1
|
Alhadidy MM, Stemmer PM, Kanaan NM. O-GlcNAc modification differentially regulates microtubule binding and pathological conformations of tau isoforms in vitro. J Biol Chem 2025; 301:108263. [PMID: 39909381 PMCID: PMC11927755 DOI: 10.1016/j.jbc.2025.108263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
Tau proteins undergo several posttranslational modifications in physiological and disease conditions. In Alzheimer's disease, O-GlcNAcylation modification of serine/threonine (S/T) residues in tau is reduced. In mouse models of tauopathy, O-GlcNAcase inhibitors lead to increased O-GlcNAcylation and decreased filamentous aggregates of tau. However, various nonfilamentous tau conformations, linked to toxicity and neurodegeneration in tauopathies, involve processes like oligomerization, misfolding, and greater exposure of the phosphatase-activating domain in the amino terminus of tau. Additionally, it is becoming clearer that posttranslational modifications may differently regulate tau pathobiology in an isoform-dependent manner. Therefore, it is crucial to investigate the effects of O-GlcNAcylation on nonfilamentous conformations of both the four-repeat (4R, e.g., hT40) and three-repeat (3R, e.g., hT39) tau isoforms. In this study, we assessed how O-GlcNAcylation impacts pathological tau conformations of the longest 4R and 3R tau isoforms (hT40 and hT39, respectively) using recombinant proteins. Mass spectrometry showed that tau is modified with O-GlcNAc at multiple S/T residues, primarily in the proline-rich domain and the C-terminal region. O-GlcNAcylation of hT40 and hT39 does not affect microtubule polymerization but has opposite effects on hT40 (increases) and hT39 (decreases) binding to preformed microtubules. Although O-GlcNAcylation interferes with forming filamentous hT40 aggregates, it does not alter the formation of pathological nonfilamentous tau conformations. On the other hand, O-GlcNAcylation increases the formation of pathological nonfilamentous hT39 conformations. These findings suggest that O-GlcNAcylation differentially modulates microtubule binding and the adoption of pathological tau conformations in the longest 4R and 3R tau isoforms.
Collapse
Affiliation(s)
- Mohammed M Alhadidy
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, United States; Neuroscience Program, Michigan State University, East Lansing, Michigan, United States
| | - Paul M Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan, United States; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, United States
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, United States; Neuroscience Program, Michigan State University, East Lansing, Michigan, United States.
| |
Collapse
|
2
|
Taza M, Schmitz TW, Spreng RN. Structural changes to the basal forebrain cholinergic system in the continuum of Alzheimer disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:81-93. [PMID: 40340069 DOI: 10.1016/b978-0-443-19088-9.00013-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
In this chapter, we review evidence, derived predominantly from in vivo human MRI studies, that the basal forebrain (BF) and its projection system undergo structural changes across the continuum of Alzheimer disease (AD) progression. We examine how these changes are detectable from the earliest presymptomatic stages and continue into the prodromal and clinical phases of AD. The chapter begins with a brief overview of BF neuroanatomy before characterizing how changes to the BF and ascending cholinergic white matter projections parallel AD progression. In subsequent sections, we describe how these structural changes are exacerbated in the presence of amyloid and tau pathology, as well as in individuals at elevated genetic risk for AD. We conclude with a review of recent findings implicating the BF as a potential origin site for AD neuropathology and discuss the transsynaptic spread hypothesis of AD progression, from the BF to cortical projection targets.
Collapse
Affiliation(s)
- Miriam Taza
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Taylor W Schmitz
- Department of Physiology and Pharmacology, Western University, London, ON, Canada; Robarts Research Institute, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada; Lawson Health Research Institute, London, ON, Canada
| | - R Nathan Spreng
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University, Montreal, QC, Canada; Department of Psychology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
3
|
Mufson EJ, Perez SE. The cholinotrophic system in Down syndrome. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:185-213. [PMID: 40340061 DOI: 10.1016/b978-0-443-19088-9.00017-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Cholinergic basal forebrain (CBF) projection neurons within the nucleus basalis and striatal cholinergic interneurons degenerate in individuals with Down syndrome (DS). However, the neuropathobiology of these diverse cholinergic phenotypes remains underinvestigated. This review summarizes the alterations of cholinergic, neurotrophic survival and cell death factors as well as tau pathology and amyloidopathy, and their effects upon these cell types in DS. In trisomy, the developing cholinergic system remains stable, whereas the neurotrophic receptors are compromised between control and DS cases. Both cholinergic neuronal phenotypes display severe cellular degeneration in both adult and the aged people with DS. Although developing cholinergic striatal neurons display a similar morphology between phenotypes, cholinergic striatal neurons appear dystrophic in adults with DS. Both cholinergic cell types display tau tangle pathology in elders with DS. Novel findings suggest that alterations in plasma and cerebral spinal fluid levels of proNGF, NGF metabolites, and select classes of neuronal genes are potential biomarkers to distinguish nondemented from demented people with DS. Compounds that target cholinergic pathways, TrkA agonists, p75NTR/proNGF small molecular antagonists, NGF metabolites, and select gene ontology classes are potential targets to slow degeneration of the CBF memory connectome in DS with translation to AD.
Collapse
Affiliation(s)
- Elliott J Mufson
- Department of Translational Neuroscience, St. Joseph's Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, United States; Department of Neurology, St. Joseph's Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, United States.
| | - Sylvia E Perez
- Department of Translational Neuroscience, St. Joseph's Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, United States
| |
Collapse
|
4
|
Moss DE, Perez RG. The phospho-tau cascade, basal forebrain neurodegeneration, and dementia in Alzheimer's disease: Anti-neurodegenerative benefits of acetylcholinesterase inhibitors. J Alzheimers Dis 2024; 102:617-626. [PMID: 39533696 DOI: 10.1177/13872877241289602] [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] [Indexed: 11/16/2024]
Abstract
A conundrum in Alzheimer's disease (AD) is why the long-term use of acetylcholinesterase (AChE) inhibitors, intended for treatment of dementia, results in slowing neurodegeneration in the cholinergic basal forebrain, hippocampus, and cortex. The phospho-tau cascade hypothesis presented here attempts to answer that question by unifying three hallmark features of AD into a specific sequence of events. It is proposed that the hyperphosphorylation of tau protein leads to the AD-associated deficit of nerve growth factor (NGF), then to atrophy of the cholinergic basal forebrain and dementia. Because the release of pro-nerve growth factor (pro-NGF) is activity-dependent and is controlled by basal forebrain projections to the hippocampus and cortex, our hypothesis is that AChE inhibitors act by increasing acetylcholine-dependent pro-NGF release and, thus, augmenting the availability of mature NGF and improving basal forebrain survival. If correct, improved central nervous system-selective AChE inhibitor therapy started prophylactically, before AD-associated basal forebrain atrophy and cognitive impairment onset, has the potential to delay not only the onset of dementia but also its rate of advancement. The phospho-tau hypothesis thus suggests that preventing hyperphosphorylation of tau protein, early should be a high priority as a strategy to help reduce dementia and its associated widespread social and economic suffering.
Collapse
Affiliation(s)
- Donald E Moss
- Professor Emeritus, University of Texas at El Paso, El Paso, TX, USA
| | | |
Collapse
|
5
|
Suzuki H, Tagai K, Ono M, Shimizu H, Endo H, Matsumoto H, Kubota M, Kataoka Y, Moriguchi S, Kurose S, Ichihashi M, Shinotoh H, Matsuoka K, Kokubo N, Tatebe H, Matsuura S, Yamamoto Y, Momota Y, Kawamura K, Zhang MR, Takado Y, Shimada H, Tokuda T, Onaya M, Mimura M, Kakita A, Sahara N, Uchida H, Higuchi M, Takahata K. Distinct tau pathologies in the nucleus basalis of Meynert between early-onset and late-onset Alzheimer's disease patients revealed by positron emission tomography. J Alzheimers Dis 2024; 102:1271-1285. [PMID: 39529384 DOI: 10.1177/13872877241297382] [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] [Indexed: 11/16/2024]
Abstract
BACKGROUND Tau accumulation in the nucleus basalis of Meynert (nbM) has been documented in Alzheimer's disease (AD), but its relationship to neuropathological changes in other brain regions and cognitive deficits remains unclear, particularly between early-onset AD (EOAD) and late-onset AD (LOAD). OBJECTIVE To evaluate tau accumulation patterns in the nbM and other brain regions in EOAD and LOAD using 18F-florzolotau PET and examine correlations with cognitive function. METHODS Thirty-eight amyloid-positive AD patients (15 EOAD, 23 LOAD) and 46 healthy controls underwent 18F-florzolotau PET. Tau levels were quantified in the nbM and Braak-staging regions. Postmortem brain samples were examined to assess 18F-florzolotau binding to tau deposits. RESULTS EOAD showed a higher overall tau burden, including in the nbM, compared with LOAD. However, nbM tau levels correlated more strongly with cognitive decline in LOAD than EOAD. The relationship between nbM tau and neocortical tau differed between EOAD and LOAD. Histopathology revealed abundant 18F-florzolotau labeling of neurofibrillary tangles (NFTs) and ghost tangles in AD nbM samples. CONCLUSIONS This study provides the first in vivo PET evidence of differential nbM tau pathology between EOAD and LOAD, with higher accumulation but weaker correlation to cognition in EOAD. The distinct relationships between nbM and cortical tau in EOAD and LOAD suggest divergent pathological trajectories. 18F-florzolotau PET successfully visualized NFTs and extracellular ghost tangles in the nbM across AD stages. These findings highlight the importance of considering age of onset when evaluating tau pathology and its clinical correlates in AD.
Collapse
Affiliation(s)
- Hisaomi Suzuki
- National Hospital Organization (NHO) Shimofusa Psychiatric Medical Center, Chiba, Japan
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Neuropychiatry, Keio University School of Medicine, Shinjuku, Tokyo, Japan
- Department of Neuropsychiatry, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Kenji Tagai
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Maiko Ono
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Hiroshi Shimizu
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hironobu Endo
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Hideki Matsumoto
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Manabu Kubota
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuko Kataoka
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Sho Moriguchi
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Neuropychiatry, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Shin Kurose
- National Hospital Organization (NHO) Shimofusa Psychiatric Medical Center, Chiba, Japan
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Neuropychiatry, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Masanori Ichihashi
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Hitoshi Shinotoh
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Kiwamu Matsuoka
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Psychiatry, Nara Medical University, Nara, Japan
| | - Naomi Kokubo
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Harutsugu Tatebe
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Sayo Matsuura
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Yasuharu Yamamoto
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Neuropychiatry, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yuki Momota
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Kazunori Kawamura
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Yuhei Takado
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Hitoshi Shimada
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Functional Neurology & Neurosurgery, Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takahiko Tokuda
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Mitsumoto Onaya
- National Hospital Organization (NHO) Shimofusa Psychiatric Medical Center, Chiba, Japan
- Department of Neuropychiatry, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Masaru Mimura
- Department of Neuropychiatry, Keio University School of Medicine, Shinjuku, Tokyo, Japan
- Center for Preventive Medicine, Keio University, Tokyo, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Naruhiko Sahara
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Hiroyuki Uchida
- Department of Neuropychiatry, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Makoto Higuchi
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Keisuke Takahata
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
- Department of Neuropychiatry, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| |
Collapse
|
6
|
Zadrozny M, Drapich P, Gasiorowska-Bien A, Niewiadomski W, Harrington CR, Wischik CM, Riedel G, Niewiadomska G. Neuroprotection of Cholinergic Neurons with a Tau Aggregation Inhibitor and Rivastigmine in an Alzheimer's-like Tauopathy Mouse Model. Cells 2024; 13:642. [PMID: 38607082 PMCID: PMC11011792 DOI: 10.3390/cells13070642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Basal forebrain cholinergic dysfunction, most likely linked with tau protein aggregation, is a characteristic feature of Alzheimer's disease (AD). Recent evidence suggests that tau protein is a putative target for the treatment of dementia, and the tau aggregation inhibitor, hydromethylthionine mesylate (HMTM), has emerged as a potential disease-modifying treatment. However, its efficacy was diminished in patients already receiving approved acetylcholinesterase inhibitors. In this study, we ask whether this negative interaction can also be mimicked in experimental tau models of AD and whether the underlying mechanism can be understood. From a previous age profiling study, 6-month-old line 1 (L1) tau transgenic mice were characterized by a severe reduction in several cholinergic markers. We therefore assessed whether long-term pre-exposure with the acetylcholinesterase inhibitor rivastigmine alone and in conjunction with the tau aggregation inhibitor HMTM can reverse cholinergic deficits in L1. Rivastigmine and HMTM, and combinations of the two compounds were administered orally for 11 weeks to both L1 and wild-type mice. The brains were sectioned with a focus on the basal forebrain, motor cortex and hippocampus. Immunohistochemical staining and quantification of choline acetyltransferase (ChAT), tyrosine kinase A (TrkA)-positive neurons and relative optical intensity (ROI) for vesicular acetylcholine transporter (VAChT), and acetylcholinesterase (AChE) reactivity confirmed reversal of the diminished cholinergic phenotype of interneurons (nucleus accumbens, striatum) and projection neurons (medial septum, nucleus basalis magnocellularis) by HMTM, to a greater extent than by rivastigmine alone in L1 mice. Combined administration did not yield additivity but, in most proxies, led to antagonistic effects in which rivastigmine decreased the benefits shown with HMTM alone. Local markers (VAChT and AChE) in target structures of the basal forebrain, motor cortex and hippocampal CA3 seemed to be normalized by HMTM, but not by rivastigmine or the combination of both drugs. HMTM, which was developed as a tau aggregation inhibitor, strongly decreased the tau load in L1 mice, however, not in combination with rivastigmine. Taken together, these data confirm a cholinergic phenotype in L1 tau transgenic mice that resembles the deficits observed in AD patients. This phenotype is reversible by HMTM, but at the same time appears to be subject to a homeostatic regulation induced by chronic pre-treatment with an acetylcholinesterase inhibitor, which interferes with the efficacy of HMTM. The strongest phenotypic reversal coincided with a normalization of the tau load in the cortex and hippocampus of L1, suggesting that tau accumulation underpins the loss of cholinergic markers in the basal forebrain and its projection targets.
Collapse
Affiliation(s)
- Maciej Zadrozny
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Patrycja Drapich
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Anna Gasiorowska-Bien
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Wiktor Niewiadomski
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Charles R. Harrington
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
- TauRx Therapeutics Ltd., Aberdeen AB24 3FX, UK
| | - Claude M. Wischik
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
- TauRx Therapeutics Ltd., Aberdeen AB24 3FX, UK
| | - Gernot Riedel
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
| | - Grazyna Niewiadomska
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
- Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| |
Collapse
|
7
|
Picton B, Wong J, Lopez AM, Solomon SS, Andalib S, Brown NJ, Dutta RR, Paff MR, Hsu FP, Oh MY. Deep Brain Stimulation as an Emerging Therapy for Cognitive Decline in Alzheimer Disease: Systematic Review of Evidence and Current Targets. World Neurosurg 2024; 184:253-266.e2. [PMID: 38141755 DOI: 10.1016/j.wneu.2023.12.083] [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: 10/21/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/25/2023]
Abstract
OBJECTIVE With no cure for Alzheimer disease (AD), current efforts involve therapeutics that prevent further cognitive impairment. Deep brain stimulation (DBS) has been studied for its potential to mitigate AD symptoms. This systematic review investigates the efficacy of current and previous targets for their ability to slow cognitive decline in treating AD. METHODS A systematic review of the literature was performed through a search of the PubMed, Scopus, and Web of Science databases. Human studies between 1994 and 2023 were included. Sample size, cognitive outcomes, and complications were recorded for each study. RESULTS Fourteen human studies were included: 7 studies with 6 distinct cohorts (n = 56) targeted the fornix, 6 studies with 3 distinct cohorts (n = 17) targeted the nucleus basalis of Meynert (NBM), and 1 study (n = 3) investigated DBS of the ventral striatum (VS). The Alzheimer's Disease Assessment Scale-Cognitive Subscale, Mini-Mental State Examination, and Clinical Dementia Rating Scale Sum of Boxes were used as the primary outcomes. In 5 of 6 cohorts where DBS targeted the fornix, cognitive decline was slowed based on the Alzheimer's Disease Assessment Scale-Cognitive Subscale or Mini-Mental State Examination scores. In 2 of 3 NBM cohorts, a similar reduction was reported. When DBS targeted the VS, the patients' Clinical Dementia Rating Scale Sum of Boxes scores indicated a slowed decline. CONCLUSIONS This review summarizes current evidence and addresses variability in study designs regarding the therapeutic benefit of DBS of the fornix, NBM, and VS. Because of varying study parameters, varying outcome measures, varying study durations, and limited cohort sizes, definitive conclusions regarding the utility of DBS for AD cannot be made. Further investigation is needed to determine the safety and efficacy of DBS for AD.
Collapse
Affiliation(s)
- Bryce Picton
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA.
| | - Joey Wong
- School of Medicine, University of California, Irvine, Orange, California, USA
| | - Alexander M Lopez
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA
| | - Sean S Solomon
- School of Medicine, University of California, Irvine, Orange, California, USA
| | - Saman Andalib
- School of Medicine, University of California, Irvine, Orange, California, USA
| | - Nolan J Brown
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA
| | - Rajeev R Dutta
- School of Medicine, University of California, Irvine, Orange, California, USA
| | - Michelle R Paff
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA
| | - Frank P Hsu
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA
| | - Michael Y Oh
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA
| |
Collapse
|
8
|
Jung Y, Damoiseaux JS. The potential of blood neurofilament light as a marker of neurodegeneration for Alzheimer's disease. Brain 2024; 147:12-25. [PMID: 37540027 PMCID: PMC11484517 DOI: 10.1093/brain/awad267] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023] Open
Abstract
Over the past several years, there has been a surge in blood biomarker studies examining the value of plasma or serum neurofilament light (NfL) as a biomarker of neurodegeneration for Alzheimer's disease. However, there have been limited efforts to combine existing findings to assess the utility of blood NfL as a biomarker of neurodegeneration for Alzheimer's disease. In addition, we still need better insight into the specific aspects of neurodegeneration that are reflected by the elevated plasma or serum concentration of NfL. In this review, we survey the literature on the cross-sectional and longitudinal relationships between blood-based NfL levels and other, neuroimaging-based, indices of neurodegeneration in individuals on the Alzheimer's continuum. Then, based on the biomarker classification established by the FDA-NIH Biomarker Working group, we determine the utility of blood-based NfL as a marker for monitoring the disease status (i.e. monitoring biomarker) and predicting the severity of neurodegeneration in older adults with and without cognitive decline (i.e. a prognostic or a risk/susceptibility biomarker). The current findings suggest that blood NfL exhibits great promise as a monitoring biomarker because an increased NfL level in plasma or serum appears to reflect the current severity of atrophy, hypometabolism and the decline of white matter integrity, particularly in the brain regions typically affected by Alzheimer's disease. Longitudinal evidence indicates that blood NfL can be useful not only as a prognostic biomarker for predicting the progression of neurodegeneration in patients with Alzheimer's disease but also as a susceptibility/risk biomarker predicting the likelihood of abnormal alterations in brain structure and function in cognitively unimpaired individuals with a higher risk of developing Alzheimer's disease (e.g. those with a higher amyloid-β). There are still limitations to current research, as discussed in this review. Nevertheless, the extant literature strongly suggests that blood NfL can serve as a valuable prognostic and susceptibility biomarker for Alzheimer's disease-related neurodegeneration in clinical settings, as well as in research settings.
Collapse
Affiliation(s)
- Youjin Jung
- Department of Psychology, Wayne State University, Detroit, MI 48202, USA
- Institute of Gerontology, Wayne State University, Detroit, MI 48202, USA
| | - Jessica S Damoiseaux
- Department of Psychology, Wayne State University, Detroit, MI 48202, USA
- Institute of Gerontology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
9
|
Shulman D, Dubnov S, Zorbaz T, Madrer N, Paldor I, Bennett DA, Seshadri S, Mufson EJ, Greenberg DS, Loewenstein Y, Soreq H. Sex-specific declines in cholinergic-targeting tRNA fragments in the nucleus accumbens in Alzheimer's disease. Alzheimers Dement 2023; 19:5159-5172. [PMID: 37158312 PMCID: PMC10632545 DOI: 10.1002/alz.13095] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/21/2023] [Indexed: 05/10/2023]
Abstract
INTRODUCTION Females with Alzheimer's disease (AD) suffer accelerated dementia and loss of cholinergic neurons compared to males, but the underlying mechanisms are unknown. Seeking causal contributors to both these phenomena, we pursued changes in transfer RNS (tRNA) fragments (tRFs) targeting cholinergic transcripts (CholinotRFs). METHODS We analyzed small RNA-sequencing (RNA-Seq) data from the nucleus accumbens (NAc) brain region which is enriched in cholinergic neurons, compared to hypothalamic or cortical tissues from AD brains; and explored small RNA expression in neuronal cell lines undergoing cholinergic differentiation. RESULTS NAc CholinotRFs of mitochondrial genome origin showed reduced levels that correlated with elevations in their predicted cholinergic-associated mRNA targets. Single-cell RNA seq from AD temporal cortices showed altered sex-specific levels of cholinergic transcripts in diverse cell types; inversely, human-originated neuroblastoma cells under cholinergic differentiation presented sex-specific CholinotRF elevations. DISCUSSION Our findings support CholinotRFs contributions to cholinergic regulation, predicting their involvement in AD sex-specific cholinergic loss and dementia.
Collapse
Affiliation(s)
- Dana Shulman
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Serafima Dubnov
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Tamara Zorbaz
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Nimrod Madrer
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Iddo Paldor
- The Neurosurgery Department, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 South Paulina, Suite 1028, Chicago, IL 60612, USA
| | - Sudha Seshadri
- UT Health Medical Arts & Research Center, San Antonio , TX 78229, USA
| | - Elliott J. Mufson
- Barrow Neurological Institute, St. Joseph's Medical Center, Phoenix, AZ, 85013, USA
| | - David S. Greenberg
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yonatan Loewenstein
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Department of Neurobiology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Department of Cognitive Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Federmann Center for the Study of Rationality, Jerusalem 9190401, Israel
| | - Hermona Soreq
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
10
|
Moore KBE, Hung TJ, Fortin JS. Hyperphosphorylated tau (p-tau) and drug discovery in the context of Alzheimer's disease and related tauopathies. Drug Discov Today 2023; 28:103487. [PMID: 36634842 PMCID: PMC9975055 DOI: 10.1016/j.drudis.2023.103487] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by intracellular neurofibrillary tangles (NFTs) and extracellular β-amyloid (βA) plaques. No disease-modifying therapy is currently available to prevent the progression of, or cure, the disease. Misfolded hyperphosphorylated tau (p-tau) is considered a pivotal point in the pathogenesis of AD and other tauopathies. Compelling evidence suggests that it is a key driver of the accumulation of NFTs and can be directly correlated with the extent of dementia in patients with AD. Therefore, inhibiting tau hyperphosphorylation-induced aggregation could be a viable strategy to discover and develop therapeutics for patients with AD.
Collapse
Affiliation(s)
- Kendall B E Moore
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA
| | - Ta-Jung Hung
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA
| | - Jessica S Fortin
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA.
| |
Collapse
|
11
|
Shulman D, Dubnov S, Zorbaz T, Madrer N, Paldor I, Bennett DA, Seshadri S, Mufson EJ, Greenberg DS, Loewenstein Y, Soreq H. Sex-specific declines in cholinergic-targeting tRNA fragments in the nucleus accumbens in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527612. [PMID: 36798311 PMCID: PMC9934682 DOI: 10.1101/2023.02.08.527612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Introduction Females with Alzheimer's disease (AD) suffer accelerated dementia and loss of cholinergic neurons compared to males, but the underlying mechanisms are unknown. Seeking causal contributors to both these phenomena, we pursued changes in tRNA fragments (tRFs) targeting cholinergic transcripts (CholinotRFs). Methods We analyzed small RNA-sequencing data from the nucleus accumbens (NAc) brain region which is enriched in cholinergic neurons, compared to hypothalamic or cortical tissues from AD brains; and explored small RNA expression in neuronal cell lines undergoing cholinergic differentiation. Results NAc CholinotRFs of mitochondrial genome origin showed reduced levels that correlated with elevations in their predicted cholinergic-associated mRNA targets. Single cell RNA seq from AD temporal cortices showed altered sex-specific levels of cholinergic transcripts in diverse cell types; inversely, human-originated neuroblastoma cells under cholinergic differentiation presented sex-specific CholinotRF elevations. Discussion Our findings support CholinotRFs contributions to cholinergic regulation, predicting their involvement in AD sex-specific cholinergic loss and dementia.
Collapse
Affiliation(s)
- Dana Shulman
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Serafima Dubnov
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Tamara Zorbaz
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Nimrod Madrer
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Iddo Paldor
- The Neurosurgery Department, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, 600 South Paulina, Suite 1028, Chicago, IL 60612, USA
| | - Sudha Seshadri
- UT Health Medical Arts & Research Center, San Antonio, TX 78229, USA
| | - Elliott J. Mufson
- Barrow Neurological Institute, St. Joseph’s Medical Center, Phoenix, AZ, 85013, USA
| | - David S. Greenberg
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yonatan Loewenstein
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Department of Neurobiology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Department of Cognitive Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Federmann Center for the Study of Rationality, Jerusalem 9190401, Israel
| | - Hermona Soreq
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
12
|
The Nerve Growth Factor Receptor (NGFR/p75 NTR): A Major Player in Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24043200. [PMID: 36834612 PMCID: PMC9965628 DOI: 10.3390/ijms24043200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Alzheimer's disease (AD) represents the most prevalent type of dementia in elderly people, primarily characterized by brain accumulation of beta-amyloid (Aβ) peptides, derived from Amyloid Precursor Protein (APP), in the extracellular space (amyloid plaques) and intracellular deposits of the hyperphosphorylated form of the protein tau (p-tau; tangles or neurofibrillary aggregates). The Nerve growth factor receptor (NGFR/p75NTR) represents a low-affinity receptor for all known mammalians neurotrophins (i.e., proNGF, NGF, BDNF, NT-3 e NT-4/5) and it is involved in pathways that determine both survival and death of neurons. Interestingly, also Aβ peptides can blind to NGFR/p75NTR making it the "ideal" candidate in mediating Aβ-induced neuropathology. In addition to pathogenesis and neuropathology, several data indicated that NGFR/p75NTR could play a key role in AD also from a genetic perspective. Other studies suggested that NGFR/p75NTR could represent a good diagnostic tool, as well as a promising therapeutic target for AD. Here, we comprehensively summarize and review the current experimental evidence on this topic.
Collapse
|
13
|
Engels-Domínguez N, Koops EA, Prokopiou PC, Van Egroo M, Schneider C, Riphagen JM, Singhal T, Jacobs HIL. State-of-the-art imaging of neuromodulatory subcortical systems in aging and Alzheimer's disease: Challenges and opportunities. Neurosci Biobehav Rev 2023; 144:104998. [PMID: 36526031 PMCID: PMC9805533 DOI: 10.1016/j.neubiorev.2022.104998] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
Primary prevention trials have shifted their focus to the earliest stages of Alzheimer's disease (AD). Autopsy data indicates that the neuromodulatory subcortical systems' (NSS) nuclei are specifically vulnerable to initial tau pathology, indicating that these nuclei hold great promise for early detection of AD in the context of the aging brain. The increasing availability of new imaging methods, ultra-high field scanners, new radioligands, and routine deep brain stimulation implants has led to a growing number of NSS neuroimaging studies on aging and neurodegeneration. Here, we review findings of current state-of-the-art imaging studies assessing the structure, function, and molecular changes of these nuclei during aging and AD. Furthermore, we identify the challenges associated with these imaging methods, important pathophysiologic gaps to fill for the AD NSS neuroimaging field, and provide future directions to improve our assessment, understanding, and clinical use of in vivo imaging of the NSS.
Collapse
Affiliation(s)
- Nina Engels-Domínguez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Elouise A Koops
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Prokopis C Prokopiou
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maxime Van Egroo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Christoph Schneider
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tarun Singhal
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
14
|
Ramirez E, Min S, Ganegamage SK, Shimanaka K, Sosa MG, Dettmer U, Rochet JC, Fortin JS. Discovery of 4-aminoindole carboxamide derivatives to curtail alpha-synuclein and tau isoform 2N4R oligomer formation. RESULTS IN CHEMISTRY 2023; 5:100938. [PMID: 37346091 PMCID: PMC10284320 DOI: 10.1016/j.rechem.2023.100938] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial, chronic neurodegenerative disease characterized by the presence of extracellular β-amyloid (Aβ) plaques, intraneuronal neurofibrillary tangles (NFTs), activated microglial cells, and an inflammatory state (involving reactive oxygen species production) in the brain. NFTs are comprised of misfolded and hyperphosphorylated forms of the microtubule-binding protein tau. Interestingly, the trimeric form of the 2N4R splice isoform of tau has been found to be more toxic than the trimeric 1N4R isoform in neuron precursor cells. Few drug discovery programs have focused on specific tau isoforms. The present drug discovery project is centered on the anti-aggregation effect of a series of seventeen 4- or 5-aminoindole carboxamides on the 2N4R isoform of tau. The selection of the best compounds was performed using α-synuclein (α-syn). The anti-oligomer and -fibril activities of newly synthesized aminoindole carboxamide derivatives were evaluated with biophysical methods, such as thioflavin T fluorescence assays, photo-induced cross-linking of unmodified proteins, and transmission electron microscopy. To evaluate the reduction of inclusions and cytoprotective effects, M17D neuroblastoma cells expressing inclusion-forming α-syn were treated with the best amide representatives. The 4-aminoindole carboxamide derivatives exhibited a better anti-fibrillar activity compared to their 5-aminoindole counterparts. The amide derivatives 2, 8, and 17 exerted anti-oligomer and anti-fibril activities on α-syn and the 2N4R isoform of tau. At a concentration of 40 μM, compound 8 reduced inclusion formation in M17D neuroblastoma cells expressing inclusion-prone αSynuclein3K::YFP. Our results demonstrate the potential of 4-aminoindole carboxamide derivatives with regard to inhibiting the oligomer formation of α-syn and tau (2N4R isoform) for further optimization prior to pre-clinical studies.
Collapse
Affiliation(s)
- Eduardo Ramirez
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University
| | - Sehong Min
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
| | | | - Kazuma Shimanaka
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Magaly Guzman Sosa
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
| | - Jessica S Fortin
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University
| |
Collapse
|
15
|
Majdi A, Deng Z, Sadigh-Eteghad S, De Vloo P, Nuttin B, Mc Laughlin M. Deep brain stimulation for the treatment of Alzheimer's disease: A systematic review and meta-analysis. Front Neurosci 2023; 17:1154180. [PMID: 37123370 PMCID: PMC10133458 DOI: 10.3389/fnins.2023.1154180] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Background One of the experimental neuromodulation techniques being researched for the treatment of Alzheimer's disease (AD) is deep brain stimulation (DBS). To evaluate the effectiveness of DBS in AD, we performed a systematic review and meta-analysis of the available evidence. Methods From the inception through December 2021, the following databases were searched: Medline via PubMed, Scopus, Embase, Cochrane Library, and Web of Science. The search phrases used were "Alzheimer's disease," "AD," "deep brain stimulation," and "DBS." The information from the included articles was gathered using a standardized data-collecting form. In the included papers, the Cochrane Collaboration methodology was used to evaluate the risk of bias. A fixed-effects model was used to conduct the meta-analysis. Results Only five distinct publications and 6 different comparisons (one study consisted of two phases) were included out of the initial 524 papers that were recruited. DBS had no impact on the cognitive ability in patients with AD [0.116 SMD, 95% confidence interval (CI), -0.236 to 0.469, p = 0.518]. The studies' overall heterogeneity was not significant (κ2 = 6.23, T 2 = 0.053, df = 5, I 2 = 19.76%, p = 0.284). According to subgroup analysis, the fornix-DBS did not improve cognitive function in patients with AD (0.145 SMD, 95%CI, -0.246 to 0.537, p = 0.467). Unfavorable neurological and non-neurological outcomes were also reported. Conclusion The inconsistencies and heterogeneity of the included publications in various target and age groups of a small number of AD patients were brought to light by this meta-analysis. To determine if DBS is useful in the treatment of AD, further studies with larger sample sizes and randomized, double-blinded, sham-controlled designs are required.
Collapse
Affiliation(s)
- Alireza Majdi
- Exp ORL, Department of Neuroscience, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Zhengdao Deng
- Research Group Experimental Neurosurgery and Neuroanatomy, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Philippe De Vloo
- Research Group Experimental Neurosurgery and Neuroanatomy, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Bart Nuttin
- Research Group Experimental Neurosurgery and Neuroanatomy, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Myles Mc Laughlin
- Exp ORL, Department of Neuroscience, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- *Correspondence: Myles Mc Laughlin
| |
Collapse
|
16
|
Liu X, Zeng Q, Luo X, Li K, Xu X, Hong L, Li J, Guan X, Xu X, Huang P, Zhang M, the Alzheimer's Disease Neuroimaging Initiative (ADNI). Effects of APOE ε2 allele on basal forebrain functional connectivity in mild cognitive impairment. CNS Neurosci Ther 2022; 29:597-608. [PMID: 36468416 PMCID: PMC9873529 DOI: 10.1111/cns.14038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 12/10/2022] Open
Abstract
BACKGROUND Basal forebrain cholinergic system (BFCS) dysfunction is associated with cognitive decline in Alzheimer's disease (AD) and mild cognitive impairment (MCI). Apolipoprotein E (APOE) ε2 is a protective genetic factor in AD and MCI, and cholinergic sprouting depends on APOE. OBJECTIVE We investigated the effect of the APOE ε2 allele on BFCS functional connectivity (FC) in cognitively normal (CN) subjects and MCI patients. METHOD We included 60 MCI patients with APOE ε3/ε3, 18 MCI patients with APOE ε2/ε3, 73 CN subjects with APOE ε3/ε3, and 36 CN subjects with APOE ε2/ε3 genotypes who had resting-state functional magnetic resonance imaging data from the Alzheimer's disease Neuroimaging Initiative. We used BFCS subregions (Ch1-3 and Ch4) as seeds and calculated the FC with other brain areas. Using a mixed-effect analysis, we explored the interaction effects of APOE ε2 allele × cognitive status on BFCS-FC. Furthermore, we examined the relationships between imaging metrics, cognitive abilities, and AD pathology markers, controlling for sex, age, and education as covariates. RESULTS An interaction effect on functional connectivity was found between the right Ch4 (RCh4) and left insula (p < 0.05, corrected), and between the RCh4 and left Rolandic operculum (p < 0.05, corrected). Among all subjects and APOE ε2 carriers, RCh4-left Insula FC was associated with early tau deposition. Furthermore, no correlation was found between imaging metrics and amyloid burden. Among all subjects and APOE ε2 carriers, FC metrics were associated with cognitive performance. CONCLUSION The APOE ε2 genotype may play a protective role during BFCS degeneration in MCI.
Collapse
Affiliation(s)
- Xiaocao Liu
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Qingze Zeng
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiao Luo
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Kaicheng Li
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiaopei Xu
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Luwei Hong
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Jixuan Li
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiaojun Guan
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiaojun Xu
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Peiyu Huang
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Min‐Ming Zhang
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | | |
Collapse
|
17
|
Shahpasand‐Kroner H, Portillo J, Lantz C, Seidler PM, Sarafian N, Loo JA, Bitan G. Three-repeat and four-repeat tau isoforms form different oligomers. Protein Sci 2022; 31:613-627. [PMID: 34902187 PMCID: PMC8862439 DOI: 10.1002/pro.4257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 11/11/2022]
Abstract
Different tauopathies are characterized by the isoform-specific composition of the aggregates found in the brain and by structurally distinct tau strains. Although tau oligomers have been implicated as important neurotoxic species, little is known about how the primary structures of the six human tau isoforms affect tau oligomerization because the oligomers are metastable and difficult to analyze. To address this knowledge gap, here, we analyzed the initial oligomers formed by the six tau isoforms in the absence of posttranslational modifications or other manipulations using dot blots probed by an oligomer-specific antibody, native-PAGE/western blots, photo-induced cross-linking of unmodified proteins, mass-spectrometry, and ion-mobility spectroscopy. We found that under these conditions, three-repeat (3R) isoforms are more prone than four-repeat (4R) isoforms to form oligomers. We also tested whether known inhibitors of tau aggregation affect its oligomerization using three small molecules representing different classes of tau aggregation inhibitors, Methylene Blue (MB), the molecular tweezer CLR01, and the all-D peptide TLKIVW, for their ability to inhibit or modulate the oligomerization of the six tau isoforms. Unlike their reported inhibitory effect on tau fibrillation, the inhibitors had little or no effect on the initial oligomerization. Our study provides novel insight into the primary-quaternary structure relationship of human tau and suggests that 3R-tau oligomers may be an important target for future development of compounds targeting pathological tau assemblies.
Collapse
Affiliation(s)
- Hedieh Shahpasand‐Kroner
- Department of NeurologyDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Jennifer Portillo
- Department of NeurologyDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Carter Lantz
- Department of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Paul M. Seidler
- Department of Pharmacology and Pharmaceutical SciencesUniversity of Southern California School of PharmacyLos AngelesCaliforniaUSA
| | - Natalie Sarafian
- Department of NeurologyDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Joseph A. Loo
- Department of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCaliforniaUSA,Molecular Biology InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA,Department of Biological ChemistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Gal Bitan
- Department of NeurologyDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA,Molecular Biology InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA,Brain Research InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
18
|
Sultzer DL, Lim AC, Gordon HL, Yarns BC, Melrose RJ. Cholinergic receptor binding in unimpaired older adults, mild cognitive impairment, and Alzheimer's disease dementia. Alzheimers Res Ther 2022; 14:25. [PMID: 35130968 PMCID: PMC8819935 DOI: 10.1186/s13195-021-00954-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 12/21/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Cholinergic neurotransmitter system dysfunction contributes to cognitive impairment in Alzheimer's disease and other syndromes. However, the specific cholinergic mechanisms and brain structures involved, time course of alterations, and relationships with specific cognitive deficits are not well understood. METHODS This study included 102 older adults: 42 cognitively unimpaired (CU), 28 with mild cognitive impairment (MCI), and 32 with Alzheimer's disease (AD) dementia. Each participant underwent a neuropsychological assessment. Regional brain α4β2 nicotinic cholinergic receptor binding (VT/fp) was measured using 2-[18F]fluoro-3-(2(S)azetidinylmethoxy)pyridine (2FA) and PET imaging. Voxel-wise analyses of group differences were performed. Relationships between receptor binding and cognition, age, and cholinesterase inhibitor medication use were assessed using binding values in six prespecified regions of interest. RESULTS SPM analysis showed the group VT/fp binding differences in the bilateral entorhinal cortex, hippocampus, insula, anterior cingulate, thalamus, and basal ganglia (p < .05, FWE-corrected). Pairwise comparisons revealed lower binding in the AD group compared to the CU group in similar regions. Binding in the entorhinal cortex was lower in the MCI group than in the CU group; binding in the hippocampus was lower in the AD group than in the MCI group. AD participants taking cholinesterase inhibitor medication had lower 2FA binding in the bilateral hippocampus and thalamus compared to those not taking medication. In the CU group, age was negatively associated with 2FA binding in each region of interest (rs = - .33 to - .59, p < .05 for each, uncorrected). Attention, immediate recall, and delayed recall scores were inversely associated with 2FA binding in most regions across the full sample. In the combined group of CU and MCI participants, attention was inversely associated with 2FA binding in most regions, beyond the effect of hippocampal volume. CONCLUSIONS Nicotinic cholinergic receptor binding in specific limbic and subcortical regions is lower in MCI and further reduced in AD dementia, compared to CU older adults, and is related to cognitive deficits. Cognitive decline with age may be a consequence of reduced cholinergic receptor density or binding affinity that may also promote vulnerability to other Alzheimer's processes. Contemporary modification of the "cholinergic deficit" of aging and AD may reveal opportunities to prevent or improve clinical symptoms.
Collapse
Affiliation(s)
- David L. Sultzer
- grid.417119.b0000 0001 0384 5381Psychiatry/Mental Health Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA ,grid.266093.80000 0001 0668 7243Department of Psychiatry and Human Behavior, School of Medicine, and Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA USA
| | - Aaron C. Lim
- grid.417119.b0000 0001 0384 5381Psychiatry/Mental Health Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA ,grid.42505.360000 0001 2156 6853Department of Family Medicine, USC Keck School of Medicine, Alhambra, CA USA
| | - Hailey L. Gordon
- grid.417119.b0000 0001 0384 5381Psychiatry/Mental Health Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA ,grid.147455.60000 0001 2097 0344Biomedical Engineering Department, Carnegie Mellon University, Pittsburgh, PA USA
| | - Brandon C. Yarns
- grid.417119.b0000 0001 0384 5381Psychiatry/Mental Health Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Rebecca J. Melrose
- grid.417119.b0000 0001 0384 5381Psychiatry/Mental Health Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| |
Collapse
|
19
|
Gonzalez S, McHugh TLM, Yang T, Syriani W, Massa SM, Longo FM, Simmons DA. Small molecule modulation of TrkB and TrkC neurotrophin receptors prevents cholinergic neuron atrophy in an Alzheimer's disease mouse model at an advanced pathological stage. Neurobiol Dis 2021; 162:105563. [PMID: 34838668 DOI: 10.1016/j.nbd.2021.105563] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/05/2021] [Accepted: 11/22/2021] [Indexed: 12/23/2022] Open
Abstract
Degeneration of basal forebrain cholinergic neurons (BFCNs) in the nucleus basalis of Meynert (NBM) and vertical diagonal band (VDB) along with their connections is a key pathological event leading to memory impairment in Alzheimer's disease (AD). Aberrant neurotrophin signaling via Trks and the p75 neurotrophin receptor (p75NTR) contributes importantly to BFCN dystrophy. While NGF/TrkA signaling has received the most attention in this regard, TrkB and TrkC signaling also provide trophic support to BFCNs and these receptors may be well located to preserve BFCN connectivity. We previously identified a small molecule TrkB/TrkC ligand, LM22B-10, that promotes cell survival and neurite outgrowth in vitro and activates TrkB/TrkC signaling in the hippocampus of aged mice when given intranasally, but shows poor oral bioavailability. An LM22B-10 derivative, PTX-BD10-2, with improved oral bioavailability has been developed and this study examined its effects on BFCN atrophy in the hAPPLond/Swe (APPL/S) AD mouse model. Oral delivery of PTX-BD10-2 was started after appreciable amyloid and cholinergic pathology was present to parallel the clinical context, as most AD patients start treatment at advanced pathological stages. PTX-BD10-2 restored cholinergic neurite integrity in the NBM and VDB, and reduced NBM neuronal atrophy in symptomatic APPL/S mice. Dystrophy of cholinergic neurites in BF target regions, including the cortex, hippocampus, and amygdala, was also reduced with treatment. Finally, PTX-BD10-2 reduced NBM tau pathology and improved the survival of cholinergic neurons derived from human induced pluripotent stem cells (iPSCs) after amyloid-β exposure. These data provide evidence that targeting TrkB and TrkC signaling with PTX-BD10-2 may be an effective disease-modifying strategy for combating cholinergic dysfunction in AD. The potential for clinical translation is further supported by the compound's reduction of AD-related degenerative processes that have progressed beyond early stages and its neuroprotective effects in human iPSC-derived cholinergic neurons.
Collapse
Affiliation(s)
- Selena Gonzalez
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Tyne L M McHugh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Wassim Syriani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Stephen M Massa
- Department of Neurology, Laboratory for Computational Neurochemistry and Drug Discovery, Veterans Affairs Health Care System and Department of Neurology, University of California-San Francisco, San Francisco, CA 94121, United States of America
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America.
| |
Collapse
|
20
|
Cantero JL, Atienza M, Lage C, Zaborszky L, Vilaplana E, Lopez-Garcia S, Pozueta A, Rodriguez-Rodriguez E, Blesa R, Alcolea D, Lleo A, Sanchez-Juan P, Fortea J. Atrophy of Basal Forebrain Initiates with Tau Pathology in Individuals at Risk for Alzheimer's Disease. Cereb Cortex 2021; 30:2083-2098. [PMID: 31799623 DOI: 10.1093/cercor/bhz224] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/30/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Evidence suggests that the basal forebrain (BF) cholinergic system degenerates early in the course of Alzheimer's disease (AD), likely due to the vulnerability of BF cholinergic neurons to tau pathology. However, it remains unclear whether the presence of tauopathy is the only requirement for initiating the BF degeneration in asymptomatic subjects at risk for AD (AR-AD), and how BF structural deficits evolve from normal aging to preclinical and prodromal AD. Here, we provide human in vivo magnetic resonance imaging evidence supporting that abnormal cerebrospinal fluid levels of phosphorylated tau (T+) are selectively associated with bilateral volume loss of the nucleus basalis of Meynert (nbM, Ch4) in AR-AD individuals. Spreading of atrophy to medial septum and vertical limb of diagonal band Broca (Ch1-Ch2) occurred in both preclinical and prodromal AD. With the exception of A+, all groups revealed significant correlations between volume reduction of BF cholinergic compartments and atrophy of their innervated regions. Overall, these results support the central role played by tauopathy in instigating the nbM degeneration in AR-AD individuals and the necessary coexistence of both AD proteinopathies for spreading damage to larger BF territories, thus affecting the core of the BF cholinergic projection system.
Collapse
Affiliation(s)
- Jose L Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, 41013 Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, 41013 Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
| | - Carmen Lage
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Laszlo Zaborszky
- Center for Molecular and Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, 07102 NJ, USA
| | - Eduard Vilaplana
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| | - Sara Lopez-Garcia
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Ana Pozueta
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Rafael Blesa
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| | - Daniel Alcolea
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| | - Alberto Lleo
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| | - Pascual Sanchez-Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Juan Fortea
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| |
Collapse
|
21
|
Singh RK. Recent Trends in the Management of Alzheimer's Disease: Current Therapeutic Options and Drug Repurposing Approaches. Curr Neuropharmacol 2021; 18:868-882. [PMID: 31989900 PMCID: PMC7569317 DOI: 10.2174/1570159x18666200128121920] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/14/2020] [Accepted: 01/27/2020] [Indexed: 01/31/2023] Open
Abstract
Alzheimer's disease is one of the most progressive forms of dementia, ultimately leading to death in aged populations. The major hallmarks of Alzheimer's disease include deposition of extracellular amyloid senile plaques and intracellular neurofibrillary tangles in brain neuronal cells. Although there are classical therapeutic options available for the treatment of the diseases, however, they provide only a symptomatic relief and do not modify the molecular pathophysiological course of the disease. Recent research advances in Alzheimer's disease have highlighted the potential role of anti-amyloid, anti-tau, and anti-inflammatory therapies. However, these therapies are still in different phases of pre-clinical/clinical development. In addition, drug repositioning/repurposing is another interesting and promising approach to explore rationalized options for the treatment of Alzheimer's disease. This review discusses the different aspects of the pathophysiological mechanism involved in the progression of Alzheimer's disease along with the limitations of current therapies. Furthermore, this review also highlights emerging investigational drugs along with recent drug repurposing approaches for Alzheimer's disease.
Collapse
Affiliation(s)
- Rakesh K Singh
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Manesar, Gurgaon-122413, Haryana, India,Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research,
Raebareli. Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India
| |
Collapse
|
22
|
Sankorrakul K, Qian L, Thangnipon W, Coulson EJ. Is there a role for the p75 neurotrophin receptor in mediating degeneration during oxidative stress and after hypoxia? J Neurochem 2021; 158:1292-1306. [PMID: 34109634 DOI: 10.1111/jnc.15451] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/21/2022]
Abstract
Cholinergic basal forebrain (cBF) neurons are particularly vulnerable to degeneration following trauma and in neurodegenerative conditions. One reason for this is their characteristic expression of the p75 neurotrophin receptor (p75NTR ), which is up-regulated and mediates neuronal death in a range of neurological and neurodegenerative conditions, including dementia, stroke and ischaemia. The signalling pathway by which p75NTR signals cell death is incompletely characterised, but typically involves activation by neurotrophic ligands and signalling through c-Jun kinase, resulting in caspase activation via mitochondrial apoptotic signalling pathways. Less well appreciated is the link between conditions of oxidative stress and p75NTR death signalling. Here, we review the literature describing what is currently known regarding p75NTR death signalling in environments of oxidative stress and hypoxia to highlight the overlap in signalling pathways and the implications for p75NTR signalling in cBF neurons. We propose that there is a causal relationship and define key questions to test this assertion.
Collapse
Affiliation(s)
- Kornraviya Sankorrakul
- School of Biomedical Sciences, Faculty of Medicine and Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Qld., Australia.,Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Lei Qian
- School of Biomedical Sciences, Faculty of Medicine and Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Qld., Australia
| | - Wipawan Thangnipon
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Elizabeth J Coulson
- School of Biomedical Sciences, Faculty of Medicine and Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Qld., Australia
| |
Collapse
|
23
|
Ingham DJ, Blankenfeld BR, Chacko S, Perera C, Oakley BR, Gamblin TC. Fungally Derived Isoquinoline Demonstrates Inducer-Specific Tau Aggregation Inhibition. Biochemistry 2021; 60:1658-1669. [PMID: 34009955 PMCID: PMC8173610 DOI: 10.1021/acs.biochem.1c00111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The microtubule-associated
protein tau promotes the stabilization
of the axonal cytoskeleton in neurons. In several neurodegenerative
diseases, such as Alzheimer’s disease, tau has been found to
dissociate from microtubules, leading to the formation of pathological
aggregates that display an amyloid fibril-like structure. Recent structural
studies have shown that the tau filaments isolated from different
neurodegenerative disorders have structurally distinct fibril cores
that are specific to the disease. These “strains” of
tau fibrils appear to propagate between neurons in a prion-like fashion
that maintains their initial template structure. In addition, the
strains isolated from diseased tissue appear to have structures that
are different from those made by the most commonly used in
vitro modeling inducer molecule, heparin. The structural
differences among strains in different diseases and in vitro-induced tau fibrils may contribute to recent failures in clinical
trials of compounds designed to target tau pathology. This study identifies
an isoquinoline compound (ANTC-15) isolated from the fungus Aspergillus nidulans that can both inhibit filaments induced
by arachidonic acid (ARA) and disassemble preformed ARA fibrils. When
compared to a tau aggregation inhibitor currently in clinical trials
(LMTX, LMTM, or TRx0237), ANTC-15 and LMTX were found to have opposing
inducer-specific activities against ARA and heparin in vitro-induced tau filaments. These findings may help explain the disappointing
results in translating potent preclinical inhibitor candidates to
successful clinical treatments.
Collapse
Affiliation(s)
- David J Ingham
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, United States
| | - Bryce R Blankenfeld
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, United States
| | - Shibin Chacko
- Synthetic Chemical Biology Core Facility, University of Kansas, Lawrence, Kansas 66047, United States
| | - Chamani Perera
- Synthetic Chemical Biology Core Facility, University of Kansas, Lawrence, Kansas 66047, United States
| | - Berl R Oakley
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, United States
| | - Truman Christopher Gamblin
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, United States.,Department of Biology, The University of Texas at San Antonio, San Antonio, Texas 78249, United States
| |
Collapse
|
24
|
Chen YS, Shu K, Kang HC. Deep Brain Stimulation in Alzheimer's Disease: Targeting the Nucleus Basalis of Meynert. J Alzheimers Dis 2021; 80:53-70. [PMID: 33492288 DOI: 10.3233/jad-201141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is becoming a prevalent disease in the elderly population. Past decades have witnessed the development of drug therapies with varying targets. However, all drugs with a single molecular target fail to reverse or ameliorate AD progression, which ultimately results in cortical and subcortical network dysregulation. Deep brain stimulation (DBS) has been proven effective for the treatment of Parkinson's disease, essential tremor, and other neurological diseases. As such, DBS has also been gradually acknowledged as a potential therapy for AD. The current review focuses on DBS of the nucleus basalis of Meynert (NBM). As a critical component of the cerebral cholinergic system and the Papez circuit in the basal ganglia, the NBM plays an indispensable role in the subcortical regulation of memory, attention, and arousal state, which makes the NBM a promising target for modulation of neural network dysfunction and AD treatment. We summarized the intricate projection relations and functionality of the NBM, current approaches for stereotactic localization and evaluation of the NBM, and the therapeutic effects of NBM-DBS both in patients and animal models. Furthermore, the current shortcomings of NBM-DBS, such as variations in cortical blood flow, increased temperature in the target area, and stimulation-related neural damage, were presented.
Collapse
Affiliation(s)
- Yu-Si Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui-Cong Kang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
25
|
Moss DE, Perez RG. Anti-Neurodegenerative Benefits of Acetylcholinesterase Inhibitors in Alzheimer's Disease: Nexus of Cholinergic and Nerve Growth Factor Dysfunction. Curr Alzheimer Res 2021; 18:1010-1022. [PMID: 34911424 PMCID: PMC8855657 DOI: 10.2174/1567205018666211215150547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/04/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is increasingly viewed as a complex multi-dimensional disease without effective treatments. Recent randomized, placebo-controlled studies have shown volume losses of ~0.7% and ~3.5% per year, respectively, in the basal cholinergic forebrain (CBF) and hippocampus in untreated suspected prodromal AD. One year of donepezil treatment reduced these annualized rates of atrophy to about half of untreated rates. Similar positive although variable results have also been found in volumetric measurements of the cortex and whole brain in patients with mild cognitive impairment as well as more advanced AD stages after treatments with all three currently available acetylcholinesterase (AChE) inhibitors (donepezil, rivastigmine, and galantamine). Here we review the anti-neurodegenerative benefits of AChE inhibitors and the expected parallel disease-accelerating impairments caused by anticholinergics, within a framework of the cholinergic hypothesis of AD and AD-associated loss of nerve growth factor (NGF). Consistent with the "loss of trophic factor hypothesis of AD," we propose that AChE inhibitors enhance acetylcholine-dependent release and uptake of NGF, thereby sustaining cholinergic neuronal viability and thus slowing AD-associated degeneration of the CBF, to ultimately delay dementia progression. We propose that improved cholinergic therapies for AD started early in asymptomatic persons, especially those with risk factors, will delay the onset, progression, or emergence of dementia. The currently available competitive and pseudo- irreversible AChE inhibitors are not CNS-selective and thus induce gastrointestinal toxicity that limits cortical AChE inhibition to ~30% (ranges from 19% to 41%) as measured by in vivo PET studies in patients undergoing therapy. These levels of inhibition are marginal relative to what is required for effective symptomatic treatment of dementia or slowing AD-associated neurodegeneration. In contrast, because of the inherently slow de novo synthesis of AChE in the CNS (about one-- tenth the rate of synthesis in peripheral tissues), irreversible AChE inhibitors produce significantly higher levels of inhibition in the CNS than in peripheral tissues. For example, methanesulfonyl fluoride, an irreversible inhibitor reduces CNS AChE activity by ~68% in patients undergoing therapy and ~80% in cortical biopsies of non-human primates. The full therapeutic benefits of AChE inhibitors, whether for symptomatic treatment of dementia or disease-slowing, thus would benefit by producing high levels of CNS inhibition. One way to obtain such higher levels of CNS AChE inhibition would be by using irreversible inhibitors.
Collapse
Affiliation(s)
- Donald E. Moss
- Department of Psychology, University of Texas at El Paso, El Paso, Texas, 79968 USA
| | - Ruth G. Perez
- Department of Molecular and Translational Medicine, Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, 79905 USA
| |
Collapse
|
26
|
Moss DE. Improving Anti-Neurodegenerative Benefits of Acetylcholinesterase Inhibitors in Alzheimer's Disease: Are Irreversible Inhibitors the Future? Int J Mol Sci 2020; 21:E3438. [PMID: 32414155 PMCID: PMC7279429 DOI: 10.3390/ijms21103438] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/01/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Decades of research have produced no effective method to prevent, delay the onset, or slow the progression of Alzheimer's disease (AD). In contrast to these failures, acetylcholinesterase (AChE, EC 3.1.1.7) inhibitors slow the clinical progression of the disease and randomized, placebo-controlled trials in prodromal and mild to moderate AD patients have shown AChE inhibitor anti-neurodegenerative benefits in the cortex, hippocampus, and basal forebrain. CNS neurodegeneration and atrophy are now recognized as biomarkers of AD according to the National Institute on Aging-Alzheimer's Association (NIA-AA) criteria and recent evidence shows that these markers are among the earliest signs of prodromal AD, before the appearance of amyloid. The current AChE inhibitors (donepezil, rivastigmine, and galantamine) have short-acting mechanisms of action that result in dose-limiting toxicity and inadequate efficacy. Irreversible AChE inhibitors, with a long-acting mechanism of action, are inherently CNS selective and can more than double CNS AChE inhibition possible with short-acting inhibitors. Irreversible AChE inhibitors open the door to high-level CNS AChE inhibition and improved anti-neurodegenerative benefits that may be an important part of future treatments to more effectively prevent, delay the onset, or slow the progression of AD.
Collapse
Affiliation(s)
- Donald E Moss
- Department of Psychology, University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
27
|
Antagonism of cysteinyl leukotrienes and their receptors as a neuroinflammatory target in Alzheimer's disease. Neurol Sci 2020; 41:2081-2093. [PMID: 32281039 DOI: 10.1007/s10072-020-04369-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 03/21/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Alzheimer's disease is a complex multifaceted neurodegenerative disorder. It is characterized by the deposition of extracellular amyloid senile plaques and intracellular neurofibrillary tangles leading to progressive dementia and death in aged adult population. Recent emerging research has highlighted a potential pharmacological role of 5-lipoxyenase-cysteinyl leukotriene pathway in molecular pathogenesis of Alzheimer's disease. OBJECTIVE Although cysteinyl leukotrienes and their receptors have a major clinical role in chronic respiratory inflammation, their roles in chronic neuroinflammation in Alzheimer's disease need a detailed and careful exploration. RESULTS AND CONCLUSION This review article highlights a novel role of cysteinyl leukotrienes and their receptors in pathophysiology of Alzheimer's disease in order to understand the underlying molecular mechanism. In addition, it summarizes the recent advances in various pre-clinical and clinical strategies used to modulate this pathway for therapeutic targeting of Alzheimer's disease.
Collapse
|
28
|
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
|
29
|
Mufson EJ, Counts SE, Ginsberg SD, Mahady L, Perez SE, Massa SM, Longo FM, Ikonomovic MD. Nerve Growth Factor Pathobiology During the Progression of Alzheimer's Disease. Front Neurosci 2019; 13:533. [PMID: 31312116 PMCID: PMC6613497 DOI: 10.3389/fnins.2019.00533] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 05/08/2019] [Indexed: 12/12/2022] Open
Abstract
The current review summarizes the pathobiology of nerve growth factor (NGF) and its cognate receptors during the progression of Alzheimer's disease (AD). Both transcript and protein data indicate that cholinotrophic neuronal dysfunction is related to an imbalance between TrkA-mediated survival signaling and the NGF precursor (proNGF)/p75NTR-mediated pro-apoptotic signaling, which may be related to alteration in the metabolism of NGF. Data indicate a spatiotemporal pattern of degeneration related to the evolution of tau pathology within cholinotrophic neuronal subgroups located within the nucleus basalis of Meynert (nbM). Despite these degenerative events the cholinotrophic system is capable of cellular resilience and/or plasticity during the prodromal and later stages of the disease. In addition to neurotrophin dysfunction, studies indicate alterations in epigenetically regulated proteins occur within cholinotrophic nbM neurons during the progression of AD, suggesting a mechanism that may underlie changes in transcript expression. Findings that increased cerebrospinal fluid levels of proNGF mark the onset of MCI and the transition to AD suggests that this proneurotrophin is a potential disease biomarker. Novel therapeutics to treat NGF dysfunction include NGF gene therapy and the development of small molecule agonists for the cognate prosurvival NGF receptor TrkA and antagonists against the pan-neurotrophin p75NTR death receptor for the treatment of AD.
Collapse
Affiliation(s)
- Elliott J. Mufson
- Department of Neurobiology and Neurology, Department of Neurobiology, and Department of Neurological Sciences, Alzheimer’s Disease Laboratory, Barrow Neurological Institute, St. Joseph’s Medical Center, Phoenix, AZ, United States
| | - Scott E. Counts
- Translational Science and Molecular Medicine Michigan State University College of Human Medicine, Grand Rapids, MI, United States
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, Department of Neuroscience, and Physiology and NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States
| | - Laura Mahady
- Department of Neurobiology and Neurology, Department of Neurobiology, and Department of Neurological Sciences, Alzheimer’s Disease Laboratory, Barrow Neurological Institute, St. Joseph’s Medical Center, Phoenix, AZ, United States
| | - Sylvia E. Perez
- Department of Neurobiology and Neurology, Department of Neurobiology, and Department of Neurological Sciences, Alzheimer’s Disease Laboratory, Barrow Neurological Institute, St. Joseph’s Medical Center, Phoenix, AZ, United States
| | - Stephen M. Massa
- Department of Neurology, San Francisco VA Health Care System, University of California, San Francisco, San Francisco, CA, United States
| | - Frank M. Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Milos D. Ikonomovic
- Department of Neurology and Department of Psychiatry, Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
30
|
Cornel iridoid glycoside induces autophagy to protect against tau oligomer neurotoxicity induced by the activation of glycogen synthase kinase-3β. J Nat Med 2019; 73:717-726. [DOI: 10.1007/s11418-019-01318-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/07/2019] [Indexed: 12/20/2022]
|
31
|
Perez SE, Miguel JC, He B, Malek-Ahmadi M, Abrahamson EE, Ikonomovic MD, Lott I, Doran E, Alldred MJ, Ginsberg SD, Mufson EJ. Frontal cortex and striatal cellular and molecular pathobiology in individuals with Down syndrome with and without dementia. Acta Neuropathol 2019; 137:413-436. [PMID: 30734106 PMCID: PMC6541490 DOI: 10.1007/s00401-019-01965-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 02/06/2023]
Abstract
Although, by age 40, individuals with Down syndrome (DS) develop amyloid-β (Aβ) plaques and tau-containing neurofibrillary tangles (NFTs) linked to cognitive impairment in Alzheimer's disease (AD), not all people with DS develop dementia. Whether Aβ plaques and NFTs are associated with individuals with DS with (DSD +) and without dementia (DSD -) is under-investigated. Here, we applied quantitative immunocytochemistry and fluorescent procedures to characterize NFT pathology using antibodies specific for tau phosphorylation (pS422, AT8), truncation (TauC3, MN423), and conformational (Alz50, MC1) epitopes, as well as Aβ and its precursor protein (APP) to frontal cortex (FC) and striatal tissue from DSD + to DSD - cases. Expression profiling of single pS422 labeled FC layer V and VI neurons was also determined using laser capture microdissection and custom-designed microarray analysis. Analysis revealed that cortical and striatal Aβ plaque burdens were similar in DSD + and DSD - cases. In both groups, most FC plaques were neuritic, while striatal plaques were diffuse. By contrast, FC AT8-positive NFTs and neuropil thread densities were significantly greater in DSD + compared to DSD -, while striatal NFT densities were similar between groups. FC pS422-positive and TauC3 NFT densities were significantly greater than Alz50-labeled NFTs in DSD + , but not DSD - cases. Putaminal, but not caudate pS422-positive NFT density, was significantly greater than TauC3-positive NFTs. In the FC, AT8 + pS422 + Alz50, TauC3 + pS422 + Alz50, pS422 + Alz50, and TauC3 + pS422 positive NFTs were more frequent in DSD + compared to DSD- cases. Single gene-array profiling of FC pS422 positive neurons revealed downregulation of 63 of a total of 864 transcripts related to Aβ/tau biology, glutamatergic, cholinergic, and monoaminergic metabolism, intracellular signaling, cell homeostasis, and cell death in DSD + compared DSD - cases. These observations suggest that abnormal tau aggregation plays a critical role in the development of dementia in DS.
Collapse
Affiliation(s)
- Sylvia E Perez
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas St, Phoenix, AZ, 85013, USA
- School of Life Sciences, College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Jennifer C Miguel
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas St, Phoenix, AZ, 85013, USA
| | - Bin He
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas St, Phoenix, AZ, 85013, USA
| | | | - Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, 15213, USA
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, 15213, USA
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ira Lott
- Departments of Pediatrics and Neurology, University of California, Irvine, CA, 92697, USA
| | - Eric Doran
- Departments of Pediatrics and Neurology, University of California, Irvine, CA, 92697, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, NYU Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10021, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, NYU Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10021, USA
- Departments of Neuroscience and Physiology, The NYU Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10021, USA
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas St, Phoenix, AZ, 85013, USA.
| |
Collapse
|
32
|
Nanowired delivery of cerebrolysin with neprilysin and p-Tau antibodies induces superior neuroprotection in Alzheimer's disease. PROGRESS IN BRAIN RESEARCH 2019; 245:145-200. [DOI: 10.1016/bs.pbr.2019.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Mahady L, Nadeem M, Malek-Ahmadi M, Chen K, Perez SE, Mufson EJ. HDAC2 dysregulation in the nucleus basalis of Meynert during the progression of Alzheimer's disease. Neuropathol Appl Neurobiol 2018; 45:380-397. [PMID: 30252960 DOI: 10.1111/nan.12518] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 08/23/2018] [Indexed: 02/06/2023]
Abstract
AIMS Alzheimer's disease (AD) is characterized by degeneration of cholinergic basal forebrain (CBF) neurons in the nucleus basalis of Meynert (nbM), which provides the major cholinergic input to the cortical mantle and is related to cognitive decline in patients with AD. Cortical histone deacetylase (HDAC) dysregulation has been associated with neuronal degeneration during AD progression. However, whether HDAC alterations play a role in CBF degeneration during AD onset is unknown. We investigated global HDAC protein levels and nuclear HDAC2 immunoreactivity in tissue containing the nbM, changes and their association with neurofibrillary tangles (NFTs) during the progression of AD. METHODS We used semi-quantitative western blotting and immunohistochemistry to evaluate HDAC and sirtuin (SIRT) levels in individuals that died with a premortem clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), mild/moderate AD (mAD) or severe AD (sAD). Quantitative immunohistochemistry was used to identify HDAC2 protein levels in individual cholinergic nbM nuclei and their colocalization with the early phosphorylated tau marker AT8, the late-stage apoptotic tau marker TauC3 and Thioflavin-S, a marker of β-pleated sheet structures in NFTs. RESULTS In AD patients, HDAC2 protein levels were dysregulated in the basal forebrain region containing cholinergic neurons of the nbM. HDAC2 nuclear immunoreactivity was reduced in individual cholinergic nbM neurons across disease stages. HDAC2 nuclear reactivity correlated with multiple cognitive domains and with NFT formation. CONCLUSIONS These findings suggest that HDAC2 dysregulation contributes to cholinergic nbM neuronal dysfunction, NFT pathology, and cognitive decline during clinical progression of AD.
Collapse
Affiliation(s)
- L Mahady
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA.,Arizona State University Interdisciplinary Graduate Program in Neuroscience, Tempe, Arizona, USA
| | - M Nadeem
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | | | - K Chen
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | - S E Perez
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - E J Mufson
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
34
|
Denver P, McClean PL. Distinguishing normal brain aging from the development of Alzheimer's disease: inflammation, insulin signaling and cognition. Neural Regen Res 2018; 13:1719-1730. [PMID: 30136683 PMCID: PMC6128051 DOI: 10.4103/1673-5374.238608] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2018] [Indexed: 12/21/2022] Open
Abstract
As populations age, prevalence of Alzheimer's disease (AD) is rising. Over 100 years of research has provided valuable insights into the pathophysiology of the disease, for which age is the principal risk factor. However, in recent years, a multitude of clinical trial failures has led to pharmaceutical corporations becoming more and more unwilling to support drug development in AD. It is possible that dependence on the amyloid cascade hypothesis as a guide for preclinical research and drug discovery is part of the problem. Accumulating evidence suggests that amyloid plaques and tau tangles are evident in non-demented individuals and that reducing or clearing these lesions does not always result in clinical improvement. Normal aging is associated with pathologies and cognitive decline that are similar to those observed in AD, making differentiation of AD-related cognitive decline and neuropathology challenging. In this mini-review, we discuss the difficulties with discerning normal, age-related cognitive decline with that related to AD. We also discuss some neuropathological features of AD and aging, including amyloid and tau pathology, synapse loss, inflammation and insulin signaling in the brain, with a view to highlighting cognitive or neuropathological markers that distinguish AD from normal aging. It is hoped that this review will help to bolster future preclinical research and support the development of clinical tools and therapeutics for AD.
Collapse
Affiliation(s)
- Paul Denver
- Greater Los Angeles Veterans Affairs Healthcare System, West Los Angeles Medical Center and Department of Neurology, University of California, Los Angeles, CA, USA
- Centre for Molecular Biosciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - Paula L. McClean
- Northern Ireland Centre for Stratified Medicine, Clinical, Translational and Research Innovation Centre (C-TRIC), University of Ulster, Derry/Londonderry, Northern Ireland, UK
| |
Collapse
|