Yang You
- Departments of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
| | - Samuel W. Hersh
- Departments of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | - Roshanak Aslebagh
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
- Mass Spectrometry FacilityUniversity of Massachusetts Medical SchoolShrewsburyMassachusettsUSA
| | - Scott A. Shaffer
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
- Mass Spectrometry FacilityUniversity of Massachusetts Medical SchoolShrewsburyMassachusettsUSA
| | - Seiko Ikezu
- Departments of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | - Jesse Mez
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | - Kathryn L. Lunetta
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Mark W. Logue
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
- Department of PsychiatryBoston University School of MedicineBostonMassachusettsUSA
- Department of Medicine (Biomedical Genetics)Boston University School of MedicineBostonMassachusettsUSA
- National Center for PTSDBehavioral Sciences DivisionVA Boston Healthcare SystemBostonMassachusettsUSA
| | - Lindsay A. Farrer
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
- Department of Medicine (Biomedical Genetics)Boston University School of MedicineBostonMassachusettsUSA
- Department of OphthalmologyBoston University School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| | - Tsuneya Ikezu
- Departments of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
- Center for Systems NeuroscienceBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
25
|
Law AD, Cassar M, Long DM, Chow ES, Giebultowicz JM, Venkataramanan A, Strauss R, Kretzschmar D. FTD-associated mutations in Tau result in a combination of dominant and recessive phenotypes. Neurobiol Dis 2022; 170:105770. [PMID: 35588988 PMCID: PMC9261467 DOI: 10.1016/j.nbd.2022.105770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 11/26/2022] Open
Abstract
Although mutations in the microtubules-associated protein Tau have long been connected with several neurodegenerative diseases, the underlying molecular mechanisms causing these tauopathies are still not fully understood. Studies in various models suggested that dominant gain-of-function effects underlie the pathogenicity of these mutants; however, there is also evidence that the loss of normal physiological functions of Tau plays a role in tauopathies. Previous studies on Tau in Drosophila involved expressing the human Tau protein in the background of the endogenous Tau gene in addition to inducing high expression levels. To study Tau pathology in more physiological conditions, we recently created Drosophila knock-in models that express either wildtype human Tau (hTauWT) or disease-associated mutant hTau (hTauV337M and hTauK369I) in place of the endogenous Drosophila Tau (dTau). Analyzing these flies as homozygotes, we could therefore detect recessive effects of the mutations while identifying dominant effects in heterozygotes. Using memory, locomotion and sleep assays, we found that homozygous mutant hTau flies showed deficits already when quite young whereas in heterozygous flies, disease phenotypes developed with aging. Homozygotes also revealed an increase in microtubule diameter, suggesting that changes in the cytoskeleton underlie the axonal degeneration we observed in these flies. In contrast, heterozygous mutant hTau flies showed abnormal axonal targeting and no detectable changes in microtubules. However, we previously showed that heterozygosity for hTauV337M interfered with synaptic homeostasis in central pacemaker neurons and we now show that heterozygous hTauK369I flies have decreased levels of proteins involved in the release of synaptic vesicles. Taken together, our results demonstrate that both mutations induce a combination of dominant and recessive disease-related phenotypes that provide behavioral and molecular insights into the etiology of Tauopathies.
Collapse
Affiliation(s)
- Alexander D Law
- Oregon Institute of Occupational Health Sciences, 3181 S.W. Sam Jackson Park Road, Portland, OR 97219, USA
| | - Marlène Cassar
- Oregon Institute of Occupational Health Sciences, 3181 S.W. Sam Jackson Park Road, Portland, OR 97219, USA
| | - Dani M Long
- Oregon Institute of Occupational Health Sciences, 3181 S.W. Sam Jackson Park Road, Portland, OR 97219, USA
| | - Eileen S Chow
- Department of Integrative Biology, Oregon State University, Corvallis, OR 97331, USA
| | | | - Anjana Venkataramanan
- Institut für Entwicklungsbiologie und Neurobiologie, Johannes Gutenberg-Universität Mainz, Hanns-Dieter-Hüsch Weg 15, 55128 Mainz, Germany
| | - Roland Strauss
- Institut für Entwicklungsbiologie und Neurobiologie, Johannes Gutenberg-Universität Mainz, Hanns-Dieter-Hüsch Weg 15, 55128 Mainz, Germany
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, 3181 S.W. Sam Jackson Park Road, Portland, OR 97219, USA.
| |
Collapse
|
26
|
Rivas F, Poblete-Aro C, Pando ME, Allel MJ, Fernandez V, Soto A, Nova P, Garcia-Diaz D. Effects of polyphenols in aging and neurodegeneration associated with oxidative stress. Curr Med Chem 2021; 29:1045-1060. [PMID: 34720075 DOI: 10.2174/0929867328666211101100632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/04/2021] [Accepted: 09/11/2021] [Indexed: 11/22/2022]
Abstract
Aging is defined as the functional loss of tissues and organs over time. This is a biological, irreversible, progressive, and universal process that results from genetic and environmental factors, such as diet, physical activity, smoking, harmful alcohol consumption, and exposure to toxins, among others. Aging is a consequence of molecular and cellular damage built up over time. This damage begins with a gradual decrease in physical and mental capacity, thus increasing the risk of neurodegenerative diseases such as Alzheimer's and Parkinson's disease. Neuronal, functional, and structural damage can be explained by an imbalance among free radicals, reactive oxygen species, reactive nitrogen species, and antioxidants, which finally lead to oxidative stress. Due to the key role of free radicals, reactive oxygen species, and reactive nitrogen species, antioxidant therapy may reduce the oxidative damage associated with neurodegeneration. Exogenous antioxidants are molecules that may help maintain the balance between the formation and elimination of free radicals, thus protecting the cell from their toxicity. Among them, polyphenols are a broad group of secondary plant metabolites with potent antioxidant properties. Here, we review several studies that show the potential role of polyphenol consumption to prevent, or slow down, harmful oxidative processes linked to neurodegenerative disorders.
Collapse
Affiliation(s)
- Francisca Rivas
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago. Chile
| | - Carlos Poblete-Aro
- Centro de Investigacion de Rehabilitacion en Salud, Universidad de las Americas, Santiago. Chile
| | - María Elsa Pando
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago. Chile
| | - María José Allel
- Escuela de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago. Chile
| | - Valentina Fernandez
- Escuela de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago. Chile
| | | | - Pablo Nova
- Unidad de Anatomia Humana Normal, Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago. Chile
| | - Diego Garcia-Diaz
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago. Chile
| |
Collapse
|
27
|
Structural mapping techniques distinguish the surfaces of fibrillar 1N3R and 1N4R human tau. J Biol Chem 2021; 297:101252. [PMID: 34592311 PMCID: PMC8551503 DOI: 10.1016/j.jbc.2021.101252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 12/02/2022] Open
Abstract
The rigid core of intracellular tau filaments from Alzheimer's disease (AD), Pick's disease (PiD), and Corticobasal disease (CBD) brains has been shown to differ in their cryo-EM atomic structure. Despite providing critical information on the intimate arrangement of a fraction of htau molecule within the fibrillar scaffold, the cryo-EM studies neither yield a complete picture of tau fibrillar assemblies structure nor contribute insights into the surfaces that define their interactions with numerous cellular components. Here, using proteomic approaches such as proteolysis and molecular covalent painting, we mapped the exposed amino acid stretches at the surface and those constituting the fibrillar core of in vitro-assembled fibrils of human htau containing one N-terminal domain and three (1N3R) or four (1N4R) C-terminal microtubule-binding repeat domains as a result of alternative splicing. Using limited proteolysis, we identified the proteolytic fragments composing the molecular “bar-code” for each type of fibril. Our results are in agreement with structural data reported for filamentous tau from AD, PiD, and CBD cases predigested with the protease pronase. Finally, we report two amino acid stretches, exposed to the solvent in 1N4R not in 1N3R htau, which distinguish the surfaces of these two kinds of fibrils. Our findings open new perspectives for the design of highly specific ligands with diagnostic and therapeutic potential.
Collapse
|
28
|
Ulugut H, Dijkstra AA, Scarioni M, Barkhof F, Scheltens P, Rozemuller AJM, Pijnenburg YAL. Right temporal variant frontotemporal dementia is pathologically heterogeneous: a case-series and a systematic review. Acta Neuropathol Commun 2021; 9:131. [PMID: 34344452 PMCID: PMC8330072 DOI: 10.1186/s40478-021-01229-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Although the right temporal variant frontotemporal dementia (rtvFTD) is characterised by distinct clinical and radiological features, its underlying histopathology remains elusive. Being considered a right-sided variant of semantic variant primary progressive aphasia (svPPA), TDP-43 type C pathology has been linked to the syndrome, but this has not been studied in detail in large cohorts. In this case report and systematic review, we report the autopsy results of five subjects diagnosed with rtvFTD from our cohort and 44 single rtvFTD subjects from the literature. Macroscopic pathological evaluation of the combined results revealed that rtvFTD demonstrated either a frontotemporal or temporal evolution, even if the degeneration started in the right temporal lobe initially. FTLD-TDP type C was the most common underlying pathology in rtvFTD, however, in 64% of rtvFTD, other underlying pathologies than FTLD-TDP type C were present, such as Tau-MAPT and FTLD-TDP type A and B. Additionally, accompanying motor neuron or corticospinal tract degeneration was observed in 28% of rtvFTD patients. Our results show that in contrast to the general assumption, rtvFTD might not be a pure FTLD-TDP type C disorder, unlike its left temporal counterpart svPPA. Large sample size pathological studies are warranted to understand the diverse pathologies of the right and left temporal variants of frontotemporal dementia.
Collapse
|
29
|
Zhong BR, Zhou GF, Song L, Wen QX, Deng XJ, Ma YL, Hu LT, Chen GJ. TUFM is involved in Alzheimer's disease-like pathologies that are associated with ROS. FASEB J 2021; 35:e21445. [PMID: 33774866 DOI: 10.1096/fj.202002461r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022]
Abstract
Mitochondrial Tu translation elongation factor (TUFM or EF-Tu) is part of the mitochondrial translation machinery. It is reported that TUFM expression is reduced in the brain of Alzheimer's disease (AD), suggesting that TUFM might play a role in the pathophysiology. In this study, we found that TUFM protein level was decreased in the hippocampus and cortex especially in the aged APP/PS1 mice, an animal model of AD. In HEK cells that stably express full-length human amyloid-β precursor protein (HEK-APP), TUFM knockdown or overexpression increased or reduced the protein levels of β-amyloid protein (Aβ) and β-amyloid converting enzyme 1 (BACE1), respectively. TUFM-mediated reduction of BACE1 was attenuated by translation inhibitor cycloheximide (CHX) or α-[2-[4-(3,4-Dichlorophenyl)-2-thiazolyl]hydrazinylidene]-2-nitro-benzenepropanoic acid (4EGI1), and in cells overexpressing BACE1 constructs deleting the 5' untranslated region (5'UTR). TUFM silencing increased the half-life of BACE1 mRNA, suggesting that RNA stability was affected by TUFM. In support, transcription inhibitor Actinomycin D (ActD) and silencing of nuclear factor κB (NFκB) failed to abolish TUFM-mediated regulation of BACE1 protein and mRNA. We further found that the mitochondria-targeted antioxidant TEMPO diminished the effects of TUFM on BACE1, suggesting that reactive oxygen species (ROS) played an important role. Indeed, cellular ROS levels were affected by TUFM knockdown or overexpression, and TUFM-mediated regulation of apoptosis and Tau phosphorylation at selective sites was attenuated by TEMPO. Collectively, TUFM protein levels were decreased in APP/PS1 mice. TUFM is involved in AD pathology by regulating BACE1 translation, apoptosis, and Tau phosphorylation, in which ROS plays an important role.
Collapse
Affiliation(s)
- Bi-Rou Zhong
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Gui-Feng Zhou
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Li Song
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Qi-Xin Wen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xiao-Juan Deng
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuan-Lin Ma
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Li-Tian Hu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China.,Department of Neurology, Nanchong Central Hospital, the Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Guo-Jun Chen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
30
|
Weston LL, Jiang S, Chisholm D, Jantzie LL, Bhaskar K. Interleukin-10 deficiency exacerbates inflammation-induced tau pathology. J Neuroinflammation 2021; 18:161. [PMID: 34275478 PMCID: PMC8286621 DOI: 10.1186/s12974-021-02211-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 07/04/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The presence of hyperphosphorylated microtubule-associated protein tau is strongly correlated with cognitive decline and neuroinflammation in Alzheimer's disease and related tauopathies. However, the role of inflammation and anti-inflammatory interventions in tauopathies is unclear. Our goal was to determine if removing anti-inflammatory interleukin-10 (IL-10) during an acute inflammatory challenge has any effect on neuronal tau pathology. METHODS We induce systemic inflammation in Il10-deficient (Il10-/-) versus Il10+/+ (Non-Tg) control mice using a single intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) to examine microglial activation and abnormal hyperphosphorylation of endogenous mouse tau protein. Tau phosphorylation was quantified by Western blotting and immunohistochemistry. Microglial morphology was quantified by skeleton analysis. Cytokine expression was determined by multiplex electro chemiluminescent immunoassay (MECI) from Meso Scale Discovery (MSD). RESULTS Our findings show that genetic deletion of Il10 promotes enhanced neuroinflammation and tau phosphorylation. First, LPS-induced tau hyperphosphorylation was significantly increased in Il10-/- mice compared to controls. Second, LPS-treated Il10-/- mice showed signs of neurodegeneration. Third, LPS-treated Il10-/- mice showed robust IL-6 upregulation and direct treatment of primary neurons with IL-6 resulted in tau hyperphosphorylation on Ser396/Ser404 site. CONCLUSIONS These data support that loss of IL-10 activates microglia, enhances IL-6, and leads to hyperphosphorylation of tau on AD-relevant epitopes in response to acute systemic inflammation.
Collapse
Affiliation(s)
- Lea L Weston
- Department of Molecular Genetics and Microbiology, University of New Mexico, MSC08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Shanya Jiang
- Department of Molecular Genetics and Microbiology, University of New Mexico, MSC08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Devon Chisholm
- Department of Molecular Genetics and Microbiology, University of New Mexico, MSC08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Lauren L Jantzie
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico, MSC08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA.
- Department of Neurology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
31
|
González C, Cartagena C, Caballero L, Melo F, Areche C, Cornejo A. The Fumarprotocetraric Acid Inhibits Tau Covalently, Avoiding Cytotoxicity of Aggregates in Cells. Molecules 2021; 26:molecules26123760. [PMID: 34205516 PMCID: PMC8234475 DOI: 10.3390/molecules26123760] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative disorders, including Tauopathies that involve tau protein, base their pathological mechanism on forming proteinaceous aggregates, which has a deleterious effect on cells triggering an inflammatory response. Moreover, tau inhibitors can exert their mechanism of action through noncovalent and covalent interactions. Thus, Michael's addition appears as a feasible type of interaction involving an α, β unsaturated carbonyl moiety to avoid pathological confirmation and further cytotoxicity. Moreover, we isolated three compounds from Antarctic lichens Cladonia cariosa and Himantormia lugubris: protolichesterinic acid (1), fumarprotocetraric acid (2), and lichesterinic acid (3). The maleimide cysteine labeling assay showed that compounds 1, 2, and 3 inhibit at 50 µM, but compounds 2 and 3 are statistically significant. Based on its inhibition capacity, we decided to test compound 2 further. Thus, our results suggest that compound 2 remodel soluble oligomers and diminish β sheet content, as demonstrated through ThT experiments. Hence, we added externally treated oligomers with compound 2 to demonstrate that they are harmless in cell culture. First, the morphology of cells in the presence of aggregates does not suffer evident changes compared to the control. Additionally, the externally added aggregates do not provoke a substantial LDH release compared to the control, indicating that treated oligomers do not provoke membrane damage in cell culture compared with aggregates alone. Thus, in the present work, we demonstrated that Michael's acceptors found in lichens could serve as a scaffold to explore different mechanisms of action to turn tau aggregates into harmless species.
Collapse
Affiliation(s)
- Camila González
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad Andres Bello, Santiago 8370071, Chile; (C.G.); (C.C.)
| | - Constanza Cartagena
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad Andres Bello, Santiago 8370071, Chile; (C.G.); (C.C.)
| | - Leonardo Caballero
- Departamento de Física, Center for Soft Matter Research, SMAT-C, Usach, Avenida Ecuador, Estación Central, Santiago 9170124, Chile; (L.C.); (F.M.)
| | - Francisco Melo
- Departamento de Física, Center for Soft Matter Research, SMAT-C, Usach, Avenida Ecuador, Estación Central, Santiago 9170124, Chile; (L.C.); (F.M.)
| | - Carlos Areche
- Departamento de Química, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile;
| | - Alberto Cornejo
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad Andres Bello, Santiago 8370071, Chile; (C.G.); (C.C.)
- Correspondence:
| |
Collapse
|
32
|
Galán-Ganga M, Rodríguez-Cueto C, Merchán-Rubira J, Hernández F, Ávila J, Posada-Ayala M, Lanciego JL, Luengo E, Lopez MG, Rábano A, Fernández-Ruiz J, Lastres-Becker I. Cannabinoid receptor CB2 ablation protects against TAU induced neurodegeneration. Acta Neuropathol Commun 2021; 9:90. [PMID: 34001284 PMCID: PMC8130522 DOI: 10.1186/s40478-021-01196-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Tauopathies are a group of neurodegenerative diseases characterized by the alteration/aggregation of TAU protein, for which there is still no effective treatment. Therefore, new pharmacological targets are being sought, such as elements of the endocannabinoid system (ECS). We analysed the occurrence of changes in the ECS in tauopathies and their implication in the pathogenesis. By integrating gene expression analysis, immunofluorescence, genetic and adeno-associated virus expressing TAU mouse models, we found a TAU-dependent increase in CB2 receptor expression in hippocampal neurons, that occurs as an early event in the pathology and was maintained until late stages. These changes were accompanied by alterations in the endocannabinoid metabolism. Remarkably, CB2 ablation in mice protects from neurodegeneration induced by hTAUP301L overexpression, corroborated at the level of cognitive behaviour, synaptic plasticity, and aggregates of insoluble TAU. At the level of neuroinflammation, the absence of CB2 did not produce significant changes in concordance with a possible neuronal location rather than its classic glial expression in these models. These findings were corroborated in post-mortem samples of patients with Alzheimer's disease, the most common tauopathy. Our results show that neurons with accumulated TAU induce the expression of the CB2 receptor, which enhances neurodegeneration. These results are important for our understanding of disease mechanisms, providing a novel therapeutic strategy to be investigated in tauopathies.
Collapse
|
33
|
Mueller RL, Combs B, Alhadidy MM, Brady ST, Morfini GA, Kanaan NM. Tau: A Signaling Hub Protein. Front Mol Neurosci 2021; 14:647054. [PMID: 33815057 PMCID: PMC8017207 DOI: 10.3389/fnmol.2021.647054] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Abstract
Over four decades ago, in vitro experiments showed that tau protein interacts with and stabilizes microtubules in a phosphorylation-dependent manner. This observation fueled the widespread hypotheses that these properties extend to living neurons and that reduced stability of microtubules represents a major disease-driving event induced by pathological forms of tau in Alzheimer’s disease and other tauopathies. Accordingly, most research efforts to date have addressed this protein as a substrate, focusing on evaluating how specific mutations, phosphorylation, and other post-translational modifications impact its microtubule-binding and stabilizing properties. In contrast, fewer efforts were made to illuminate potential mechanisms linking physiological and disease-related forms of tau to the normal and pathological regulation of kinases and phosphatases. Here, we discuss published work indicating that, through interactions with various kinases and phosphatases, tau may normally act as a scaffolding protein to regulate phosphorylation-based signaling pathways. Expanding on this concept, we also review experimental evidence linking disease-related tau species to the misregulation of these pathways. Collectively, the available evidence supports the participation of tau in multiple cellular processes sustaining neuronal and glial function through various mechanisms involving the scaffolding and regulation of selected kinases and phosphatases at discrete subcellular compartments. The notion that the repertoire of tau functions includes a role as a signaling hub should widen our interpretation of experimental results and increase our understanding of tau biology in normal and disease conditions.
Collapse
Affiliation(s)
- Rebecca L Mueller
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Benjamin Combs
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Mohammed M Alhadidy
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Scott T Brady
- Department of Anatomy and Cell Biology, The University of Illinois at Chicago, Chicago, IL, United States.,Marine Biological Laboratory, Woods Hole, MA, United States
| | - Gerardo A Morfini
- Department of Anatomy and Cell Biology, The University of Illinois at Chicago, Chicago, IL, United States.,Marine Biological Laboratory, Woods Hole, MA, United States
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, United States
| |
Collapse
|
34
|
Zeng Y, Yang J, Zhang B, Gao M, Su Z, Huang Y. The structure and phase of tau: from monomer to amyloid filament. Cell Mol Life Sci 2021; 78:1873-1886. [PMID: 33078207 PMCID: PMC11073437 DOI: 10.1007/s00018-020-03681-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/20/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Tau is a microtubule-associated protein involved in regulation of assembly and spatial organization of microtubule in neurons. However, in pathological conditions, tau monomers assemble into amyloid filaments characterized by the cross-β structures in a number of neurodegenerative diseases known as tauopathies. In this review, we summarize recent progression on the characterization of structures of tau monomer and filament, as well as the dynamic liquid droplet assembly. Our aim is to reveal how post-translational modifications, amino acid mutations, and interacting molecules modulate the conformational ensemble of tau monomer, and how they accelerate or inhibit tau assembly into aggregates. Structure-based aggregation inhibitor design is also discussed in the context of dynamics and heterogeneity of tau structures.
Collapse
Affiliation(s)
- Yifan Zeng
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Jing Yang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Bailing Zhang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Meng Gao
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Zhengding Su
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Yongqi Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China.
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
35
|
Caenorhabditis elegans Models to Investigate the Mechanisms Underlying Tau Toxicity in Tauopathies. Brain Sci 2020; 10:brainsci10110838. [PMID: 33187241 PMCID: PMC7697895 DOI: 10.3390/brainsci10110838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
The understanding of the genetic, biochemical, and structural determinants underlying tau aggregation is pivotal in the elucidation of the pathogenic process driving tauopathies and the design of effective therapies. Relevant information on the molecular basis of human neurodegeneration in vivo can be obtained using the nematode Caenorhabditis elegans (C. elegans). To this end, two main approaches can be applied: the overexpression of genes/proteins leading to neuronal dysfunction and death, and studies in which proteins prone to misfolding are exogenously administered to induce a neurotoxic phenotype. Thanks to the easy generation of transgenic strains expressing human disease genes, C. elegans allows the identification of genes and/or proteins specifically associated with pathology and the specific disruptions of cellular processes involved in disease. Several transgenic strains expressing human wild-type or mutated tau have been developed and offer significant information concerning whether transgene expression regulates protein production and aggregation in soluble or insoluble form, onset of the disease, and the degenerative process. C. elegans is able to specifically react to the toxic assemblies of tau, thus developing a neurodegenerative phenotype that, even when exogenously administered, opens up the use of this assay to investigate in vivo the relationship between the tau sequence, its folding, and its proteotoxicity. These approaches can be employed to screen drugs and small molecules that can interact with the biogenesis and dynamics of formation of tau aggregates and to analyze their interactions with other cellular proteins.
Collapse
|
36
|
Elmaleh DR, Farlow MR, Conti PS, Tompkins RG, Kundakovic L, Tanzi RE. Developing Effective Alzheimer's Disease Therapies: Clinical Experience and Future Directions. J Alzheimers Dis 2020; 71:715-732. [PMID: 31476157 PMCID: PMC6839593 DOI: 10.3233/jad-190507] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) clinical trials, focused on disease modifying drugs and conducted in patients with mild to moderate AD, as well as prodromal (early) AD, have failed to reach efficacy endpoints in improving cognitive function in most cases to date or have been terminated due to adverse events. Drugs that have reached clinical stage were reviewed using web resources (such as clinicaltrials.gov, alzforum.org, company press releases, and peer reviewed literature) to identify late stage (Phase II and Phase III) efficacy clinical trials and summarize reasons for their failure. For each drug, only the latest clinical trials and ongoing trials that aimed at improving cognitive function were included in the analysis. Here we highlight the potential reasons that have hindered clinical success, including clinical trial design and choice of outcome measures, heterogeneity of patient populations, difficulties in diagnosing and staging the disease, drug design, mechanism of action, and toxicity related to the long-term use. We review and suggest approaches for AD clinical trial design aimed at improving our ability to identify novel therapies for this devastating disease.
Collapse
Affiliation(s)
- David R Elmaleh
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.,AZTherapies Inc., Boston, MA, USA
| | - Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter S Conti
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ronald G Tompkins
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
37
|
Alyenbaawi H, Allison WT, Mok SA. Prion-Like Propagation Mechanisms in Tauopathies and Traumatic Brain Injury: Challenges and Prospects. Biomolecules 2020; 10:E1487. [PMID: 33121065 PMCID: PMC7692808 DOI: 10.3390/biom10111487] [Citation(s) in RCA: 5] [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: 09/22/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022] Open
Abstract
The accumulation of tau protein in the form of filamentous aggregates is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). These dementias share traumatic brain injury (TBI) as a prominent risk factor. Tau aggregates can transfer between cells and tissues in a "prion-like" manner, where they initiate the templated misfolding of normal tau molecules. This enables the spread of tau pathology to distinct parts of the brain. The evidence that tauopathies spread via prion-like mechanisms is considerable, but work detailing the mechanisms of spread has mostly used in vitro platforms that cannot fully reveal the tissue-level vectors or etiology of progression. We review these issues and then briefly use TBI and CTE as a case study to illustrate aspects of tauopathy that warrant further attention in vivo. These include seizures and sleep/wake disturbances, emphasizing the urgent need for improved animal models. Dissecting these mechanisms of tauopathy progression continues to provide fresh inspiration for the design of diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hadeel Alyenbaawi
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Medical Laboratories, Majmaah University, Majmaah 11952, Saudi Arabia
| | - W. Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Sue-Ann Mok
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
38
|
Gambino G, Rizzo V, Giglia G, Ferraro G, Sardo P. Microtubule Dynamics and Neuronal Excitability: Advances on Cytoskeletal Components Implicated in Epileptic Phenomena. Cell Mol Neurobiol 2020; 42:533-543. [PMID: 32929563 PMCID: PMC8891195 DOI: 10.1007/s10571-020-00963-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/05/2020] [Indexed: 12/14/2022]
Abstract
Extensive researches have deepened knowledge on the role of synaptic components in epileptogenesis, but limited attention has been devoted to the potential implication of the cytoskeleton. The study of the development of epilepsy and hyperexcitability states involves molecular, synaptic, and structural alterations of neuronal bioelectric activity. In this paper we aim to explore the neurobiological targets involved in microtubule functioning and cytoskeletal transport, i.e. how dynamic scaffolding of microtubules can influence neuronal morphology and excitability, in order to suggest a potential role for microtubule dynamics in the processes turning a normal neuronal network in a hyperexcited one. Pathophysiological alterations of microtubule dynamics inducing neurodegeneration, network remodeling and relative impairment on synaptic transmission were overviewed. Recent researches were reported on the phosphorylation state of microtubule-associated proteins such as tau in neurodegenerative diseases and epileptic states, but also on the effect of microtubule-active agents influencing cytoskeleton destabilization in epilepsy models. The manipulation of microtubule polymerization was found effective in the modulation of hyperexcitability. In addition, it was considered the importance of microtubules and related neurotrophic factors during neural development since they are essential for the formation of a properly functional neuronal network. Otherwise, this can lead to cognitive deficits, hyperexcitability phenomena and neurodevelopmental disorders. Lastly, we evaluated the role of microtubule dynamics on neuronal efficiency considering their importance in the transport of mitochondria, cellular elements fulfilling energy requirements for neuronal activity, and a putative influence on cannabinoid-mediated neuroprotection. This review provides novel perspectives for the implication of microtubule dynamics in the development of epileptic phenomena.
Collapse
Affiliation(s)
- Giuditta Gambino
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione Di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Valerio Rizzo
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione Di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Giuseppe Giglia
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione Di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy.
| | - Giuseppe Ferraro
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione Di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Pierangelo Sardo
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione Di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| |
Collapse
|
39
|
Shutinoski B, Hakimi M, Harmsen IE, Lunn M, Rocha J, Lengacher N, Zhou YY, Khan J, Nguyen A, Hake-Volling Q, El-Kodsi D, Li J, Alikashani A, Beauchamp C, Majithia J, Coombs K, Shimshek D, Marcogliese PC, Park DS, Rioux JD, Philpott DJ, Woulfe JM, Hayley S, Sad S, Tomlinson JJ, Brown EG, Schlossmacher MG. Lrrk2 alleles modulate inflammation during microbial infection of mice in a sex-dependent manner. Sci Transl Med 2020; 11:11/511/eaas9292. [PMID: 31554740 DOI: 10.1126/scitranslmed.aas9292] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 12/27/2018] [Accepted: 05/11/2019] [Indexed: 12/20/2022]
Abstract
Variants in the leucine-rich repeat kinase-2 (LRRK2) gene are associated with Parkinson's disease, leprosy, and Crohn's disease, three disorders with inflammation as an important component. Because of its high expression in granulocytes and CD68-positive cells, LRRK2 may have a function in innate immunity. We tested this hypothesis in two ways. First, adult mice were intravenously inoculated with Salmonella typhimurium, resulting in sepsis. Second, newborn mouse pups were intranasally infected with reovirus (serotype 3 Dearing), which induced encephalitis. In both mouse models, wild-type Lrrk2 expression was protective and showed a sex effect, with female Lrrk2-deficient animals not controlling infection as well as males. Mice expressing Lrrk2 carrying the Parkinson's disease-linked p.G2019S mutation controlled infection better, with reduced bacterial growth and longer animal survival during sepsis. This gain-of-function effect conferred by the p.G2019S mutation was mediated by myeloid cells and was abolished in animals expressing a kinase-dead Lrrk2 variant, p.D1994S. Mouse pups with reovirus-induced encephalitis that expressed the p.G2019S Lrrk2 mutation showed increased mortality despite lower viral titers. The p.G2019S mutant Lrrk2 augmented immune cell chemotaxis and generated more reactive oxygen species during virulent infection. Reovirus-infected brains from mice expressing the p.G2019S mutant Lrrk2 contained higher concentrations of α-synuclein. Animals expressing one or two p.D1994S Lrrk2 alleles showed lower mortality from reovirus-induced encephalitis. Thus, Lrrk2 alleles may alter the course of microbial infections by modulating inflammation, and this may be dependent on the sex and genotype of the host as well as the type of pathogen.
Collapse
Affiliation(s)
- Bojan Shutinoski
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Mansoureh Hakimi
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Irene E Harmsen
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Michaela Lunn
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Juliana Rocha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Nathalie Lengacher
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Yi Yuan Zhou
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Jasmine Khan
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Angela Nguyen
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Quinton Hake-Volling
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Daniel El-Kodsi
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Juan Li
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Azadeh Alikashani
- Research Centre, Montreal Heart Institute, Montréal, QC, Canada.,Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Claudine Beauchamp
- Research Centre, Montreal Heart Institute, Montréal, QC, Canada.,Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Jay Majithia
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Kevin Coombs
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Derya Shimshek
- Novartis Institutes of BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Paul C Marcogliese
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - David S Park
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - John D Rioux
- Research Centre, Montreal Heart Institute, Montréal, QC, Canada.,Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - John M Woulfe
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Julianna J Tomlinson
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Earl G Brown
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Michael G Schlossmacher
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.,Division of Neurology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
40
|
Yadikar H, Torres I, Aiello G, Kurup M, Yang Z, Lin F, Kobeissy F, Yost R, Wang KK. Screening of tau protein kinase inhibitors in a tauopathy-relevant cell-based model of tau hyperphosphorylation and oligomerization. PLoS One 2020; 15:e0224952. [PMID: 32692785 PMCID: PMC7373298 DOI: 10.1371/journal.pone.0224952] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 06/15/2020] [Indexed: 12/15/2022] Open
Abstract
Tauopathies are a class of neurodegenerative disorders characterized by abnormal deposition of post-translationally modified tau protein in the human brain. Tauopathies are associated with Alzheimer's disease (AD), chronic traumatic encephalopathy (CTE), and other diseases. Hyperphosphorylation increases tau tendency to aggregate and form neurofibrillary tangles (NFT), a pathological hallmark of AD. In this study, okadaic acid (OA, 100 nM), a protein phosphatase 1/2A inhibitor, was treated for 24h in mouse neuroblastoma (N2a) and differentiated rat primary neuronal cortical cell cultures (CTX) to induce tau-hyperphosphorylation and oligomerization as a cell-based tauopathy model. Following the treatments, the effectiveness of different kinase inhibitors was assessed using the tauopathy-relevant tau antibodies through tau-immunoblotting, including the sites: pSer202/pThr205 (AT8), pThr181 (AT270), pSer202 (CP13), pSer396/pSer404 (PHF-1), and pThr231 (RZ3). OA-treated samples induced tau phosphorylation and oligomerization at all tested epitopes, forming a monomeric band (46-67 kDa) and oligomeric bands (170 kDa and 240 kDa). We found that TBB (a casein kinase II inhibitor), AR and LiCl (GSK-3 inhibitors), cyclosporin A (calcineurin inhibitor), and Saracatinib (Fyn kinase inhibitor) caused robust inhibition of OA-induced monomeric and oligomeric p-tau in both N2a and CTX culture. Additionally, a cyclin-dependent kinase 5 inhibitor (Roscovitine) and a calcium chelator (EGTA) showed contrasting results between the two neuronal cultures. This study provides a comprehensive view of potential drug candidates (TBB, CsA, AR, and Saracatinib), and their efficacy against tau hyperphosphorylation and oligomerization processes. These findings warrant further experimentation, possibly including animal models of tauopathies, which may provide a putative Neurotherapy for AD, CTE, and other forms of tauopathy-induced neurodegenerative diseases.
Collapse
Affiliation(s)
- Hamad Yadikar
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, United States of America
- Department of Biological Sciences, Faculty of Science, Kuwait University, Safat, Kuwait
- Department of Chemistry, Chemistry Laboratory Building, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| | - Isabel Torres
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, United States of America
| | - Gabrielle Aiello
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, United States of America
| | - Milin Kurup
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, United States of America
| | - Zhihui Yang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, United States of America
| | - Fan Lin
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, United States of America
| | - Firas Kobeissy
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, United States of America
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Richard Yost
- Department of Chemistry, Chemistry Laboratory Building, University of Florida, Gainesville, FL, United States of America
| | - Kevin K. Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, United States of America
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, Gainesville, FL, United States of America
| |
Collapse
|
41
|
Casillas‐Espinosa PM, Ali I, O'Brien TJ. Neurodegenerative pathways as targets for acquired epilepsy therapy development. Epilepsia Open 2020; 5:138-154. [PMID: 32524040 PMCID: PMC7278567 DOI: 10.1002/epi4.12386] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/13/2020] [Accepted: 02/24/2020] [Indexed: 12/16/2022] Open
Abstract
There is a growing body of clinical and experimental evidence that neurodegenerative diseases and epileptogenesis after an acquired brain insult may share common etiological mechanisms. Acquired epilepsy commonly develops as a comorbid condition in patients with neurodegenerative diseases such as Alzheimer's disease, although it is likely much under diagnosed in practice. Progressive neurodegeneration has also been described after traumatic brain injury, stroke, and other forms of brain insults. Moreover, recent evidence has shown that acquired epilepsy is often a progressive disorder that is associated with the development of drug resistance, cognitive decline, and worsening of other neuropsychiatric comorbidities. Therefore, new pharmacological therapies that target neurobiological pathways that underpin neurodegenerative diseases have potential to have both an anti-epileptogenic and disease-modifying effect on the seizures in patients with acquired epilepsy, and also mitigate the progressive neurocognitive and neuropsychiatric comorbidities. Here, we review the neurodegenerative pathways that are plausible targets for the development of novel therapies that could prevent the development or modify the progression of acquired epilepsy, and the supporting published experimental and clinical evidence.
Collapse
Affiliation(s)
- Pablo M. Casillas‐Espinosa
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
| | - Idrish Ali
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
| | - Terence J. O'Brien
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
- Department of NeurologyThe Alfred HospitalMelbourneVic.Australia
- Department of NeurologyThe Royal Melbourne HospitalParkvilleVic.Australia
| |
Collapse
|
42
|
Pražienková V, Schirmer C, Holubová M, Železná B, Kuneš J, Galas MC, Maletínská L. Lipidized Prolactin-Releasing Peptide Agonist Attenuates Hypothermia-Induced Tau Hyperphosphorylation in Neurons. J Alzheimers Dis 2020; 67:1187-1200. [PMID: 30689580 DOI: 10.3233/jad-180837] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases, characterized by the accumulation of extracellular amyloid plaques and intraneuronal neurofibrillary tangles. These tangles mainly consist of hyperphosphorylated tau protein. As it induces tau hyperphosphorylation in vitro and in vivo, hypothermia is a useful tool for screening potential neuroprotective compounds that ameliorate tau pathology. In this study, we examined the effect of prolactin-releasing peptide (PrRP), its lipidized analog palm11-PrRP31 and glucagon-like-peptide-1 agonist liraglutide, substances with anorexigenic and antidiabetic properties, on tau phosphorylation and on the main kinases and phosphatases involved in AD development. Our study was conducted in a neuroblastoma cell line SH-SY5Y and rat primary neuronal cultures under normothermic and hypothermic conditions. Hypothermia induced a significant increase in tau phosphorylation at the pThr212 and pSer396/pSer404 epitopes. The palmitoylated analogs liraglutide and palm11-PrRP31 attenuated tau hyperphosphorylation, suggesting their potential use in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Claire Schirmer
- Université Lille, INSERM, CHU Lille, UMR - S 1172 - Jean Pierre Aubert Research Centre, Alzheimer and Tauopathies, Lille, France
| | - Martina Holubová
- Institute of Organic Chemistry and Biochemistry, AS CR, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, AS CR, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, AS CR, Prague, Czech Republic.,Institute of Physiology, AS CR, Prague, Czech Republic
| | - Marie-Christine Galas
- Université Lille, INSERM, CHU Lille, UMR - S 1172 - Jean Pierre Aubert Research Centre, Alzheimer and Tauopathies, Lille, France
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, AS CR, Prague, Czech Republic
| |
Collapse
|
43
|
Cassar M, Law AD, Chow ES, Giebultowicz JM, Kretzschmar D. Disease-Associated Mutant Tau Prevents Circadian Changes in the Cytoskeleton of Central Pacemaker Neurons. Front Neurosci 2020; 14:232. [PMID: 32292325 PMCID: PMC7118733 DOI: 10.3389/fnins.2020.00232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/02/2020] [Indexed: 01/10/2023] Open
Abstract
A hallmark feature of Alzheimer's disease (AD) and other Tauopathies, like Frontotemporal Dementia with Parkinsonism linked to chromosome 17 (FTDP-17), is the accumulation of neurofibrillary tangles composed of the microtubule-associated protein Tau. As in AD, symptoms of FTDP-17 include cognitive decline, neuronal degeneration, and disruptions of sleep patterns. However, mechanisms by which Tau may lead to these disturbances in sleep and activity patterns are unknown. To identify such mechanisms, we have generated novel Drosophila Tauopathy models by replacing endogenous fly dTau with normal human Tau (hTau) or the FTDP-17 causing hTauV337M mutation. This mutation is localized in one of the microtubule-binding domains of hTau and has a dominant effect. Analyzing heterozygous flies, we found that aged hTauV337M flies show neuronal degeneration and locomotion deficits when compared to wild type or hTauWT flies. Furthermore, hTauV337M flies are hyperactive and they show a fragmented sleep pattern. These changes in the sleep/activity pattern are accompanied by morphological changes in the projection pattern of the central pacemaker neurons. These neurons show daily fluctuations in their connectivity, whereby synapses are increased during the day and reduced during sleep. Synapse formation requires cytoskeletal changes that can be detected by the accumulation of the end-binding protein 1 (EB1) at the site of synapse formation. Whereas, hTauWT flies show the normal day/night changes in EB1 accumulation, hTauV337M flies do not show this fluctuation. This suggests that hTauV337M disrupts sleep patterns by interfering with the cytoskeletal changes that are required for the synaptic homeostasis of central pacemaker neurons.
Collapse
Affiliation(s)
- Marlène Cassar
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| | - Alexander D Law
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| | - Eileen S Chow
- Department of Integrative Biology, Oregon State University, Corvallis, OR, United States
| | - Jadwiga M Giebultowicz
- Department of Integrative Biology, Oregon State University, Corvallis, OR, United States
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
44
|
Genetic architecture of neurodegenerative dementias. Neuropharmacology 2020; 168:108014. [PMID: 32097768 DOI: 10.1016/j.neuropharm.2020.108014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/03/2020] [Accepted: 02/14/2020] [Indexed: 12/14/2022]
Abstract
Molecular genetics has been an invaluable tool to help understand the molecular basis of neurodegenerative dementias. In this review, we provide an overview of the genetic architecture underlying some of the most prevalent causes of dementia, including Alzheimer's dementia, frontotemporal lobar degeneration, Lewy body dementia, and prion diseases. We also discuss the complexity of the human genome and how the novel technologies have revolutionized and accelerated the way we screen the variety of our DNA. Finally, we also provide some examples about how this genetic knowledge is being transferred into the clinic through personalized medicine. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
|
45
|
Alquezar C, Felix JB, McCandlish E, Buckley BT, Caparros-Lefebvre D, Karch CM, Golbe LI, Kao AW. Heavy metals contaminating the environment of a progressive supranuclear palsy cluster induce tau accumulation and cell death in cultured neurons. Sci Rep 2020; 10:569. [PMID: 31953414 PMCID: PMC6969162 DOI: 10.1038/s41598-019-56930-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/05/2019] [Indexed: 11/09/2022] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disorder characterized by the presence of intracellular aggregates of tau protein and neuronal loss leading to cognitive and motor impairment. Occurrence is mostly sporadic, but rare family clusters have been described. Although the etiopathology of PSP is unknown, mutations in the MAPT/tau gene and exposure to environmental toxins can increase the risk of PSP. Here, we used cell models to investigate the potential neurotoxic effects of heavy metals enriched in a highly industrialized region in France with a cluster of sporadic PSP cases. We found that iPSC-derived iNeurons from a MAPT mutation carrier tend to be more sensitive to cell death induced by chromium (Cr) and nickel (Ni) exposure than an isogenic control line. We hypothesize that genetic variations may predispose to neurodegeneration induced by those heavy metals. Furthermore, using an SH-SY5Y neuroblastoma cell line, we showed that both heavy metals induce cell death by an apoptotic mechanism. Interestingly, Cr and Ni treatments increased total and phosphorylated tau levels in both cell types, implicating Cr and Ni exposure in tau pathology. Overall, this study suggests that chromium and nickel could contribute to the pathophysiology of tauopathies such as PSP by promoting tau accumulation and neuronal cell death.
Collapse
Affiliation(s)
- Carolina Alquezar
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, 94158, USA
| | - Jessica B Felix
- Graduate Program, Department of Molecular and Cellular Biology Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Elizabeth McCandlish
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, 170, Frelinghuysen Road Piscataway NJ, 08854, New Brunswick, NJ, United States
| | - Brian T Buckley
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, 170, Frelinghuysen Road Piscataway NJ, 08854, New Brunswick, NJ, United States
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Lawrence I Golbe
- Division of Movement Disorders. Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Aimee W Kao
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, 94158, USA.
| |
Collapse
|
46
|
Pyridine-2,3-dicarboxylate, quinolinic acid, induces 1N4R Tau amyloid aggregation in vitro: Another evidence for the detrimental effect of the inescapable endogenous neurotoxin. Chem Biol Interact 2019; 315:108884. [PMID: 31678113 DOI: 10.1016/j.cbi.2019.108884] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/18/2019] [Accepted: 10/26/2019] [Indexed: 01/19/2023]
Abstract
Quinolinic acid (QA) known as a neuro-active metabolite associated with the kynurenine pathway. At high concentrations, QA is often involved in the initiation and development of several human neurologic diseases, like Alzheimer's disease. Because of the QA action as the NMDA receptor, it is considered as a potent excitotoxin in vivo. Since it is probable that different mechanisms are employed by QA, activation of NMDA receptors cannot fully explain the revealed toxicity and it is even believed that there are multiple unknown mechanisms/targets leading to QA cytotoxicity. Herein we report accelerated amyloid oligomerization of 1N4R Tau under the effect of QA, in vitro, then the molecular structure, morphology and toxicity of the protein aggregate were documented by using various theoretical/experimental approaches. The possible mechanism of action of QA-induced Tau oligomerization has also been explored.
Collapse
|
47
|
Novel Peptidomic Approach for Identification of Low and High Molecular Weight Tauopathy Peptides Following Calpain Digestion, and Primary Culture Neurotoxic Challenges. Int J Mol Sci 2019; 20:ijms20205213. [PMID: 31640160 PMCID: PMC6829287 DOI: 10.3390/ijms20205213] [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: 08/03/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/29/2022] Open
Abstract
Tauopathy is a class of a neurodegenerative disorder linked with tau hyperphosphorylation, proteolysis, and aggregation. Tau can be subjected to proteolysis upon calpain activation in Alzheimer disease (AD), and traumatic brain injury (TBI). We and others have extensively researched calpain-mediated tau breakdown products (Tau-BDP; 45K, 35K, and 17K). Tau proteolysis might also generate low molecular weight (LMW ≤10K) proteolytic peptides after neurodegenerative damage. In this study, we have subjected purified tau protein (phospho and non-phospho) and mouse brain lysate to calpain-1 digestion to characterize the LMW generated by nano-liquid chromatography coupled to electrospray ionization to tandem mass spectrometry (nano-LC-ESI-MS/MS). We have also challenged differentiated primary cerebrocortical neuronal cultures (CTX) with neurotoxic agents (calcium ionophore calcimycin (A23187), staurosporine (STS), N-methyl-D-aspartate (NMDA), and Maitotoxin (MTX)) that mimic neurodegeneration to investigate the peptidome released into the conditioned cell media. We used a simple workflow in which we fractionate LMW calpain-mediated tau peptides by ultrafiltration (molecular weight cut-off value (MWCO) of 10K) and subject filtrate fractions to nano-LC-MS/MS analysis. The high molecular weight (HMW) peptides and intact proteins retained on the filter were analyzed separately by western blotting using total and phospho-specific tau antibodies. We have identified several novel proteolytic tau peptides (phosphorylated and non-phosphorylated) that are only present in samples treated with calpain or cell-based calpain activation model (particularly N- and C-terminal peptides). Our findings can help in developing future research strategies emphasizing on the suppression of tau proteolysis as a target.
Collapse
|
48
|
Clinical Laboratory Tests Used To Aid in Diagnosis of Human Prion Disease. J Clin Microbiol 2019; 57:JCM.00769-19. [PMID: 31366689 DOI: 10.1128/jcm.00769-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Prion diseases are a group of rapidly progressive and always fatal neurodegenerative disorders caused by misfolded prion protein in the brain. Although autopsy remains the gold-standard diagnostic tool, antemortem laboratory testing can be performed to aid in the diagnosis of prion disease. This review is meant to help laboratory directors and physicians in their interpretation of test results. Laboratory assays to detect both nonspecific biomarkers of prion disease and prion-specific biomarkers can be used. The levels of nonspecific biomarkers in cerebrospinal fluid (CSF) are elevated when rapid neurodegeneration is occurring in the patient, and these markers include 14-3-3, tau, neuron-specific enolase, S100B, and alpha-synuclein. These markers have various sensitivities and specificities but are overall limited, as the levels of any of these analytes can be elevated in nonprion disease that is causing rapid damage of brain tissue. Prion-specific assays used in clinical laboratory testing are currently limited to two options. The first option is second-generation real-time quaking-induced conversion (RT-QuIC) performed on CSF, and the second option is Western blotting of a brain biopsy specimen used to detect protease-resistant prion protein. Although both tests have exquisite specificity, RT-QuIC has a sensitivity of 92 to 97.2% in symptomatic individuals, compared to the brain biopsy Western blot sensitivity of 20 to 60%. RT-QuIC was added to the Centers for Disease Control and Prevention's diagnostic criteria for prion disease in 2018. Other caveats of laboratory testing need to be considered, as sporadic, genetic, and acquired forms of prion disease have different clinical and laboratory presentations, and these caveats are discussed. Laboratory testing plays an important role in the diagnosis of prion disease, which is often challenging to diagnose.
Collapse
|
49
|
Xiang XJ, Song L, Deng XJ, Tang Y, Min Z, Luo B, Wen QX, Li KY, Chen J, Ma YL, Zhu BL, Yan Z, Chen GJ. Mitochondrial methionine sulfoxide reductase B2 links oxidative stress to Alzheimer's disease-like pathology. Exp Neurol 2019; 318:145-156. [DOI: 10.1016/j.expneurol.2019.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/10/2019] [Accepted: 05/08/2019] [Indexed: 01/25/2023]
|
50
|
Wang L, Liu J, Wang Q, Jiang H, Zeng L, Li Z, Liu R. MicroRNA-200a-3p Mediates Neuroprotection in Alzheimer-Related Deficits and Attenuates Amyloid-Beta Overproduction and Tau Hyperphosphorylation via Coregulating BACE1 and PRKACB. Front Pharmacol 2019; 10:806. [PMID: 31379578 PMCID: PMC6658613 DOI: 10.3389/fphar.2019.00806] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/21/2019] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by two landmark pathologies, the overproduction of amyloid-beta peptides (Aβ), predominated by the β-amyloid protein precursor cleaving enzyme 1 (BACE1), and hyperphosphorylation of the microtubule protein, tau, because of an imbalance in a kinase/phosphatase system that involves the activation of the protein kinase A (PKA). Current evidence indicates that brain microRNAs participate in multiple aspects of AD pathology. Here, the role and underlying molecular mechanisms of microRNA-200a-3p (miR-200a-3p) in mediating neuroprotection against AD-related deficits were investigated. The expression of miR-200a-3p was measured in the hippocampus of APP/PS1 and SAMP8 mice and in an AD cell model in vitro, as well as in blood plasma extracted from AD patients. The targets of miR-200a-3p were determined using bioinformatics and dual-luciferase assay analyses. In addition, cell apoptosis was detected using flow cytometry, and related protein levels were measured using Western blot and enzyme-linked immunosorbent assay (ELISA) techniques. miR-200a-3p was confirmed to be depressed in microarray miRNA profile analysis in vitro and in vivo, suggesting that miR-200a-3p is a potential biomarker of AD. Subsequently, miR-200a-3p was demonstrated to inhibit cell apoptosis accompanied by the inactivation of the Bax/caspase-3 axis and downregulation of Aβ1-42 and tau phosphorylation levels in vitro. Further mechanistic studies revealed that miR-200a-3p reduced the production of Aβ1-42 and decreased hyperphosphorylation of tau by regulating the protein translocation of BACE1 and the protein kinase cAMP-activated catalytic subunit beta (PRKACB) associated with the three prime untranslated regions, respectively. Importantly, the function of miR-200a-3p was reversed by overexpression of BACE1 or PRKACB in cultured cells. This resulted in an elevation in cell apoptosis and increases in Aβ1-42 and tau hyperphosphorylation levels, involving the epitopes threonine 205 and serine 202, 214, 396, and 356, the favorable phosphorylated sites of PKA. In conclusion, our study suggests that miR-200a-3p is implicated in the pathology of AD, exerting neuroprotective effects against Aβ-induced toxicity by two possible mechanisms: one involving the inhibition of Aβ overproduction via suppression of the expression of BACE1 and synergistically decreasing the hyperphosphorylation of tau via attenuation of the expression of PKA.
Collapse
Affiliation(s)
- Linlin Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianghong Liu
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Qian Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hailun Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Li Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhuorong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|