1
|
Hohberger B, Prüss H, Mardin C, Lämmer R, Müller J, Wallukat G. Glaucoma and Alzheimer: Neurodegenerative disorders show an adrenergic dysbalance. PLoS One 2022; 17:e0272811. [PMID: 36201426 PMCID: PMC9536590 DOI: 10.1371/journal.pone.0272811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/26/2022] [Indexed: 11/06/2022] Open
Abstract
Glaucoma disease is characterized by an increased intraocular pressure (IOP), glaucomatous alterations of the optic disc and corresponding visual field defects. Even lowering the main risk factor IOP until an individual target level does not prevent this neurodegenerative disorder from proceeding. Several autoimmune mechanisms were discovered, partly showing a functionality. One of these autoimmune phenomena targets the ß2-adrenergic receptor (ß2-AR; i.e. agonistic autoantibodies; ß2-agAAb) and is linked to an elevated IOP and an impaired retinal microcirculation. As neurodegenerative disorder, Alzheimer's Disease (AD) is postulated to share a common molecular mechanism with glaucoma. In the present study we investigated autoimmune phenomena targeting the ß2-AR in patients with AD. Sera of the patients were analyzed in a rat cardiomyocyte bioassay for the presence of functional autoantibodies against ß2-AR. In addition, different species of amyloid beta (Aß) monomers were tested (Aß1-14, Aß10-25, Aβ10-37 Aß1-40, Aß1-42, Aβ28-40, and Aß-[Pyr]3-43). Our results demonstrate that none of the short-chain Aß (Aß1-14, Aß10-25, or Aβ28-40) showed any agonistic or inhibitory effect on ß2-AR. Contrary, long-chain Aß-[Pyr]3-43, representing a major neurogenic plaque component, exerted an activation that after blocking by the ß2-AR antagonist ICI118.551, could be identified as that the effect was realized via the ß2-AR. Moreover, the long chain Aß1-40, Aβ1-42, and Aβ10-37, yet not the short-chain Aß peptides prevented the clenbuterol induced desensitization of the ß2-AR. In addition, we identified functional autoantibodies in the sera of AD patients, activating the ß2-AR, like the ß2-agAAb found in patients with glaucoma. As autoimmune mechanisms were reportedly involved in the pathogenesis of glaucoma and Alzheimer's Disease, we postulate that overstimulation of the ß2-AR pathway can induce an adrenergic overdrive, that may play an important role in the multifactorial interplay of neurodegenerative disorders.
Collapse
Affiliation(s)
- Bettina Hohberger
- Department of Ophthalmology, Universität of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Harald Prüss
- Department of Neurology, Charite´-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Mardin
- Department of Ophthalmology, Universität of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Lämmer
- Department of Ophthalmology, Universität of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | |
Collapse
|
2
|
Consens ME, Chen Y, Menon V, Wang Y, Schneider JA, De Jager PL, Bennett DA, Tripathy SJ, Felsky D. Bulk and Single-Nucleus Transcriptomics Highlight Intra-Telencephalic and Somatostatin Neurons in Alzheimer's Disease. Front Mol Neurosci 2022; 15:903175. [PMID: 35754708 PMCID: PMC9231610 DOI: 10.3389/fnmol.2022.903175] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Cortical neuron loss is a pathological hallmark of late-onset Alzheimer's disease (AD). However, it remains unclear which neuronal subtypes beyond broad excitatory and inhibitory classes are most vulnerable. Here, we analyzed cell subtype proportion differences in AD compared to non-AD controls using 1037 post-mortem brain samples from six neocortical regions. We identified the strongest associations of AD with fewer somatostatin (SST) inhibitory neurons (β = -0.48, p bonf = 8.98 × 10-9) and intra-telencephalic (IT) excitatory neurons (β = -0.45, p bonf = 4.32 × 10-7). Replication in three AD case-control single-nucleus RNAseq datasets most strongly supported the bulk tissue association of fewer SST neurons in AD. In depth analyses of cell type proportions with specific AD-related neuropathological and cognitive phenotypes revealed fewer SST neurons with greater brain-wide post-mortem tau and beta amyloid, as well as a faster rate of antemortem cognitive decline. In contrast, greater IT neuron proportions were associated with a slower rate of cognitive decline as well as greater residual cognition-a measure of cognitive resilience-but not canonical AD neuropathology. Our findings implicate somatostatin inhibitory and intra-telencephalic excitatory neuron subclasses in the pathogenesis of AD and in cognitive resilience to AD pathology, respectively.
Collapse
Affiliation(s)
- Micaela E. Consens
- The Krembil Centre for Neuroinformatics (KCNI), Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Yuxiao Chen
- The Krembil Centre for Neuroinformatics (KCNI), Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Vilas Menon
- The Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, United States
| | - Yanling Wang
- The Rush Alzheimer’s Disease Center, Rush University, Chicago, IL, United States
| | - Julie A. Schneider
- The Rush Alzheimer’s Disease Center, Rush University, Chicago, IL, United States
| | - Philip L. De Jager
- The Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, United States
| | - David A. Bennett
- The Rush Alzheimer’s Disease Center, Rush University, Chicago, IL, United States
| | - Shreejoy J. Tripathy
- The Krembil Centre for Neuroinformatics (KCNI), Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Daniel Felsky
- The Krembil Centre for Neuroinformatics (KCNI), Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Singh A, Allen D, Fracassi A, Tumurbaatar B, Natarajan C, Scaduto P, Woltjer R, Kayed R, Limon A, Krishnan B, Taglialatela G. Functional Integrity of Synapses in the Central Nervous System of Cognitively Intact Individuals with High Alzheimer's Disease Neuropathology Is Associated with Absence of Synaptic Tau Oligomers. J Alzheimers Dis 2021; 78:1661-1678. [PMID: 33185603 PMCID: PMC7836055 DOI: 10.3233/jad-200716] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Certain individuals, here referred to as Non-Demented with Alzheimer Neuropathology (NDAN), do not show overt neurodegeneration (N-) and remain cognitively intact despite the presence of plaques (A+) and tangles (T+) that would normally be consistent with fully symptomatic Alzheimer's disease (AD). OBJECTIVE The existence of NDAN (A + T+N-) subjects suggests that the human brain utilizes intrinsic mechanisms that can naturally evade cognitive decline normally associated with the symptomatic stages of AD (A + T+N+). Deciphering the underlying mechanisms would prove relevant to develop complementing therapeutics to prevent progression of AD-related cognitive decline. METHODS Previously, we have reported that NDAN present with preserved neurogenesis and synaptic integrity paralleled by absence of amyloid oligomers at synapses. Using postmortem brain samples from age-matched control subjects, demented AD patients and NDAN individuals, we performed immunofluorescence, western blots, micro transplantation of synaptic membranes in Xenopus oocytes followed by twin electrode voltage clamp electrophysiology and fluorescence assisted single synaptosome-long term potentiation studies. RESULTS We report decreased tau oligomers at synapses in the brains of NDAN subjects. Furthermore, using novel approaches we report, for the first time, that such absence of tau oligomers at synapses is associated with synaptic functional integrity in NDAN subjects as compared to demented AD patients. CONCLUSION Overall, these results give further credence to tau oligomers as primary actors of synaptic destruction underscoring cognitive demise in AD and support their targeting as a viable therapeutic strategy for AD and related tauopathies.
Collapse
Affiliation(s)
- Ayush Singh
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Dyron Allen
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Batbayar Tumurbaatar
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Chandramouli Natarajan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Pietro Scaduto
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Randy Woltjer
- Department of Pathology, Oregon Health and Science University, Portland, OR, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Agenor Limon
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Balaji Krishnan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA,Correspondence to: Giulio Taglialatela, PhD, Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA. Tel.: +1 409 772 1679; Fax: +1 409 772 0015; E-mail: . and Balaji Krishnan, PhD, Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA. Tel.: +1 409 772 8069; Fax: +1 409 772 0015; E-mail:
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA,Correspondence to: Giulio Taglialatela, PhD, Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA. Tel.: +1 409 772 1679; Fax: +1 409 772 0015; E-mail: . and Balaji Krishnan, PhD, Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA. Tel.: +1 409 772 8069; Fax: +1 409 772 0015; E-mail:
| |
Collapse
|
4
|
Defining early changes in Alzheimer's disease from RNA sequencing of brain regions differentially affected by pathology. Sci Rep 2021; 11:4865. [PMID: 33649380 PMCID: PMC7921390 DOI: 10.1038/s41598-021-83872-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 02/03/2021] [Indexed: 01/01/2023] Open
Abstract
Tau pathology in Alzheimer’s disease (AD) spreads in a predictable pattern that corresponds with disease symptoms and severity. At post-mortem there are cortical regions that range from mildly to severely affected by tau pathology and neuronal loss. A comparison of the molecular signatures of these differentially affected areas within cases and between cases and controls may allow the temporal modelling of disease progression. Here we used RNA sequencing to explore differential gene expression in the mildly affected primary visual cortex and moderately affected precuneus of ten age-, gender- and RNA quality-matched post-mortem brains from AD patients and healthy controls. The two regions in AD cases had similar transcriptomic signatures but there were broader abnormalities in the precuneus consistent with the greater tau load. Both regions were characterised by upregulation of immune-related genes such as those encoding triggering receptor expressed on myeloid cells 2 and membrane spanning 4-domains A6A and milder changes in insulin/IGF1 signalling. The precuneus in AD was also characterised by changes in vesicle secretion and downregulation of the interneuronal subtype marker, somatostatin. The ‘early’ AD transcriptome is characterised by perturbations in synaptic vesicle secretion on a background of neuroimmune dysfunction. In particular, the synaptic deficits that characterise AD may begin with the somatostatin division of inhibitory neurotransmission.
Collapse
|
5
|
Curcumin as Scaffold for Drug Discovery against Neurodegenerative Diseases. Biomedicines 2021; 9:biomedicines9020173. [PMID: 33572457 PMCID: PMC7916200 DOI: 10.3390/biomedicines9020173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases (NDs) are one of major public health problems and their impact is continuously growing. Curcumin has been proposed for the treatment of several of these pathologies, such as Alzheimer's disease (AD) and Parkinson's disease (PD) due to the ability of this molecule to reduce inflammation and aggregation of involved proteins. Nevertheless, the poor metabolic stability and bioavailability of curcumin reduce the possibilities of its practical use. For these reasons, many curcumin derivatives were synthetized in order to overcome some limitations. In this review will be highlighted recent results on modification of curcumin scaffold in the search of new effective therapeutic agents against NDs, with particular emphasis on AD.
Collapse
|
6
|
Mavilio A, Sisto D, Prete F, Guadalupi V, Dammacco R, Alessio G. RE-PERG in early-onset Alzheimer's disease: A double-blind, electrophysiological pilot study. PLoS One 2020; 15:e0236568. [PMID: 32790788 PMCID: PMC7425894 DOI: 10.1371/journal.pone.0236568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 07/08/2020] [Indexed: 12/31/2022] Open
Abstract
PURPOSE To evaluate the ability of re-test pattern electroretinogram (RE-PERG), a non-invasive and fast steady-state PERG, to detect inner retinal bioelectric function anomalies in patients with early-onset Alzheimer's disease (AD). METHODS The study population consisted of 17 patients with AD-related mild cognitive impairment (MCI), 16 patients with vascular dementia (VD)-related MCI, both assessed using the neuropsychological Mini-Mental State Examination (MMSE) and by structural magnetic resonance imaging, and 19 healthy, age-matched normal controls (NC). All participants were visually asymptomatic, had normal or near-normal general cognitive functioning and no or minimal impairments in daily life activities. Visual field (VF) test, optical coherence tomography (OCT) and RE-PERG, sampled in five consecutive blocks of 130 events, were performed. RESULTS There was no statistically significant difference among the three groups with respect to age, VF parameters (mean and pattern standard deviations) and OCT parameters (ganglion cell complex thickness and retinal nerve fiber layer thickness). The mean amplitude in the RE-PERG was significantly lower, but only weakly in the AD group than in NC (p = 0.1) whereas the intrinsic variability of the 2nd harmonic phase was significantly higher in the AD group than in either the VD or NC group (p<0.001). CONCLUSIONS RE-PERG is altered in early-stage AD, showing a reduced amplitude with high intrinsic phase variability. It also allows the discrimination of AD from VD. A high intrinsic variability in the PERG signal, determined using RE-PERG, may thus be a new promising test for neurodegenerative diseases.
Collapse
Affiliation(s)
- Alberto Mavilio
- Social Health District, Glaucoma Center, Azienda Sanitaria Locale–Brindisi, Brindisi, Italy
| | - Dario Sisto
- Department of Neurosciences, Institute of Ophthalmology, University of Bari, Bari, Italy
| | - Florenza Prete
- Social Health District, Alzheimer Evaluation Units, Azienda Sanitaria Locale—Brindisi, Brindisi, Italy
| | - Viviana Guadalupi
- Social Health District, Alzheimer Evaluation Units, Azienda Sanitaria Locale—Brindisi, Brindisi, Italy
| | - Rosanna Dammacco
- Department of Neurosciences, Institute of Ophthalmology, University of Bari, Bari, Italy
| | - Giovanni Alessio
- Department of Neurosciences, Institute of Ophthalmology, University of Bari, Bari, Italy
| |
Collapse
|
7
|
Sen S, Saxena R, Tripathi M, Vibha D, Dhiman R. Neurodegeneration in Alzheimer's disease and glaucoma: overlaps and missing links. Eye (Lond) 2020; 34:1546-1553. [PMID: 32152519 PMCID: PMC7608361 DOI: 10.1038/s41433-020-0836-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 12/17/2019] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
The eye is said to be the window into the brain. Alzheimer’s disease (AD) and glaucoma both being diseases of the elderly, have several epidemiological and histological overlaps in pathogenesis. Both these diseases are neurodegenerative conditions. Over the years, a consensus has developed that both may be two ends of a singular spectrum of diseases. Epidemiological studies have shown that more Alzheimer’s patients may be suffering from glaucoma than general healthy population. Retinal ganglion cell damage is a characteristic of both diseases, along with discovery of amyloid-β and tau protein deposition in the retina and aqueous humor of eye. The latter two proteins are known to be pathognomonic of AD. Other pathways such as the insulin receptor pathway also seem to be affected in both diseases similarly. In spite of these overlaps, there are few missing links which still need more evidence, namely, intraocular pressure mechanisms, cerebrospinal fluid pressure and trans-lamina cribrosa pressure gradients, vascular autoregulation factors, etc. Several factors point towards a common pathogenesis at some level for both diseases and prospective studies are necessary to study the natural course of both diseases.
Collapse
Affiliation(s)
- Sagnik Sen
- Department of Ophthalmology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Rohit Saxena
- Department of Ophthalmology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India.
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Deepti Vibha
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Rebika Dhiman
- Department of Ophthalmology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
8
|
Sullivan EV, Pfefferbaum A. Brain-behavior relations and effects of aging and common comorbidities in alcohol use disorder: A review. Neuropsychology 2019; 33:760-780. [PMID: 31448945 PMCID: PMC7461729 DOI: 10.1037/neu0000557] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Alcohol use disorder (AUD) is a complex, dynamic condition that waxes and wanes with unhealthy drinking episodes and varies in drinking patterns and effects on brain structure and function with age. Its excessive use renders chronically heavy drinkers vulnerable to direct alcohol toxicity and a variety of comorbidities attributable to nonalcohol drug misuse, viral infections, and accelerated or premature aging. AUD affects widespread brain systems, commonly, frontolimbic, frontostriatal, and frontocerebellar networks. METHOD AND RESULTS Multimodal assessment using selective neuropsychological testing and whole-brain neuroimaging provides evidence for AUD-related specific brain structure-function relations established with double dissociations. Longitudinal study using noninvasive imaging provides evidence for brain structural and functional improvement with sustained sobriety and further decline with relapse. Functional imaging suggests the possibility that some alcoholics in recovery can compensate for impairment by invoking brain systems typically not used for a target task but that can enable normal-level performance. CONCLUSIONS Evidence for AUD-aging interactions, indicative of accelerated aging, together with increasing alcohol consumption in middle-age and older adults, put aging drinkers at special risk for developing cognitive decline and possibly dementia. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
Affiliation(s)
- Edith V. Sullivan
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Adolf Pfefferbaum
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
- Center for Health Sciences, SRI International, Menlo Park, CA
| |
Collapse
|
9
|
d'Orange M, Aurégan G, Cheramy D, Gaudin-Guérif M, Lieger S, Guillermier M, Stimmer L, Joséphine C, Hérard AS, Gaillard MC, Petit F, Kiessling MC, Schmitz C, Colin M, Buée L, Panayi F, Diguet E, Brouillet E, Hantraye P, Bemelmans AP, Cambon K. Potentiating tangle formation reduces acute toxicity of soluble tau species in the rat. Brain 2019; 141:535-549. [PMID: 29253129 PMCID: PMC5837551 DOI: 10.1093/brain/awx342] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 10/23/2017] [Indexed: 12/21/2022] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by the aggregation of tau protein. These pathologies exhibit a wide variety of clinical and anatomo-pathological presentations, which may result from different pathological mechanisms. Although tau inclusions are a common feature in all these diseases, recent evidence instead implicates small oligomeric aggregates as drivers of tau-induced toxicity. Hence in vivo model systems displaying either soluble or fibrillary forms of wild-type or mutant tau are needed to better identify their respective pathological pathways. Here we used adeno-associated viruses to mediate gene transfer of human tau to the rat brain to develop models of pure tauopathies. Two different constructs were used, each giving rise to a specific phenotype developing in less than 3 months. First, hTAUWT overexpression led to a strong hyperphosphorylation of the protein, which was associated with neurotoxicity in the absence of any significant aggregation. In sharp contrast, its co-expression with the pro-aggregation peptide TauRD-ΔK280 in the hTAUProAggr group strongly promoted its aggregation into Gallyas-positive neurofibrillary tangles, while preserving neuronal survival. Our results support the hypothesis that soluble tau species are key players of tau-induced neurodegeneration.
Collapse
Affiliation(s)
- Marie d'Orange
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Gwénaelle Aurégan
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Dimitri Cheramy
- Institut de Recherches Servier, DRD-RDNPS, 125 chemin de ronde, 78 290 Croissy sur Seine, France
| | - Mylène Gaudin-Guérif
- Institut de Recherches Servier, DRD-RDNPS, 125 chemin de ronde, 78 290 Croissy sur Seine, France
| | - Sarah Lieger
- Inserm, UMR-S 1172, Lille, France.,Université Lille 2, Faculté de Médecine, IMPRT, JPARC, Lille, France.,CMRR, CHR, Lille, France
| | - Martine Guillermier
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Lev Stimmer
- MIRCen, INSERM-CEA, Platform for experimental pathology, U1169 and US27, F- 92265 Fontenay-aux-Roses, France
| | - Charlène Joséphine
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Anne-Sophie Hérard
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Marie-Claude Gaillard
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Fanny Petit
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | | | - Christoph Schmitz
- Department of Neuroanatomy, Ludwig-Maximilians-University, Munich, Germany
| | - Morvane Colin
- Inserm, UMR-S 1172, Lille, France.,Université Lille 2, Faculté de Médecine, IMPRT, JPARC, Lille, France.,CMRR, CHR, Lille, France
| | - Luc Buée
- Inserm, UMR-S 1172, Lille, France.,Université Lille 2, Faculté de Médecine, IMPRT, JPARC, Lille, France.,CMRR, CHR, Lille, France
| | - Fany Panayi
- Institut de Recherches Servier, DRD-RDNPS, 125 chemin de ronde, 78 290 Croissy sur Seine, France
| | - Elsa Diguet
- Institut de Recherches Servier, DRD-RDNPS, 125 chemin de ronde, 78 290 Croissy sur Seine, France
| | - Emmanuel Brouillet
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Philippe Hantraye
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Alexis-Pierre Bemelmans
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Karine Cambon
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| |
Collapse
|
10
|
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder affecting millions of people worldwide. Therefore, finding effective interventions and therapies is extremely important. AD is one of over 20 different disorders known as tauopathies, characterized by the pathological aggregation and accumulation of tau, a microtubule-associated protein. Tau aggregates are heterogeneous and can be divided into two major groups: large metastable fibrils, including neurofibrillary tangles, and oligomers. The smaller, soluble and dynamic tau oligomers have been shown to be more toxic with more proficient seeding properties for the propagation of tau pathology as compared to the fibrillar Paired Helical Filaments (PHFs). Therefore, developing small molecules that target and interact with toxic tau oligomers can be beneficial to modulate their aggregation pathways and toxicity, preventing progression of the pathology. In this study, we show that Azure C (AC) is capable of modulating tau oligomer aggregation pathways at micromolar concentrations and rescues tau oligomers-induced toxicity in cell culture. We used both biochemical and biophysical in vitro techniques to characterize preformed tau oligomers in the presence and absence of AC. Interestingly, AC prevents toxicity not by disassembling the oligomers but rather by converting them into clusters of aggregates with nontoxic conformation.
Collapse
Affiliation(s)
- Filippa Lo Cascio
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Experimental Biomedicine and Clinical Neuroscience, University of Palermo, 90127 Palermo, Italy
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
11
|
Tau Accumulation, Altered Phosphorylation, and Missorting Promote Neurodegeneration in Glaucoma. J Neurosci 2017; 36:5785-98. [PMID: 27225768 DOI: 10.1523/jneurosci.3986-15.2016] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/13/2016] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Glaucoma, the leading cause of irreversible blindness worldwide, is characterized by the selective death of retinal ganglion cells (RGCs). Ocular hypertension is the most significant known risk factor for developing the disease, but the mechanism by which elevated pressure damages RGCs is currently unknown. The axonal-enriched microtubule-associated protein tau is a key mediator of neurotoxicity in Alzheimer's disease and other tauopathies. Using a well characterized in vivo rat glaucoma model, we show an age-related increase in endogenous retinal tau that was markedly exacerbated by ocular hypertension. Early alterations in tau phosphorylation, characterized by epitope-dependent hyperphosphorylation and hypophosphorylation, correlated with the appearance of tau oligomers in glaucomatous retinas. Our data demonstrate the mislocalization of tau in the somatodendritic compartment of RGCs subjected to high intraocular pressure. In contrast, tau was depleted from RGC axons in the optic nerve of glaucomatous eyes. Importantly, intraocular administration of short interfering RNA against tau effectively reduced retinal tau accumulation and promoted robust survival of RGC somas and axons, supporting a critical role for tau alterations in ocular hypertension-induced neuronal damage. Our study reveals that glaucoma displays signature pathological features of tauopathies, including tau accumulation, altered phosphorylation, and missorting; and identifies tau as a novel target to counter RGC neurodegeneration in glaucoma and prevalent optic neuropathies. SIGNIFICANCE STATEMENT In this study, we investigated the role of tau in retinal ganglion cell (RGC) damage in glaucoma. We demonstrate that high intraocular pressure leads to a rapid increase in endogenous retinal tau with altered phosphorylation profile and the formation of tau oligomers. Tau accumulation was primarily observed in RGC dendrites, while tau in RGC axons within the optic nerve was depleted. Attenuation of endogenous retinal tau using a targeted siRNA led to striking protection of RGC somas and axons from hypertension-induced damage. Our study identifies novel and substantial alterations of endogenous tau protein in glaucoma, including abnormal subcellular distribution, an altered phosphorylation profile, and neurotoxicity.
Collapse
|
12
|
Schaeffer EL, Catanozi S, West MJ, Gattaz WF. Stereological investigation of the CA1 pyramidal cell layer in untreated and lithium-treated 3xTg-AD and wild-type mice. Ann Anat 2017; 209:51-60. [DOI: 10.1016/j.aanat.2016.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/03/2016] [Accepted: 10/11/2016] [Indexed: 12/25/2022]
|
13
|
Nilson AN, English KC, Gerson JE, Barton Whittle T, Nicolas Crain C, Xue J, Sengupta U, Castillo-Carranza DL, Zhang W, Gupta P, Kayed R. Tau Oligomers Associate with Inflammation in the Brain and Retina of Tauopathy Mice and in Neurodegenerative Diseases. J Alzheimers Dis 2017; 55:1083-1099. [PMID: 27716675 PMCID: PMC5147514 DOI: 10.3233/jad-160912] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2016] [Indexed: 12/13/2022]
Abstract
It is well-established that inflammation plays an important role in Alzheimer's disease (AD) and frontotemporal lobar dementia (FTLD). Inflammation and synapse loss occur in disease prior to the formation of larger aggregates, but the contribution of tau to inflammation has not yet been thoroughly investigated. Tau pathologically aggregates to form large fibrillar structures known as tangles. However, evidence suggests that smaller soluble aggregates, called oligomers, are the most toxic species and form prior to tangles. Furthermore, tau oligomers can spread to neighboring cells and between anatomically connected brain regions. In addition, recent evidence suggests that inspecting the retina may be a window to brain pathology. We hypothesized that there is a relationship between tau oligomers and inflammation, which are hallmarks of early disease. We conducted immunofluorescence and biochemical analyses on tauopathy mice, FTLD, and AD subjects. We showed that oligomers co-localize with astrocytes, microglia, and HMGB1, a pro-inflammatory cytokine. Additionally, we show that tau oligomers are present in the retina and are associated with inflammatory cells suggesting that the retina may be a valid non-invasive biomarker for brain pathology. These results suggest that there may be a toxic relationship between tau oligomers and inflammation. Therefore, the ability of tau oligomers to spread may initiate a feed-forward cycle in which tau oligomers induce inflammation, leading to neuronal damage, and thus more inflammation. Further mechanistic studies are warranted in order to understand this relationship, which may have critical implications for improving the treatment of tauopathies.
Collapse
Affiliation(s)
- Ashley N. Nilson
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kelsey C. English
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Julia E. Gerson
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - T. Barton Whittle
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - C. Nicolas Crain
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Judy Xue
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Diana L. Castillo-Carranza
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Praveena Gupta
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
14
|
Mutated tau, amyloid and neuroinflammation in Alzheimer disease—A brief review. ACTA ACUST UNITED AC 2016; 51:1-8. [PMID: 26851150 DOI: 10.1016/j.proghi.2016.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/14/2016] [Accepted: 01/14/2016] [Indexed: 01/08/2023]
|
15
|
Guerrero-Muñoz MJ, Gerson J, Castillo-Carranza DL. Tau Oligomers: The Toxic Player at Synapses in Alzheimer's Disease. Front Cell Neurosci 2015; 9:464. [PMID: 26696824 PMCID: PMC4667007 DOI: 10.3389/fncel.2015.00464] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 11/16/2015] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive disorder in which the most noticeable symptoms are cognitive impairment and memory loss. However, the precise mechanism by which those symptoms develop remains unknown. Of note, neuronal loss occurs at sites where synaptic dysfunction is observed earlier, suggesting that altered synaptic connections precede neuronal loss. The abnormal accumulation of amyloid-β (Aβ) and tau protein is the main histopathological feature of the disease. Several lines of evidence suggest that the small oligomeric forms of Aβ and tau may act synergistically to promote synaptic dysfunction in AD. Remarkably, tau pathology correlates better with the progression of the disease than Aβ. Recently, a growing number of studies have begun to suggest that missorting of tau protein from the axon to the dendrites is required to mediate the detrimental effects of Aβ. In this review we discuss the novel findings regarding the potential mechanisms by which tau oligomers contribute to synaptic dysfunction in AD.
Collapse
Affiliation(s)
- Marcos J Guerrero-Muñoz
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston TX, USA ; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston TX, USA
| | - Julia Gerson
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston TX, USA ; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston TX, USA
| | - Diana L Castillo-Carranza
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston TX, USA ; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston TX, USA
| |
Collapse
|
16
|
Coppola G, Di Renzo A, Ziccardi L, Martelli F, Fadda A, Manni G, Barboni P, Pierelli F, Sadun AA, Parisi V. Optical Coherence Tomography in Alzheimer's Disease: A Meta-Analysis. PLoS One 2015; 10:e0134750. [PMID: 26252902 PMCID: PMC4529274 DOI: 10.1371/journal.pone.0134750] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/13/2015] [Indexed: 12/24/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a neurodegenerative disorder, which is likely to start as mild cognitive impairment (MCI) several years before the its full-blown clinical manifestation. Optical coherence tomography (OCT) has been used to detect a loss in peripapillary retina nerve fiber layer (RNFL) and a reduction in macular thickness and volume of people affected by MCI or AD. Here, we performed an aggregate meta-analysis combining results from different studies. Methods and Findings Data sources were case-control studies published between January 2001 and August 2014 (identified through PubMed and Google Scholar databases) that examined the RNFL thickness by means of OCT in AD and MCI patients compared with cognitively healthy controls. Results 11 studies were identified, including 380 patients with AD, 68 with MCI and 293 healthy controls (HC). The studies suggest that the mean RNFL thickness is reduced in MCI (weighted mean differences in μm, WMD = -13.39, 95% CI: -17.34 to -9.45, p = 0.031) and, even more so, in AD (WMD = -15.95, 95% CI: -21.65 to -10.21, p<0.0001) patients compared to HC. RNFL in the 4 quadrants were all significantly thinner in AD superior (superior WMD = -24.0, 95% CI: -34.9 to -13.1, p<0.0001; inferior WMD = -20.8, 95% CI: -32.0 to -9.7, p<0.0001; nasal WMD = -14.7, 95% CI: -23.9 to -5.5, p<0.0001; and temporal WMD = -10.7, 95% CI: -19.9 to -1.4, p<0.0001); the same significant reduction in quadrant RNFL was observed in MCI patients compared with HC (Inferior WMD = -20.22, 95% CI: -30.41 to -10.03, p = 0.0001; nasal WMD = -7.4, 95% CI: -10.08 to -4.7, p = 0.0000; and temporal WMD = -6.88, 95% CI: -12.62 to -1.13, p = 0.01), with the exception of superior quadrant (WMD = -19.45, 95% CI: -40.23 to 1.32, p = 0.06). Conclusion Results from the meta-analysis support the important role of OCT for RNFL analysis in monitoring the progression of AD and in assessing the effectiveness of purported AD treatments.
Collapse
Affiliation(s)
- Gianluca Coppola
- G.B. Bietti Foundation-IRCCS, Department of Neurophysiology of Vision and Neurophthalmology, Rome, Italy
- * E-mail:
| | - Antonio Di Renzo
- G.B. Bietti Foundation-IRCCS, Department of Neurophysiology of Vision and Neurophthalmology, Rome, Italy
| | - Lucia Ziccardi
- G.B. Bietti Foundation-IRCCS, Department of Neurophysiology of Vision and Neurophthalmology, Rome, Italy
| | - Francesco Martelli
- Istituto Superiore di Sanità, Dipartimento Tecnologie e Salute, Rome, Italy
| | - Antonello Fadda
- Istituto Superiore di Sanità, Dipartimento Tecnologie e Salute, Rome, Italy
| | - Gianluca Manni
- Tor Vergata University of Rome, Department of Clinical Sciences and Translation Medicine; Rome, Italy
| | - Piero Barboni
- IRCCS Istituto Scientifico San Raffaele, Milan, Italy
| | - Francesco Pierelli
- Sapienza University of Rome Polo Pontino, Department of Medico-Surgical Sciences and Biotechnologies, Latina, Italy
- IRCCS-Neuromed, Pozzilli (IS), Italy
| | - Alfredo A. Sadun
- Doheny Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Vincenzo Parisi
- G.B. Bietti Foundation-IRCCS, Department of Neurophysiology of Vision and Neurophthalmology, Rome, Italy
| |
Collapse
|
17
|
Salama M, Mohamed WM. NDUFA12L mitochondrial complex-I assembly factor: Implications for taupathies. Appl Transl Genom 2015; 5:37-9. [PMID: 26937358 PMCID: PMC4745364 DOI: 10.1016/j.atg.2015.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 05/29/2015] [Indexed: 12/02/2022]
Abstract
There is a strong correlation between taupathies and the development and progression of neurodegenerative disorders. Abnormal tau becomes hyperphosphorylated and dissociated from microtubules with the aggregation of intracellular tau aggregates within the patient's brain. The current review is divided into two broad sections. In the first section we discuss the molecular biology and the clinicopathologic features of taupathies. While in the second section we discuss the relationship between mitochondrial complex-I and taupathies. Polymorphism in NDUFA12L may be a crucial factor for development of neurodegenerative taupathies. Thus NDUFA12L screening may be an early biomarker for identifying risk groups for such disorders.
Collapse
Affiliation(s)
| | - Wael M.Y. Mohamed
- Clinical Pharmacology Dept, Menoufia Medical School, Menoufia University, Egypt
| |
Collapse
|
18
|
Yang Y, Shepherd C, Halliday G. Aneuploidy in Lewy body diseases. Neurobiol Aging 2014; 36:1253-60. [PMID: 25595497 DOI: 10.1016/j.neurobiolaging.2014.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 12/13/2014] [Accepted: 12/13/2014] [Indexed: 12/13/2022]
Abstract
An increase in DNA content is associated with neuronal degeneration in Alzheimer's disease but has not been evaluated in Lewy body diseases. Using stereological principles, flow cytometry, and standard histopathologic methods, we evaluated the number and DNA content of neurons and all cells and the severity of Lewy and Alzheimer pathologies, in brain regions affected at different stages in Lewy body diseases compared with controls. An increase in neuronal DNA content was observed in all the affected brain regions examined, although this change was related to different pathologies. In the substantia nigra, increased neuronal DNA content related to neuronal loss, whereas in the cortex and hippocampus, increased neuronal DNA content related to Alzheimer pathologies. Of note, increased neuronal DNA content did not relate to the deposition of Lewy bodies in any region examined. These data support the concept that increased DNA content increases neuronal susceptibility to degeneration and Alzheimer pathologies.
Collapse
Affiliation(s)
- Yue Yang
- Neuroscience Research Australia, Randwick, New South Wales, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Claire Shepherd
- Neuroscience Research Australia, Randwick, New South Wales, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Glenda Halliday
- Neuroscience Research Australia, Randwick, New South Wales, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia.
| |
Collapse
|
19
|
Gerson JE, Castillo-Carranza DL, Kayed R. Advances in therapeutics for neurodegenerative tauopathies: moving toward the specific targeting of the most toxic tau species. ACS Chem Neurosci 2014; 5:752-69. [PMID: 25075869 DOI: 10.1021/cn500143n] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative disease is one of the greatest health concerns today and with no effective treatment in sight, it is crucial that researchers find a safe and successful therapeutic. While neurofibrillary tangles are considered the primary tauopathy hallmark, more evidence continues to come to light to suggest that soluble, intermediate tau aggregates--tau oligomers--are the most toxic species in disease. These intermediate tau species may also be responsible for the spread of pathology, suggesting that oligomeric tau may be the best therapeutic target. Here, we summarize results for the modulation of tau by molecular chaperones, small molecules and aggregation inhibitors, post-translational modifications, immunotherapy, other techniques, and future directions.
Collapse
Affiliation(s)
- Julia E. Gerson
- Department
of Neurology, George and Cynthia Mitchell
Center for Alzheimer’s Disease Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Diana L. Castillo-Carranza
- Department
of Neurology, George and Cynthia Mitchell
Center for Alzheimer’s Disease Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Rakez Kayed
- Department
of Neurology, George and Cynthia Mitchell
Center for Alzheimer’s Disease Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
20
|
Gerson JE, Sengupta U, Lasagna-Reeves CA, Guerrero-Muñoz MJ, Troncoso J, Kayed R. Characterization of tau oligomeric seeds in progressive supranuclear palsy. Acta Neuropathol Commun 2014; 2:73. [PMID: 24927818 PMCID: PMC4229782 DOI: 10.1186/2051-5960-2-73] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/10/2014] [Indexed: 02/01/2023] Open
Abstract
Background Progressive supranuclear palsy (PSP) is a neurodegenerative tauopathy which is primarily defined by the deposition of tau into globose-type neurofibrillary tangles (NFT). Tau in its native form has important functions for microtubule dynamics. Tau undergoes alternative splicing in exons 2, 3, and 10 which results in six different isoforms. Products of splicing on exon 10 are the most prone to mutations. Three repeat (3R) and four repeat (4R) tau, like other disease-associated amyloids, can form oligomers which may then go on to further aggregate and form fibrils. Recent studies from our laboratory and others have provided evidence that tau oligomers, not NFTs, are the most toxic species in neurodegenerative tauopathies and seed the pathological spread of tau. Results Analysis of PSP brain sections revealed globose-type NFTs, as well as both phosphorylated and unphosphorylated tau oligomers. Analysis of PSP brains via Western blot and ELISA revealed the presence of increased levels of tau oligomers compared to age-matched control brains. Oligomers were immunoprecipitated from PSP brain and were capable of seeding the oligomerization of both 3R and 4R tau isoforms. Conclusions This is the first time tau oligomers have been characterized in PSP. These results indicate that tau oligomers are an important component of PSP pathology, along with NFTs. The ability of PSP brain-derived tau oligomers to seed 3R and 4R tau suggests that these oligomers represent the pathological species responsible for disease propagation and the presence of oligomers in a pure neurodegenerative tauopathy implies a common neuropathological process for tau seen in diseases with other amyloid proteins.
Collapse
|
21
|
Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J Neurosci 2014; 34:4260-72. [PMID: 24647946 DOI: 10.1523/jneurosci.3192-13.2014] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recent findings suggest that tau oligomers, which form before neurofibrillary tangles (NFTs), are the true neurotoxic tau entities in neurodegenerative tauopathies, including Alzheimer's disease (AD). Studies in animal models of tauopathy suggest that tau oligomers play a key role in eliciting behavioral and cognitive impairments. Here, we used a novel tau oligomer-specific monoclonal antibody (TOMA) for passive immunization in mice expressing mutant human tau. A single dose of TOMA administered either intravenously or intracerebroventricularly was sufficient to reverse both locomotor and memory deficits in a mouse model of tauopathy for 60 d, coincident with rapid reduction of tau oligomers but not phosphorylated NFTs or monomeric tau. Our data demonstrate that antibody protection is mediated by extracellular and rapid peripheral clearance. These findings provide the first direct evidence in support of a critical role for tau oligomers in disease progression and validate tau oligomers as a target for the treatment of AD and other neurodegenerative tauopathies.
Collapse
|
22
|
Beauquis J, Vinuesa A, Pomilio C, Pavía P, Galván V, Saravia F. Neuronal and glial alterations, increased anxiety, and cognitive impairment before hippocampal amyloid deposition in PDAPP mice, model of Alzheimer's disease. Hippocampus 2014; 24:257-69. [PMID: 24132937 DOI: 10.1002/hipo.22219] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2013] [Indexed: 11/05/2022]
Abstract
In the context of Alzheimer's disease (AD), hippocampal alterations have been well described in advanced stages of the pathology, when amyloid deposition, inflammation and glial activation occur, but less attention has been directed to studying early brain and behavioral changes. Using an animal model of AD, the transgenic PDAPP-J20 mouse at 5 months of age, when no amyloid plaques are present and low cerebral levels of amyloid peptides are detectable, we found structural, morphological, and cellular alterations in the hippocampus. Young transgenic mice showed a reduced hippocampal volume with less number of pyramidal and granular neurons, which additionally exhibited cell atrophy. The neurogenic capability in this zone, measured as DCX+ cells, was strongly diminished and associated to alterations in cell maturity. A decrease in presynaptic synaptophysin optical density was detected in mossy fibers reaching CA3 subfield but not in Golgi stained- CA1 dendritic spine density. Employing confocal microscopy and accurate stereological tools we also found a reduction in the number of GFAP+ cells, along with decreased astrocyte complexity, suggesting a potential detriment of neural support. According with untimely neuroglial alterations, young PDAPP mice failed in the novel location recognition test, that depends on hippocampal function. Moreover, multivariate statistical analysis of the behavioral outcome in the open-field test evidenced an elevated anxiety score in Tg mice compared with age-matched control mice. In line with this, the transgenic group showed a higher number of c-Fos+ nuclei in central and basolateral amygdala, a result that supports the early involvement of the emotionality factor in AD pathology. Applying an integrative approach, this work focuses on early structural, morphological and functional changes and provides new and compelling evidence of behavioral alterations that precede manifest AD.
Collapse
Affiliation(s)
- Juan Beauquis
- Instituto de Biología y Medicina Experimental CONICET and Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
23
|
Transient focal cerebral ischemia/reperfusion induces early and chronic axonal changes in rats: its importance for the risk of Alzheimer's disease. PLoS One 2012; 7:e33722. [PMID: 22457786 PMCID: PMC3311543 DOI: 10.1371/journal.pone.0033722] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 02/15/2012] [Indexed: 11/19/2022] Open
Abstract
The dementia of Alzheimer's type and brain ischemia are known to increase at comparable rates with age. Recent advances suggest that cerebral ischemia may contribute to the pathogenesis of Alzheimer's disease (AD), however, the neuropathological relationship between these two disorders is largely unclear. It has been demonstrated that axonopathy, mainly manifesting as impairment of axonal transport and swelling of the axon and varicosity, is a prominent feature in AD and may play an important role in the neuropathological mechanisms in AD. In this study, we investigated the early and chronic changes of the axons of neurons in the different brain areas (cortex, hippocampus and striatum) using in vivo tracing technique and grading analysis method in a rat model of transient focal cerebral ischemia/reperfusion (middle cerebral artery occlusion, MCAO). In addition, the relationship between the changes of axons and the expression of β-amyloid 42 (Aβ42) and hyperphosphorylated Tau, which have been considered as the key neuropathological processes of AD, was analyzed by combining tracing technique with immunohistochemistry or western blotting. Subsequently, we found that transient cerebral ischemia/reperfusion produced obvious swelling of the axons and varicosities, from 6 hours after transient cerebral ischemia/reperfusion even up to 4 weeks. We could not observe Aβ plaques or overexpression of Aβ42 in the ischemic brain areas, however, the site-specific hyperphosphorylated Tau could be detected in the ischemic cortex. These results suggest that transient cerebral ischemia/reperfusion induce early and chronic axonal changes, which may be an important mechanism affecting the clinical outcome and possibly contributing to the development of AD after stroke.
Collapse
|
24
|
Augustinack JC, Huber KE, Postelnicu GM, Kakunoori S, Wang R, van der Kouwe AJW, Wald LL, Stein TD, Frosch MP, Fischl B. Entorhinal verrucae geometry is coincident and correlates with Alzheimer's lesions: a combined neuropathology and high-resolution ex vivo MRI analysis. Acta Neuropathol 2012; 123:85-96. [PMID: 22160360 DOI: 10.1007/s00401-011-0929-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 10/14/2022]
Abstract
Entorhinal cortex displays a distinctive organization in layer II and forms small elevations on its surface called entorhinal verrucae. In Alzheimer's disease, the verrucae disappear due to neurofibrillary tangle formation and neuronal death. Isosurface models were reconstructed from high-resolution ex vivo MRI volumes scanned at 7.0 T and individual verruca were measured quantitatively for height, width, volume, and surface area on control and mild Alzheimer's cases. Mean verruca height was 0.13 ± 0.04 mm for our cognitively normal (controls) sample set whereas for mild AD samples mean height was 0.11 mm ± 0.05 mm (p < 0.001) in entorhinal cortex (n = 10 cases). These quantitative methods were validated by a significant correlation of verrucae height and volume with qualitative verrucae ratings (n = 36 cases). Entorhinal surfaces were significantly different from other cortical heights such as, cingulate, frontal, occipital, parietal and temporal cortices. Colocalization of verrucae with entorhinal islands was confirmed in ex vivo MRI and, moreover, verrucae ratings were negatively correlated to Braak and Braak pathological stage. This study characterizes novel methods to measure individual entorhinal verruca size, and shows that verrucae size correlates to Alzheimer's pathology. Taken together, these results suggest that verrucae may have the potential to serve as an early and specific morphological marker for mild cognitive impairment and Alzheimer's disease.
Collapse
Affiliation(s)
- Jean C Augustinack
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Building 149-13th St., Room 2301, Charlestown, MA 02129, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gemmell E, Bosomworth H, Allan L, Hall R, Khundakar A, Oakley AE, Deramecourt V, Polvikoski TM, O'Brien JT, Kalaria RN. Hippocampal neuronal atrophy and cognitive function in delayed poststroke and aging-related dementias. Stroke 2011; 43:808-14. [PMID: 22207507 DOI: 10.1161/strokeaha.111.636498] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE We have previously shown delayed poststroke dementia in elderly (≥75 years old) stroke survivors is associated with medial temporal lobe atrophy; however, the basis of the structural and functional changes is unknown. METHODS Using 3-dimensional stereological methods, we quantified hippocampal pyramidal neuronal volumes and densities in a total of 95 postmortem samples from demented and nondemented poststroke survivors within our prospective Cognitive Function after Stroke study and subjects pathologically diagnosed with vascular dementia, Alzheimer disease, and mixed Alzheimer disease and vascular dementia syndrome. RESULTS Hippocampal CA1 but not CA2 subfield neuron density was affected in poststroke, Alzheimer disease, vascular dementia, and mixed dementia groups relative to control subjects (P<0.05). Neuronal volume was reduced in the poststroke dementia relative to poststroke nondemented group in both CA1 and CA2, although there were no apparent differences in neuronal density. Poststroke nondemented neuronal volumes were similar to control subjects but greater than in all dementias (P<0.05). Neuronal volumes positively correlated with global cognitive function and memory function in both CA1 and CA2 in poststroke subjects (P<0.01). Degrees of neuronal atrophy and loss were similar in the poststroke dementia and vascular dementia groups. However, in the entorhinal cortex layer V, neuronal volumes were only impaired in the mixed and Alzheimer disease groups (P<0.05). CONCLUSIONS Our results suggest hippocampal neuronal atrophy is an important substrate for dementia in both cerebrovascular and neurodegenerative disease.
Collapse
Affiliation(s)
- Elizabeth Gemmell
- Centre for Brain Ageing and Vitality, Institute for Ageing and Health, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Spires-Jones TL, Kopeikina KJ, Koffie RM, de Calignon A, Hyman BT. Are tangles as toxic as they look? J Mol Neurosci 2011; 45:438-44. [PMID: 21638071 PMCID: PMC3173560 DOI: 10.1007/s12031-011-9566-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 05/23/2011] [Indexed: 12/28/2022]
Abstract
Neurofibrillary tangles are intracellular accumulations of hyperphosphorylated and misfolded tau protein characteristic of Alzheimer's disease and other tauopathies. Classic cross-sectional studies of Alzheimer patient brains showed associations of tangle accumulation with neuronal loss, synapse loss, and dementia, which led to the supposition that tangles are toxic to neurons. More recent advances in imaging techniques and mouse models have allowed the direct exploration of the question of toxicity of aggregated versus soluble tau and have surprisingly challenged the view of tangles as toxic species in the brain. Here, we review these recent experiments on the nature of the toxicity of tau with particular emphasis on our experiments imaging tangles in the intact brain through a cranial window, which allows observation of tangle formation and longitudinal imaging of the fate of tangle-bearing neurons.
Collapse
Affiliation(s)
- Tara L Spires-Jones
- Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA.
| | | | | | | | | |
Collapse
|
27
|
Kayed R, Jackson GR, Estes DM, Barrett ADT. Alzheimers disease: review of emerging treatment role for intravenous immunoglobulins. J Cent Nerv Syst Dis 2011; 3:67-73. [PMID: 23861639 PMCID: PMC3663607 DOI: 10.4137/jcnsd.s5018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder. Currently available therapies are symptomatic but do not alter underlying disease progression. Immunotherapeutic approaches such as anti Aβ peptide active vaccination trials have had limited success to date. Intravenous immunoblobulin (IVIg) is widely used in immune-mediated neurological disorders such myasthenia gravis and Guillain-Barre syndrome. These preparations have been obtained from the pooled plasma of healthy human donors and contain natural anti-amyloid antibodies and are well tolerated. A small pilot study of passive immunotherapy using IVIg has suggested cognitive improvement. A multicenter phase III trial is ongoing and will determine whether or not this treatment can ameliorate cognitive deficits in mild-to-moderate AD. Here, we briefly review the pathogenic role of amyloid and tau in AD, as well as immunotherapeutic efforts to date. We also summarize what is known about naturally occurring anti-Aβ and tau antibodies in IVIg with a view toward explaining potential mechanisms underlying their therapeutic effects.
Collapse
Affiliation(s)
- Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA. ; Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA. ; Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | |
Collapse
|
28
|
Wu G, Sankaranarayanan S, Hsieh SHK, Simon AJ, Savage MJ. Decrease in brain soluble amyloid precursor protein β (sAPPβ) in Alzheimer's disease cortex. J Neurosci Res 2011; 89:822-32. [PMID: 21433051 DOI: 10.1002/jnr.22618] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 12/23/2010] [Accepted: 01/11/2011] [Indexed: 11/10/2022]
Abstract
Amyloid-β peptide (Aβ) is generated by sequential cleavage of the amyloid precursor protein (APP) by β-site amyloid precursor protein cleaving enzyme 1 (β-secretase, or BACE1) and γ-secretase. Several reports demonstrate increased BACE1 enzymatic activity in brain and cerebrospinal fluid (CSF) from Alzheimer's disease (AD) subjects, suggesting that an increase in BACE1-mediated cleavage of APP drives amyloid pathophysiology in AD. BACE1 cleavage of APP leads to the generation of a secreted N-terminal fragment of APP (sAPPβ). To relate BACE1 activity better to endogenous APP processing in AD and control brains, we have directly measured brain sAPPβ levels using a novel APP β-site specific enzyme-linked immunosorbent assay. We demonstrate a significant reduction in brain cortical sAPPβ levels in AD compared with control subjects. In the same brain samples, BACE1 activity was unchanged, full-length APP and sAPPα levels were significantly reduced, and Aβ peptides were significantly elevated. In conclusion, a reduction in cortical brain sAPPβ together with unchanged BACE1 activity suggests that this is due to reduced full-length APP substrate in late-stage AD subjects. These results highlight the need for multiparameter analysis of the amyloidogenic process to understand better AD pathophysiology in early vs. late-stage AD.
Collapse
Affiliation(s)
- Guoxin Wu
- Department of Neurology, Merck Research Laboratory, West Point, Pennsylvania.
| | | | | | | | | |
Collapse
|
29
|
Variations in the neuropathology of familial Alzheimer's disease. Acta Neuropathol 2009; 118:37-52. [PMID: 19306098 DOI: 10.1007/s00401-009-0521-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 03/10/2009] [Accepted: 03/11/2009] [Indexed: 12/25/2022]
Abstract
Mutations in the amyloid precursor protein (APP), presenilin 1 (PSEN1) and presenilin 2 (PSEN2) genes cause autosomal dominant familial Alzheimer's disease (AD). PSEN1 and PSEN2 are essential components of the gamma-secretase complex, which cleaves APP to affect Abeta processing. Disruptions in Abeta processing have been hypothesised to be the major cause of AD (the amyloid cascade hypothesis). These genetic cases exhibit all the classic hallmark pathologies of AD including neuritic plaques, neurofibrillary tangles (NFT), tissue atrophy, neuronal loss and inflammation, often in significantly enhanced quantities. In particular, these cases have average greater hippocampal atrophy and NFT, more significant cortical Abeta42 plaque deposition and more substantial inflammation. Enhanced cerebral Abeta40 angiopathy is a feature of many cases, but particularly those with APP mutations where it can be the dominant pathology. Additional frontotemporal neuronal loss in association with increased tau pathology appears unique to PSEN mutations, with mutations in exons 8 and 9 having enlarged cotton wool plaques throughout their cortex. The mechanisms driving these pathological differences in AD are discussed.
Collapse
|