1
|
Wang M, Hu S, Fu X, Zhou H, Yang S, Yang C. Neurosteroids: A potential target for neuropsychiatric disorders. J Steroid Biochem Mol Biol 2024; 239:106485. [PMID: 38369032 DOI: 10.1016/j.jsbmb.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Neurosteroids are steroids produced by endocrine glands and subsequently entering the brain, and also include steroids synthesis in the brain. It has been widely known that neurosteroids influence many neurological functions, including neuronal signaling, synaptic adaptations, and neuroprotective effects. In addition, abnormality in the synthesis and function of neurosteroids has been closely linked to neuropsychiatric disorders, such as Alzheimer's disease (AD), schizophrenia (SZ), and epilepsy. Given their important role in brain pathophysiology and disorders, neurosteroids offer potential therapeutic targets for a variety of neuropsychiatric diseases, and that therapeutic strategies targeting neurosteroids probably exert beneficial effects. We therefore summarized the role of neurosteroids in brain physiology and neuropsychiatric disorders, and introduced the recent findings of synthetic neurosteroid analogues for potential treatment of neuropsychiatric disorders, thereby providing insights for further research in the future.
Collapse
Affiliation(s)
- Mengyu Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xinghuo Fu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Huixuan Zhou
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
2
|
Abstract
As men grow older, circulating testosterone concentrations decline, while prevalence of cognitive impairment and dementia increase. Epidemiological studies of middle-aged and older men have demonstrated associations of lower testosterone concentrations with higher prevalence and incidence of cognitive decline and dementia, including Alzheimer's disease. In observational studies, men with prostate cancer treated by androgen deprivation therapy had a higher risk of dementia. Small intervention studies of testosterone using different measures of cognitive function have provided inconsistent results, with some suggesting improvement. A randomised placebo-controlled trial of one year's testosterone treatment conducted in 788 men aged ≥ 65 years, baseline testosterone < 9.54 nmol/L, showed an improvement in sexual function, but no improvement in cognitive function. There is a known association between diabetes and dementia risk. A randomised placebo-controlled trial of two year's testosterone treatment in 1,007 men aged 50-74 years, waist circumference ≥ 95 cm, baseline testosterone ≤ 14 nmol/L, showed an effect of testosterone in reducing type 2 diabetes risk. There were no cognitive endpoints in that trial. Additional research is warranted but at this stage lower testosterone concentrations in ageing men should be regarded as a biomarker rather than a proven therapeutic target for risk reduction of cognitive decline and dementia, including Alzheimer's disease.
Collapse
Affiliation(s)
- Bu B Yeap
- Medical School, University of Western Australia, Perth, Australia.
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Australia.
| | - Leon Flicker
- Medical School, University of Western Australia, Perth, Australia
- Western Australian Centre for Health and Ageing, University of Western Australia, Perth, Australia
- Department of Geriatric Medicine, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
3
|
Moretto E, Stuart S, Surana S, Vargas JNS, Schiavo G. The Role of Extracellular Matrix Components in the Spreading of Pathological Protein Aggregates. Front Cell Neurosci 2022; 16:844211. [PMID: 35573838 PMCID: PMC9100790 DOI: 10.3389/fncel.2022.844211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Several neurodegenerative diseases are characterized by the accumulation of aggregated misfolded proteins. These pathological agents have been suggested to propagate in the brain via mechanisms similar to that observed for the prion protein, where a misfolded variant is transferred from an affected brain region to a healthy one, thereby inducing the misfolding and/or aggregation of correctly folded copies. This process has been characterized for several proteins, such as α-synuclein, tau, amyloid beta (Aβ) and less extensively for huntingtin and TDP-43. α-synuclein, tau, TDP-43 and huntingtin are intracellular proteins, and their aggregates are located in the cytosol or nucleus of neurons. They have been shown to spread between cells and this event occurs, at least partially, via secretion of these protein aggregates in the extracellular space followed by re-uptake. Conversely, Aβ aggregates are found mainly extracellularly, and their spreading occurs in the extracellular space between brain regions. Due to the inherent nature of their spreading modalities, these proteins are exposed to components of the extracellular matrix (ECM), including glycans, proteases and core matrix proteins. These ECM components can interact with or process pathological misfolded proteins, potentially changing their properties and thus regulating their spreading capabilities. Here, we present an overview of the documented roles of ECM components in the spreading of pathological protein aggregates in neurodegenerative diseases with the objective of identifying the current gaps in knowledge and stimulating further research in the field. This could potentially lead to the identification of druggable targets to slow down the spreading and/or progression of these pathologies.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, National Research Council, CNR, Milan, Italy
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- *Correspondence: Edoardo Moretto,
| | - Skye Stuart
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sunaina Surana
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Jose Norberto S. Vargas
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
- Giampietro Schiavo,
| |
Collapse
|
4
|
Zhao Y, Wang X, Liu Y, Wang HY, Xiang J. The effects of estrogen on targeted cancer therapy drugs. Pharmacol Res 2022; 177:106131. [DOI: 10.1016/j.phrs.2022.106131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/29/2022] [Accepted: 02/10/2022] [Indexed: 10/19/2022]
|
5
|
Muthu SJ, Lakshmanan G, Shimray KW, Kaliyappan K, Sathyanathan SB, Seppan P. Testosterone Influence on Microtubule-Associated Proteins and Spine Density in Hippocampus: Implications on Learning and Memory. Dev Neurosci 2022; 44:498-507. [PMID: 35609517 DOI: 10.1159/000525038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
The thorny protrusions or spines increase the neuronal surface area, facilitate synaptic interconnections among neurons, and play an essential role in the hippocampus. Increasing evidence suggests that testosterone, the gonadal hormone, plays an important role in neurogenesis and synaptic plasticity. The role of testosterone on microtubule-associated proteins on dendritic neurite stability in the hippocampus and its impact on learning disability is not elucidated. Adult male Wistar albino rats were randomly selected for the control, castrated, castrated + testosterone, and control + testosterone groups. Bilateral orchidectomy was done, and the testosterone propionate was administered during the entire trial period, i.e., 14 days. The learning assessments were done using working/reference memory versions of the 8-arm radial maze and hippocampal tissues processed for histological and protein expressions. There were reduced expressions of microtubule-associated protein 2 (MAP2), postsynaptic density protein 95 (PSD95), and androgen receptor (AR) and increased expression of pTau in the castrated group. Conversely, the expression of MAP2, PSD95, and AR was increased, and the pTau expression was reduced in the hippocampus of the castrated rat administrated with testosterone. Androgen-depleted rats showed impaired synaptic plasticity in the hippocampus associated with contracted microtubule dynamics. Along with learning disability, there was an increased number of reference memory errors and working memory errors in castrated rats. Observations suggest that androgen regulates expression of neural tissue-specific MAPs and plays a vital role in hippocampus synaptic plasticity and that a similar mechanism may underlie neurological disorders in aging and hypogonadal men.
Collapse
Affiliation(s)
- Sakthi Jothi Muthu
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | - Ganesh Lakshmanan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | - Khayinmi Wungpam Shimray
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | - Kathiravan Kaliyappan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | | | - Prakash Seppan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| |
Collapse
|
6
|
The formation of small aggregates contributes to the neurotoxic effects of tau 45-230. Neurochem Int 2022; 152:105252. [PMID: 34856321 PMCID: PMC8712401 DOI: 10.1016/j.neuint.2021.105252] [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: 06/07/2021] [Revised: 11/08/2021] [Accepted: 11/28/2021] [Indexed: 01/03/2023]
Abstract
Intracellular deposits of hyperphosphorylated tau are commonly detected in tauopathies. Furthermore, these aggregates seem to play an important role in the pathobiology of these diseases. In the present study, we determined whether the recently identified neurotoxic tau45-230 fragment also formed aggregates in neurodegenerative disorders. The presence of such aggregates was examined in brain samples obtained from Alzheimer's disease (AD) subjects by means of Western blot analysis performed under non-denaturing conditions. Our results showed that a mixture of tau45-230 oligomers of different sizes was easily detectable in brain samples obtained from AD subjects. Our data also suggested that tau45-230 oligomers could be internalized by cultured hippocampal neurons, mainly through a clathrin-mediated mechanism, triggering their degeneration. In addition, in vitro aggregation studies showed that tau45-230 modulated full-length tau aggregation thereby inducing the formation of smaller, and potentially more toxic, aggregates of this microtubule-associated protein. Together, these data identified alternative mechanisms underlying the toxic effects of tau45-230.
Collapse
|
7
|
Limorenko G, Lashuel HA. Revisiting the grammar of Tau aggregation and pathology formation: how new insights from brain pathology are shaping how we study and target Tauopathies. Chem Soc Rev 2021; 51:513-565. [PMID: 34889934 DOI: 10.1039/d1cs00127b] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Converging evidence continues to point towards Tau aggregation and pathology formation as central events in the pathogenesis of Alzheimer's disease and other Tauopathies. Despite significant advances in understanding the morphological and structural properties of Tau fibrils, many fundamental questions remain about what causes Tau to aggregate in the first place. The exact roles of cofactors, Tau post-translational modifications, and Tau interactome in regulating Tau aggregation, pathology formation, and toxicity remain unknown. Recent studies have put the spotlight on the wide gap between the complexity of Tau structures, aggregation, and pathology formation in the brain and the simplicity of experimental approaches used for modeling these processes in research laboratories. Embracing and deconstructing this complexity is an essential first step to understanding the role of Tau in health and disease. To help deconstruct this complexity and understand its implication for the development of effective Tau targeting diagnostics and therapies, we firstly review how our understanding of Tau aggregation and pathology formation has evolved over the past few decades. Secondly, we present an analysis of new findings and insights from recent studies illustrating the biochemical, structural, and functional heterogeneity of Tau aggregates. Thirdly, we discuss the importance of adopting new experimental approaches that embrace the complexity of Tau aggregation and pathology as an important first step towards developing mechanism- and structure-based therapies that account for the pathological and clinical heterogeneity of Alzheimer's disease and Tauopathies. We believe that this is essential to develop effective diagnostics and therapies to treat these devastating diseases.
Collapse
Affiliation(s)
- Galina Limorenko
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|
8
|
Stimmell AC, Xu Z, Moseley SC, Benthem SD, Fernandez DM, Dang JV, Santos-Molina LF, Anzalone RA, Garcia-Barbon CL, Rodriguez S, Dixon JR, Wu W, Wilber AA. Tau Pathology Profile Across a Parietal-Hippocampal Brain Network Is Associated With Spatial Reorientation Learning and Memory Performance in the 3xTg-AD Mouse. FRONTIERS IN AGING 2021; 2. [PMID: 34746919 PMCID: PMC8570590 DOI: 10.3389/fragi.2021.655015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In early Alzheimer's disease (AD) spatial navigation is one of the first impairments to emerge; however, the precise cause of this impairment is unclear. Previously, we showed that, in a mouse model of tau and amyloid beta (Aβ) aggregation, getting lost represents, at least in part, a failure to use distal cues to get oriented in space and that impaired parietal-hippocampal network level plasticity during sleep may underlie this spatial disorientation. However, the relationship between tau and amyloid beta aggregation in this brain network and impaired spatial orientation has not been assessed. Therefore, we used several approaches, including canonical correlation analysis and independent components analysis tools, to examine the relationship between pathology profile across the parietal-hippocampal brain network and spatial reorientation learning and memory performance. We found that consistent with the exclusive impairment in 3xTg-AD 6-month female mice, only 6-month female mice had an ICA identified pattern of tau pathology across the parietal-hippocampal network that were positively correlated with behavior. Specifically, a higher density of pTau positive cells predicted worse spatial learning and memory. Surprisingly, despite a lack of impairment relative to controls, 3-month female, as well as 6- and 12- month male mice all had patterns of tau pathology across the parietal-hippocampal brain network that are predictive of spatial learning and memory performance. However, the direction of the effect was opposite, a negative correlation, meaning that a higher density of pTau positive cells predicted better performance. Finally, there were not significant group or region differences in M78 density at any of the ages examined and ICA analyses were not able to identify any patterns of 6E10 staining across brain regions that were significant predictors of behavioral performance. Thus, the pattern of pTau staining across the parietal-hippocampal network is a strong predictor of spatial learning and memory performance, even for mice with low levels of tau accumulation and intact spatial re-orientation learning and memory. This suggests that AD may cause spatial disorientation as a result of early tau accumulation in the parietal-hippocampal network.
Collapse
Affiliation(s)
- Alina C Stimmell
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Zishen Xu
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| | - Shawn C Moseley
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Sarah D Benthem
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Diana M Fernandez
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Jessica V Dang
- Department of Psychology, University of Florida, Gainesville, FL, United States
| | - Luis F Santos-Molina
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Rosina A Anzalone
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Carolina L Garcia-Barbon
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Stephany Rodriguez
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Jessica R Dixon
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Wei Wu
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| | - Aaron A Wilber
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
9
|
Means JC, Lopez AA, Koulen P. Estrogen Protects Optic Nerve Head Astrocytes Against Oxidative Stress by Preventing Caspase-3 Activation, Tau Dephosphorylation at Ser 422 and the Formation of Tau Protein Aggregates. Cell Mol Neurobiol 2021; 41:449-458. [PMID: 32385548 PMCID: PMC7648721 DOI: 10.1007/s10571-020-00859-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/28/2020] [Indexed: 12/15/2022]
Abstract
Glaucoma is a neurodegenerative disorder that leads to the slow degeneration of retinal ganglion cells, and results in damage to the optic nerve and concomitant vision loss. As in other disorders affecting the viability of central nervous system neurons, neurons affected by glaucoma do not have the ability to regenerate after injury. Recent studies indicate a critical role for optic nerve head astrocytes (ONHAs) in this process of retinal ganglion cell degeneration. Cleavage of tau, a microtubule stabilizing protein and constituent of neurofibrillary tangles (NFT), plays a major part in the mechanisms that lead to toxicity in CNS neurons and astrocytes. Here, we tested the hypothesis that estrogen, a pleiotropic neuro- and cytoprotectant with high efficacy in the CNS, prevents tau cleavage, and hence, protects ONHAs against cell damage caused by oxidative stress. Our results indicate that estrogen prevents caspase-3 mediated tau cleavage, and thereby decreases the levels of the resulting form of proteolytically cleaved tau protein, which leads to a decrease in NFT formation, which requires proteolytically cleaved tau protein. Overall, our data propose that by stopping the reduction of estrogen levels involved with aging the sensitivity of the optic nerve to glaucomatous damage might be reduced. Furthermore, our data suggest that therapeutic use of estrogen may be beneficial in slowing or preventing the onset or severity of neurodegenerative diseases such as glaucoma and potentially also other degenerative diseases of the CNS through direct control of posttranslational modifications of tau protein.
Collapse
Affiliation(s)
- John C Means
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO, 64108, USA
| | - Adam A Lopez
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO, 64108, USA
| | - Peter Koulen
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO, 64108, USA.
- Department of Biomedical Sciences, School of Medicine, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO, USA.
| |
Collapse
|
10
|
Guha S, Johnson GVW, Nehrke K. The Crosstalk Between Pathological Tau Phosphorylation and Mitochondrial Dysfunction as a Key to Understanding and Treating Alzheimer's Disease. Mol Neurobiol 2020; 57:5103-5120. [PMID: 32851560 PMCID: PMC7544674 DOI: 10.1007/s12035-020-02084-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common progressive neurodegenerative disorder. A defining hallmark of the AD brain is the presence of intraneuronal neurofibrillary tangles (NFTs) which are made up of abnormally modified tau, with aberrant phosphorylation being the most studied posttranslational modification (PTM). Although the accumulation of tau as NFTs is an invariant feature of the AD brain, it has become evident that these insoluble aggregates are likely not the primary pathogenic form of tau, rather soluble forms of tau with abnormal PTMs are the mediators of toxicity. The most prevalent PTM on tau is phosphorylation, with the abnormal modification of specific residues on tau playing a key role in its toxicity. Even though it is widely accepted that tau with aberrant PTMs facilitates neurodegeneration, the precise cellular mechanisms remain unknown. Nonetheless, there is an evolving conceptual framework that an important contributing factor may be selective pathological tau species compromising mitochondrial biology. Understanding the mechanisms by which tau with site-specific PTM impacts mitochondria is crucial for understanding the role tau plays in AD. Here, we provide a brief introduction to tau and its phosphorylation and function in a physiological context, followed by a discussion of the impact of soluble phosphorylated tau species on neuronal processes in general and mitochondria more specifically. We also discuss how therapeutic strategies that attenuate pathological tau species in combination with treatments that improve mitochondrial biology could be a potential therapeutic avenue to mitigate disease progression in AD and other tauopathies.
Collapse
Affiliation(s)
- Sanjib Guha
- Department of Anesthesiology & Perioperative Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| | - Gail V W Johnson
- Department of Anesthesiology & Perioperative Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester, Rochester, 14642, NY, USA
| |
Collapse
|
11
|
Gamache J, Yun Y, Chiba-Falek O. Sex-dependent effect of APOE on Alzheimer's disease and other age-related neurodegenerative disorders. Dis Model Mech 2020; 13:dmm045211. [PMID: 32859588 PMCID: PMC7473656 DOI: 10.1242/dmm.045211] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The importance of apolipoprotein E (APOE) in late-onset Alzheimer's disease (LOAD) has been firmly established, but the mechanisms through which it exerts its pathogenic effects remain elusive. In addition, the sex-dependent effects of APOE on LOAD risk and endophenotypes have yet to be explained. In this Review, we revisit the different aspects of APOE involvement in neurodegeneration and neurological diseases, with particular attention to sex differences in the contribution of APOE to LOAD susceptibility. We discuss the role of APOE in a broader range of age-related neurodegenerative diseases, and summarize the biological factors linking APOE to sex hormones, drawing on supportive findings from rodent models to identify major mechanistic themes underlying the exacerbation of LOAD-associated neurodegeneration and pathology in the female brain. Additionally, we list sex-by-genotype interactions identified across neurodegenerative diseases, proposing APOE variants as a shared etiology for sex differences in the manifestation of these diseases. Finally, we present recent advancements in 'omics' technologies, which provide a new platform for more in-depth investigations of how dysregulation of this gene affects the development and progression of neurodegenerative diseases. Collectively, the evidence summarized in this Review highlights the interplay between APOE and sex as a key factor in the etiology of LOAD and other age-related neurodegenerative diseases. We emphasize the importance of careful examination of sex as a contributing factor in studying the underpinning genetics of neurodegenerative diseases in general, but particularly for LOAD.
Collapse
Affiliation(s)
- Julia Gamache
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Young Yun
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
12
|
Vegeto E, Villa A, Della Torre S, Crippa V, Rusmini P, Cristofani R, Galbiati M, Maggi A, Poletti A. The Role of Sex and Sex Hormones in Neurodegenerative Diseases. Endocr Rev 2020; 41:5572525. [PMID: 31544208 PMCID: PMC7156855 DOI: 10.1210/endrev/bnz005] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDs) are a wide class of disorders of the central nervous system (CNS) with unknown etiology. Several factors were hypothesized to be involved in the pathogenesis of these diseases, including genetic and environmental factors. Many of these diseases show a sex prevalence and sex steroids were shown to have a role in the progression of specific forms of neurodegeneration. Estrogens were reported to be neuroprotective through their action on cognate nuclear and membrane receptors, while adverse effects of male hormones have been described on neuronal cells, although some data also suggest neuroprotective activities. The response of the CNS to sex steroids is a complex and integrated process that depends on (i) the type and amount of the cognate steroid receptor and (ii) the target cell type-either neurons, glia, or microglia. Moreover, the levels of sex steroids in the CNS fluctuate due to gonadal activities and to local metabolism and synthesis. Importantly, biochemical processes involved in the pathogenesis of NDs are increasingly being recognized as different between the two sexes and as influenced by sex steroids. The aim of this review is to present current state-of-the-art understanding on the potential role of sex steroids and their receptors on the onset and progression of major neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, and the peculiar motoneuron disease spinal and bulbar muscular atrophy, in which hormonal therapy is potentially useful as disease modifier.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze della Salute (DiSS), Università degli Studi di Milano, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Valeria Crippa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Paola Rusmini
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Riccardo Cristofani
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Mariarita Galbiati
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| |
Collapse
|
13
|
Yan Y, Yang H, Xie Y, Ding Y, Kong D, Yu H. Research Progress on Alzheimer's Disease and Resveratrol. Neurochem Res 2020; 45:989-1006. [PMID: 32162143 DOI: 10.1007/s11064-020-03007-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), a common irreversible neurodegenerative disease characterized by amyloid-β plaques, neurofibrillary tangles, and changes in tau phosphorylation, is accompanied by memory loss and symptoms of cognitive dysfunction. Increases in disease incidence due to the ageing of the population have placed a great burden on society. To date, the mechanism of AD and the identities of adequate drugs for AD prevention and treatment have eluded the medical community. It has been confirmed that phytochemicals have certain neuroprotective effects against AD. For example, some progress has been made in research on the use of resveratrol, a natural polyphenolic phytochemical, for the prevention and treatment of AD in recent years. Elucidation of the pathogenesis of AD will create a solid foundation for drug treatment. In addition, research on resveratrol, including its mechanism of action, the roles of signalling pathways and its therapeutic targets, will provide new ideas for AD treatment, which is of great significance. In this review, we discuss the possible relationships between AD and the following factors: synapses, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors (AMPARs), silent information regulator 1 (SIRT1), and estrogens. We also discuss the findings of previous studies regarding these relationships in the context of AD treatment and further summarize research progress related to resveratrol treatment.
Collapse
Affiliation(s)
- Yan Yan
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Huihuang Yang
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuxun Xie
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuanlin Ding
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Danli Kong
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| | - Haibing Yu
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
14
|
Du J, Li S, Su Y, Wang H, Liu H, Cui H. The role of extracellular matrix metalloproteinase inducer on the action of dihydrotestosterone against the cellular damage induced by Aβ 42. Mol Cell Endocrinol 2019; 498:110536. [PMID: 31401349 DOI: 10.1016/j.mce.2019.110536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 11/24/2022]
Abstract
Clinical studies have revealed that the risk of Alzheimer's disease (AD) in men is increased by age-related androgen depletion. The level of β-amyloid (Aβ) is elevated in the brains of AD patients, and Aβ is believed to play a critical role in the pathology of AD. Some studies have indicated that androgens affect AD risk by regulating the metabolism of Aβ by an unclear mechanism. In this study, we investigated the role of the extracellular matrix metalloproteinase inducer (CD147) in this action. Initially, we demonstrated that androgens positively regulate the expression of CD147 in adult male rats and SH-SY5Y cells. Furthermore, this regulation may involve androgen receptor (AR). Additionally, interference of CD147 expression decreased the clearance of Aβ in culture medium and reduced cell viability. It also affected the morphology of the cells and the expression of apoptosis-related proteins. Finally, we found that interference of CD147 expression blocked the dihydrotestosterone (DHT)-induced reduction in Aβ and the protection of cells. DHT regulates MMP-2's expression through CD147. Together, these results imply that androgen regulation of Aβ and cell protection may be affected by interfering with the expression of CD147.
Collapse
Affiliation(s)
- Juan Du
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China; Human Brain Bank, Hebei Medical University, Shijiazhuang, China
| | - Sha Li
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China; Human Brain Bank, Hebei Medical University, Shijiazhuang, China
| | - Yuhong Su
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Haidong Wang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
| | - Hanjie Liu
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huixian Cui
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China; Human Brain Bank, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
15
|
Afreen S, Ferreira A. Altered Cytoskeletal Composition and Delayed Neurite Elongation in tau 45-230-Expressing Hippocampal Neurons. Neuroscience 2019; 412:1-15. [PMID: 31158440 DOI: 10.1016/j.neuroscience.2019.05.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/15/2022]
Abstract
Calpain-mediated tau cleavage into the neurotoxic tau45-230 fragment plays an important role in Alzheimer's disease (AD). This tau fragment accumulates mainly in the cytoplasm of degenerating neurons. However, subcellular localization studies indicated that a pool of tau45-230 associates with the cytoskeleton in hippocampal neurons. In the present study, we assessed whether such localization could underlie tau45-230 neurotoxic effects. Quantitative Western blot analysis showed decreased levels of full-length tau bound to microtubules in tau45-230-expressing hippocampal neurons when compared to controls. In addition, the presence of this tau fragment induced a transient increase in tyrosinated tubulin, a marker of unstable microtubules, followed by a significant decrease in the levels of this tubulin isoform. The data obtained also showed a significant reduction in actin filaments in tau45-230-expressing neurons. These changes in microtubules and actin filaments correlated with delayed neurite elongation and axonal differentiation in the presence of this tau fragment. Together, these results suggest that tau45-230 could exert its toxic effects, at least in part, by modifying the composition of the neuronal cytoskeleton and impairing neurite elongation in neurons undergoing degeneration.
Collapse
Affiliation(s)
- Sana Afreen
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Adriana Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
16
|
Gölz C, Kirchhoff FP, Westerhorstmann J, Schmidt M, Hirnet T, Rune GM, Bender RA, Schäfer MKE. Sex hormones modulate pathogenic processes in experimental traumatic brain injury. J Neurochem 2019; 150:173-187. [PMID: 30790293 DOI: 10.1111/jnc.14678] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/26/2022]
Abstract
Clinical and animal studies have revealed sex-specific differences in histopathological and neurological outcome after traumatic brain injury (TBI). The impact of perioperative administration of sex steroid inhibitors on TBI is still elusive. Here, we subjected male and female C57Bl/6N mice to the controlled cortical impact (CCI) model of TBI and applied pharmacological inhibitors of steroid hormone synthesis, that is, letrozole (LET, inhibiting estradiol synthesis by aromatase) and finasteride (FIN, inhibiting dihydrotestosterone synthesis by 5α-reductase), respectively, starting 72 h prior CCI, and continuing for a further 48 h after CCI. Initial gene expression analyses showed that androgen (Ar) and estrogen receptors (Esr1) were sex-specifically altered 72 h after CCI. When examining brain lesion size, we found larger lesions in male than in female mice, but did not observe effects of FIN or LET treatment. However, LET treatment exacerbated neurological deficits 24 and 72 h after CCI. On the molecular level, FIN administration reduced calpain-dependent spectrin breakdown products, a proxy of excitotoxicity and disturbed Ca2+ homeostasis, specifically in males, whereas LET increased the reactive astrocyte marker glial fibrillary acid protein specifically in females. Examination of neurotrophins (brain-derived neurotrophic factor, neuronal growth factor, NT-3) and their receptors (p75NTR , TrkA, TrkB, TrkC) revealed CCI-induced down-regulation of TrkB and TrkC protein expression, which was reduced by LET in both sexes. Interestingly, FIN decreased neuronal growth factor mRNA expression and protein levels of its receptor TrkA only in males. Taken together, our data suggest a sex-specific impact on pathogenic processes in the injured brain after TBI. Sex hormones may thus modulate pathogenic processes in experimental TBI.
Collapse
Affiliation(s)
- Christina Gölz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Florian Paul Kirchhoff
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | | | - Matthias Schmidt
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Tobias Hirnet
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center, Hamburg, Germany
| | - Roland A Bender
- Institute of Neuroanatomy, University Medical Center, Hamburg, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Focus Program Translational Neurosciences, Mainz, Germany.,Research Center for Immunotherapy (FZI), Mainz, Germany
| |
Collapse
|
17
|
Mahaman YAR, Huang F, Kessete Afewerky H, Maibouge TMS, Ghose B, Wang X. Involvement of calpain in the neuropathogenesis of Alzheimer's disease. Med Res Rev 2018; 39:608-630. [PMID: 30260518 PMCID: PMC6585958 DOI: 10.1002/med.21534] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/11/2018] [Accepted: 07/29/2018] [Indexed: 01/02/2023]
Abstract
Alzheimer’s disease (AD) is the most common (60% to 80%) age‐related disease associated with dementia and is characterized by a deterioration of behavioral and cognitive capacities leading to death in few years after diagnosis, mainly due to complications from chronic illness. The characteristic hallmarks of the disease are extracellular senile plaques (SPs) and intracellular neurofibrillary tangles (NFTs) with neuropil threads, which are a direct result of amyloid precursor protein (APP) processing to Aβ, and τ hyperphosphorylation. However, many indirect underlying processes play a role in this event. One of these underlying mechanisms leading to these histological hallmarks is the uncontrolled hyperactivation of a family of cysteine proteases called calpains. Under normal physiological condition calpains participate in many processes of cells’ life and their activation is tightly controlled. However, with an increase in age, increased oxidative stress and other excitotoxicity assaults, this regulatory system becomes impaired and result in increased activation of these proteases involving them in the pathogenesis of various diseases including neurodegeneration like AD. Reviewed here is a pool of data on the implication of calpains in the pathogenesis of AD, the underlying molecular mechanism, and the potential of targeting these enzymes for AD therapeutics.
Collapse
Affiliation(s)
- Yacoubou Abdoul Razak Mahaman
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Huang
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Henok Kessete Afewerky
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tanko Mahamane Salissou Maibouge
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bishwajit Ghose
- Department of Social Medicine and Health Management, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Division of Neurodegenerative Disorders, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
18
|
Cao L, Liang Y, Liu Y, Xu Y, Wan W, Zhu C. Pseudo-phosphorylation at AT8 epitopes regulates the tau truncation at aspartate 421. Exp Cell Res 2018; 370:103-115. [PMID: 29908160 DOI: 10.1016/j.yexcr.2018.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/10/2018] [Accepted: 06/12/2018] [Indexed: 12/21/2022]
Abstract
Tau pathology in Alzheimer's disease (AD) includes hyperphosphorylation and truncation of tau. Phosphorylation at S422 is found to suppress truncation of tau at D421 that leading to the generation of ΔTau. However, the interrelation between hyperphosphorylation and generation of ΔTau in AD remains elusive. In current study, staurosporine (Stau) induced ΔTau generation by caspases in SH-SY5Y cells with tau overexpression was found to be accompanied by a dramatic dephosphorylation at S422 and the epitope of the diagnostic antibody AT8 (S199 + S202 + T205), but a moderate dephosphorylation of PHF1 (S396 + S404) epitope. Therefore, to explore the effect of AT8 epitope on tau truncation, the residues in AT8 epitope were mutated to produce "pseudo-phosphorylated" (AT8E) or "pseudo-unphosphorylated" (AT8A) tau constructs. With Stau treatment, the generation of ΔTau from tau-AT8E was significantly attenuated comparing with that from tau-AT8A, which was S422-independent in that addition of S422A mutation still preserved this effect. Interestingly, this modulatory effect was able to be reversed by addition of PHF1E mutation. Moreover, treating the crude tau extracts with recombinant caspase-3 in vitro, also showed that ΔTau level was suppressed by AT8E, and potentiated by AT8E + PHF1E. The results primarily revealed the modulating effects of phosphorylation on ΔTau generation which may have potential implications in tau pathological processes and therapeutic intervention.
Collapse
Affiliation(s)
- Lan Cao
- State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Liang
- State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunsheng Liu
- State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuxia Xu
- State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenbin Wan
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Cuiqing Zhu
- State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Manassero G, Guglielmotto M, Monteleone D, Vasciaveo V, Butenko O, Tamagno E, Arancio O, Tabaton M. Dual Mechanism of Toxicity for Extracellular Injection of Tau Oligomers versus Monomers in Human Tau Mice. J Alzheimers Dis 2018; 59:743-751. [PMID: 28671129 DOI: 10.3233/jad-170298] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mechanism of tau toxicity is still unclear. Here we report that recombinant tau oligomers and monomers, intraventricularly injected in mice with a pure human tau background, foster tau pathology through different mechanisms. Oligomeric forms of tau alter the conformation of tau in a paired helical filament-like manner. This effect occurs without tau hyperphosphorylation as well as activation of specific kinases, suggesting that oligomers of tau induce tau assembly through a nucleation effect. Monomers, in turn, induce neurodegeneration through a calpain-mediated tau cleavage that leads to accumulation of a 17 kDa neurotoxic peptide and induction of apoptotic cell death.
Collapse
Affiliation(s)
- Giusi Manassero
- Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Torino, Italy.,Department of Internal Medicine and Medical Specialties (DIMI), Unit of Geriatric Medicine, University of Genova, Genova, Italy.,IRCS San Martino-IST, University of Genova, Genova, Italy
| | - Michela Guglielmotto
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Torino, Italy.,Department of Internal Medicine and Medical Specialties (DIMI), Unit of Geriatric Medicine, University of Genova, Genova, Italy
| | - Debora Monteleone
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Torino, Italy
| | - Valeria Vasciaveo
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Torino, Italy
| | - Olena Butenko
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Torino, Italy
| | - Elena Tamagno
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Torino, Italy
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Massimo Tabaton
- Department of Internal Medicine and Medical Specialties (DIMI), Unit of Geriatric Medicine, University of Genova, Genova, Italy
| |
Collapse
|
20
|
Kulbe JR, Hall ED. Chronic traumatic encephalopathy-integration of canonical traumatic brain injury secondary injury mechanisms with tau pathology. Prog Neurobiol 2017; 158:15-44. [PMID: 28851546 PMCID: PMC5671903 DOI: 10.1016/j.pneurobio.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
In recent years, a new neurodegenerative tauopathy labeled Chronic Traumatic Encephalopathy (CTE), has been identified that is believed to be primarily a sequela of repeated mild traumatic brain injury (TBI), often referred to as concussion, that occurs in athletes participating in contact sports (e.g. boxing, American football, Australian football, rugby, soccer, ice hockey) or in military combatants, especially after blast-induced injuries. Since the identification of CTE, and its neuropathological finding of deposits of hyperphosphorylated tau protein, mechanistic attention has been on lumping the disorder together with various other non-traumatic neurodegenerative tauopathies. Indeed, brains from suspected CTE cases that have come to autopsy have been confirmed to have deposits of hyperphosphorylated tau in locations that make its anatomical distribution distinct for other tauopathies. The fact that these individuals experienced repetitive TBI episodes during their athletic or military careers suggests that the secondary injury mechanisms that have been extensively characterized in acute TBI preclinical models, and in TBI patients, including glutamate excitotoxicity, intracellular calcium overload, mitochondrial dysfunction, free radical-induced oxidative damage and neuroinflammation, may contribute to the brain damage associated with CTE. Thus, the current review begins with an in depth analysis of what is known about the tau protein and its functions and dysfunctions followed by a discussion of the major TBI secondary injury mechanisms, and how the latter have been shown to contribute to tau pathology. The value of this review is that it might lead to improved neuroprotective strategies for either prophylactically attenuating the development of CTE or slowing its progression.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States
| | - Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States.
| |
Collapse
|
21
|
Falfushynska HI, Gnatyshyna LL, Horyn O, Stoliar OB. Vulnerability of marsh frog Pelophylax ridibundus to the typical wastewater effluents ibuprofen, triclosan and estrone, detected by multi-biomarker approach. Comp Biochem Physiol C Toxicol Pharmacol 2017; 202:26-38. [PMID: 28757214 DOI: 10.1016/j.cbpc.2017.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/23/2017] [Accepted: 07/25/2017] [Indexed: 11/24/2022]
Abstract
Pharmaceutical and personal care products (PPCPs) are the environmental pollutants of growing concern. The aim of this study was to indicate the effects of typical PPCPs on the marsh frog Pelophylax ridibundus. We treated male frogs with waterborne ibuprofen (IBU, 250ng·L-1), triclosan (TCS, 500ng·L-1), or estrone (E1, 100ng·L-1) for 14days. Common vulnerability of the frogs was detected from dramatic decrease of Zn, total and metalated metallothionein (MT) concentrations, Zn/Cu ratio, the elevation of activity of glutathione-S-transferase, cathepsin D and DNA instability in the liver, the depletion of cholinesterase in the brain and cortisol in the blood plasma in all exposures. Nevertheless, lipofuscin concentration in the liver was always decreased. The groups were best distinguished by cytochrome P450 (CYP450) activity determined by ELISA. The exposure to IBU caused lesser damage, but elevated the levels of oxyradicals and glutathione (GSH and GSSG) and lysosomal membrane instability. Exposures to TCS and E1 provoked the endocrine disturbance (increased levels of vitellogenin and thyrotropin in blood plasma), decreased lactate dehydrogenase activity and increased level of pyruvate in the liver. TCS caused the increase of GSSG by 7.3 times and lactate levels. Only E1 lead to decrease of deiodinase activity in the liver, activation of CYP450 and caspase-3 and efflux of cathepsin D from lysosomes. Spectrophotometric and ELISA assays of MTs and CYP450 gave distinct results in E1-group. Broad disruption of the hormonal pathways caused by E1 could be of concern for the health status of frogs in their habitats.
Collapse
Affiliation(s)
- Halina I Falfushynska
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil National Pedagogical University, 2, M. Kryvonosa Str., Ternopil 46027, Ukraine
| | - Lesya L Gnatyshyna
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil National Pedagogical University, 2, M. Kryvonosa Str., Ternopil 46027, Ukraine; I.Ya. Horbachevsky Ternopil State Medical University, 1, Maidan Voli, Ternopil 46001, Ukraine
| | - Oksana Horyn
- I.Ya. Horbachevsky Ternopil State Medical University, 1, Maidan Voli, Ternopil 46001, Ukraine
| | - Oksana B Stoliar
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil National Pedagogical University, 2, M. Kryvonosa Str., Ternopil 46027, Ukraine.
| |
Collapse
|
22
|
Massa MG, David C, Jörg S, Berg J, Gisevius B, Hirschberg S, Linker RA, Gold R, Haghikia A. Testosterone Differentially Affects T Cells and Neurons in Murine and Human Models of Neuroinflammation and Neurodegeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2017. [PMID: 28634006 DOI: 10.1016/j.ajpath.2017.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The high female-to-male sex ratio of multiple sclerosis (MS) prevalence has continuously confounded researchers, especially in light of male patients' accelerated disease course at later stages of MS. Although multiple studies have concentrated on estrogenic mechanisms of disease modulation, fairly little attention has been paid to androgenic effects in a female system, and even fewer studies have attempted to dissociate hormonal effects on the neurodegenerative and neuroinflammatory processes of MS. Herein, we demonstrate the differential effects of hormone treatment on the acute inflammatory and chronic neurodegenerative phases of murine experimental autoimmune encephalomyelitis. Although s.c. treatment with testosterone and aromatase inhibitor applied beginning on the day of immunization ameliorated initial course of disease, similar treatment administered therapeutically exacerbated chronic disease course. Spinal cord analyses of axonal densities reflected the clinical scores of the chronic phase. In vitro, testosterone treatment not only decreased Th1 and Th17 differentiation in an aromatase-independent fashion, but also exacerbated cell death in induced pluripotent stem cell-derived primary human neurons under oxidative stress conditions in an aromatase inhibitor-dependent manner. Thus, through the alleviation of inflammatory processes and the exacerbation of neurodegenerative processes, androgens may contribute to the epidemiologic sex differentials observed in MS prevalence and course.
Collapse
Affiliation(s)
- Megan G Massa
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Christina David
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Stefanie Jörg
- Department of Neurology, Friedrich-Alexander University-Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Berg
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Barbara Gisevius
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Sarah Hirschberg
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Ralf A Linker
- Department of Neurology, Friedrich-Alexander University-Erlangen-Nuremberg, Erlangen, Germany
| | - Ralf Gold
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany.
| |
Collapse
|
23
|
Rubenstein R, Chang B, Yue JK, Chiu A, Winkler EA, Puccio AM, Diaz-Arrastia R, Yuh EL, Mukherjee P, Valadka AB, Gordon WA, Okonkwo DO, Davies P, Agarwal S, Lin F, Sarkis G, Yadikar H, Yang Z, Manley GT, Wang KKW, Cooper SR, Dams-O'Connor K, Borrasso AJ, Inoue T, Maas AIR, Menon DK, Schnyer DM, Vassar MJ. Comparing Plasma Phospho Tau, Total Tau, and Phospho Tau-Total Tau Ratio as Acute and Chronic Traumatic Brain Injury Biomarkers. JAMA Neurol 2017; 74:1063-1072. [PMID: 28738126 DOI: 10.1001/jamaneurol.2017.0655] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Importance Annually in the United States, at least 3.5 million people seek medical attention for traumatic brain injury (TBI). The development of therapies for TBI is limited by the absence of diagnostic and prognostic biomarkers. Microtubule-associated protein tau is an axonal phosphoprotein. To date, the presence of the hypophosphorylated tau protein (P-tau) in plasma from patients with acute TBI and chronic TBI has not been investigated. Objective To examine the associations between plasma P-tau and total-tau (T-tau) levels and injury presence, severity, type of pathoanatomic lesion (neuroimaging), and patient outcomes in acute and chronic TBI. Design, Setting, and Participants In the TRACK-TBI Pilot study, plasma was collected at a single time point from 196 patients with acute TBI admitted to 3 level I trauma centers (<24 hours after injury) and 21 patients with TBI admitted to inpatient rehabilitation units (mean [SD], 176.4 [44.5] days after injury). Control samples were purchased from a commercial vendor. The TRACK-TBI Pilot study was conducted from April 1, 2010, to June 30, 2012. Data analysis for the current investigation was performed from August 1, 2015, to March 13, 2017. Main Outcomes and Measures Plasma samples were assayed for P-tau (using an antibody that specifically recognizes phosphothreonine-231) and T-tau using ultra-high sensitivity laser-based immunoassay multi-arrayed fiberoptics conjugated with rolling circle amplification. Results In the 217 patients with TBI, 161 (74.2%) were men; mean (SD) age was 42.5 (18.1) years. The P-tau and T-tau levels and P-tau-T-tau ratio in patients with acute TBI were higher than those in healthy controls. Receiver operating characteristic analysis for the 3 tau indices demonstrated accuracy with area under the curve (AUC) of 1.000, 0.916, and 1.000, respectively, for discriminating mild TBI (Glasgow Coma Scale [GCS] score, 13-15, n = 162) from healthy controls. The P-tau level and P-tau-T-tau ratio were higher in individuals with more severe TBI (GCS, ≤12 vs 13-15). The P-tau level and P-tau-T-tau ratio outperformed the T-tau level in distinguishing cranial computed tomography-positive from -negative cases (AUC = 0.921, 0.923, and 0.646, respectively). Acute P-tau levels and P-tau-T-tau ratio weakly distinguished patients with TBI who had good outcomes (Glasgow Outcome Scale-Extended GOS-E, 7-8) (AUC = 0.663 and 0.658, respectively) and identified those with poor outcomes (GOS-E, ≤4 vs >4) (AUC = 0.771 and 0.777, respectively). Plasma samples from patients with chronic TBI also showed elevated P-tau levels and a P-tau-T-tau ratio significantly higher than that of healthy controls, with both P-tau indices strongly discriminating patients with chronic TBI from healthy controls (AUC = 1.000 and 0.963, respectively). Conclusions and Relevance Plasma P-tau levels and P-tau-T-tau ratio outperformed T-tau level as diagnostic and prognostic biomarkers for acute TBI. Compared with T-tau levels alone, P-tau levels and P-tau-T-tau ratios show more robust and sustained elevations among patients with chronic TBI.
Collapse
Affiliation(s)
- Richard Rubenstein
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, State University of New York Downstate Medical Center, Brooklyn
| | - Binggong Chang
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, State University of New York Downstate Medical Center, Brooklyn
| | - John K Yue
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - Allen Chiu
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, State University of New York Downstate Medical Center, Brooklyn
| | - Ethan A Winkler
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco
| | - Ava M Puccio
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Esther L Yuh
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California.,Department of Radiology, University of California, San Francisco
| | - Pratik Mukherjee
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California.,Department of Radiology, University of California, San Francisco
| | - Alex B Valadka
- Department of Neurosurgery, Virginia Commonwealth University, Richmond
| | - Wayne A Gordon
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - David O Okonkwo
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Peter Davies
- Litwin-Zucker Center for Research in Alzheimer's Disease, Feinstein Institute for Medical Research, Manhasset, New York
| | - Sanjeev Agarwal
- Department of Orthopedic Surgery and Rehabilitation Medicine, State University of New York Downstate Medical Center, Brooklyn
| | - Fan Lin
- Program for Neurotrauma, Neuroproteomics, and Biomarker Research, Department of Emergency Medicine, Psychiatry and Chemistry, University of Florida, Gainesville
| | - George Sarkis
- Program for Neurotrauma, Neuroproteomics, and Biomarker Research, Department of Emergency Medicine, Psychiatry and Chemistry, University of Florida, Gainesville.,Department of Chemistry, Faculty of Science, Alexandria University, Ibrahimia, Alexandria, Egypt
| | - Hamad Yadikar
- Program for Neurotrauma, Neuroproteomics, and Biomarker Research, Department of Emergency Medicine, Psychiatry and Chemistry, University of Florida, Gainesville.,Department of Biochemistry, Kuwait University, Khadiya, Kuwait
| | - Zhihui Yang
- Program for Neurotrauma, Neuroproteomics, and Biomarker Research, Department of Emergency Medicine, Psychiatry and Chemistry, University of Florida, Gainesville
| | - Geoffrey T Manley
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics, and Biomarker Research, Department of Emergency Medicine, Psychiatry and Chemistry, University of Florida, Gainesville
| | | | - Shelly R Cooper
- Department of Psychology, Washington University, St Louis, Missouri
| | - Kristen Dams-O'Connor
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Allison J Borrasso
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Tomoo Inoue
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco
| | - Andrew I R Maas
- Department of Neurosurgery, Antwerp University Hospital, Edegem, Belgium
| | - David K Menon
- Departments of Anesthesia and Neurocritical Care, University of Cambridge, Cambridge, England
| | | | - Mary J Vassar
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco
| |
Collapse
|
24
|
Bojar I, Pinkas J, Gujski M, Owoc A, Raczkiewicz D, Gustaw-Rothenberg K. Postmenopausal cognitive changes and androgen levels in the context of apolipoprotein E polymorphism. Arch Med Sci 2017; 13:1148-1159. [PMID: 28883857 PMCID: PMC5575214 DOI: 10.5114/aoms.2016.62869] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/25/2016] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION The focus of this study was to assess cognitive functions in relation to androgens and specifically testosterone and dehydroepiandrosterone in postmenopausal women as well as the correlation between cognitive functions and these two androgens according to polymorphism of the apolipoprotein E gene (APOE). MATERIAL AND METHODS A group of 402 women was recruited to the study (minimum 2 years after the last menstruation, follicle-stimulating hormone (FSH) more than 30 U/ml and no dementia signs on Montreal Cognitive Assessment). The computerized battery of the Central Nervous System Vital Signs test was used to diagnose cognitive functions. APOE genotyping was performed by multiplex polymerase chain reaction (PCR). Testosterone (TTE) and dehydroepiandrosterone (DHEA) in the blood serum were assessed for further statistical correlations analysis. RESULTS In the group of postmenopausal women, higher testosterone concentration was associated with lower scores for Neurocognition Index (NCI) (p = 0.028), memory (p = 0.008) and psychomotor speed (p < 0.001). Presence of at least one APOE ε4 allele potentiated testosterone's negative influence on cognitive functions (p < 0.05). Woman with a high normal level of DHEA scored significantly better in verbal (p = 0.027) and visual memory (p < 0.001) than other participants. APOE polymorphism did not modify the relationship between DHEA concentration and scores for cognitive functions. CONCLUSIONS Hormonal balance variations after menopause may influence brain processes concerned with cognition, especially memory and psychomotor speed. The observed effects may be related to androgens' influence on higher cortical functions in the changed hormonal dynamics of the postmenopausal period.
Collapse
Affiliation(s)
- Iwona Bojar
- Department for Women Health, Institute of Rural Health, Lublin, Poland
| | - Jarosław Pinkas
- School of Public Health, Center of Postgraduate Medical Education, Warsaw, Poland
| | - Mariusz Gujski
- Department of Prevention of Environmental Hazards and Allergology, Medical University of Warsaw, Warsaw, Poland
| | - Alfred Owoc
- Center for Public Health and Health Promotion, Institute of Rural Health, Lublin, Poland
| | - Dorota Raczkiewicz
- Institute of Statistics and Demography, Warsaw School of Economics, Warsaw, Poland
| | - Kasia Gustaw-Rothenberg
- Lou Ruvo Brain Wellness Center, Neurological Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Neurodegenerative Diseases, Institute of Rural Health, Lublin, Poland
| |
Collapse
|
25
|
Abstract
Several tau posttranslational modifications have been implicated in neuronal degeneration. Among them, tau fragmentation has been identified not only in brain samples obtained from Alzheimer's disease (AD) and related disorder subjects but also in AD culture and animal models. Some of these tau fragments have not been extensively studied. In contrast, data obtained recently showed that tau fragmentation mediated by enhanced or abnormal calpain, caspase 2, caspase 3, and asparagine endopeptidase activity results in the formation of toxic fragments. These cleaved tau forms induce neuronal death, synapse loss, and/or behavioral deficits. Here, we described protease activity assays and methods to study the effects of tau fragments on neuronal viability.
Collapse
Affiliation(s)
- Adriana Ferreira
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Sana Afreen
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
26
|
Song J, Jung C, Kim OY. The Novel Implication of Androgen in Diabetes-induced Alzheimer's Disease. J Lipid Atheroscler 2017. [DOI: 10.12997/jla.2017.6.2.66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Gwangju, Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju, Korea
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong-A University, Busan, Korea
| |
Collapse
|
27
|
Vintilescu CR, Afreen S, Rubino AE, Ferreira A. The Neurotoxic TAU 45-230 Fragment Accumulates in Upper and Lower Motor Neurons in Amyotrophic Lateral Sclerosis Subjects. Mol Med 2016; 22:477-486. [PMID: 27496042 PMCID: PMC5072411 DOI: 10.2119/molmed.2016.00095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/23/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and lethal neurodegenerative disease characterized by the loss of upper and lower motor neurons leading to muscle paralysis in affected individuals. Numerous mechanisms have been implicated in the death of these neurons. However, the pathobiology of this disease has not been completely elucidated. In the present study, we investigated to what extent tau cleavage and the generation of the neurotoxic tau45-230 fragment is associated with ALS. Quantitative Western blot analysis indicated that high levels of tau45-230 accumulated in lumbar and cervical spinal cord specimens obtained from ALS subjects. This neurotoxic tau fragment was also detected in ALS upper motor neurons located in the precentral gyrus. Our results also showed that tau45-230 aggregates were present in the spinal cord of ALS patients. On the other hand, this neurotoxic fragment was not generated in a mouse model of a familial form of this disease. Together, these results suggest a potential role for this neurotoxic tau fragment in the mechanisms leading to the degeneration of motor neurons in the context of sporadic ALS.
Collapse
Affiliation(s)
- Claudia R Vintilescu
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, IL 60611
| | - Sana Afreen
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, IL 60611
| | - Ashlee E Rubino
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, IL 60611
| | - Adriana Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, IL 60611
| |
Collapse
|
28
|
Cartagena CM, Mountney A, Hwang H, Swiercz A, Rammelkamp Z, Boutte AM, Shear DA, Tortella FC, Schmid KE. Subacute Changes in Cleavage Processing of Amyloid Precursor Protein and Tau following Penetrating Traumatic Brain Injury. PLoS One 2016; 11:e0158576. [PMID: 27428544 PMCID: PMC4948774 DOI: 10.1371/journal.pone.0158576] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 06/19/2016] [Indexed: 01/22/2023] Open
Abstract
Traumatic brain injury (TBI) is an established risk factor for the development of Alzheimer's disease (AD). Here the effects of severe penetrating TBI on APP and tau cleavage processing were investigated in a rodent model of penetrating ballistic-like brain injury (PBBI). PBBI was induced by stereotactically inserting a perforated steel probe through the right frontal cortex of the anesthetized rat and rapidly inflating/deflating the probe's elastic tubing into an elliptical shaped balloon to 10% of total rat brain volume causing temporary cavitation injury. Separate animals underwent probe injury (PrI) alone without balloon inflation. Shams underwent craniectomy. Brain tissue was collected acutely (4h, 24h, 3d) and subacutely (7d) post-injury and analyzed by immunoblot for full length APP (APP-FL) and APP beta c-terminal fragments (βCTFs), full length tau (tau-FL) and tau truncation fragments and at 7d for cytotoxic Beta amyloid (Aβ) peptides Aβ40 and Aβ42 analysis. APP-FL was significantly decreased at 3d and 7d following PBBI whereas APP βCTFs were significantly elevated by 4h post-injury and remained elevated through 7d post-injury. Effects on βCTFs were mirrored with PrI, albeit to a lesser extent. Aβ40 and Aβ42 were significantly elevated at 7d following PBBI and PrI. Tau-FL decreased substantially 3d and 7d post-PBBI and PrI. Importantly, a 22 kDa tau fragment (tau22), similar to that found in AD, was significantly elevated by 4h and remained elevated through 7d post-injury. Thus both APP and tau cleavage was dramatically altered in the acute and subacute periods post-injury. As cleavage of these proteins has also been implicated in AD, TBI pathology shown here may set the stage for the later development of AD or other tauopathies.
Collapse
Affiliation(s)
- Casandra M. Cartagena
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Andrea Mountney
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Hye Hwang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Adam Swiercz
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Zoe Rammelkamp
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Angela M. Boutte
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Deborah A. Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Frank C. Tortella
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Kara E. Schmid
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| |
Collapse
|
29
|
Xu Y, Martini-Stoica H, Zheng H. A seeding based cellular assay of tauopathy. Mol Neurodegener 2016; 11:32. [PMID: 27112488 PMCID: PMC4845507 DOI: 10.1186/s13024-016-0100-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/19/2016] [Indexed: 01/07/2023] Open
Abstract
Background Tauopathy is characterized by neurofibrillary tangles composed of insoluble hyperphosphorylated tau protein. Currently, cellular models that mimic neurofibrillary tangles in vitro are lacking. Previous studies indicate that neurofibrillary tangles form via a prion replication mechanism. In the present work, we establish a seeding based cellular model according to the prion hypothesis. Results We show that cellular soluble tau can be converted to insoluble tau by seeds from the brain lysate of rTg4510 mice or synthetically generated preformed tau fibrils (PFFs). The cellular insoluble tau exhibits classic features of neurofibrillary tangles. Using genetic and pharmacological methods, we demonstrate that inhibition of autophagy increases whereas enhancement of autophagy reduces insoluble tau in our seeding based cellular model. The insoluble tau can be detected and quantified by thioflavin-S staining, thus allowing us to adapt our cellular model to a high-content image-based screening platform. Conclusions Our seeding based cellular model reproduces neurofibrillary tangle pathology in vitro and serves as a useful tool for studying tauopathy and identifying tau modulators.
Collapse
Affiliation(s)
- Yin Xu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Heidi Martini-Stoica
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Interdepartmental Program of Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA.,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA. .,Interdepartmental Program of Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
30
|
Wang KKW, Yang Z, Chiu A, Lin F, Rubenstein R. Examining the Neural and Astroglial Protective Effects of Cellular Prion Protein Expression and Cell Death Protease Inhibition in Mouse Cerebrocortical Mixed Cultures. Mol Neurobiol 2015; 53:4821-32. [PMID: 26337296 DOI: 10.1007/s12035-015-9407-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 08/20/2015] [Indexed: 12/14/2022]
Abstract
Overexpression of cellular prion protein, PrP(C), has cytoprotective effects against neuronal injuries. Inhibition of cell death-associated proteases such as necrosis-linked calpain and apoptosis-linked caspase are also neuroprotective. Here, we systematically studied how PrP(C) expression levels and cell death protease inhibition affect cytotoxic challenges to both neuronal and glial cells in mouse cerebrocortical mixed cultures (CCM). Primary CCM derived from three mouse lines expressing no (PrP(C) knockout mice (PrPKO)), normal (wild-type (wt)), or high (tga20) levels of PrP(C) were subjected to necrotic challenge (calcium ionophore A23187) and apoptotic challenge (staurosporine (STS)). CCM which originated from tga20 mice provided the most robust neuron-astroglia protective effects against necrotic and early apoptotic cell death (lactate dehydrogenase (LDH) release) at 6 h but subsequently lost its cytoprotective effects. In contrast, PrPKO-derived cultures displayed elevated A23187- and STS-induced cell death at 24 h. Calpain inhibitor SNJ-1945 protected against A23187 challenge at 6 h in CCM from all three mouse lines but protected only against A23187 and STS treatments by 24 h in the PrPKO line. In parallel, caspase inhibitor Z-D-DCB protected against pro-apoptotic STS challenge at 6 and 24 h. Furthermore, we also examined αII-spectrin breakdown products (primarily from neurons) and glial fibrillary acidic protein (GFAP) breakdown products (from astroglia) as cytoskeletal proteolytic biomarkers. Overall, it appeared that both neurons and astroglial cells were less vulnerable to proteolytic attack during A23187 and STS challenges in tga20-derived cultures but more vulnerable in PrPKO-derived cultures. In addition, calpain and caspase inhibitors provide further protection against respective protease attacks on these neuronal and glial cytoskeletal proteins in CCM regardless of mouse-line origin. Lastly, some synergistic cytoprotective effects between PrP(C) expression and addition of cell death-linked protease inhibitors were also observed.
Collapse
Affiliation(s)
- Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics and Biomarkers Research, Departments of Psychiatry, Neuroscience and Physiological Science, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA.
| | - Zhihui Yang
- Program for Neurotrauma, Neuroproteomics and Biomarkers Research, Departments of Psychiatry, Neuroscience and Physiological Science, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Allen Chiu
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Avenue, Box #1213, Brooklyn, NY, 11203-2098, USA
| | - Fan Lin
- Program for Neurotrauma, Neuroproteomics and Biomarkers Research, Departments of Psychiatry, Neuroscience and Physiological Science, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Richard Rubenstein
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Avenue, Box #1213, Brooklyn, NY, 11203-2098, USA.
| |
Collapse
|
31
|
Zhang XF, Zhao YF, Zhu SW, Huang WJ, Luo Y, Chen QY, Ge LJ, Li RS, Wang JF, Sun M, Xiao ZC, Fan GH. CXCL1 Triggers Caspase-3 Dependent Tau Cleavage in Long-Term Neuronal Cultures and in the Hippocampus of Aged Mice: Implications in Alzheimer’s Disease. J Alzheimers Dis 2015; 48:89-104. [DOI: 10.3233/jad-150041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Xiao-Fang Zhang
- The Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China
| | - Yan-Feng Zhao
- Neuroinflammation DPU, GlaxoSmithKline R&D Center, Shanghai, China
| | - Shun-Wei Zhu
- Neurodegeneration DPU, GlaxoSmithKline R&D Center, Shanghai, China
| | - Wei-Jie Huang
- Neurodegeneration DPU, GlaxoSmithKline R&D Center, Shanghai, China
| | - Yan Luo
- Neurodegeneration DPU, GlaxoSmithKline R&D Center, Shanghai, China
| | - Qing-Ying Chen
- Neurodegeneration DPU, GlaxoSmithKline R&D Center, Shanghai, China
| | - Li-Jun Ge
- Department of Laboratory Animal Sciences, Platform Technology Sciences, GlaxoSmithKline R&D Center, Shanghai, China
| | - Run-Sheng Li
- Neuroinflammation DPU, GlaxoSmithKline R&D Center, Shanghai, China
| | - Jian-Fei Wang
- Department of Laboratory Animal Sciences, Platform Technology Sciences, GlaxoSmithKline R&D Center, Shanghai, China
| | - Mu Sun
- Neurodegeneration DPU, GlaxoSmithKline R&D Center, Shanghai, China
| | - Zhi-Cheng Xiao
- The Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China
- Shunxi-Monash Immune Regeneration and Neuroscience Laboratories, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Guo-Huang Fan
- Neuroinflammation DPU, GlaxoSmithKline R&D Center, Shanghai, China
- Tongji University School of Life Sciences and Technology, Shanghai, China
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
32
|
Taniuchi N, Niidome T, Sugimoto H. [Fundamental study of memory impairment and non-cognitive behavioral alterations in APPswe/PS1dE9 mice]. YAKUGAKU ZASSHI 2015; 135:323-9. [PMID: 25747232 DOI: 10.1248/yakushi.14-00226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In addition to cognitive decline, Alzheimer's disease patients also exhibit non-cognitive symptoms commonly referred to as behavioral and psychological symptoms of dementia, or BPSD. These symptoms have a serious impact on the quality of life of these patients, as well as that of their caregivers, but there are currently no effective therapies. The amyloid β-peptide (Aβ) is suspected to play a central role in the cascade leading to Alzheimer's disease, but the precise mechanisms are still incompletely known. To assess the influence of Aβ pathology on cognitive and non-cognitive behaviors, we examined locomotor activity, motor coordination, and spatial memory in male and female APPswePS1dE9 mice (Alzheimer's disease model, double transgenic mice expressing an amyloid precursor protein with Swedish mutation and a presenilin-1 with deletion of exon 9) at 5 months of age, when the mice had subtle Aβ deposits, and again at 9 months of age, when the mice had numerous Aβ deposits. Compared to wild-type mice, the male and female APPswe/PS1dE9 mice showed normal motor coordination in the rotarod test at both 5 and 9 months. In the Morris water maze test, male and female APPswe/PS1dE9 mice showed impaired spatial memory at 9 months; however, no such deficits were found at 5 months. In a locomotor activity test, male APPswe/PS1dE9 mice exhibited locomotor hyperactivity at 9 months, while females exhibited locomotor hyperactivity at both 5 and 9 months compared to the control mice. Together, these results indicate that APPswe/PS1dE9 mice developed spatial memory impairment and BPSD-like behavioral alterations resulting from Aβ accumulation.
Collapse
Affiliation(s)
- Nobuhiko Taniuchi
- Department of Neuroscience for Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University
| | | | | |
Collapse
|
33
|
Lang AE, Riherd Methner DN, Ferreira A. Neuronal degeneration, synaptic defects, and behavioral abnormalities in tau₄₅₋₂₃₀ transgenic mice. Neuroscience 2014; 275:322-39. [PMID: 24952329 DOI: 10.1016/j.neuroscience.2014.06.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 01/08/2023]
Abstract
The complement of mechanisms underlying tau pathology in neurodegenerative disorders has yet to be elucidated. Among these mechanisms, abnormal tau phosphorylation has received the most attention because neurofibrillary tangles present in Alzheimer's disease (AD) and related disorders known as tauopathies are composed of hyperphosphorylated forms of this microtubule-associated protein. More recently, we showed that calpain-mediated cleavage leading to the generation of the 17kDa tau₄₅₋₂₃₀ fragment is a conserved mechanism in these diseases. To obtain insights into the role of this fragment in neurodegeneration, we generated transgenic mice that express tau₄₅₋₂₃₀ and characterized their phenotype. Our results showed a significant increase in cell death in the hippocampal pyramidal cell layer of transgenic tau₄₅₋₂₃₀ mice when compared to wild-type controls. In addition, significant synapse loss was detected as early as six months after birth in transgenic hippocampal neurons. These synaptic changes were accompanied by alterations in the expression of the N-methyl-d-aspartate glutamate (NMDA) receptor subunits. Furthermore, functional abnormalities were detected in the transgenic mice using Morris Water Maze and fear conditioning tests. These results suggest that the accumulation of tau₄₅₋₂₃₀ is responsible, at least in part, for neuronal degeneration and some behavioral changes in AD and other tauopathies. Collectively, these data provide the first direct evidence of the toxic effects of a tau fragment biologically produced in the context of these diseases in vertebrate neurons that develop in situ.
Collapse
Affiliation(s)
- A E Lang
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - D N Riherd Methner
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - A Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States.
| |
Collapse
|
34
|
Seven A, Yüksel B, Kılıç S, Esen H, Keskin U, Ulubay M, Ozekinci M. Effect of injectable medroxyprogesterone acetate and etonogestrel implants on GABA-A and serotonin receptors in white and gray matter of the brain: experimental study in rats. Gynecol Endocrinol 2014; 30:320-4. [PMID: 24460500 DOI: 10.3109/09513590.2014.880417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to evaluate the time-dependent effect of progesterone-only contraceptives on the brain and to obtain an improved understanding of mood disorders experienced under this medication. A total of 66 Wistar albino rats were divided into three groups: etonogestrel (ENG) implant (group 1, n = 30); depot medroxyprogesterone acetate (MPA)-injectable (group 2, n = 30); and control (group 3, n = 6) groups. Groups 1 and 2 were each divided into five subgroups, which were examined every 10 d for up to 50 d after medication administration, to evaluate its time-dependent effect. There was no difference in terms of gamma-aminobutyric acid (GABA) and serotonin immunohistochemical staining in white and gray matter among the subgroups of group 1. In group 2, there was a significant decrease in serotonin receptor staining intensity in white and gray matter on day 50, when compared to the control group (p = 0.041). When the subgroups of group 2 were compared, there was a significant decrease in serotonin receptor staining intensity in white and gray matter on days 40 and 50 when compared to day 10. In conclusion, we showed that ENG and MPA have no effect on apoptosis and GABA-A receptors in the brain. We also showed that MPA has time-dependent effects on serotonin receptors, which may be a possible mechanism involved in mood disorders during long-term usage of injectable progesterone-only contraceptives.
Collapse
Affiliation(s)
- Ali Seven
- Department of Obstetrics and Gynaecology, Dumlupinar University, Faculty of Medicine , Kutahya , Turkey
| | | | | | | | | | | | | |
Collapse
|
35
|
Panizzon MS, Hauger R, Xian H, Vuoksimaa E, Spoon KM, Mendoza SP, Jacobson KC, Vasilopoulos T, Rana BK, McKenzie R, McCaffery JM, Lyons MJ, Kremen WS, Franz CE. Interaction of APOE genotype and testosterone on episodic memory in middle-aged men. Neurobiol Aging 2013; 35:1778.e1-8. [PMID: 24444806 DOI: 10.1016/j.neurobiolaging.2013.12.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 12/19/2013] [Accepted: 12/21/2013] [Indexed: 01/12/2023]
Abstract
Age-related changes in testosterone are believed to be a key component of the processes that contribute to cognitive aging in men. The APOE-ε4 allele may interact with testosterone and moderate the hormone's association with cognition. The goals of the present study were to examine the degree to which free testosterone is associated with episodic memory in a community-based sample of middle-aged men, and examine the potential interaction between free testosterone and the APOE-ε4 allele. Data were used from 717 participants in the Vietnam Era Twin Study of Aging. Average age was 55.4 years (standard deviation = 2.5). Significant positive associations were observed between free testosterone level and verbal episodic memory, as well as a significant interaction between free testosterone and APOE-ε4 status. In ε4 carriers free testosterone was positively associated with verbal episodic memory performance (story recall), whereas no association was observed in ε4 noncarriers. Results support the hypothesis that APOE-ε4 status increases susceptibility to other risk factors, such as low testosterone, which may ultimately contribute to cognitive decline or dementia.
Collapse
Affiliation(s)
- Matthew S Panizzon
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA.
| | - Richard Hauger
- Department of Psychiatry, University of California, San Diego, CA, USA; VA San Diego Healthcare System, CA, USA; Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | - Hong Xian
- Department of Biostatistics, St. Louis University, College for Public Health & Social Justice, St. Louis, MO, USA; Research Service, St. Louis Veterans Affairs Medical Center, St. Louis, MO
| | - Eero Vuoksimaa
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Kelly M Spoon
- Computational Science Research Center, San Diego State University, San Diego, CA, USA
| | - Sally P Mendoza
- Department of Psychology, University of California, Davis, CA, USA
| | | | | | - Brinda K Rana
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA
| | - Ruth McKenzie
- Department of Psychology, Boston University, Boston, MA, USA
| | - Jeanne M McCaffery
- Department of Psychiatry and Human Behavior, The Miriam Hospital and Warren Alpert School of Medicine at Brown University, Providence, RI, USA
| | - Michael J Lyons
- Department of Psychology, Boston University, Boston, MA, USA
| | - William S Kremen
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA; VA San Diego Healthcare System, CA, USA; Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | - Carol E Franz
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA
| |
Collapse
|
36
|
Kim B, Backus C, Oh S, Feldman EL. Hyperglycemia-induced tau cleavage in vitro and in vivo: a possible link between diabetes and Alzheimer's disease. J Alzheimers Dis 2013; 34:727-39. [PMID: 23254634 DOI: 10.3233/jad-121669] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multiple lines of evidence link the incidence of diabetes to the development of Alzheimer's disease (AD). Patients with diabetes have a 50 to 75% increased risk of developing AD. In parallel, AD patients have a higher than normal tendency to develop type 2 diabetes or impaired fasting glucose. Tau is the major component of neurofibrillary tangles, one of the hallmarks of AD pathology. The current study examined the effect of hyperglycemia on tau modification. Glucose treatment of rat embryonic cortical neurons results in concentration-dependent apoptosis and caspase-3 activation. These changes are well correlated with glucose time- and concentration-dependent tau cleavage. Aβ treatment induces tau cleavage and when added together with glucose, there is an additive effect on caspase activation, apoptosis, and tau cleavage. Tau cleavage is partially blocked by the caspase inhibitor, ZVAD. Cleaved tau displays a punctate staining along the neurites and colocalizes with cleaved caspase-3 in the cytoplasm. Both type 1 and type 2 diabetic mice display increased tau phosphorylation in the brain. In agreement with the effects of glucose on tau modifications in vitro, there is increased tau cleavage in the brains of ob/ob mice; however, tau cleavage is not observed in type 1 diabetic mouse brains. Our study demonstrates that hyperglycemia is one of major factors that induce tau modification in both in vitro and in vivo models of diabetes. We speculate that tau cleavage in diabetic conditions (especially in type 2 diabetes) may be a key link for the increased incidence of AD in diabetic patients.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA.
| | | | | | | |
Collapse
|
37
|
Xiao Z, Huang C, Wu J, Sun L, Hao W, Leung LK, Huang J. The neuroprotective effects of ipriflavone against H2O2 and amyloid beta induced toxicity in human neuroblastoma SH-SY5Y cells. Eur J Pharmacol 2013; 721:286-93. [DOI: 10.1016/j.ejphar.2013.09.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/25/2013] [Accepted: 09/06/2013] [Indexed: 01/15/2023]
|
38
|
Vest RS, Pike CJ. Gender, sex steroid hormones, and Alzheimer's disease. Horm Behav 2013; 63:301-7. [PMID: 22554955 PMCID: PMC3413783 DOI: 10.1016/j.yhbeh.2012.04.006] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 03/24/2012] [Accepted: 04/11/2012] [Indexed: 02/07/2023]
Abstract
Age-related loss of sex steroid hormones is a established risk factor for the development of Alzheimer's disease (AD) in women and men. While the relationships between the sex steroid hormones and AD are not fully understood, findings from both human and experimental paradigms indicate that depletion of estrogens in women and androgens in men increases vulnerability of the aging brain to AD pathogenesis. We review evidence of a wide range of beneficial neural actions of sex steroid hormones that may contribute to their hypothesized protective roles against AD. Both estrogens and androgens exert general neuroprotective actions relevant to a several neurodegenerative conditions, some in a sex-specific manner, including protection from neuron death and promotion of select aspects of neural plasticity. In addition, estrogens and androgens regulate key processes implicated in AD pathogenesis, in particular the accumulation of β-amyloid protein. We discuss evidence of hormone-specific mechanisms related to the regulation of the production and clearance of β-amyloid as critical protective pathways. Continued elucidation of these pathways promises to yield effective hormone-based strategies to delay development of AD.
Collapse
Affiliation(s)
- Rebekah S Vest
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | | |
Collapse
|
39
|
Ferreira A. Calpain dysregulation in Alzheimer's disease. ISRN BIOCHEMISTRY 2012; 2012:728571. [PMID: 25969760 PMCID: PMC4393001 DOI: 10.5402/2012/728571] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 09/12/2012] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease (AD) is characterized by the presence of senile plaques and neurofibrillary tangles in the neocortex and hippocampus of AD patients. In addition, a marked decrease in synaptic contacts has been detected in these affected brain areas. Due to its prevalence in the aging population, this disease has been the focus of numerous studies. The data obtained from those studies suggest that the mechanisms leading to the formation of the hallmark lesions of AD might be linked. One of such mechanisms seems to be the dysregulation of calcium homeostasis that results in the abnormal activation of calpains. Calpains are a family of Ca(2+)-dependent cysteine proteases that play a key role in multiple cell functions including cell development, differentiation and proliferation, axonal guidance, growth cone motility, and cell death, among others. In this paper, we briefly reviewed data on the structure of these proteases and their regulation under normal conditions. We also summarized data underscoring the participation of calpains in the neurodegenerative mechanisms associated with AD.
Collapse
Affiliation(s)
- Adriana Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Ward 8-140, Chicago, IL 60611, USA
| |
Collapse
|
40
|
Ge C, Yu M, Zhang C. G protein-coupled receptor 30 mediates estrogen-induced proliferation of primordial germ cells via EGFR/Akt/β-catenin signaling pathway. Endocrinology 2012; 153:3504-16. [PMID: 22635679 DOI: 10.1210/en.2012-1200] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In vertebrates, estrogens are required for the normal development and function of postnatal gonads. However, it remains unclear whether estrogens are able to modulate development of the fetal germ cells. Here, we show that, unexpectedly, chicken primordial germ cells (PGC) lacking estrogen receptor α/β still proliferate in response to 17β-estradiol (E(2)). This is due to the capacity of G protein-coupled receptor 30 (GPR30), existing on PGC, to directly bind E(2). Knockdown experiments suggest that GPR30 is required for E(2)-stimulated PGC proliferation. Furthermore, this estrogen-induced activation of GPR30 is revealed to occur through the Gβγ-subunit protein-dependent and through the matrix metalloproteinase-dependent transactivation of the epidermal growth factor receptor. Epidermal growth factor receptor activation results in a series of intracellular events, including activation of the phosphatidylinositol 3-kinase/serine-threonine kinase/β-catenin pathway, which are followed by the induction of c-fos, c-myc, cyclin D1/E, and B-cell lymphoma 2 expression, and the inhibition of B-cell lymphoma 2-associated X protein expression and caspase3/9 activity. This eventually leads to decreased apoptosis and increased PGC proliferation. Collectively, these findings offer novel insights into the dynamic mechanism of estrogen action on PGC proliferation and suggest that E(2)/GPR30 signaling might play an important role in regulating fetal germ cell development, particularly at the stage before sexual differentiation.
Collapse
Affiliation(s)
- Chutian Ge
- Department of Veterinary Medicine , College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, People's Republic of China
| | | | | |
Collapse
|
41
|
Wang Y, Sørensen MG, Zheng Q, Zhang C, Karsdal MA, Henriksen K. Will posttranslational modifications of brain proteins provide novel serological markers for dementias? Int J Alzheimers Dis 2012; 2012:209409. [PMID: 22779024 PMCID: PMC3388459 DOI: 10.1155/2012/209409] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/26/2012] [Indexed: 11/23/2022] Open
Abstract
Drug development for dementias is significantly hampered by the lack of easily accessible biomarkers. Fluid biomarkers of dementias provide indications of disease stage, but have little prognostic value, cannot detect early pathological changes, and can only be measured in CSF (cerebrospinal fluid) which significantly limits their applicability. In contrast, imaging based biomarkers can provide indications of probability of disease progression, yet are limited in applicability due to cost, radiation and radio-tracers. These aspects highlight the need for other approaches to the development of biomarkers of dementia, which should focus on not only providing information about pathological changes, but also on being measured easily and reproducibly. For other diseases, focus on development of assays monitoring highly specific protease-generated cleavage fragments of proteins has provided assays, which in serum or plasma have the ability to predict early pathological changes. Proteolytic processing of brain proteins, such as tau, APP, and α-synuclein, is a key pathological event in dementias. Here, we speculate that aiming biomarker development for dementias at detecting small brain protein degradation fragments of generated by brain-derived proteases specifically in blood samples could lead to the development of novel markers of disease progression, stage and importantly of treatment efficacy.
Collapse
Affiliation(s)
- Y. Wang
- Department of Biomarker Development, Nordic Bioscience A/S, Beijing 102206, China
| | - M. G. Sørensen
- Neurodegenerative Diseases, Nordic Bioscience A/S, Herlev Hovedgade 207, 2730 Herlev, Denmark
| | - Q. Zheng
- Department of Biomarker Development, Nordic Bioscience A/S, Beijing 102206, China
| | - C. Zhang
- Neurodegenerative Diseases, Nordic Bioscience A/S, Herlev Hovedgade 207, 2730 Herlev, Denmark
| | - M. A. Karsdal
- Neurodegenerative Diseases, Nordic Bioscience A/S, Herlev Hovedgade 207, 2730 Herlev, Denmark
| | - K. Henriksen
- Neurodegenerative Diseases, Nordic Bioscience A/S, Herlev Hovedgade 207, 2730 Herlev, Denmark
| |
Collapse
|
42
|
Rial D, Piermartiri TC, Duarte FS, Tasca CI, Walz R, Prediger RD. Overexpression of cellular prion protein (PrP(C)) prevents cognitive dysfunction and apoptotic neuronal cell death induced by amyloid-β (Aβ₁₋₄₀) administration in mice. Neuroscience 2012; 215:79-89. [PMID: 22537845 DOI: 10.1016/j.neuroscience.2012.04.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/02/2012] [Accepted: 04/07/2012] [Indexed: 11/28/2022]
Abstract
The cellular prion protein (PrP(C)) is a neuronal-anchored glycoprotein that has been associated with several functions in the CNS such as synaptic plasticity, learning and memory and neuroprotection. There is great interest in understanding the role of PrP(C) in the deleterious effects induced by the central accumulation of amyloid-β (Aβ) peptides, a pathological hallmark of Alzheimer's disease, but the existent results are still controversial. Here we compared the effects of a single intracerebroventricular (i.c.v.) injection of aggregated Aβ(1-40) peptide (400pmol/mouse) on the spatial learning and memory performance as well as hippocampal cell death biomarkers in adult wild type (Prnp(+/+)), PrP(C) knockout (Prnp(0/0)) and the PrP(C) overexpressing Tg-20 mice. Tg-20 mice, which present a fivefold increase in PrP(C) expression in comparison to wild type mice, were resistant to the Aβ(1-40)-induced spatial learning and memory impairments as indicated by reduced escape latencies to find the platform and higher percentage of time spent in the correct quadrant during training and probe test sessions of the water maze task. The protection against Aβ(1-40)-induced cognitive impairments observed in Tg-20 mice was accompanied by a significant decrease in the hippocampal expression of the activated caspase-3 protein and Bax/Bcl-2 ratio as well as reduced hippocampal cell damage assessed by MTT and propidium iodide incorporation assays. These findings indicate that the overexpression of PrP(C) prevents Aβ(1-40)-induced spatial learning and memory deficits in mice and that this response is mediated, at least in part, by the modulation of programed cell death pathways.
Collapse
Affiliation(s)
- D Rial
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, UFSC, Florianópolis, SC, Brazil
| | | | | | | | | | | |
Collapse
|
43
|
Nicholson AM, Wold LA, Walsh DM, Ferreira A. β-Amyloid carrying the Dutch mutation has diverse effects on calpain-mediated toxicity in hippocampal neurons. Mol Med 2012; 18:178-85. [PMID: 22160219 DOI: 10.2119/molmed.2011.00366] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 11/28/2011] [Indexed: 11/06/2022] Open
Abstract
Hereditary cerebral hemorrhage with amyloidosis-Dutch type is a disorder associated with a missense mutation (E693Q) in the β-amyloid (Aβ)-coding region of the amyloid precursor protein (APP). This familial disease is characterized by cognitive deficits secondary to intracerebral hemorrhage and, in some cases, progressive Alzheimer's disease (AD)-like dementia. Although this mutation was the first ever reported in the human APP gene, little is known about the molecular mechanisms underlying the direct toxic effects of this mutated Aβ on central neurons. In the present study, we assessed the role of calpain-mediated toxicity in such effects using an AD primary culture model system. Our results showed that Dutch mutant Aβ (E22Q) induced calpain-mediated cleavage of dynamin 1 and a significant decrease in synaptic contacts in mature hippocampal cultures. These synaptic deficits were similar to those induced by wild-type (WT) Aβ. In contrast, calpain-mediated tau cleavage leading to the generation of a 17-kDa neurotoxic fragment, as well as neuronal death, were significantly reduced in E22Q Aβ-treated neurons when compared with WT Aβ-treated ones. This complex regulation of the calpain-mediated toxicity pathway by E22Q Aβ could have some bearing in the pathobiology of this familial AD form.
Collapse
Affiliation(s)
- Alexandra M Nicholson
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | | | | | | |
Collapse
|
44
|
Vicencio JM, Estrada M, Galvis D, Bravo R, Contreras AE, Rotter D, Szabadkai G, Hill JA, Rothermel BA, Jaimovich E, Lavandero S. Anabolic androgenic steroids and intracellular calcium signaling: a mini review on mechanisms and physiological implications. Mini Rev Med Chem 2011; 11:390-8. [PMID: 21443511 DOI: 10.2174/138955711795445880] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 01/21/2011] [Indexed: 02/05/2023]
Abstract
Increasing evidence suggests that nongenomic effects of testosterone and anabolic androgenic steroids (AAS) operate concertedly with genomic effects. Classically, these responses have been viewed as separate and independent processes, primarily because nongenomic responses are faster and appear to be mediated by membrane androgen receptors, whereas long-term genomic effects are mediated through cytosolic androgen receptors regulating transcriptional activity. Numerous studies have demonstrated increases in intracellular Ca2+ in response to AAS. These Ca2+ mediated responses have been seen in a diversity of cell types, including osteoblasts, platelets, skeletal muscle cells, cardiac myocytes and neurons. The versatility of Ca2+ as a second messenger provides these responses with a vast number of pathophysiological implications. In cardiac cells, testosterone elicits voltage-dependent Ca2+ oscillations and IP3R-mediated Ca2+ release from internal stores, leading to activation of MAPK and mTOR signaling that promotes cardiac hypertrophy. In neurons, depending upon concentration, testosterone can provoke either physiological Ca2+ oscillations, essential for synaptic plasticity, or sustained, pathological Ca2+ transients that lead to neuronal apoptosis. We propose therefore, that Ca2+ acts as an important point of crosstalk between nongenomic and genomic AAS signaling, representing a central regulator that bridges these previously thought to be divergent responses.
Collapse
Affiliation(s)
- J M Vicencio
- Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences/Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Reinecke JB, DeVos SL, McGrath JP, Shepard AM, Goncharoff DK, Tait DN, Fleming SR, Vincent MP, Steinhilb ML. Implicating calpain in tau-mediated toxicity in vivo. PLoS One 2011; 6:e23865. [PMID: 21858230 PMCID: PMC3157467 DOI: 10.1371/journal.pone.0023865] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 07/26/2011] [Indexed: 11/19/2022] Open
Abstract
Alzheimer's disease and other related neurodegenerative disorders known as tauopathies are characterized by the accumulation of abnormally phosphorylated and aggregated forms of the microtubule-associated protein tau. Several laboratories have identified a 17 kD proteolytic fragment of tau in degenerating neurons and in numerous cell culture models that is generated by calpain cleavage and speculated to contribute to tau toxicity. In the current study, we employed a Drosophila tauopathy model to investigate the importance of calpain-mediated tau proteolysis in contributing to tau neurotoxicity in an animal model of human neurodegenerative disease. We found that mutations that disrupted endogenous calpainA or calpainB activity in transgenic flies suppressed tau toxicity. Expression of a calpain-resistant form of tau in Drosophila revealed that mutating the putative calpain cleavage sites that produce the 17 kD fragment was sufficient to abrogate tau toxicity in vivo. Furthermore, we found significant toxicity in the fly retina associated with expression of only the 17 kD tau fragment. Collectively, our data implicate calpain-mediated proteolysis of tau as an important pathway mediating tau neurotoxicity in vivo.
Collapse
Affiliation(s)
- James B. Reinecke
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Sarah L. DeVos
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - James P. McGrath
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Amanda M. Shepard
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Dustin K. Goncharoff
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Don N. Tait
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Samantha R. Fleming
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Michael P. Vincent
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Michelle L. Steinhilb
- Department of Biology, Central Michigan University, Mount Pleasant, Michigan, United States of America
- * E-mail:
| |
Collapse
|
46
|
Ferreira A, Bigio EH. Calpain-mediated tau cleavage: a mechanism leading to neurodegeneration shared by multiple tauopathies. Mol Med 2011; 17:676-85. [PMID: 21442128 DOI: 10.2119/molmed.2010.00220] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Accepted: 03/18/2011] [Indexed: 12/14/2022] Open
Abstract
Tau dysfunction has been associated with a host of neurodegenerative diseases called tauopathies. These diseases share, as a common pathological hallmark, the presence of intracellular aggregates of hyperphosphorylated tau in affected brain areas. Aside from tau hyperphosphorylation, little is known about the role of other posttranslational modifications in tauopathies. Recently, we obtained data suggesting that calpain-mediated tau cleavage leading to the generation of a neurotoxic tau fragment might play an important role in Alzheimer's disease. In the current study, we assessed the presence of this tau fragment in several tauopathies. Our results show high levels of the 17-kDa tau fragment and enhanced calpain activity in the temporal cortex of AD patients and in brain samples obtained from patients with other tauopathies. In addition, our data suggest that this fragment could partially inhibit tau aggregation. Conversely, tau aggregation might prevent calpain-mediated cleavage, establishing a feedback circuit that might lead to the accumulation of this toxic tau fragment. Collectively, these data suggest that the mechanism underlying the generation of the 17-kDa neurotoxic tau fragment might be part of a conserved pathologic process shared by multiple tauopathies.
Collapse
Affiliation(s)
- Adriana Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| | | |
Collapse
|
47
|
Nakajima T, Ochi S, Oda C, Ishii M, Ogawa K. Ischemic preconditioning attenuates of ischemia-induced degradation of spectrin and tau: implications for ischemic tolerance. Neurol Sci 2010; 32:229-39. [DOI: 10.1007/s10072-010-0359-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 06/15/2010] [Indexed: 10/19/2022]
|
48
|
Nicholson AM, Ferreira A. CHOLESTEROL AND NEURONAL SUSCEPTIBILITY TO BETA-AMYLOID TOXICITY. COGNITIVE SCIENCES 2010; 5:35-56. [PMID: 25339981 PMCID: PMC4203449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Alzheimer's disease (AD) is a devastating neurocognitive disorder rapidly growing across the elderly population. Although few cases arise due to genetic mutations, sporadic AD is the most common form of this disease. Therefore, there is a continuing research effort to discover a unifying cause of this form of AD. To date, the only strong genetic correlate to the sporadic AD is inheritance of the apolipoprotein E4 (ApoE4) allele, whose encoded protein is involved in cholesterol transport in the central nervous system. This genetic link has prompted a series of studies on the potential molecular mechanisms by which cholesterol could modulate neuronal degeneration in the context of AD. In this review, we discussed the involvement of cholesterol in the production of the pathological hallmarks of the disease and how it might alter the susceptibility of cells to AD-related insult. Finally, we discussed the use of cholesterol-lowering drugs as a potential preventative approach in AD.
Collapse
Affiliation(s)
| | - Adriana Ferreira
- Correspondence should be addressed to: Adriana Ferreira, M.D., Ph. D., Cell and Molecular Biology Department Northwestern University, Ward Building 8-140, 303 East Chicago Avenue, Chicago, Illinois 60611, Phone: (312) 503-0597, Fax: (312) 503-7345,
| |
Collapse
|
49
|
de Calignon A, Fox LM, Pitstick R, Carlson GA, Bacskai BJ, Spires-Jones TL, Hyman BT. Caspase activation precedes and leads to tangles. Nature 2010; 464:1201-4. [PMID: 20357768 DOI: 10.1038/nature08890] [Citation(s) in RCA: 390] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 02/10/2010] [Indexed: 01/07/2023]
Abstract
Studies of post-mortem tissue have shown that the location of fibrillar tau deposits, called neurofibrillary tangles (NFT), matches closely with regions of massive neuronal death, severe cytological abnormalities, and markers of caspase activation and apoptosis, leading to the idea that tangles cause neurodegeneration in Alzheimer's disease and tau-related frontotemporal dementia. However, using in vivo multiphoton imaging to observe tangles and activation of executioner caspases in living tau transgenic mice (Tg4510 strain), we find the opposite: caspase activation occurs first, and precedes tangle formation by hours to days. New tangles form within a day. After a new tangle forms, the neuron remains alive and caspase activity seems to be suppressed. Similarly, introduction of wild-type 4-repeat tau (tau-4R) into wild-type animals triggered caspase activation, tau truncation and tau aggregation. Adeno-associated virus-mediated expression of a construct mimicking caspase-cleaved tau into wild-type mice led to the appearance of intracellular aggregates, tangle-related conformational- and phospho-epitopes, and the recruitment of full-length endogenous tau to the aggregates. On the basis of these data, we propose a new model in which caspase activation cleaves tau to initiate tangle formation, then truncated tau recruits normal tau to misfold and form tangles. Because tangle-bearing neurons are long-lived, we suggest that tangles are 'off pathway' to acute neuronal death. Soluble tau species, rather than fibrillar tau, may be the critical toxic moiety underlying neurodegeneration.
Collapse
Affiliation(s)
- Alix de Calignon
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Alzheimer's Disease Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Saito M, Chakraborty G, Mao RF, Paik SM, Vadasz C, Saito M. Tau phosphorylation and cleavage in ethanol-induced neurodegeneration in the developing mouse brain. Neurochem Res 2010; 35:651-9. [PMID: 20049527 DOI: 10.1007/s11064-009-0116-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2009] [Indexed: 11/29/2022]
Abstract
Previous studies indicated that ethanol-induced neurodegeneration in postnatal day 7 (P7) mice, widely used as a model for the fetal alcohol spectrum disorders, was accompanied by glycogen synthase kinase-3beta (GSK-3beta) and caspase-3 activation. Presently, we examined whether tau, a microtubule associated protein, is modified by GSK-3beta and caspase-3 in ethanol-treated P7 mouse forebrains. We found that ethanol increased phosphorylated tau recognized by the paired helical filament (PHF)-1 antibody and by the antibody against tau phosphorylated at Ser199. Ethanol also generated tau fragments recognized by an antibody against caspase-cleaved tau (C-tau). C-tau was localized in neurons bearing activated caspase-3 and fragmented nuclei. Over time, cell debris and degenerated projections containing C-tau appeared to be engulfed by activated microglia. A caspase-3 inhibitor partially blocked C-tau formation. Lithium, a GSK-3beta inhibitor, blocked ethanol-induced caspase-3 activation, phosphorylated tau elevation, C-tau formation, and microglial activation. These results indicate that tau is phosphorylated by GSK-3beta and cleaved by caspase-3 during ethanol-induced neurodegeneration in the developing brain.
Collapse
Affiliation(s)
- Mariko Saito
- Laboratory of Neurobehavior Genetics, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | | | | | | | | | | |
Collapse
|