1
|
Mittra N, He S, Bao H, Bhattacharjee A, Dodds SG, Dupree JL, Han X. Sulfatide deficiency-induced astrogliosis and myelin lipid dyshomeostasis are independent of Trem2-mediated microglial activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623651. [PMID: 39605561 PMCID: PMC11601472 DOI: 10.1101/2024.11.14.623651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Disrupted lipid homeostasis and neuroinflammation often co-exist in neurodegenerative disorders including Alzheimer's disease (AD). However, the intrinsic connection and causal relationship between these deficits remain elusive. Our previous studies show that the loss of sulfatide (ST), a class of myelin-enriched lipids, causes AD-like neuroinflammatory responses, cognitive impairment, bladder enlargement, as well as lipid dyshomeostasis. To better understand the relationship between neuroinflammation and lipid disruption induced by ST deficiency, we established a ST-deficient mouse model with constitutive Trem2 knockout and studied the impact of Trem2 in regulating ST deficiency-induced microglia-mediated neuroinflammation, astrocyte activation and lipid disruption. Our study demonstrates that Trem2 regulates ST deficiency-induced microglia-mediated neuroinflammatory pathways and astrogliosis at the transcriptomic level, but not astrocyte activation at the protein level, suggesting that Trem2 is indispensable for ST deficiency-induced microglia-mediated neuroinflammation but not astrogliosis. Meanwhile, ST loss-induced lipidome disruption and free water retention were consistently observed in the absence of Trem2 . Collectively, these results emphasize the essential role of Trem2 in mediating lipid loss-associated microglia-mediated neuroinflammation, but not both astrogliosis and myelin lipid disruption. Moreover, we demonstrated that attenuating neuroinflammation has a limited impact on brain ST loss-induced lipidome alteration or AD-like peripheral disorders. Our findings suggest that preserving lipidome and astrocyte balance may be crucial in decelerating the progression of AD.
Collapse
|
2
|
McKenna MC, Kleinerova J, Power A, Garcia-Gallardo A, Tan EL, Bede P. Quantitative and Computational Spinal Imaging in Neurodegenerative Conditions and Acquired Spinal Disorders: Academic Advances and Clinical Prospects. BIOLOGY 2024; 13:909. [PMID: 39596864 PMCID: PMC11592215 DOI: 10.3390/biology13110909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
Introduction: Quantitative spinal cord imaging has facilitated the objective appraisal of spinal cord pathology in a range of neurological conditions both in the academic and clinical setting. Diverse methodological approaches have been implemented, encompassing a range of morphometric, diffusivity, susceptibility, magnetization transfer, and spectroscopy techniques. Advances have been fueled both by new MRI platforms and acquisition protocols as well as novel analysis pipelines. The quantitative evaluation of specific spinal tracts and grey matter indices has the potential to be used in diagnostic and monitoring applications. The comprehensive characterization of spinal disease burden in pre-symptomatic cohorts, in carriers of specific genetic mutations, and in conditions primarily associated with cerebral disease, has contributed important academic insights. Methods: A narrative review was conducted to examine the clinical and academic role of quantitative spinal cord imaging in a range of neurodegenerative and acquired spinal cord disorders, including hereditary spastic paraparesis, hereditary ataxias, motor neuron diseases, Huntington's disease, and post-infectious or vascular disorders. Results: The clinical utility of specific methods, sample size considerations, academic role of spinal imaging, key radiological findings, and relevant clinical correlates are presented in each disease group. Conclusions: Quantitative spinal cord imaging studies have demonstrated the feasibility to reliably appraise structural, microstructural, diffusivity, and metabolic spinal cord alterations. Despite the notable academic advances, novel acquisition protocols and analysis pipelines are yet to be implemented in the clinical setting.
Collapse
Affiliation(s)
- Mary Clare McKenna
- Computational Neuroimaging Group, Trinity College Dublin, 152-160 Pearse St, 2 D02 R590 Dublin, Ireland
- Department of Neurology, St James’s Hospital, James St, 8 D08 NHY1 Dublin, Ireland
| | - Jana Kleinerova
- Computational Neuroimaging Group, Trinity College Dublin, 152-160 Pearse St, 2 D02 R590 Dublin, Ireland
| | - Alan Power
- Computational Neuroimaging Group, Trinity College Dublin, 152-160 Pearse St, 2 D02 R590 Dublin, Ireland
- Department of Neurology, St James’s Hospital, James St, 8 D08 NHY1 Dublin, Ireland
| | - Angela Garcia-Gallardo
- Computational Neuroimaging Group, Trinity College Dublin, 152-160 Pearse St, 2 D02 R590 Dublin, Ireland
- Department of Neurology, St James’s Hospital, James St, 8 D08 NHY1 Dublin, Ireland
| | - Ee Ling Tan
- Computational Neuroimaging Group, Trinity College Dublin, 152-160 Pearse St, 2 D02 R590 Dublin, Ireland
| | - Peter Bede
- Computational Neuroimaging Group, Trinity College Dublin, 152-160 Pearse St, 2 D02 R590 Dublin, Ireland
- Department of Neurology, St James’s Hospital, James St, 8 D08 NHY1 Dublin, Ireland
| |
Collapse
|
3
|
Emmenegger TM, Seiler R, Unschuld PG, Freund P, Klohs J. Progressive cervical cord atrophy parallels cognitive decline in Alzheimer's disease. Sci Rep 2024; 14:21595. [PMID: 39284823 PMCID: PMC11405669 DOI: 10.1038/s41598-024-67389-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 07/10/2024] [Indexed: 09/22/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive episodic memory dysfunction. A prominent hallmark of AD is gradual brain atrophy. Despite extensive research on brain pathology, the understanding of spinal cord pathology in AD and its association with cognitive decline remains understudied. We analyzed serial magnetic resonance imaging (MRI) scans from the ADNI data repository to assess whether progressive cord atrophy is associated with clinical worsening. Cervical cord morphometry was measured in 45 patients and 49 cognitively normal controls (CN) at two time points over 1.5 years. Regression analysis examined associations between cord atrophy rate and cognitive worsening. Cognitive and functional activity performance declined in patients during follow-up. Compared with controls, patients showed a greater rate of decline of the anterior-posterior width of the cross-sectional cord area per month (- 0.12%, p = 0.036). Worsening in the mini-mental state examination (MMSE), clinical dementia rating (CDR), and functional assessment questionnaire (FAQ) was associated with faster rates of cord atrophy (MMSE: r = 0.320, p = 0.037; CDR: r = - 0.361, p = 0.017; FAQ: r = - 0.398, p = 0.029). Progressive cord atrophy occurs in AD patients; its rate over time being associated with cognitive and functional activity decline.
Collapse
Affiliation(s)
- Tim M Emmenegger
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland
| | - Raoul Seiler
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Wolfgang-Pauli-Strasse 27, 8093, Zurich, Switzerland
| | - Paul G Unschuld
- Department of Psychiatry, University of Geneva (UniGE), 1205, Geneva, Switzerland
- Division of Geriatric Psychiatry, University Hospitals of Geneva (HUG), 1226, Thônex, Switzerland
| | - Patrick Freund
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland.
- Zurich Neuroscience Center (ZNZ), Winterthurer Strasse 190, 8057, Zürich, Switzerland.
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Wolfgang-Pauli-Strasse 27, 8093, Zurich, Switzerland.
- Zurich Neuroscience Center (ZNZ), Winterthurer Strasse 190, 8057, Zürich, Switzerland.
| |
Collapse
|
4
|
Liao K, Lou Q. Alzheimer's disease increases the risk of erectile dysfunction independent of cardiovascular diseases: A mendelian randomization study. PLoS One 2024; 19:e0303338. [PMID: 38870203 PMCID: PMC11175418 DOI: 10.1371/journal.pone.0303338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/23/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Previous research has underscored the correlation between Alzheimer's disease (AD) and erectile dysfunction (ED). However, due to inherent limitations of observational studies, the causative relationship remains inconclusive. METHODS Utilizing publicly available data from genome-wide association studies (GWAS) summary statistics, this study probed the potential causal association between AD and ED using univariate Mendelian randomization (MR). Further, the multivariable MR assessed the confounding effects of six cardiovascular diseases (CVDs). The primary approach employed was inverse variance weighted (IVW), supplemented by three additional methods. A series of sensitivity analyses were conducted to ensure the robustness of the results. RESULTS In the forward MR analysis, the IVW method revealed causal evidence of genetically predicted AD being a risk factor for ED (OR = 1.077, 95% CI 1.007∼1.152, P = 0.031). Reverse analysis did not demonstrate any causal evidence linking ED to AD (OR = 1.018, 95% CI 0.974∼1.063, P = 0.430). Multivariable MR analysis showed that after adjusting for coronary heart disease (OR = 1.082, 95% CI 0.009∼1.160, P = 0.027), myocardial infarction (OR = 1.085, 95% CI 1.012∼1.163, P = 0.022), atrial fibrillation (OR = 1.076, 95% CI 1.002∼1.154, P = 0.043), heart failure (OR = 1.103, 95% CI 1.024∼1.188, P = 0.010), ischemic stroke (OR = 1.079, 95% CI 1.009∼1.154, P = 0.027), hypertension (OR = 1.092, 95% CI 1.011∼1.180, P = 0.025), and all models (OR = 1.115, 95% CI 1.024∼1.214, P = 0.012), the causal association between AD and ED persisted. Sensitivity analyses confirmed the absence of pleiotropy, heterogeneity, and outliers, validating the robustness of our results (P > 0.05). CONCLUSIONS This MR study consistently evidences a causal effect of genetically predicted AD on the risk of ED, independent of certain CVDs, yet offers no evidence for a reverse effect from ED.
Collapse
Affiliation(s)
- Kaisen Liao
- Department of Urology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Qiang Lou
- Department of Andrology, the Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
5
|
Zhang S, Chen Y, Wang Y, Wang H, Yao D, Chen G. Tau Accumulation in the Spinal Cord Contributes to Chronic Inflammatory Pain by Upregulation of IL-1β and BDNF. Neurosci Bull 2024; 40:466-482. [PMID: 38148427 PMCID: PMC11003936 DOI: 10.1007/s12264-023-01152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/09/2023] [Indexed: 12/28/2023] Open
Abstract
Microtubule-associated protein Tau is responsible for the stabilization of neuronal microtubules under normal physiological conditions. Much attention has been focused on Tau's contribution to cognition, but little research has explored its role in emotions such as pain, anxiety, and depression. In the current study, we found a significant increase in the levels of p-Tau (Thr231), total Tau, IL-1β, and brain-derived neurotrophic factor (BDNF) on day 7 after complete Freund's adjuvant (CFA) injection; they were present in the vast majority of neurons in the spinal dorsal horn. Microinjection of Mapt-shRNA recombinant adeno-associated virus into the spinal dorsal cord alleviated CFA-induced inflammatory pain and inhibited CFA-induced IL-1β and BDNF upregulation. Importantly, Tau overexpression was sufficient to induce hyperalgesia by increasing the expression of IL-1β and BDNF. Furthermore, the activation of glycogen synthase kinase 3 beta partly contributed to Tau accumulation. These findings suggest that Tau in the dorsal horn could be a promising target for chronic inflammatory pain therapy.
Collapse
Affiliation(s)
- Shuxia Zhang
- Department of Anesthesiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Yeru Chen
- Department of Anesthesiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Yongjie Wang
- Key Laboratory of Elemene Anti-Cancer Medicine of Zhejiang Province and Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, 311121, China
| | - Hongwei Wang
- Department of Anesthesiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Dandan Yao
- Department of Anesthesiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Gang Chen
- Department of Anesthesiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
6
|
Sabbir MG. Cholinergic Receptor Muscarinic 1 Co-Localized with Mitochondria in Cultured Dorsal Root Ganglion Neurons, and Its Deletion Disrupted Mitochondrial Ultrastructure in Peripheral Neurons: Implications in Alzheimer's Disease. J Alzheimers Dis 2024; 98:247-264. [PMID: 38427478 DOI: 10.3233/jad-230883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Loss of Cholinergic Receptor Muscarinic 1 (CHRM1) has been linked to the pathogenesis of Alzheimer's disease (AD). Our recent study found significantly lower CHRM1 protein levels in AD patient cortices, linked to reduced survival. Furthermore, using knockout mice (Chrm1-/-) we demonstrated that deletion of Chrm1 alters cortical mitochondrial structure and function, directly establishing a connection between its loss and mitochondrial dysfunction in the context of AD. While CHRM1's role in the brain has been extensively investigated, its impact on peripheral neurons in AD remains a crucial area of research, especially considering reported declines in peripheral nerve conduction among AD patients. Objective The objective was to characterize Chrm1 localization and mitochondrial deficits in Chrm1-/- dorsal root ganglion (DRG) neurons. Methods Recombinant proteins tagged with Green or Red Fluorescent Protein (GFP/RFP) were transiently expressed to investigate the localization of Chrm1 and mitochondria, as well as mitochondrial movement in the neurites of cultured primary mouse DRG neurons, using confocal time-lapse live cell imaging. Transmission electron microscopy was performed to examine the ultrastructure of mitochondria in both wild-type and Chrm1-/- DRGs. Results Fluorescence imaging revealed colocalization and comigration of N-terminal GFP-tagged Chrm1 and mitochondrial localization signal peptide-tagged RFP-labelled mitochondria in the DRGs neurons. A spectrum of mitochondrial structural abnormalities, including disruption and loss of cristae was observed in 87% neurons in Chrm1-/- DRGs. Conclusions This study suggests that Chrm1 may be localized in the neuronal mitochondria and loss of Chrm1 in peripheral neurons causes sever mitochondrial structural aberrations resembling AD pathology.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Department of Psychology and Neuroscience, Collegeof Psychology, Nova Southeastern University, Fort Lauderdale, FL, USA
- Alzo Biosciences Inc., San Diego, CA, USA
| |
Collapse
|
7
|
Hamsafar Y, Chen Q, Borowsky AD, Beach TG, Serrano GE, Sue LI, Adler CH, Walker DG, Dugger BN. Biochemical analyses of tau and other neuronal markers in the submandibular gland and frontal cortex across stages of Alzheimer disease. Neurosci Lett 2023; 810:137330. [PMID: 37330193 PMCID: PMC11006283 DOI: 10.1016/j.neulet.2023.137330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 02/14/2023] [Accepted: 06/05/2023] [Indexed: 06/19/2023]
Abstract
Hyperphosphorylation of the microtubule-associated protein tau is hypothesized to lead to the development of neurofibrillary tangles in select brain regions during normal aging and in Alzheimer disease (AD). The distribution of neurofibrillary tangles is staged by its involvement starting in the transentorhinal regions of the brain and in final stages progress to neocortices. However, it has also been determined neurofibrillary tangles can extend into the spinal cord and select tau species are found in peripheral tissues and this may be depended on AD disease stage. To further understand the relationships of peripheral tissues to AD, we utilized biochemical methods to evaluate protein levels of total tau and phosphorylated tau (p-tau) as well as other neuronal proteins (i.e., tyrosine hydroxylase (TH), neurofilament heavy chain (NF-H), and microtubule-associated protein 2 (MAP2)) in the submandibular gland and frontal cortex of human cases across different clinicopathological stages of AD (n = 3 criteria not met or low, n = 6 intermediate, and n = 9 high likelihood that dementia is due to AD based on National Institute on Aging-Reagan criteria). We report differential protein levels based on the stage of AD, anatomic specific tau species, as well as differences in TH and NF-H. In addition, exploratory findings were made of the high molecular weight tau species big tau that is unique to peripheral tissues. Although sample sizes were small, these findings are, to our knowledge, the first comparison of these specific protein changes in these tissues.
Collapse
Affiliation(s)
- Yamah Hamsafar
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 4400 V Street, Sacramento, CA 95817, USA
| | - Qian Chen
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 4400 V Street, Sacramento, CA 95817, USA
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 4400 V Street, Sacramento, CA 95817, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr., Sun City, AZ 95351, USA
| | - Geidy E Serrano
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr., Sun City, AZ 95351, USA
| | - Lucia I Sue
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr., Sun City, AZ 95351, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E. Shea Blvd., Scottsdale, AZ 85259, USA
| | - Douglas G Walker
- School of Life Sciences and Biodesign Institute, Arizona State University, 1151 S. Forest Ave., Tempe, AZ 85281, USA
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 4400 V Street, Sacramento, CA 95817, USA.
| |
Collapse
|
8
|
He S, Qiu S, Pan M, Palavicini JP, Wang H, Li X, Bhattacharjee A, Barannikov S, Bieniek KF, Dupree JL, Han X. Central nervous system sulfatide deficiency as a causal factor for bladder disorder in Alzheimer's disease. Clin Transl Med 2023; 13:e1332. [PMID: 37478300 PMCID: PMC10361545 DOI: 10.1002/ctm2.1332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/02/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Despite being a brain disorder, Alzheimer's disease (AD) is often accompanied by peripheral organ dysregulations (e.g., loss of bladder control in late-stage AD), which highly rely on spinal cord coordination. However, the causal factor(s) for peripheral organ dysregulation in AD remain elusive. METHODS The central nervous system (CNS) is enriched in lipids. We applied quantitative shotgun lipidomics to determine lipid profiles of human AD spinal cord tissues. Additionally, a CNS sulfatide (ST)-deficient mouse model was used to study the lipidome, transcriptome and peripheral organ phenotypes of ST loss. RESULTS We observed marked myelin lipid reduction in the spinal cord of AD subjects versus cognitively normal individuals. Among which, levels of ST, a myelin-enriched lipid class, were strongly and negatively associated with the severity of AD. A CNS myelin-specific ST-deficient mouse model was used to further identify the causes and consequences of spinal cord lipidome changes. Interestingly, ST deficiency led to spinal cord lipidome and transcriptome profiles highly resembling those observed in AD, characterized by decline of multiple myelin-enriched lipid classes and enhanced inflammatory responses, respectively. These changes significantly disrupted spinal cord function and led to substantial enlargement of urinary bladder in ST-deficient mice. CONCLUSIONS Our study identified CNS ST deficiency as a causal factor for AD-like lipid dysregulation, inflammation response and ultimately the development of bladder disorders. Targeting to maintain ST levels may serve as a promising strategy for the prevention and treatment of AD-related peripheral disorders.
Collapse
Affiliation(s)
- Sijia He
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Shulan Qiu
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Meixia Pan
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Juan P. Palavicini
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health San AntonioSan AntonioTexasUSA
- Division of DiabetesDepartment of MedicineUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Hu Wang
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Xin Li
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Anindita Bhattacharjee
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Savannah Barannikov
- Department of PathologyGlenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Kevin F. Bieniek
- Department of PathologyGlenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Jeffrey L. Dupree
- Department of Anatomy and NeurobiologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Research DivisionMcGuire Veterans Affairs Medical CenterRichmondVirginiaUSA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health San AntonioSan AntonioTexasUSA
- Division of DiabetesDepartment of MedicineUniversity of Texas Health San AntonioSan AntonioTexasUSA
| |
Collapse
|
9
|
Ponnusamy M, Wang S, Yuksel M, Hansen MT, Blazier DM, McMillan JD, Zhang X, Dammer EB, Collier L, Thinakaran G. Loss of forebrain BIN1 attenuates hippocampal pathology and neuroinflammation in a tauopathy model. Brain 2023; 146:1561-1579. [PMID: 36059072 PMCID: PMC10319775 DOI: 10.1093/brain/awac318] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/08/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Bridging integrator 1 (BIN1) is the second most prevalent genetic risk factor identified by genome-wide association studies (GWAS) for late-onset Alzheimer's disease. BIN1 encodes an adaptor protein that regulates membrane dynamics in the context of endocytosis and neurotransmitter vesicle release. In vitro evidence suggests that BIN1 can directly bind to tau in the cytosol. In addition, BIN1's function limits extracellular tau seed uptake by endocytosis and subsequent propagation as well as influences tau release through exosomes. However, the in vivo roles of BIN1 in tau pathogenesis and tauopathy-mediated neurodegeneration remain uncharacterized. We generated conditional knockout mice with a selective loss of Bin1 expression in the forebrain excitatory neurons and oligodendrocytes in P301S human tau transgenic background (line PS19). PS19 mice develop age-dependent tau neuropathology and motor deficits and are commonly used to study Alzheimer's disease tau pathophysiology. The severity of motor deficits and neuropathology was compared between experimental and control mice that differ with respect to forebrain BIN1 expression. BIN1's involvement in tau pathology and neuroinflammation was quantified by biochemical methods and immunostaining. Transcriptome changes were profiled by RNA-sequencing analysis to gain molecular insights. The loss of forebrain BIN1 expression in PS19 mice exacerbated tau pathology in the somatosensory cortex, thalamus, spinal cord and sciatic nerve, accelerated disease progression and caused early death. Intriguingly, the loss of BIN1 also mitigated tau neuropathology in select regions, including the hippocampus, entorhinal/piriform cortex, and amygdala, thus attenuating hippocampal synapse loss, neuronal death, neuroinflammation and brain atrophy. At the molecular level, the loss of forebrain BIN1 elicited complex neuronal and non-neuronal transcriptomic changes, including altered neuroinflammatory gene expression, concomitant with an impaired microglial transition towards the disease-associated microglial phenotype. These results provide crucial new information on in vivo BIN1 function in the context of tau pathogenesis. We conclude that forebrain neuronal BIN1 expression promotes hippocampal tau pathogenesis and neuroinflammation. Our findings highlight an exciting region specificity in neuronal BIN1 regulation of tau pathogenesis and reveal cell-autonomous and non-cell-autonomous mechanisms involved in BIN1 modulation of tau neuropathology.
Collapse
Affiliation(s)
- Moorthi Ponnusamy
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Shuai Wang
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Melike Yuksel
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Mitchell T Hansen
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Danielle M Blazier
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Joseph D McMillan
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Xiaolin Zhang
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University, Atlanta, GA 30322, USA
| | - Lisa Collier
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Gopal Thinakaran
- Byrd Alzheimer’s Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
10
|
Sturchio A, Espay AJ. The theoretical problems of "prodrome" and "phenoconversion" in neurodegeneration. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:155-167. [PMID: 36796940 DOI: 10.1016/b978-0-323-85538-9.00002-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The recognition of and approach to prodromal symptoms, those which manifest before a diagnosis can be ascertained at the bedside, are of increasing interest in neurodegenerative research. A prodrome is conceived of as an early window into a disease, a critical time when putative disease-modifying interventions may be best suited for examination. Several challenges affect research in this area. Prodromal symptoms are highly prevalent in the population, can be nonprogressive for years or decades, and exhibit limited specificity in predicting conversion versus nonconversion into a neurodegenerative category within a time window feasible for most longitudinal clinical studies. In addition, there is a large range of biological alterations subsumed within each prodromal syndrome, forced to converge into the unifying nosology of each neurodegenerative disorder. Initial prodromal subtyping efforts have been developed but given the scarcity of prodrome-to-disease longitudinal studies, it is not yet clear whether any prodromal subtype can be predicted to evolve into the corresponding subtype of manifesting disease - a form of construct validity. As current subtypes generated from one clinical population are not faithfully replicated to others, it is likely that, lacking biological or molecular anchors, prodromal subtypes may only be applicable to the cohorts within which they were developed. Furthermore, as clinical subtypes have not aligned with a consistent pattern of pathology or biology, such might also be the fate of prodromal subtypes. Finally, the threshold defining the change from prodrome to disease for most neurodegenerative disorders remains clinical (e.g., a motor change in gait becoming noticeable to a clinician or measurable with portable technologies), not biological. As such, a prodrome can be viewed as a disease state not yet overt to a clinician. Efforts into identifying biological subtypes of disease, regardless of clinical phenotype or disease stage, may best serve future disease-modifying therapeutic strategies deployed not for a prodromal symptom but for a defined biological derangement as soon as it can be determined to lead to clinical changes, prodromal or not.
Collapse
Affiliation(s)
- Andrea Sturchio
- James J. and Joan A. Gardner Family Center for Parkinson's disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States; Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, Stockholm, Sweden.
| | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States.
| |
Collapse
|
11
|
Buchman AS, Leurgans SE, Kim N, Agrawal S, Oveisgharan S, Zammit AR, VanderHorst V, Nag S, Bennett DA. Alzheimer's Disease Pathology Outside of the Cerebrum Is Related to a Higher Odds of Dementia. J Alzheimers Dis 2023; 96:563-578. [PMID: 37840485 PMCID: PMC11406461 DOI: 10.3233/jad-230223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
BACKGROUND Assessments of Alzheimer's disease pathology do not routinely include lower brainstem, olfactory bulb, and spinal cord. OBJECTIVE Test if amyloid-β (Aβ) and paired helical filament (PHF) tau-tangles outside the cerebrum are associated with the odds of dementia. METHODS Autopsies were obtained in decedents with cognitive testing (n = 300). Aβ plaques and PHF tau-tangles were assessed in 24 sites: cerebrum (n = 14), brainstem (n = 5), olfactory bulb, and four spinal cord levels. Since spinal Aβ were absent in the first 165 cases, it was not assessed in the remaining cases. RESULTS Age at death was 91 years old. About 90% had Aβ in cerebrum and of these, half had Aβ in the brainstem. Of the latter, 85% showed Aβ in the olfactory bulb. All but one participant had tau-tangles in the cerebrum and 86% had brainstem tau-tangles. Of the latter, 80% had tau-tangles in olfactory bulb and 36% tau-tangles in one or more spinal cord levels. About 90% of adults with tau-tangles also had Aβ in one or more regions. In a logistic model controlling for demographics, Aβ and tau-tangles within the cerebrum, the presence of Aβ in olfactory bulb [OR, 1.74(1.00, 3.05)]; tau-tangles in brainstem [OR, 4.00(1.1.57,10.21)]; and spinal cord [OR, 1.87 (1.21,3.11)] were independently associated with higher odds of dementia. CONCLUSION Regional differences in Aβ and tau-tangle accumulation extend beyond cerebrum to spinal cord and their presence outside the cerebrum are associated with a higher odds of dementia. Further studies are needed to clarify the extent, burden, and consequences of AD pathology outside of cerebrum.
Collapse
Affiliation(s)
- Aron S Buchman
- Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Sue E Leurgans
- Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Namhee Kim
- Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
| | - Sonal Agrawal
- Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Shahram Oveisgharan
- Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Andrea R Zammit
- Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | | | - Sukrit Nag
- Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
12
|
Sartoretti T, Ganley RP, Ni R, Freund P, Zeilhofer HU, Klohs J. Structural MRI Reveals Cervical Spinal Cord Atrophy in the P301L Mouse Model of Tauopathy: Gender and Transgene-Dosing Effects. Front Aging Neurosci 2022; 14:825996. [PMID: 35585865 PMCID: PMC9108240 DOI: 10.3389/fnagi.2022.825996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
In primary tauopathies, the deposition of tau neurofibrillary tangles and threads as well as neurodegenerative changes have been found within the brain and spinal cord. While degenerative changes have been intensively studied in the brain using structural magnetic resonance imaging (MRI), MRI studies investigating the spinal cord are still scarce. In the present study, we acquired ex vivo high resolution structural MRI of the cervical spinal cord of 8.5–9 month old hemizygous and homozygous P301L mice and non-transgenic littermates of both genders. We assessed the total cross-sectional area, and the gray and white matter anterior-posterior width and left-right width that are established imaging marker of spinal cord degeneration. We observed significant tissue-specific reductions in these parameters in female P301L mice that were stronger in homozygous than in hemizygous P301L mice, indicating both an effect of gender and transgene expression on cervical spinal cord atrophy. Moreover, atrophy was stronger in the gray matter than in the white matter. Immunohistochemical analysis revealed neurodegenerative and neuroinflammatory changes in the cervical spinal cord in both the gray and white matter of P301L mice. Collectively, our results provide evidence for cervical spinal cord atrophy that may directly contribute to the motor signs associated with tauopathy.
Collapse
Affiliation(s)
- Thomas Sartoretti
- Institute for Biomedical Engineering, ETH & University of Zurich, Zurich, Switzerland
| | - Robert P. Ganley
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| | - Patrick Freund
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
- Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
- Institute for Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, ETH & University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
- *Correspondence: Jan Klohs
| |
Collapse
|
13
|
Singh A, Dawson TM, Kulkarni S. Neurodegenerative disorders and gut-brain interactions. J Clin Invest 2021; 131:e143775. [PMID: 34196307 DOI: 10.1172/jci143775] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders (NDs) affect essential functions not only in the CNS, but also cause persistent gut dysfunctions, suggesting that they have an impact on both CNS and gut-innervating neurons. Although the CNS biology of NDs continues to be well studied, how gut-innervating neurons, including those that connect the gut to the brain, are affected by or involved in the etiology of these debilitating and progressive disorders has been understudied. Studies in recent years have shown how CNS and gut biology, aided by the gut-brain connecting neurons, modulate each other's functions. These studies underscore the importance of exploring the gut-innervating and gut-brain connecting neurons of the CNS and gut function in health, as well as the etiology and progression of dysfunction in NDs. In this Review, we discuss our current understanding of how the various gut-innervating neurons and gut physiology are involved in the etiology of NDs, including Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, to cause progressive CNS and persistent gut dysfunction.
Collapse
Affiliation(s)
- Alpana Singh
- Center for Neurogastroenterology, Division of Gastroenterology and Hepatology, Department of Medicine
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering.,Department of Neurology.,Solomon H. Snyder Department of Neuroscience, and.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, USA
| | - Subhash Kulkarni
- Center for Neurogastroenterology, Division of Gastroenterology and Hepatology, Department of Medicine
| |
Collapse
|
14
|
Kiper K, Freeman JL. Use of Zebrafish Genetic Models to Study Etiology of the Amyloid-Beta and Neurofibrillary Tangle Pathways in Alzheimer's Disease. Curr Neuropharmacol 2021; 20:524-539. [PMID: 34030617 DOI: 10.2174/1570159x19666210524155944] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/09/2021] [Accepted: 05/16/2021] [Indexed: 11/22/2022] Open
Abstract
The prevalence of neurodegenerative diseases is increasing globally, with an imperative need to identify and expand the availability of pharmaceutical treatment strategies. Alzheimer's disease is the most common neurodegenerative disease for which there is no cure or has limited treatments. Rodent models are primarily used in Alzheimer's disease research to investigate causes, pathology, molecular mechanisms, and pharmaceutical therapies. However, there is a lack of a comprehensive understanding of Alzheimer's disease causes, pathogenesis, and optimal treatments due in part to some limitations of using rodents, including higher economic cost, which can influence sample size and ultimately statistical power. It is necessary to expand our animal model toolbox to provide alternative strategies in Alzheimer's disease research. The zebrafish application in neurodegenerative disease research and neuropharmacology is greatly expanding due to several vital strengths spanning lower economic costs, the smaller size of the organism, a sequenced characterized genome, and well described anatomical structures. These characteristics are coupled to the conserved molecular function and disease pathways in humans. The existence of orthologs for genes associated with Alzheimer's disease in zebrafish is also confirmed. While wild-type zebrafish appear to lack some of the neuropathological features of Alzheimer's disease, the advent of genetic editing technologies has expanded evaluation of the amyloid and neurofibrillary tangle hypotheses using the zebrafish and exploration of pharmaceutical molecular targets. An overview of how genetic editing technologies are being used with the zebrafish to create models to investigate the causes, pathology, molecular mechanisms, and pharmaceutical targets of Alzheimer's disease is detailed.
Collapse
Affiliation(s)
- Keturah Kiper
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States
| |
Collapse
|
15
|
Hitrec T, Squarcio F, Cerri M, Martelli D, Occhinegro A, Piscitiello E, Tupone D, Amici R, Luppi M. Reversible Tau Phosphorylation Induced by Synthetic Torpor in the Spinal Cord of the Rat. Front Neuroanat 2021; 15:592288. [PMID: 33603651 PMCID: PMC7884466 DOI: 10.3389/fnana.2021.592288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/11/2021] [Indexed: 12/21/2022] Open
Abstract
Tau is a key protein in neurons, where it affects the dynamics of the microtubule system. The hyperphosphorylation of Tau (PP-Tau) commonly leads to the formation of neurofibrillary tangles, as it occurs in tauopathies, a group of neurodegenerative diseases, including Alzheimer's. Hypothermia-related accumulation of PP-Tau has been described in hibernators and during synthetic torpor (ST), a torpor-like condition that has been induced in rats, a non-hibernating species. Remarkably, in ST PP-Tau is reversible and Tau de-phosphorylates within a few hours following the torpor bout, apparently not evolving into pathology. These observations have been limited to the brain, but in animal models of tauopathies, PP-Tau accumulation also appears to occur in the spinal cord (SpCo). The aim of the present work was to assess whether ST leads to PP-Tau accumulation in the SpCo and whether this process is reversible. Immunofluorescence (IF) for AT8 (to assess PP-Tau) and Tau-1 (non-phosphorylated Tau) was carried out on SpCo coronal sections. AT8-IF was clearly expressed in the dorsal horns (DH) during ST, while in the ventral horns (VH) no staining was observed. The AT8-IF completely disappeared after 6 h from the return to euthermia. Tau-1-IF disappeared in both DH and VH during ST, returning to normal levels during recovery. To shed light on the cellular process underlying the PP-Tau pattern observed, the inhibited form of the glycogen-synthase kinase 3β (the main kinase acting on Tau) was assessed using IF: VH (i.e., in motor neurons) were highly stained mainly during ST, while in DH there was no staining. Since tauopathies are also related to neuroinflammation, microglia activation was also assessed through morphometric analyses, but no ST-induced microglia activation was found in the SpCo. Taken together, the present results show that, in the DH of SpCo, ST induces a reversible accumulation of PP-Tau. Since during ST there is no motor activity, the lack of AT8-IF in VH may result from an activity-related process at a cellular level. Thus, ST demonstrates a newly-described physiological mechanism that is able to resolve the accumulation of PP-Tau and apparently avoid the neurodegenerative outcome.
Collapse
Affiliation(s)
- Timna Hitrec
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Fabio Squarcio
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Matteo Cerri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Davide Martelli
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Occhinegro
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Emiliana Piscitiello
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Domenico Tupone
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy.,Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | - Roberto Amici
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Luppi
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Müller K, Fröhlich S, Germano AMC, Kondragunta J, Agoitia Hurtado MFDC, Rudisch J, Schmidt D, Hirtz G, Stollmann P, Voelcker-Rehage C. Sensor-based systems for early detection of dementia (SENDA): a study protocol for a prospective cohort sequential study. BMC Neurol 2020; 20:84. [PMID: 32145744 PMCID: PMC7060588 DOI: 10.1186/s12883-020-01666-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
Background Dementia and cognitive decline are serious social and economic burdens. An increase in the population of older people, as well as longer lifespans mean that numbers of dementia cases are exponentially rising. Neuropathological changes associated with dementia are thought to appear before the clinical manifestation of cognitive symptoms, i.e., memory impairments. Further, some older adults (OA) experience cognitive decline before it can be objectively diagnosed. For optimal care of these patients, it is necessary to detect cognitive decline and dementia at an early stage. In this vein, motor, sensory, and neurophysiological declines could be promising factors if found to be present before the onset of cognitive impairment. Hence, the objective of the SENDA study is to develop a multi-dimensional sensor-based instrument that allows early detection of cognitive decline or dementia in OA with the help of cognitive, sensory, motor, and neurophysiological parameters before its clinical manifestation. Methods/design In the cohort sequential study, participants are assigned to one of three study groups depending on their cognitive status: 1. cognitively healthy individuals (CHI), 2. subjectively cognitively impaired persons (SCI), or 3. (possible) mildly cognitively impaired persons (pMCI, MCI). All groups take part in the same cognitive (e.g., executive function tests), motor (e.g., gait analyses, balance tests), sensory (e.g., vibration perception threshold test, proprioception tests), and neurophysiological (e.g., electroencephalograms) measurements. Depending on the time at which participants are included into the study, all measurements are repeated up to four times in intervals of 8 months within 3 years to identify associations with cognitive changes over time. Discussion This study aims to detect possible motor, sensory, neurophysiological, and cognitive predictors to develop an early screening tool for dementia and its pre-stages in OA. Thus, affected persons could receive optimal health care at an earlier time point to maintain their health resources. Trial status The study is ongoing. The recruitment of participants will be continued until May 2020.
Collapse
Affiliation(s)
- Katrin Müller
- Department of Sports Psychology (with focus on prevention and rehabilitation), Institute of Human Movement Science and Health, Faculty of Behavioural and Social Sciences, Chemnitz University of Technology, Thüringer Weg 11, 09126, Chemnitz, Germany.
| | - Stephanie Fröhlich
- Department of Sports Psychology (with focus on prevention and rehabilitation), Institute of Human Movement Science and Health, Faculty of Behavioural and Social Sciences, Chemnitz University of Technology, Thüringer Weg 11, 09126, Chemnitz, Germany
| | - Andresa M C Germano
- Department of Human Locomotion, Institute of Human Movement Science and Health, Faculty of Behavioural and Social Sciences, Chemnitz University of Technology, Chemnitz, Germany
| | - Jyothsna Kondragunta
- Department of Digital Signal Processing and Circuit Technology, Faculty of Electrical Engineering and Information Technology, Chemnitz University of Technology, Chemnitz, Germany
| | | | - Julian Rudisch
- Department of Sports Psychology (with focus on prevention and rehabilitation), Institute of Human Movement Science and Health, Faculty of Behavioural and Social Sciences, Chemnitz University of Technology, Thüringer Weg 11, 09126, Chemnitz, Germany.,Department of Neuromotor Behavior and Exercise, University of Münster, Münster, Germany
| | - Daniel Schmidt
- Department of Human Locomotion, Institute of Human Movement Science and Health, Faculty of Behavioural and Social Sciences, Chemnitz University of Technology, Chemnitz, Germany
| | - Gangolf Hirtz
- Department of Digital Signal Processing and Circuit Technology, Faculty of Electrical Engineering and Information Technology, Chemnitz University of Technology, Chemnitz, Germany
| | - Peter Stollmann
- Department of Analysis, Faculty of Mathematics, Chemnitz University of Technology, Chemnitz, Germany
| | - Claudia Voelcker-Rehage
- Department of Sports Psychology (with focus on prevention and rehabilitation), Institute of Human Movement Science and Health, Faculty of Behavioural and Social Sciences, Chemnitz University of Technology, Thüringer Weg 11, 09126, Chemnitz, Germany.,Department of Neuromotor Behavior and Exercise, University of Münster, Münster, Germany
| |
Collapse
|
17
|
Lorenzi RM, Palesi F, Castellazzi G, Vitali P, Anzalone N, Bernini S, Cotta Ramusino M, Sinforiani E, Micieli G, Costa A, D’Angelo E, Gandini Wheeler-Kingshott CAM. Unsuspected Involvement of Spinal Cord in Alzheimer Disease. Front Cell Neurosci 2020; 14:6. [PMID: 32082122 PMCID: PMC7002560 DOI: 10.3389/fncel.2020.00006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/10/2020] [Indexed: 11/13/2022] Open
Abstract
Objective: Brain atrophy is an established biomarker for dementia, yet spinal cord involvement has not been investigated to date. As the spinal cord is relaying sensorimotor control signals from the cortex to the peripheral nervous system and vice-versa, it is indeed a very interesting question to assess whether it is affected by atrophy due to a disease that is known for its involvement of cognitive domains first and foremost, with motor symptoms being clinically assessed too. We, therefore, hypothesize that in Alzheimer's disease (AD), severe atrophy can affect the spinal cord too and that spinal cord atrophy is indeed an important in vivo imaging biomarker contributing to understanding neurodegeneration associated with dementia. Methods: 3DT1 images of 31 AD and 35 healthy control (HC) subjects were processed to calculate volume of brain structures and cross-sectional area (CSA) and volume (CSV) of the cervical cord [per vertebra as well as the C2-C3 pair (CSA23 and CSV23)]. Correlated features (ρ > 0.7) were removed, and the best subset identified for patients' classification with the Random Forest algorithm. General linear model regression was used to find significant differences between groups (p ≤ 0.05). Linear regression was implemented to assess the explained variance of the Mini-Mental State Examination (MMSE) score as a dependent variable with the best features as predictors. Results: Spinal cord features were significantly reduced in AD, independently of brain volumes. Patients classification reached 76% accuracy when including CSA23 together with volumes of hippocampi, left amygdala, white and gray matter, with 74% sensitivity and 78% specificity. CSA23 alone explained 13% of MMSE variance. Discussion: Our findings reveal that C2-C3 spinal cord atrophy contributes to discriminate AD from HC, together with more established features. The results show that CSA23, calculated from the same 3DT1 scan as all other brain volumes (including right and left hippocampi), has a considerable weight in classification tasks warranting further investigations. Together with recent studies revealing that AD atrophy is spread beyond the temporal lobes, our result adds the spinal cord to a number of unsuspected regions involved in the disease. Interestingly, spinal cord atrophy explains also cognitive scores, which could significantly impact how we model sensorimotor control in degenerative diseases with a primary cognitive domain involvement. Prospective studies should be purposely designed to understand the mechanisms of atrophy and the role of the spinal cord in AD.
Collapse
Affiliation(s)
| | - Fulvia Palesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Neuroradiology Unit, Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Gloria Castellazzi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
- Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Paolo Vitali
- Neuroradiology Unit, Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Sara Bernini
- Laboratory of Neuropsychology, IRCCS Mondino Foundation, Pavia, Italy
| | - Matteo Cotta Ramusino
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Unit of Behavioral Neurology, IRCCS Mondino Foundation, Pavia, Italy
| | - Elena Sinforiani
- Laboratory of Neuropsychology, IRCCS Mondino Foundation, Pavia, Italy
| | - Giuseppe Micieli
- Department of Emergency Neurology, IRCCS Mondino Foundation, Pavia, Italy
| | - Alfredo Costa
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Unit of Behavioral Neurology, IRCCS Mondino Foundation, Pavia, Italy
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Brain Connectivity Center (BCC), IRCCS Mondino Foundation, Pavia, Italy
| | - Claudia A. M. Gandini Wheeler-Kingshott
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
- Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
18
|
Abstract
Staging of neurodegenerative diseases is based chiefly on the topographical or anatomical extent of aggregated proteinaceous inclusions, and the density or severity of the lesions in a given region is usually assessed semiquantitatively. Associated phenomena, such as cell loss and synapse loss, are evaluated but not staged. This article reviews the development of neuropathological staging of the sporadic Alzheimer's and sporadic Parkinson's diseases. It considers challenges for staging systems, and it poses the question whether neuropathological staging as practiced up to now is still relevant.
Collapse
|
19
|
Xie Q, Zhao WJ, Ou GY, Xue WK. An Overview of Experimental and Clinical Spinal Cord Findings in Alzheimer's Disease. Brain Sci 2019; 9:E168. [PMID: 31319495 PMCID: PMC6681410 DOI: 10.3390/brainsci9070168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that occurs mainly in the elderly and presenile life stages. It is estimated that by the year 2050, 135 million people will be affected by AD worldwide, representing a huge burden to society. The pathological hallmarks of AD mainly include intracellular neurofibrillary tangles (NFTs) caused by hyperphosphorylation of tau protein, formation of extracellular amyloid plaques, and massive neural cell death in the affected nervous system. The pathogenesis of AD is very complicated, and recent scientific research on AD is mainly concentrated on the cortex and hippocampus. Although the spinal cord is a pivotal part of the central nervous system, there are a limited number of studies focusing on the spinal cord. As an extension of the brain, the spinal cord functions as the bridge between the brain and various parts of the body. However, pathological changes in the spinal cord in AD have not been comprehensively and systematically studied at present. We here review the existing progress on the pathological features of AD in the spinal cord.
Collapse
Affiliation(s)
- Qing Xie
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China.
| | - Guan-Yong Ou
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| | - Wei-Kang Xue
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| |
Collapse
|
20
|
Zhu K, Wang X, Sun B, Wu J, Lu H, Zhang X, Liang H, Zhang D, Liu C. Primary Age-Related Tauopathy in Human Subcortical Nuclei. Front Neurosci 2019; 13:529. [PMID: 31191227 PMCID: PMC6549797 DOI: 10.3389/fnins.2019.00529] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/07/2019] [Indexed: 12/29/2022] Open
Abstract
The present study aimed to determine the spatial distribution patterns of hyperphosphorylated tau-immunoreactive cells in subcortical nuclei of post-mortem human brain with primary age-related tauopathy (PART). Subcortical tauopathy has important pathological and clinical implications. Expression of tau was examined in different subcortical regions of definite PART cases with a Braak neurofibrillary tangle stage >0 and ≤IV, and with a Thal phase 0 (no beta-amyloid present). Post-mortem brain tissue of PART was studied using immunohistochemistry and subsequent semi-quantitative assessment with Braak NFT stage -matched pre-Alzheimer’s disease (AD) and AD cases as a control. Expression of tau was frequently found in subcortical nuclei including the substantia nigra, inferior colliculus, locus coeruleus, medulla oblongata in the brainstem, the caudate, putamen, nucleus globus pallidus in the striatum, the hypothalamus, thalamus, subthalamus in the diencephalon, and the cervical spinal cord in both PART and AD, but not in the dentate nucleus of the cerebellum. A positive correlation was found between the Braak NFT stage and the tau distribution (qualitative)/tau density (quantitative) in PART and AD. Brainstem nuclei were commonly involved in early PART with NFT Braak stage I/II, there was no preference among the substantia nigra, inferior colliculus, locus caeruleus and medulla oblongata. The prevalence and severity of tau pathology in subcortical nuclei of PART and AD were positively correlated with NFT Braak stage, suggesting that these nuclei were increasingly involved as PART and AD progressed. Subcortical nuclei were likely the sites initially affected by aging associated tau pathology, especially the brainstem nuclei including the substantia nigra, inferior colliculus, locus caeruleus and medulla oblongata.
Collapse
Affiliation(s)
- Keqing Zhu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Wang
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Sun
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Juanli Wu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Lu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoling Zhang
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Huazheng Liang
- Brain Structure and Function, Neuroscience Research Australia, Randwick, NSW, Australia.,Department of Neurology, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Dandan Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Chong Liu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
21
|
Braak H, Del Tredici K. Top-Down Projections Direct the Gradual Progression of Alzheimer-Related Tau Pathology Throughout the Neocortex. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:291-303. [PMID: 32096045 DOI: 10.1007/978-981-32-9358-8_22] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In sporadic Alzheimer's disease (sAD), tau pathology gradually but relentlessly progresses from the transentorhinal region of the temporal lobe into both the allocortex and temporal high order association areas of the neocortex. From there, it ultimately reaches the primary sensory and motor fields of the neocortex. The brunt of the changes seen during neurofibrillary stages (NFT) I-VI is borne by top-down projection neurons that contribute to cortico-cortical connectivities between different neocortical fields. Very early changes develop in isolated pyramidal cells in layers III and V, and these cells are targets of top-down projections terminating in association areas of the first temporal gyrus or in peristriate regions of the occipital lobe. Neurofibrillary pathology in these regions is routinely associated with late NFT stages. Sequential changes occur in different cell compartments (dendritic, somatic, axonal) of these early-involved neurons. Tau pathology first develops in distal segments of basal dendrites, then in proximal dendrites, the soma, and, finally, in the axon of affected pyramidal neurons. This sequence of abnormal changes supports the concept that axons of cortico-cortical top-down neurons may carry and spread abnormal tau seeds in a focused manner (transsynaptically) into the distal dendritic segments of nerve cells directly following in the neuronal chain, thereby sustaining tau-seeded templating in sAD.
Collapse
Affiliation(s)
- Heiko Braak
- Clinical Neuroanatomy Section/Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| | - Kelly Del Tredici
- Clinical Neuroanatomy Section/Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany.
| |
Collapse
|
22
|
Dugger BN, Hoffman BR, Scroggins A, Serrano GE, Adler CH, Shill HA, Belden CM, Sabbagh MN, Caviness JN, Driver Dunckley E, Beach TG. Tau immunoreactivity in peripheral tissues of human aging and select tauopathies. Neurosci Lett 2018; 696:132-139. [PMID: 30579993 DOI: 10.1016/j.neulet.2018.12.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 11/18/2022]
Abstract
Many studies have been directed at understanding mechanisms of tau aggregation and therapeutics, nearly all focusing on the brain. It is critical to understand the presence of tau in peripheral tissues since this may provide new insights into disease progression and selective vulnerability. The current study sought to determine the presence of select tau species in peripheral tissues in elderly individuals and across an array of tauopathies. Using formalin fixed paraffin embedded sections, we examined abdominal skin, submandibular gland, and sigmoid colon among 69 clinicopathologically defined cases: 19 lacking a clinical neuropathological diagnosis (normal controls), 26 progressive supranuclear palsy (PSP), 21 Alzheimer's disease (AD), and 3 with corticobasal degeneration (CBD). Immunohistochemistry was performed using antibodies for "total" tau (HT7) and two phosphorylated tau species (AT8 and pT231). HT7 staining of abdominal skin revealed immunoreactivity of potential nerve elements in 5% of cases (1 AD, 1 AD/PSP, and 1 CBD out of 55 cases examined); skin sections lacked AT8 and pT231 immunoreactive nerve elements. Submandibular glands from all cases had HT7 immunoreactive nerve elements; while pT231 was present in 92% of cases, and AT8 in only 3 cases (2 AD and one AD/PSP case). In sigmoid colon, HT7 immunoreactivity was present in all but 2 cases (97%), pT231 in 54%, and AT8 was present in only 5/62 cases (8%). These data suggest select tau species in CNS tauopathies do not have a high propensity to spread to the periphery and this may hold clues for the understanding of CNS tau pathogenicity and vulnerability.
Collapse
Affiliation(s)
- Brittany N Dugger
- Department of Pathology and Laboratory Medicine University of California, Davis, Sacramento, CA, United States.
| | | | - Alex Scroggins
- Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Geidy E Serrano
- Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Charles H Adler
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic, Scottsdale, AZ, United States
| | - Holly A Shill
- Barrow Neurological Institute, Phoenix, AZ, United States
| | | | - Marwan N Sabbagh
- Barrow Neurological Institute, Phoenix, AZ, United States; Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
| | - John N Caviness
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic, Scottsdale, AZ, United States
| | - Erika Driver Dunckley
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic, Scottsdale, AZ, United States
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, AZ, United States
| |
Collapse
|
23
|
Tau seeding activity begins in the transentorhinal/entorhinal regions and anticipates phospho-tau pathology in Alzheimer's disease and PART. Acta Neuropathol 2018; 136:57-67. [PMID: 29752551 PMCID: PMC6015098 DOI: 10.1007/s00401-018-1855-6] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/27/2018] [Accepted: 04/29/2018] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is characterized by accumulation of tau neurofibrillary tangles (NFTs) and, according to the prion model, transcellular propagation of pathological "seeds" may underlie its progression. Staging of NFT pathology with phospho-tau antibody is useful to classify AD and primary age-related tauopathy (PART) cases. The locus coeruleus (LC) shows the earliest phospho-tau signal, whereas other studies suggest that pathology begins in the transentorhinal/entorhinal cortices (TRE/EC). The relationship of tau seeding activity, phospho-tau pathology, and progression of neurodegeneration remains obscure. Consequently, we employed an established cellular biosensor assay to quantify tau seeding activity in fixed human tissue, in parallel with AT8 phospho-tau staining of immediately adjacent sections. We studied four brain regions from each of n = 247 individuals across a range of disease stages. We detected the earliest and most robust seeding activity in the TRE/EC. The LC did not uniformly exhibit seeding activity until later NFT stages. We also detected seeding activity in the superior temporal gyrus (STG) and primary visual cortex (VC) at stages before NFTs and/or AT8-immunopositivity were detectable. AD and putative PART cases exhibited similar patterns of seeding activity that anticipated histopathology across all NFT stages. Our findings are consistent with the prion model and suggest that pathological seeding activity begins in the TRE/EC rather than in the LC. In the analysis of tauopathy, quantification of seeding activity may offer an important addition to classical histopathology.
Collapse
|
24
|
Ayaki T, Ito H, Komure O, Kamada M, Nakamura M, Wate R, Kusaka H, Yamaguchi Y, Li F, Kawakami H, Urushitani M, Takahashi R. Multiple Proteinopathies in Familial ALS Cases With Optineurin Mutations. J Neuropathol Exp Neurol 2017; 77:128-138. [DOI: 10.1093/jnen/nlx109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
25
|
Axonal and myelinic pathology in 5xFAD Alzheimer's mouse spinal cord. PLoS One 2017; 12:e0188218. [PMID: 29176903 PMCID: PMC5703477 DOI: 10.1371/journal.pone.0188218] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/02/2017] [Indexed: 01/14/2023] Open
Abstract
As an extension of the brain, the spinal cord has unique properties which could allow us to gain a better understanding of CNS pathology. The brain and cord share the same cellular components, yet the latter is simpler in cytoarchitecture and connectivity. In Alzheimer's research, virtually all focus is on brain pathology, however it has been shown that transgenic Alzheimer's mouse models accumulate beta amyloid plaques in spinal cord, suggesting that the cord possesses the same molecular machinery and conditions for plaque formation. Here we report a spatial-temporal map of plaque load in 5xFAD mouse spinal cord. We found that plaques started to appear at 11 weeks, then exhibited a time dependent increase and differential distribution along the cord. More plaques were found in cervical than other spinal levels at all time points examined. Despite heavy plaque load at 6 months, the number of cervical motor neurons in 5xFAD mice is comparable to wild type littermates. On detailed microscopic examination, fine beta amyloid-containing and beta sheet-rich thread-like structures were found in the peri-axonal space of many axons. Importantly, these novel structures appear before any plaque deposits are visible in young mice spinal cord and they co-localize with axonal swellings at later stages, suggesting that these thread-like structures might represent the initial stages of plaque formation, and could play a role in axonal damage. Additionally, we were able to demonstrate increasing myelinopathy in aged 5xFAD mouse spinal cord using the lipid probe Nile Red with high resolution. Collectively, we found significant amyloid pathology in grey and white matter of the 5xFAD mouse spinal cord which indicates that this structure maybe a useful platform to study mechanisms of Alzheimer's pathology and disease progression.
Collapse
|
26
|
Caprelli MT, Mothe AJ, Tator CH. CNS Injury: Posttranslational Modification of the Tau Protein as a Biomarker. Neuroscientist 2017; 25:8-21. [PMID: 29283022 DOI: 10.1177/1073858417742125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The ideal biomarker for central nervous system (CNS) trauma in patients would be a molecular marker specific for injured nervous tissue that would provide a consistent and reliable assessment of the presence and severity of injury and the prognosis for recovery. One candidate biomarker is the protein tau, a microtubule-associated protein abundant in the axonal compartment of CNS neurons. Following axonal injury, tau becomes modified primarily by hyperphosphorylation of its various amino acid residues and cleavage into smaller fragments. These posttrauma products can leak into the cerebrospinal fluid or bloodstream and become candidate biomarkers of CNS injury. This review examines the primary molecular changes that tau undergoes following traumatic brain injury and spinal cord injury, and reviews the current literature in traumatic CNS biomarker research with a focus on the potential for hyperphosphorylated and cleaved tau as sensitive biomarkers of injury.
Collapse
Affiliation(s)
- Mitchell T Caprelli
- 1 Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,2 Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Andrea J Mothe
- 2 Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Charles H Tator
- 1 Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,2 Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada.,3 Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
A novel approach for assigning levels to monkey and human lumbosacral spinal cord based on ventral horn morphology. PLoS One 2017; 12:e0177243. [PMID: 28542213 PMCID: PMC5443490 DOI: 10.1371/journal.pone.0177243] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/24/2017] [Indexed: 12/11/2022] Open
Abstract
Proper identification of spinal cord levels is crucial for clinical-pathological and imaging studies in humans, but can be a challenge given technical limitations. We have previously demonstrated in non-primate models that the contours of the spinal ventral horn are determined by the position of motoneuron pools. These positions are preserved within and among individuals and can be used to identify lumbosacral spinal levels. Here we tested the hypothesis that this approach can be extended to identify monkey and human spinal levels. In 7 rhesus monkeys, we retrogradely labeled motoneuron pools that represent rostral, middle and caudal landmarks of the lumbosacral enlargement. We then aligned the lumbosacral enlargements among animals using absolute length, segmental level or a relative scale based upon rostral and caudal landmarks. Inter-animal matching of labeled motoneurons across the lumbosacral enlargement was most precise when using internal landmarks. We then reconstructed 3 human lumbosacral spinal cords, and aligned these based upon homologous internal landmarks. Changes in shape of the ventral horn were consistent among human subjects using this relative scale, despite marked differences in absolute length or age. These data suggest that the relative position of spinal motoneuron pools is conserved across species, including primates. Therefore, in clinical-pathological or imaging studies in humans, one can assign spinal cord levels to even single sections by matching ventral horn shape to standardized series.
Collapse
|
28
|
Iatrou A, Kenis G, Rutten BPF, Lunnon K, van den Hove DLA. Epigenetic dysregulation of brainstem nuclei in the pathogenesis of Alzheimer's disease: looking in the correct place at the right time? Cell Mol Life Sci 2017; 74:509-523. [PMID: 27628303 PMCID: PMC5241349 DOI: 10.1007/s00018-016-2361-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/15/2016] [Accepted: 09/07/2016] [Indexed: 12/20/2022]
Abstract
Even though the etiology of Alzheimer's disease (AD) remains unknown, it is suggested that an interplay among genetic, epigenetic and environmental factors is involved. An increasing body of evidence pinpoints that dysregulation in the epigenetic machinery plays a role in AD. Recent developments in genomic technologies have allowed for high throughput interrogation of the epigenome, and epigenome-wide association studies have already identified unique epigenetic signatures for AD in the cortex. Considerable evidence suggests that early dysregulation in the brainstem, more specifically in the raphe nuclei and the locus coeruleus, accounts for the most incipient, non-cognitive symptomatology, indicating a potential causal relationship with the pathogenesis of AD. Here we review the advancements in epigenomic technologies and their application to the AD research field, particularly with relevance to the brainstem. In this respect, we propose the assessment of epigenetic signatures in the brainstem as the cornerstone of interrogating causality in AD. Understanding how epigenetic dysregulation in the brainstem contributes to AD susceptibility could be of pivotal importance for understanding the etiology of the disease and for the development of novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- A Iatrou
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - G Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - B P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - K Lunnon
- University of Exeter Medical School, RILD, University of Exeter, Barrack Road, Devon, UK
| | - D L A van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands.
- Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Fuechsleinstrasse 15, 97080, Würzburg, Germany.
| |
Collapse
|
29
|
Dugger BN, Whiteside CM, Maarouf CL, Walker DG, Beach TG, Sue LI, Garcia A, Dunckley T, Meechoovet B, Reiman EM, Roher AE. The Presence of Select Tau Species in Human Peripheral Tissues and Their Relation to Alzheimer's Disease. J Alzheimers Dis 2016; 51:345-56. [PMID: 26890756 DOI: 10.3233/jad-150859] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tau becomes excessively phosphorylated in Alzheimer's disease (AD) and is widely studied within the brain. Further examination of the extent and types of tau present in peripheral tissues and their relation to AD is warranted given recent publications on pathologic spreading. Cases were selected based on the presence of pathological tau spinal cord deposits (n = 18). Tissue samples from sigmoid colon, scalp, abdominal skin, liver, and submandibular gland were analyzed by western blot and enzyme-linked immunosorbent assays (ELISAs) for certain tau species; frontal cortex gray matter was used for comparison. ELISAs revealed brain to have the highest total tau levels, followed by submandibular gland, sigmoid colon, liver, scalp, and abdominal skin. Western blots with antibodies recognizing tau phosphorylated at threonine 231(pT231), serine 396 and 404 (PHF-1), and an unmodified total human tau between residues 159 and 163 (HT7) revealed multiple banding patterns, some of which predominated in peripheral tissues. As submandibular gland had the highest levels of peripheral tau, a second set of submandibular gland samples were analyzed (n = 36; 19 AD, 17 non-demented controls). ELISAs revealed significantly lower levels of pS396 (p = 0.009) and pT231 (p = 0.005) in AD cases but not total tau (p = 0.18). Furthermore, pT231 levels in submandibular gland inversely correlated with Braak neurofibrillary tangle stage (p = 0.04), after adjusting for age at death, gender, and postmortem interval. These results provide evidence that certain tau species are present in peripheral tissues. Of potential importance, submandibular gland pT231 is progressively less abundant with increasing Braak neurofibrillary tangle stage.
Collapse
Affiliation(s)
- Brittany N Dugger
- Banner Sun Health Research Institute, Sun City, AZ, USA.,Arizona Alzheimer's Consortium
| | - Charisse M Whiteside
- Banner Sun Health Research Institute, Sun City, AZ, USA.,Arizona Alzheimer's Consortium
| | - Chera L Maarouf
- Banner Sun Health Research Institute, Sun City, AZ, USA.,Arizona Alzheimer's Consortium
| | - Douglas G Walker
- Banner Sun Health Research Institute, Sun City, AZ, USA.,Arizona Alzheimer's Consortium
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, AZ, USA.,Arizona Alzheimer's Consortium
| | - Lucia I Sue
- Banner Sun Health Research Institute, Sun City, AZ, USA.,Arizona Alzheimer's Consortium
| | - Angelica Garcia
- Banner Sun Health Research Institute, Sun City, AZ, USA.,Arizona Alzheimer's Consortium
| | - Travis Dunckley
- Translational Genomics Research Institute, Phoenix, AZ, USA.,Arizona Alzheimer's Consortium
| | - Bessie Meechoovet
- Translational Genomics Research Institute, Phoenix, AZ, USA.,Arizona Alzheimer's Consortium
| | - Eric M Reiman
- Banner Alzheimer's Institute, Phoenix, AZ, USA.,Arizona Alzheimer's Consortium
| | - Alex E Roher
- Banner Sun Health Research Institute, Sun City, AZ, USA.,Arizona Alzheimer's Consortium
| |
Collapse
|
30
|
Arendt T, Stieler JT, Holzer M. Tau and tauopathies. Brain Res Bull 2016; 126:238-292. [PMID: 27615390 DOI: 10.1016/j.brainresbull.2016.08.018] [Citation(s) in RCA: 425] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022]
|
31
|
|
32
|
Guo Y, Wang L, Zhu M, Zhang H, Hu Y, Han Z, Liu J, Zhao W, Wang D. Detection of hyperphosphorylated tau protein and α-synuclein in spinal cord of patients with Alzheimer's disease. Neuropsychiatr Dis Treat 2016; 12:445-52. [PMID: 27013875 PMCID: PMC4777227 DOI: 10.2147/ndt.s90735] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The aim of this study was to investigate the neuropathological features of the spinal cord in patients suffering with Alzheimer's disease (AD). Spinal cord tissue collected from three AD patients and eight controls was selected for the study. Data were collected at T2, T8, T10, L4, and S2 spinal levels. The sections were subjected to hematoxylin and eosin and Gallyas-Braak staining methods and then were immunostained with antibodies such as phosphorylated tau protein (AT8), α-synuclein, Aβ, amyloid precursor protein, ubiquitin, and TDP-43. Pathological changes exhibited by the biomarkers were detected by microscopy. Neurofibrillary tangles (NFTs) were detectable in spinal anterior horn motor neurons in two of the three AD patients. AT8-positive axons or axon-like structures and AT8 expression in glial cells were detected in all three AD cases. Hyperphosphorylation of tau protein was detected in spinal anterior horn cells, glial cells, and axons, and its severity was associated with NFTs in the brain tissue. α-Synuclein-positive Lewy bodies and scattered Lewy-like neuritis were detected in the medial horn of the thoracic spinal cord and ventral sacral gray matter, respectively, in one patient who had AD with Lewy bodies. Neither amyloid deposition nor amyloid precursor protein and TDP-43 expression was detected in the spinal cord of AD patients. Spinal cord of AD patients was observed to contain phosphorylated tau protein and α-synuclein immunoreactive structures, which may play a role in dyskinesia and autonomic dysfunction in advanced AD.
Collapse
Affiliation(s)
- Yanjun Guo
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China; Department of Geriatric Neurology, PLA General Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Luning Wang
- Department of Geriatric Neurology, PLA General Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Mingwei Zhu
- Department of Geriatric Neurology, PLA General Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Honghong Zhang
- Institute of Geriatrics, Chinese PLA General Hospital & Chinese PLA Medical Academy, Capital Medical University, Beijing, People's Republic of China
| | - Yazhuo Hu
- Institute of Geriatrics, Chinese PLA General Hospital & Chinese PLA Medical Academy, Capital Medical University, Beijing, People's Republic of China
| | - Zhitao Han
- Institute of Geriatrics, Chinese PLA General Hospital & Chinese PLA Medical Academy, Capital Medical University, Beijing, People's Republic of China
| | - Jia Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Weiqin Zhao
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Dexin Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
33
|
Braak H, Del Tredici K. The preclinical phase of the pathological process underlying sporadic Alzheimer’s disease. Brain 2015; 138:2814-33. [DOI: 10.1093/brain/awv236] [Citation(s) in RCA: 293] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/21/2015] [Indexed: 12/13/2022] Open
|
34
|
Iverson GL, Gardner AJ, McCrory P, Zafonte R, Castellani RJ. A critical review of chronic traumatic encephalopathy. Neurosci Biobehav Rev 2015; 56:276-93. [PMID: 26183075 DOI: 10.1016/j.neubiorev.2015.05.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 04/14/2015] [Accepted: 05/08/2015] [Indexed: 12/14/2022]
Abstract
Chronic traumatic encephalopathy (CTE) has been described in the literature as a neurodegenerative disease with: (i) localized neuronal and glial accumulations of phosphorylated tau (p-tau) involving perivascular areas of the cerebral cortex, sulcal depths, and with a preference for neurons within superficial cortical laminae; (ii) multifocal axonal varicosities and axonal loss involving deep cortex and subcortical white matter; (iii) relative absence of beta-amyloid deposits; (iv) TDP-43 immunoreactive inclusions and neurites; and (v) broad and diverse clinical features. Some of the pathological findings reported in the literature may be encountered with age and other neurodegenerative diseases. However, the focality of the p-tau cortical findings in particular, and the regional distribution, are believed to be unique to CTE. The described clinical features in recent cases are very similar to how depression manifests in middle-aged men and with frontotemporal dementia as the disease progresses. It has not been established that the described tau pathology, especially in small amounts, can cause complex changes in behavior such as depression, substance abuse, suicidality, personality changes, or cognitive impairment. Future studies will help determine the extent to which the neuropathology is causally related to the diverse clinical features.
Collapse
Affiliation(s)
- Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, MassGeneral Hospital for Children Sports Concussion Program, & Red Sox Foundation and Massachusetts General Hospital Home Base Program, Boston, MA, USA.
| | - Andrew J Gardner
- Hunter New England Local Health District Sports Concussion Program; & Centre for Translational Neuroscience and Mental Health, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Paul McCrory
- The Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre - Austin Campus, Heidelberg, Victoria, Australia
| | - Ross Zafonte
- Department of Physical Medicine and Rehabilitation, Harvard Medical School; Spaulding Rehabilitation Hospital; Brigham and Women's Hospital; & Red Sox Foundation and Massachusetts General Hospital Home Base Program, Boston, MA, USA
| | - Rudy J Castellani
- Division of Neuropathology, University of Maryland School of Medicine, USA
| |
Collapse
|
35
|
Duyckaerts C, Braak H, Brion JP, Buée L, Del Tredici K, Goedert M, Halliday G, Neumann M, Spillantini MG, Tolnay M, Uchihara T. PART is part of Alzheimer disease. Acta Neuropathol 2015; 129:749-56. [PMID: 25628035 PMCID: PMC4405349 DOI: 10.1007/s00401-015-1390-7] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 12/16/2022]
Abstract
It has been proposed that tau aggregation confined to entorhinal cortex and hippocampus, with no or only minimal Aβ deposition, should be considered as a 'primary age-related tauopathy' (PART) that is not integral to the continuum of sporadic Alzheimer disease (AD). Here, we examine the evidence that PART has a pathogenic mechanism and a prognosis which differ from those of AD. We contend that no specific property of the entorhinal-hippocampal tau pathology makes it possible to predict either a limited progression or the development of AD, and that biochemical differences await an evidence base. On the other hand, entorhinal-hippocampal tau pathology is an invariant feature of AD and is always associated with its development. Rather than creating a separate disease entity, we recommend the continued use of an analytical approach based on NFT stages and Aβ phases with no inference about hypothetical disease processes.
Collapse
Affiliation(s)
- Charles Duyckaerts
- Laboratoire de Neuropathologie Escourolle, AP-HP, Hôpital de la Salpêtrière, 47 Bd de l'Hôpital, 75651, Paris Cedex 13, France,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Crary JF, Trojanowski JQ, Schneider JA, Abisambra JF, Abner EL, Alafuzoff I, Arnold SE, Attems J, Beach TG, Bigio EH, Cairns NJ, Dickson DW, Gearing M, Grinberg LT, Hof PR, Hyman BT, Jellinger K, Jicha GA, Kovacs GG, Knopman DS, Kofler J, Kukull WA, Mackenzie IR, Masliah E, McKee A, Montine TJ, Murray ME, Neltner JH, Santa-Maria I, Seeley WW, Serrano-Pozo A, Shelanski ML, Stein T, Takao M, Thal DR, Toledo JB, Troncoso JC, Vonsattel JP, White CL, Wisniewski T, Woltjer RL, Yamada M, Nelson PT. Primary age-related tauopathy (PART): a common pathology associated with human aging. Acta Neuropathol 2014; 128:755-66. [PMID: 25348064 DOI: 10.1007/s00401-014-1349-0] [Citation(s) in RCA: 1061] [Impact Index Per Article: 96.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/26/2014] [Accepted: 09/28/2014] [Indexed: 01/31/2023]
Abstract
We recommend a new term, "primary age-related tauopathy" (PART), to describe a pathology that is commonly observed in the brains of aged individuals. Many autopsy studies have reported brains with neurofibrillary tangles (NFTs) that are indistinguishable from those of Alzheimer's disease (AD), in the absence of amyloid (Aβ) plaques. For these "NFT+/Aβ-" brains, for which formal criteria for AD neuropathologic changes are not met, the NFTs are mostly restricted to structures in the medial temporal lobe, basal forebrain, brainstem, and olfactory areas (bulb and cortex). Symptoms in persons with PART usually range from normal to amnestic cognitive changes, with only a minority exhibiting profound impairment. Because cognitive impairment is often mild, existing clinicopathologic designations, such as "tangle-only dementia" and "tangle-predominant senile dementia", are imprecise and not appropriate for most subjects. PART is almost universally detectable at autopsy among elderly individuals, yet this pathological process cannot be specifically identified pre-mortem at the present time. Improved biomarkers and tau imaging may enable diagnosis of PART in clinical settings in the future. Indeed, recent studies have identified a common biomarker profile consisting of temporal lobe atrophy and tauopathy without evidence of Aβ accumulation. For both researchers and clinicians, a revised nomenclature will raise awareness of this extremely common pathologic change while providing a conceptual foundation for future studies. Prior reports that have elucidated features of the pathologic entity we refer to as PART are discussed, and working neuropathological diagnostic criteria are proposed.
Collapse
|
37
|
Nutrition and the brain: what advice should we give? Neurobiol Aging 2014; 35 Suppl 2:S79-83. [PMID: 24925810 DOI: 10.1016/j.neurobiolaging.2014.02.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/27/2014] [Indexed: 11/23/2022]
Abstract
The knowledge base of nutrition and the brain is steadily expanding. Much of the research is aimed at ways to protect the brain from damage. In adults, the major causes of brain damage are aging and dementia. The most prominent dementia, and the condition that grabs the most public attention, is Alzheimer's disease. The assumption in the field is that possibly some change in nutrition could protect the brain and prevent, delay, or minimize Alzheimer's disease damage. Presented here is a framework for understanding the implications of this research. There is a gap between publishing research results and change in public nutrition behavior. Several influencing elements intervene. These include regulatory agencies and all the organizations and people who advise the public, all with their own perspectives. In considering what advice to give, advisors may consider effectiveness, research model, persuasiveness, and risks, among other factors. Advice about nutrition and Alzheimer's disease today requires several caveats.
Collapse
|
38
|
McCluskey LF, Geser F, Elman LB, Van Deerlin VM, Robinson JL, Lee VMY, Trojanowski JQ. Atypical Alzheimer's disease in an elderly United States resident with amyotrophic lateral sclerosis and pathological tau in spinal motor neurons. Amyotroph Lateral Scler Frontotemporal Degener 2014; 15:466-72. [PMID: 24809433 DOI: 10.3109/21678421.2014.903973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|