1
|
Fu H, Li J, Zhang C, Gao G, Ge Q, Guan X, Cui D. Pathological axonal enlargement in connection with amyloidosis, lysosome destabilization, and bleeding is a major defect in Alzheimer's disease. Neural Regen Res 2026; 21:790-799. [PMID: 40326989 DOI: 10.4103/nrr.nrr-d-24-01440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00047/figure1/v/2025-05-05T160104Z/r/image-tiff Alzheimer's disease is a multi-amyloidosis disease characterized by amyloid-β deposits in brain blood vessels, microaneurysms, and senile plaques. How amyloid-β deposition affects axon pathology has not been examined extensively. We used immunohistochemistry and immunofluorescence staining to analyze the forebrain tissue slices of Alzheimer's disease patients. Widespread axonal amyloidosis with distinctive axonal enlargement was observed in patients with Alzheimer's disease. On average, amyloid-β-positive axon diameters in Alzheimer's disease brains were 1.72 times those of control brain axons. Furthermore, axonal amyloidosis was associated with microtubule-associated protein 2 reduction, tau phosphorylation, lysosome destabilization, and several blood-related markers, such as apolipoprotein E, alpha-hemoglobin, glycosylated hemoglobin type A1C, and hemin. Lysosome destabilization in Alzheimer's disease was also clearly identified in the neuronal soma, where it was associated with the co-expression of amyloid-β, Cathepsin D, alpha-hemoglobin, actin alpha 2, and collagen type IV. This suggests that exogenous hemorrhagic protein intake influences neural lysosome stability. Additionally, the data showed that amyloid-β-containing lysosomes were 2.23 times larger than control lysosomes. Furthermore, under rare conditions, axonal breakages were observed, which likely resulted in Wallerian degeneration. In summary, axonal enlargement associated with amyloidosis, micro-bleeding, and lysosome destabilization is a major defect in patients with Alzheimer's disease. This finding suggests that, in addition to the well-documented neural soma and synaptic damage, axonal damage is a key component of neuronal defects in Alzheimer's disease.
Collapse
Affiliation(s)
- Hualin Fu
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Marine Equipment, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jilong Li
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chunlei Zhang
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guo Gao
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiqi Ge
- Institute of Marine Equipment, Shanghai Jiao Tong University, Shanghai, China
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China
| | - Xinping Guan
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China
- The Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
2
|
Reijner N, Frigerio I, Bouwman MMA, Boon BDC, Guizard N, Jubault T, Hoozemans JJM, Rozemuller AJM, Bouwman FH, Barkhof F, Gordon E, van de Berg WDJ, Jonkman LE. Clinical phenotypes of Alzheimer's disease: investigating atrophy patterns and their pathological correlates. Alzheimers Res Ther 2025; 17:93. [PMID: 40281562 PMCID: PMC12032798 DOI: 10.1186/s13195-025-01727-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND In Alzheimer's disease (AD), MRI atrophy patterns can distinguish between amnestic (typical) and non-amnestic (atypical) clinical phenotypes and are increasingly used for diagnosis and outcome measures in clinical trials. However, understanding how protein accumulation and other key features of neurodegeneration influence these imaging measurements, are lacking. The current study aimed to assess regional MRI patterns of cortical atrophy across clinical AD phenotypes, and their association with amyloid-beta (Aβ), phosphorylated tau (pTau), neuro-axonal degeneration and microvascular deterioration. METHODS Post-mortem in-situ 3DT1 3 T-MRI data was obtained from 33 AD (17 typical, 16 atypical) and 16 control brain donors. Additionally, ante-mortem 3DT1 3 T-MRI scans of brain donors were collected if available. Regional volumes were obtained from MRI scans using an atlas based parcellation software. Eight cortical brain regions were selected from formalin-fixed right hemispheres of brain donors and then immunostained for Aβ, pTau, neurofilament light, and collagen IV. Group comparisons and volume-pathology associations were analyzed using linear mixed models corrected for age, sex, post-mortem delay, and intracranial volume. RESULTS Compared to controls, both typical and atypical AD showed volume loss in the temporo-occipital cortex, while typical AD showed additional volume loss in the parietal cortex. Posterior cingulate volume was lower in typical AD compared to atypical AD (- 6.9%, p = 0.043). In AD, a global positive association between MRI cortical volume and Aβ load (βs = 0.21, p = 0.010), and a global negative association with NfL load (βs = - 0.18, p = 0.018) were observed. Regionally, higher superior parietal gyrus volume was associated with higher Aβ load in typical AD (βs = 0.47, p = 0.004), lower middle frontal gyrus volume associated with higher NfL load in atypical AD (βs = - 0.50, p < 0.001), and lower hippocampal volume associated with higher COLIV load in typical AD (βs = - 1.69, p < 0.001). Comparing post-mortem with ante-mortem scans showed minimal volume differences at scan-intervals within 2 years, highlighting the translational aspect of this study. CONCLUSION For both clinical phenotypes, cortical volume is affected by Aβ and neuro-axonal damage, but in opposing directions. Differences in volume-pathology relationships between clinical phenotypes are region-specific. The findings of this study could improve the interpretation of MRI datasets in heterogenous AD cohorts, both in research and clinical settings.
Collapse
Affiliation(s)
- Niels Reijner
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands.
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands.
- Programs of Brain Imaging, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands.
| | - I Frigerio
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Programs of Brain Imaging, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
| | - M M A Bouwman
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Programs of Brain Imaging, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
| | - B D C Boon
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, USA
| | - N Guizard
- Qynapse, 2 - 10 Rue d'Oradour-Sur-Glane, Paris, 75015, France
| | - T Jubault
- Qynapse, 2 - 10 Rue d'Oradour-Sur-Glane, Paris, 75015, France
| | - J J M Hoozemans
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - A J M Rozemuller
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - F H Bouwman
- Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - F Barkhof
- Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Institutes of Neurology and Healthcare Engineering, University College London, Gower Street, London, UK
| | - E Gordon
- Qynapse, 2 - 10 Rue d'Oradour-Sur-Glane, Paris, 75015, France
| | - W D J van de Berg
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
| | - L E Jonkman
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Programs of Brain Imaging, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
| |
Collapse
|
3
|
Joshi N, Vaidya B, Sharma SS. Transient receptor potential channels as an emerging target for the treatment of Alzheimer's disease: Unravelling the potential of pharmacological interventions. Basic Clin Pharmacol Toxicol 2024; 135:375-400. [PMID: 39209323 DOI: 10.1111/bcpt.14073] [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: 03/01/2024] [Revised: 07/09/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Alzheimer's disease (AD) is a devastating disorder with a multifaceted aetiology characterized by dementia, which later progresses to cognitive impairment. Significant efforts have been made to develop pharmacological interventions that slow down the pathogenesis of AD. However, conventional drugs have failed to satisfactorily treat AD and are more focussed towards symptomatic management. Thus, there is a gap in the literature regarding novel targets and modulators targeting them for the effective treatment of AD. Recent studies have demonstrated that modulation of transient receptor potential (TRP) channels has the potential to halt AD pathogenesis at an early stage and rescue hippocampal neurons from death. Amongst several members, TRP channels like TRPA1, TRPC6, TRPM2 and TRPV2 have shown promising results in the attenuation of neurobehavioural cognitive deficits as well as signalling pathways governing such cognitive decline. Furthermore, as these channels govern the ionic balance in the cell, their beneficial effects have also been known to maintain the homeostasis of Ca2+, which is the major culprit eliciting the vicious cycle of excitotoxicity, mitochondrial dysfunction, ROS generation and neurodegeneration. Despite such tremendous potential of TRP channel modulators, their clinical investigation remains elusive. Therefore, in the present review, we have discussed such agents in the light of TRP channels as molecular targets for the amelioration of AD both at the preclinical and clinical levels.
Collapse
Affiliation(s)
- Nishit Joshi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| | - Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| |
Collapse
|
4
|
Li J, Yang M, Wei R, Cao Y, Fan X, Zhang S. The Predictive Ability of Blood Neurofilament Light Chain in Predicting Cognitive Decline in the Alzheimer's Disease Continuum: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2024; 97:1589-1620. [PMID: 38306045 DOI: 10.3233/jad-231080] [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: 02/03/2024]
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease with insidious onset. Identifying candidate predictors to forecast AD dementia risk before disease onset is crucial for early diagnosis and treatment. Objective We aimed to assess the predictive ability of blood neurofilament light (NfL) chain in anticipating cognitive decline in the AD continuum. Methods We systematically searched PubMed, Web of Science, and Embase from inception until April 7, 2023. Longitudinal observational studies examining the association between baseline blood NfL and cognitive decline or clinical disease conversion were included based on inclusion/exclusion criteria. The final effect size was represented by adjusted hazard ratios (HR) or standardized beta (s.β) coefficients with a 95% confidence interval (CI). Results A total of 2,862 articles were identified, and 26 studies were included in this meta-analysis. The results indicated that baseline blood NfL could predict cognitive decline, with MMSE [s.β= -0.17, 95% CI (-0.26, -0.07)]; PACC [s.β= -0.09, 95% CI (-0.16, -0.03)]; ADAS-cog [s.β= 0.21, 95% CI (0.13, 0.29)]; CDR-SOB [s.β= 0.27, 95% CI (0.03, 0.50)]; Global cognitive composite [s.β= -0.05, 95% CI (-0.08, -0.01)]; Memory subdomain [s.β= -0.06, 95% CI (-0.09, -0.03)]; Language subdomain [s.β= -0.07, 95% CI (-0.10, -0.05)]; Executive function subdomain [s.β= -0.02, 95% CI (-0.03, -0.01)]; Visuospatial subdomain [s.β= -0.06, 95% CI (-0.08, -0.04)]. Additionally, baseline blood NfL could predict disease progression (conversion from CU/SCD/MCI to MCI/AD) in the AD continuum [Adjust HR = 1.32, 95% CI (1.12, 1.56)]. Conclusions Baseline blood NfL demonstrated predictive capabilities for global cognition and its memory, language, executive function, visuospatial subdomains decline in the AD continuum. Moreover, it exhibited the potential to predict disease progression in non-AD dementia participants.
Collapse
Affiliation(s)
- Jianhong Li
- Fujian Key Laboratory of Aptamers Technology, 900TH hospital of Joint Logistics Support Force, People's Liberation Army (PLA), Fuzhou, Fujian, China
| | - Minguang Yang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Renli Wei
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yue Cao
- Fujian Key Laboratory of Aptamers Technology, 900TH hospital of Joint Logistics Support Force, People's Liberation Army (PLA), Fuzhou, Fujian, China
| | - Xu Fan
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Shenghang Zhang
- Fujian Key Laboratory of Aptamers Technology, 900TH hospital of Joint Logistics Support Force, People's Liberation Army (PLA), Fuzhou, Fujian, China
| |
Collapse
|
5
|
Lapp HS, Freigang M, Hagenacker T, Weiler M, Wurster CD, Günther R. Biomarkers in 5q-associated spinal muscular atrophy-a narrative review. J Neurol 2023; 270:4157-4178. [PMID: 37289324 PMCID: PMC10421827 DOI: 10.1007/s00415-023-11787-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023]
Abstract
5q-associated spinal muscular atrophy (SMA) is a rare genetic disease caused by mutations in the SMN1 gene, resulting in a loss of functional SMN protein and consecutive degeneration of motor neurons in the ventral horn. The disease is clinically characterized by proximal paralysis and secondary skeletal muscle atrophy. New disease-modifying drugs driving SMN gene expression have been developed in the past decade and have revolutionized SMA treatment. The rise of treatment options led to a concomitant need of biomarkers for therapeutic guidance and an improved disease monitoring. Intensive efforts have been undertaken to develop suitable markers, and numerous candidate biomarkers for diagnostic, prognostic, and predictive values have been identified. The most promising markers include appliance-based measures such as electrophysiological and imaging-based indices as well as molecular markers including SMN-related proteins and markers of neurodegeneration and skeletal muscle integrity. However, none of the proposed biomarkers have been validated for the clinical routine yet. In this narrative review, we discuss the most promising candidate biomarkers for SMA and expand the discussion by addressing the largely unfolded potential of muscle integrity markers, especially in the context of upcoming muscle-targeting therapies. While the discussed candidate biomarkers hold potential as either diagnostic (e.g., SMN-related biomarkers), prognostic (e.g., markers of neurodegeneration, imaging-based markers), predictive (e.g., electrophysiological markers) or response markers (e.g., muscle integrity markers), no single measure seems to be suitable to cover all biomarker categories. Hence, a combination of different biomarkers and clinical assessments appears to be the most expedient solution at the time.
Collapse
Affiliation(s)
- H S Lapp
- Department of Neurology, University Hospital Carl Gustav Carus at TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - M Freigang
- Department of Neurology, University Hospital Carl Gustav Carus at TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - T Hagenacker
- Department of Neurology and Center for Translational Neuro- and Behavioral Science (C-TNBS), University Medicine Essen, Essen, Germany
| | - M Weiler
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - C D Wurster
- Department of Neurology, University Hospital Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE) Ulm, Ulm, Germany
| | - René Günther
- Department of Neurology, University Hospital Carl Gustav Carus at TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
| |
Collapse
|
6
|
Overmeyer C, Jorgensen K, Vohra BPS. The Translocase of the Outer Mitochondrial Membrane (TOM40) is required for mitochondrial dynamics and neuronal integrity in Dorsal Root Ganglion Neurons. Mol Cell Neurosci 2023; 125:103853. [PMID: 37100265 DOI: 10.1016/j.mcn.2023.103853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/28/2023] Open
Abstract
Polymorphisms and altered expression of the Translocase of the Outer Mitochondrial Membrane - 40 kD (Tom40) are observed in neurodegenerative disease subjects. We utilized in vitro cultured dorsal root ganglion (DRG) neurons to investigate the association of TOM40 depletion to neurodegeneration, and to unravel the mechanism of neurodegeneration induced by decreased levels of TOM40 protein. We provide evidence that severity of neurodegeneration induced in the TOM40 depleted neurons increases with the increase in the depletion of TOM40 and is exacerbated by an increase in the duration of TOM40 depletion. We also demonstrate that TOM40 depletion causes a surge in neuronal calcium levels, decreases mitochondrial motility, increases mitochondrial fission, and decreases neuronal ATP levels. We observed that alterations in the neuronal calcium homeostasis and mitochondrial dynamics precede BCL-xl and NMNAT1 dependent neurodegenerative pathways in the TOM40 depleted neurons. This data also suggests that manipulation of BCL-xl and NMNAT1 may be of therapeutic value in TOM40 associated neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Kylie Jorgensen
- Department of Biology, William Jewell College Liberty, MO 64068
| | | |
Collapse
|
7
|
Brain region-specific myelinogenesis is not directly linked to amyloid-β in APP/PS1 transgenic mice. Exp Neurol 2023; 362:114344. [PMID: 36736651 DOI: 10.1016/j.expneurol.2023.114344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
Alzheimer's disease (AD) is characterized by aggregating amyloid beta-protein (Aβ). Recent evidence has shown that insufficient myelinogenesis contributes to AD-related functional deficits. However, it remains unclear whether Aβ, in either plaque or soluble form, could alter myelinogenesis in AD brains. By cell-lineage tracing and labeling, we found both myelinogenesis and Aβ deposits displayed a region-specific pattern in the 13-month-old APP/PS1 transgenic mouse brains. Aβ plaques cause focal demyelination, but only about 15% Aβ plaques are closely associated with newly formed myelin in the APP/PS1 brains. Further, the Aβ plaque total area and the amount of new myelin are not linearly correlated across different cortical regions, suggesting that Aβ plaques induce demyelination but may not exclusively trigger remyelination. To understand the role of soluble Aβ in regulating myelinogenesis, we chose to observe the visual system, wherein soluble Aβ is detectable but without the presence of Aβ plaques in the APP/PS1 retina, optic nerve, and optic tract. Interestingly, newly-formed myelin density was not significantly altered in the APP/PS1 optic nerves and optic tracts as compared to the wildtype controls, suggesting soluble Aβ probably does not change myelinogenesis. Further, treatment of purified oligodendrocyte precursor cells (OPCs) with soluble Aβ (oligomers) for 48 h did not change the cell densities of MBP positive cells and PDGFRα positive OPCs in vitro. Consistently, injection of soluble Aβ into the lateral ventricles did not alter myelinogenesis in the corpus callosum of NG2-CreErt; Tau-mGFP mice significantly. Together, these findings indicate that the region-dependent myelinogenesis in AD brains is not directly linked to Aβ, but rather probably a synergic result in adapting to AD pathology.
Collapse
|
8
|
Mesoscopic Mapping of Visual Pathway in a Female 5XFAD Mouse Model of Alzheimer's Disease. Cells 2022; 11:cells11233901. [PMID: 36497159 PMCID: PMC9740259 DOI: 10.3390/cells11233901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
Amyloid-β (Aβ) deposition and Aβ-induced neurodegeneration appear in the retina and retinorecipient areas in the early stages of Alzheimer's disease (AD). Although these Aβ-related changes in the retina cause damage to the visual functions, no studies have yet revealed the alterations in the visual pathways of AD. Therefore, we investigated the alterations of visual circuits in the AD mouse model using anterograde tracer cholera toxin β subunits (CTβ). Moreover, we investigated the Aβ accumulation in the retina and retinorecipient areas and the neuronal loss, and synaptic degeneration in retinorecipient areas by immunofluorescent staining of 4- and 12-month-old female 5XFAD transgenic mice. Our results demonstrated that Aβ accumulation and neurodegeneration occurred in the retina and retinorecipient regions of early and late stages of the 5XFAD mice. Retinal efferents to the suprachiasmatic nucleus and lateral geniculate nucleus were impaired in the early stage of AD. Moreover, retinal connections to the dorsal lateral geniculate nucleus and superior colliculus were degenerated in the late-stage of AD. These findings reveal the Aβ-related pathology induced visual circuit disturbances at the mesoscale level in both the early and late stages of AD and provide anatomical and functional insights into the visual circuitry of AD.
Collapse
|
9
|
Interaction between TRPML1 and p62 in Regulating Autophagosome-Lysosome Fusion and Impeding Neuroaxonal Dystrophy in Alzheimer’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8096009. [PMID: 35116093 PMCID: PMC8807035 DOI: 10.1155/2022/8096009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022]
Abstract
The loss of transient receptor potential mucolipin 1 (TRPML1), an endosomal and lysosomal Ca2+-releasing channel, has been implicated in neurodegenerative disorders. Mounting evidence have shown that TRPML1 could clear intraneuronal amyloid-β (Aβ), which triggers a hypothesis that TRPML1 activation may be beneficial for axonal transport in Alzheimer's disease (AD). In this work, the functional roles of TRPML1 were studied in the APP/PS1 transgenic mice and Aβ1-42-stimulated hippocampal neurons HT22. We found that lentivirus-mediated overexpression of TRPML1 was shown to promote an accumulation of autolysosomes and increase brain-derived neurotrophic factor (BDNF) transportation to the nucleus, suggesting an axon-protective function. More importantly, we found that TRPML1 also increased p62 that interacted with dynein. Lentivirus-mediated knockdown of p62 or inhibition of dynein by ciliobrevin D stimulation was found to reduce autolysosome formation and nuclear accumulation of BDNF in HT22 cells with Aβ1-42 stimulation. Inhibition of p62 by XRK3F2 stimulation was observed to promote the death of hippocampal neurons of the APP/PS1 transgenic mice. TRPML1 recruited dynein by interacting with p62 to promote the autophagosome-lysosome fusion to mediate BDNF nuclear translocation to impede axon dystrophy in mice with Alzheimer-like phenotypes. In summary, these results demonstrate the presence of a TRPML1/p62/dynein regulatory network in AD, and activation of TRPML1 is required for axon protection to prevent neuroaxonal dystrophy.
Collapse
|
10
|
Mu L, Cai J, Gu B, Yu L, Li C, Liu QS, Zhao L. Treadmill Exercise Prevents Decline in Spatial Learning and Memory in 3×Tg-AD Mice through Enhancement of Structural Synaptic Plasticity of the Hippocampus and Prefrontal Cortex. Cells 2022; 11:244. [PMID: 35053360 PMCID: PMC8774241 DOI: 10.3390/cells11020244] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/22/2021] [Accepted: 01/08/2022] [Indexed: 01/27/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by deficits in learning and memory. A pathological feature of AD is the alterations in the number and size of synapses, axon length, dendritic complexity, and dendritic spine numbers in the hippocampus and prefrontal cortex. Treadmill exercise can enhance synaptic plasticity in mouse or rat models of stroke, ischemia, and dementia. The aim of this study was to examine the effects of treadmill exercise on learning and memory, and structural synaptic plasticity in 3×Tg-AD mice, a mouse model of AD. Here, we show that 12 weeks treadmill exercise beginning in three-month-old mice improves spatial working memory in six-month-old 3×Tg-AD mice, while non-exercise six-month-old 3×Tg-AD mice exhibited impaired spatial working memory. To investigate potential mechanisms for the treadmill exercise-induced improvement of spatial learning and memory, we examined structural synaptic plasticity in the hippocampus and prefrontal cortex of six-month-old 3×Tg-AD mice that had undergone 12 weeks of treadmill exercise. We found that treadmill exercise led to increases in synapse numbers, synaptic structural parameters, the expression of synaptophysin (Syn, a presynaptic marker), the axon length, dendritic complexity, and the number of dendritic spines in 3×Tg-AD mice and restored these parameters to similar levels of non-Tg control mice without treadmill exercise. In addition, treadmill exercise also improved these parameters in non-Tg control mice. Strengthening structural synaptic plasticity may represent a potential mechanism by which treadmill exercise prevents decline in spatial learning and memory and synapse loss in 3×Tg-AD mice.
Collapse
Affiliation(s)
- Lianwei Mu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Jiajia Cai
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Boya Gu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Laikang Yu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Cui Li
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
- School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou 450001, China
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Li Zhao
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| |
Collapse
|
11
|
Baek MS, Lee MJ, Kim HK, Lyoo CH. Temporal trajectory of biofluid markers in Parkinson's disease. Sci Rep 2021; 11:14820. [PMID: 34285331 PMCID: PMC8292456 DOI: 10.1038/s41598-021-94345-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/09/2021] [Indexed: 11/18/2022] Open
Abstract
Full dynamics of biofluid biomarkers have been unknown in patients with Parkinson’s disease (PD). Using data from 396 PD patients and 182 controls in the Parkinson's Progression Markers Initiative (PPMI) database, we estimated long-term temporal trajectories of CSF α-synuclein (α-syn), amyloid-β (Aβ), total tau (t-tau), phosphorylated tau (p-tau) and serum neurofilament light chain (NfL) by integrating function between the baseline levels and annual changes. At baseline, PD patients showed lower CSF α-syn, Aβ, t-tau and p-tau levels than those of the controls. In all PD patients, CSF α-syn and Aβ decreased in a negative exponential pattern before the onset of motor symptoms, whereas CSF t-tau and p-tau, and serum NfL increased. Patients with cognitive impairment exhibited faster decline of Aβ and α-syn and faster rise of t-tau, p-tau and NfL, when compared to those without. Similarly, low Aβ group showed earlier decline of α-syn, faster rise of t-tau, p-tau and NfL, and faster decline of cognitive performances, when compared to high Aβ group. Our results suggest that longitudinal changes in biomarkers can be influenced by cognitive impairment and Aβ burden at baseline. PD patients with Aβ pathology may be associated with early appearance of α-synuclein pathology, rapid progression of axonal degeneration and neurodegeneration, and consequently greater cognitive decline.
Collapse
Affiliation(s)
- Min Seok Baek
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon do, Republic of Korea.,Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Gudeok-ro 179, Seo-gu, Busan, 49241, Republic of Korea.
| | - Han-Kyeol Kim
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| |
Collapse
|
12
|
Fross S, Mansel C, McCormick M, Vohra BPS. Tributyltin Alters Calcium Levels, Mitochondrial Dynamics, and Activates Calpains Within Dorsal Root Ganglion Neurons. Toxicol Sci 2021; 180:342-355. [PMID: 33481012 DOI: 10.1093/toxsci/kfaa193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tributyltin (TBT) remains a global health concern. The primary route of human exposure to TBT is either through ingestion or skin absorption, but TBT's effects on the peripheral nervous system have still not been investigated. Therefore, we exposed in vitro sensory dorsal root ganglion (DRG) neurons to TBT at a concentration of 50-200 nM, which is similar to the observed concentrations of TBT in human blood samples. We observed that TBT causes extensive axon degeneration and neuronal death in the DRG neurons. Furthermore, we discovered that TBT causes an increase in both cytosolic and mitochondrial calcium levels, disrupts mitochondrial dynamics, decreases neuronal ATP levels, and leads to the activation of calpains. Additional experiments demonstrated that inhibition of calpain activation prevented TBT-induced fragmentation of neuronal cytoskeletal proteins and neuronal cell death. Thus, we conclude that calpain activation is the key executioner of TBT-induced peripheral neurodegeneration.
Collapse
Affiliation(s)
- Shaneann Fross
- Department of Biology, William Jewell College, Liberty, Missouri 64068, USA
| | - Clayton Mansel
- Department of Biology, William Jewell College, Liberty, Missouri 64068, USA
| | - Madison McCormick
- Department of Biology, William Jewell College, Liberty, Missouri 64068, USA
| | | |
Collapse
|
13
|
Arruda GLM, Vigerelli H, Bufalo MC, Longato GB, Veloso RV, Zambelli VO, Picolo G, Cury Y, Morandini AC, Marques AC, Sciani JM. Box Jellyfish (Cnidaria, Cubozoa) Extract Increases Neuron's Connection: A Possible Neuroprotector Effect. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8855248. [PMID: 33748281 PMCID: PMC7954621 DOI: 10.1155/2021/8855248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/14/2021] [Accepted: 02/23/2021] [Indexed: 01/29/2023]
Abstract
Neurodegenerative diseases are one of the major causes of death worldwide, characterized by neurite atrophy, neuron apoptosis, and synapse loss. No effective treatment has been indicated for such diseases so far, and the search for new drugs is being increased in the last years. Animal venoms' secretion/venom can be an alternative for the discovery of new molecules, which could be the prototype for a new treatment. Here, we present the biochemical characterization and activity of the extract from the box jellyfish Chiropsalmus quadrumanus (Cq) on neurites. The Cq methanolic extract was obtained and incubated to human SH-SY5Y neurons, and neurite parameters were evaluated. The extract was tested in other cell types to check its cytotoxicity and was submitted to biochemical analysis by mass spectrometry in order to check its composition. We could verify that the Cq extract increased neurite outgrowth length and branching junctions, amplifying the contact between SH-SY5Y neurons, without affecting cell body and viability. The extract action was selective for neurons, as it did not cause any effects on other cell types, such as tumor line, nontumor line, and red blood cells. Moreover, mass spectrometry analysis revealed that there are no proteins but several low molecular mass compounds and peptides. Three peptides, characterized as cryptides, and 14 low molecular mass compounds were found to be related to cytoskeleton reorganization, cell membrane expansion, and antioxidant/neuroprotective activity, which act together to increase neuritogenesis. After this evaluation, we conclude that the Cq extract is a promising tool for neuronal connection recovery, an essential condition for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Gian Lucas M. Arruda
- Laboratório Multidisciplinar de Pesquisa, Universidade São Francisco, Bragança Paulista 12916-900, Brazil
| | - Hugo Vigerelli
- Laboratório de Genética, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Michelle C. Bufalo
- Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Giovanna B. Longato
- Laboratório de Pesquisa em Farmacologia Molecular e Compostos Bioativos, Universidade São Francisco, Bragança Paulista 12916-900, Brazil
| | - Rodinei V. Veloso
- Laboratório Multidisciplinar de Pesquisa, Universidade São Francisco, Bragança Paulista 12916-900, Brazil
| | - Vanessa O. Zambelli
- Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Gisele Picolo
- Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Yara Cury
- Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo 05503-900, Brazil
| | - André C. Morandini
- Departamento de Zoologia, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
- Centro de Biologia Marinha, Universidade de São Paulo, São Sebastião 11612-109, Brazil
| | - Antonio Carlos Marques
- Departamento de Zoologia, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Juliana Mozer Sciani
- Laboratório Multidisciplinar de Pesquisa, Universidade São Francisco, Bragança Paulista 12916-900, Brazil
| |
Collapse
|
14
|
Young JK. Neurogenesis Makes a Crucial Contribution to the Neuropathology of Alzheimer's Disease. J Alzheimers Dis Rep 2020; 4:365-371. [PMID: 33163897 PMCID: PMC7592839 DOI: 10.3233/adr-200218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
One unexplained feature of Alzheimer’s disease (AD) is that the lateral entorhinal cortex undergoes neurodegeneration before other brain areas. However, this brain region does not have elevated levels of amyloid peptides in comparison with undamaged regions. What is the cause of this special vulnerability of the entorhinal cortex? One special feature of the lateral entorhinal cortex is that it projects to newborn neurons that have undergone adult neurogenesis in the dentate gyrus of the hippocampus. Neurogenesis is abnormal in human AD brains, and modulation of neurogenesis in experimental animals influences the course of AD. This complex process of neurogenesis may expose axon terminals originating from neurons of the entorhinal cortex to a unique combination of molecules that can enhance toxic effects of amyloid. Retrograde degeneration of neurons with axons terminating in the dentate gyrus provides a likely explanation for the spatial patterns of neuronal cell death seen in AD. Specialized astrocytes in the dentate gyrus participate in adult neurogenesis and produce fatty acid binding protein7 (FABP7). These FABP7+ cells undergo an aging-related mitochondrial pathology that likely impairs their functions. This age-related abnormality may contribute to the impairment in neurogenesis seen in aging and Alzheimer’s disease. Also, a compromised function of these astrocytes likely results in local elevations of palmitic acid, iron, copper, and glucose, which all enhance the toxicity of amyloid peptides. Treatments that modulate neurogenesis or diminish the production of these toxic substances may prove more successful than treatments that are solely aimed at reducing the amyloid burden alone.
Collapse
Affiliation(s)
- John K Young
- Professor Emeritus, Department of Anatomy, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
15
|
Wang H, Zhang C, Yang LE, Yang Z. Hederagenin Modulates M1 Microglial Inflammatory Responses and Neurite Outgrowth. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20946252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder. Neurite atrophy and synaptic loss initiate the onset of neuronal death, while the activated M1 microglia-induced neuroinflammatory microenvironment inhibits neurite regeneration and exacerbates neuronal loss. Thus, optimizing the brain microenvironment using small compounds through suppressing activated M1 microglia and promoting neurite regrowth might be an effective therapeutic strategy for AD. We found that hederagenin (HED), a naturally occurring triterpene compound, inhibited lipopolysaccharide-induced nitric oxide generation and downregulated expression of proinflammatory cytokines, such as tumor necrosis factor-α, interleukin-1β (IL-1β), and IL-6. Further investigation of primary microglia confirmed that HED inhibited Iba-1 positive M1 microglia. However, no changes were seen in CD206 positive M2 microglia polarization. HED remarkably suppressed phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells subunit p65 signaling. In addition, HED ameliorated Aβ25-35-induced neuritic atrophy and neuronal death. Therefore, HED might be a therapeutic candidate for AD.
Collapse
Affiliation(s)
- Hua Wang
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, P. R. China
| | - Cai Zhang
- Department of Nutrition and Marine Drugs, College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Ocean University, Zhanjiang, P. R. China
| | - Long-en Yang
- Department of Nutrition and Marine Drugs, College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Ocean University, Zhanjiang, P. R. China
| | - Zhiyou Yang
- Department of Nutrition and Marine Drugs, College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Ocean University, Zhanjiang, P. R. China
- Department of Neuropharmacology, Shenzhen Institute of Guangdong Ocean University, Shenzhen, P. R. China
| |
Collapse
|
16
|
Sagnou M, Mavroidi B, Kaminari A, Boukos N, Pelecanou M. Novel Isatin Thiosemicarbazone Derivatives as Potent Inhibitors of β-Amyloid Peptide Aggregation and Toxicity. ACS Chem Neurosci 2020; 11:2266-2276. [PMID: 32598129 DOI: 10.1021/acschemneuro.0c00208] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Inhibition of β-amyloid peptide (Αβ) aggregation in Alzheimer's disease (AD) is among the therapeutic approaches against AD which still attracts scientific research interest. In the search for compounds that interact with Aβ and disrupt its typical aggregation course toward oligomeric or polymeric toxic assemblies, small organic molecules of natural origin, combining low molecular weight (necessary blood-brain barrier penetration) and low toxicity (necessary for pharmacological application), are greatly sought after. Isatin (1H-indoline-2,3-dione), a natural endogenous indole, and many of its derivatives exhibit a wide spectrum of neuropharmacological and chemotherapeutic properties. The synthesis and biological evaluation of four new isatins as inhibitors of Aβ aggregation is presented herein. In these derivatives, the N-phenyl thiosemicarbazide moiety is joined at the 3-oxo position of isatin through Schiff base formation, and substitutions are present at the indole nitrogen and position 5 of the isatin core. Biophysical studies employing circular dichroism, thioflavin T fluorescence assay, and transmission electron microscopy reveal the potential of the isatin thiosemicarbazones (ITSCs) to alter the course of Αβ aggregation, with two of the derivatives exhibiting outstanding inhibition of the aggregation process, preventing completely the formation of amyloid fibrils. Furthermore, in in vitro studies in primary neuronal cell cultures, the ITSCs were found to inhibit the Aβ-induced neurotoxicity and reactive oxygen species production at concentrations as low as 1 μM. Taken all together, the novel ITSCs can be considered as privileged structures for further development as potential AD therapeutics.
Collapse
Affiliation(s)
- Marina Sagnou
- Institute of Biosciences & Applications, National Centre for Scientific Research “Demokritos”, 15310 Athens, Greece
| | - Barbara Mavroidi
- Institute of Biosciences & Applications, National Centre for Scientific Research “Demokritos”, 15310 Athens, Greece
| | - Archontia Kaminari
- Institute of Biosciences & Applications, National Centre for Scientific Research “Demokritos”, 15310 Athens, Greece
| | - Nikos Boukos
- Institute of Nanoscience and Nanotechnology, National Centre for Scientific Research “Demokritos”, 15310 Athens, Greece
| | - Maria Pelecanou
- Institute of Biosciences & Applications, National Centre for Scientific Research “Demokritos”, 15310 Athens, Greece
| |
Collapse
|
17
|
Kuboyama T, Yang X, Tohda C. Natural Medicines and Their Underlying Mechanisms of Prevention and Recovery from Amyloid Β-Induced Axonal Degeneration in Alzheimer's Disease. Int J Mol Sci 2020; 21:E4665. [PMID: 32630004 PMCID: PMC7369795 DOI: 10.3390/ijms21134665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 01/26/2023] Open
Abstract
In Alzheimer's disease (AD), amyloid β (Aβ) induces axonal degeneration, neuronal network disruption, and memory impairment. Although many candidate drugs to reduce Aβ have been clinically investigated, they failed to recover the memory function in AD patients. Reportedly, Aβ deposition occurred before the onset of AD. Once neuronal networks were disrupted by Aβ, they could hardly be recovered. Therefore, we speculated that only removal of Aβ was not enough for AD therapy, and prevention and recovery from neuronal network disruption were also needed. This review describes the challenges related to the condition of axons for AD therapy. We established novel in vitro models of Aβ-induced axonal degeneration. Using these models, we found that several traditional medicines and their constituents prevented or helped recover from Aβ-induced axonal degeneration. These drugs also prevented or helped recover from memory impairment in in vivo models of AD. One of these drugs ameliorated memory decline in AD patients in a clinical study. These results indicate that prevention and recovery from axonal degeneration are possible strategies for AD therapy.
Collapse
Affiliation(s)
- Tomoharu Kuboyama
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan; (T.K.); (X.Y.)
- Laboratory of Pharmacognosy, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan
| | - Ximeng Yang
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan; (T.K.); (X.Y.)
| | - Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan; (T.K.); (X.Y.)
| |
Collapse
|
18
|
Platycodigenin as Potential Drug Candidate for Alzheimer's Disease via Modulating Microglial Polarization and Neurite Regeneration. Molecules 2019; 24:molecules24183207. [PMID: 31487775 PMCID: PMC6767002 DOI: 10.3390/molecules24183207] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022] Open
Abstract
Neuroinflammatory microenvironment, regulating neurite regrowth and neuronal survival, plays a critical role in Alzheimer’s disease (AD). During neuroinflammation, microglia are activated, inducing the release of inflammatory or anti-inflammatory factors depending on their polarization into classical M1 microglia or alternative M2 phenotype. Therefore, optimizing brain microenvironment by small molecule-targeted microglia polarization and promoting neurite regeneration might be a potential therapeutic strategy for AD. In this study, we found platycodigenin, a naturally occurring triterpenoid, promoted M2 polarization and inhibited M1 polarization in lipopolysaccharide (LPS)-stimulated BV2 and primary microglia. Platycodigenin downregulated pro-inflammatory molecules such as interleukin (IL)-1β, tumor necrosis factor (TNF)-α, IL-6 and nitric oxide (NO), while upregulated anti-inflammatory cytokine IL-10. Further investigation confirmed that platycodigenin inhibited cyclooxygenase-2 (Cox2) positive M1 but increased Ym1/2 positive M2 microglial polarization in primary microglia. In addition, platycodigenin significantly decreased LPS-induced the hyperphosphorylation of mitogen-activated protein kinase (MAPK) p38 and nuclear factor-κB (NF-κB) p65 subunits. Furthermore, the inactivation of peroxisome proliferators-activated receptor γ (PPARγ) induced by LPS was completely ameliorated by platycodigenin. Platycodigenin also promoted neurite regeneration and neuronal survival after Aβ treatment in primary cortical neurons. Taken together, our study for the first time clarified that platycodigenin effectively ameliorated LPS-induced inflammation and Aβ-induced neurite atrophy and neuronal death.
Collapse
|
19
|
Pottorf T, Mann A, Fross S, Mansel C, Vohra BPS. Nicotinamide Mononucleotide Adenylyltransferase 2 maintains neuronal structural integrity through the maintenance of golgi structure. Neurochem Int 2018; 121:86-97. [PMID: 30278188 DOI: 10.1016/j.neuint.2018.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/18/2018] [Accepted: 09/28/2018] [Indexed: 10/28/2022]
Abstract
Golgi fragmentation and loss of Nicotinamide Mononucleotide Adenylyltransferase 2 (NMNAT2) are the early key features of many neurodegenerative disorders. We investigated the link between NMNAT2 loss, Golgi fragmentation and axon degeneration. Golgi fragmentation in the cultured dorsal root ganglion (DRG) neurons resulted in caspase dependent axon degeneration and neuronal cell death. NMNAT2 depletion in the DRG neurons caused Golgi fragmentation and caspase dependent axon degeneration. NMNAT2 depletion did not cause ATP loss in the axons. These results indicate that NMNAT2 is required for maintenance of Golgi structure. Loss of Golgi structure or Nmnat2 depletion causes caspase dependent neurodegeneration. cytNmnat1 overexpression inhibited the axon degeneration induced by Golgi fragmentation or NMNAT2 depletion. These results also suggest that these degeneration signals converge on a common cytNmnat1 mediated axon protective program and are distinct from the SARM1 mediated caspase independent axon degeneration.
Collapse
Affiliation(s)
- Tana Pottorf
- William Jewell College, Department of Biology, Liberty, MO, USA
| | - Alexis Mann
- William Jewell College, Department of Biology, Liberty, MO, USA
| | - Shaneann Fross
- William Jewell College, Department of Biology, Liberty, MO, USA
| | - Clayton Mansel
- William Jewell College, Department of Biology, Liberty, MO, USA
| | | |
Collapse
|
20
|
Virga DM, Capps J, Vohra BPS. Enteric Neurodegeneration is Mediated Through Independent Neuritic and Somal Mechanisms in Rotenone and MPP+ Toxicity. Neurochem Res 2018; 43:2288-2303. [PMID: 30259276 DOI: 10.1007/s11064-018-2649-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/18/2018] [Accepted: 09/24/2018] [Indexed: 01/09/2023]
Abstract
Gut motility malfunction and pathological changes in the enteric nervous system (ENS) are observed in the early stages of Parkinson's disease (PD). In many cases disturbances in the autonomous functions such as gut motility precedes the observed loss of central motor functions in PD. However, the mechanism by which ENS degeneration occurs in PD is unknown. We show that parkinsonian mimetics rotenone and MPP+ induce neurite degeneration that precedes cell death in primary enteric neurons cultured in vitro. If the neuronal death signals originate from degenerating neurites, neuronal death should be prevented by inhibiting neurite degeneration. Our data demonstrate that overexpression of cytNmnat1, an axon protector, maintains healthy neurites in enteric neurons treated with either of the parkinsonian mimetics, but cannot protect the soma. We also demonstrate that neurite protection via cytNmnat1 is independent of mitochondrial dynamics or ATP levels. Overexpression of Bcl-xl, an anti-apoptotic factor, protects both the neuronal cell body and the neurites in both rotenone and MPP+ treated enteric neurons. Our data reveals that Bcl-xl and cytNmnat1 act through separate mechanisms to protect enteric neurites. Our findings suggest that neurite protection alone is not sufficient to inhibit enteric neuronal degeneration in rotenone or MPP+ toxicity, and enteric neurodegeneration in PD may be occurring through independent somatic and neuritic mechanisms. Thus, therapies targeting both axonal and somal protection can be important in finding interventions for enteric symptoms in PD.
Collapse
Affiliation(s)
- Daniel M Virga
- Biology Department, William Jewell College, Liberty, MO, 64068, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Jessica Capps
- Biology Department, William Jewell College, Liberty, MO, 64068, USA
| | | |
Collapse
|
21
|
Chen X, Jiang XM, Zhao LJ, Sun LL, Yan ML, Tian Y, Zhang S, Duan MJ, Zhao HM, Li WR, Hao YY, Wang LB, Xiong QJ, Ai J. MicroRNA-195 prevents dendritic degeneration and neuron death in rats following chronic brain hypoperfusion. Cell Death Dis 2017; 8:e2850. [PMID: 28569780 PMCID: PMC5520902 DOI: 10.1038/cddis.2017.243] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/16/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023]
Abstract
Impaired synaptic plasticity and neuron loss are hallmarks of Alzheimer’s disease and vascular dementia. Here, we found that chronic brain hypoperfusion (CBH) by bilateral common carotid artery occlusion (2VO) decreased the total length, numbers and crossings of dendrites and caused neuron death in rat hippocampi and cortices. It also led to increase in N-terminalβ-amyloid precursor protein (N-APP) and death receptor-6 (DR6) protein levels and in the activation of caspase-3 and caspase-6. Further study showed that DR6 protein was downregulated bymiR-195overexpression, upregulated bymiR-195inhibition, and unchanged by binding-site mutation and miR-masks. Knockdown of endogenousmiR-195by lentiviral vector-mediated overexpression of its antisense molecule (lenti-pre-AMO-miR-195) decreased the total length, numbers and crossings of dendrites and neuron death, upregulated N-APP and DR6 levels, and elevated cleaved caspase-3 and caspase-6 levels. Overexpression ofmiR-195using lenti-pre-miR-195prevented these changes triggered by 2VO. We conclude thatmiR-195is involved in CBH-induced dendritic degeneration and neuron death through activation of the N-APP/DR6/caspase pathway.
Collapse
Affiliation(s)
- Xin Chen
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China.,Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xue-Mei Jiang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Lin-Jing Zhao
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Lin-Lin Sun
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Mei-Ling Yan
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - You Tian
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Shuai Zhang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Ming-Jing Duan
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Hong-Mei Zhao
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Wen-Rui Li
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Yang-Yang Hao
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Li-Bo Wang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| | - Qiao-Jie Xiong
- Department of Neurobiology and Behavior, SUNY at Stony Brook, Stony Brook, NY 1794, USA
| | - Jing Ai
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
22
|
Hoy AR, Ly M, Carlsson CM, Okonkwo OC, Zetterberg H, Blennow K, Sager MA, Asthana S, Johnson SC, Alexander AL, Bendlin BB. Microstructural white matter alterations in preclinical Alzheimer's disease detected using free water elimination diffusion tensor imaging. PLoS One 2017; 12:e0173982. [PMID: 28291839 PMCID: PMC5349685 DOI: 10.1371/journal.pone.0173982] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/16/2017] [Indexed: 11/27/2022] Open
Abstract
Brain changes associated with Alzheimer's disease (AD) begin decades before disease diagnosis. While β-amyloid plaques and neurofibrillary tangles are defining features of AD, neuronal loss and synaptic pathology are closely related to the cognitive dysfunction. Brain imaging methods that are tuned to assess degeneration of myelinated nerve fibers in the brain (collectively called white matter) include diffusion tensor imaging (DTI) and related techniques, and are expected to shed light on disease-related loss of structural connectivity. Participants (N = 70, ages 47-76 years) from the Wisconsin Registry for Alzheimer's Prevention study underwent DTI and hybrid diffusion imaging to determine a free-water elimination (FWE-DTI) model. The study assessed the extent to which preclinical AD pathology affects brain white matter. Preclinical AD pathology was determined using cerebrospinal fluid (CSF) biomarkers. The sample was enriched for AD risk (APOE ε4 and parental history of AD). AD pathology assessed by CSF analyses was significantly associated with altered microstructure on both DTI and FWE-DTI. Affected regions included frontal, parietal, and especially temporal white matter. The f-value derived from the FWE-DTI model appeared to be the most sensitive to the relationship between the CSF AD biomarkers and microstructural alterations in white matter. These findings suggest that white matter degeneration is an early pathological feature of AD that may have utility both for early disease detection and as outcome measures for clinical trials. More complex models of microstructural diffusion properties including FWE-DTI may provide increased sensitivity to early brain changes associated with AD over standard DTI.
Collapse
Affiliation(s)
- Andrew R. Hoy
- Lieutenant, Medical Service Corp, United States Navy, Falls Church, Virginia, United States of America
- Department of Medical Physics, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Radiology and Radiologic Sciences, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Martina Ly
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veteran's Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
- Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Cynthia M. Carlsson
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veteran's Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Ozioma C. Okonkwo
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veteran's Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Mark A. Sager
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Sanjay Asthana
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veteran's Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Sterling C. Johnson
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veteran's Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Andrew L. Alexander
- Department of Medical Physics, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Psychiatry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Barbara B. Bendlin
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veteran's Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| |
Collapse
|
23
|
Li LS, Lu YL, Nie J, Xu YY, Zhang W, Yang WJ, Gong QH, Lu YF, Lu Y, Shi JS. Dendrobium nobile Lindl alkaloid, a novel autophagy inducer, protects against axonal degeneration induced by Aβ 25-35 in hippocampus neurons in vitro. CNS Neurosci Ther 2017; 23:329-340. [PMID: 28261990 DOI: 10.1111/cns.12678] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/12/2017] [Accepted: 01/16/2017] [Indexed: 12/19/2022] Open
Abstract
AIMS Axonal degeneration is a pathological symbol in the early stage of Alzheimer's disease (AD), which can be triggered by amyloid-β (Aβ) peptide deposition. Growing evidence indicates that deficit of autophagy eventually leads to the axonal degeneration. Our previous studies have shown that Dendrobium nobile Lindl alkaloid (DNLA) had protective effect on neuron impairment in vivo and in vitro; however, the underlying mechanisms is still unclear. METHODS We exposed cultured hippocampus neurons to Aβ25-35 to investigate the effect of DNLA in vitro. Axonal degeneration was evaluated by immunofluorescence staining and MTT assay. Neurons overexpressing GFP-LC3B were used to measure the formation of autophagosome. Autophagosome-lysosome fusion, the lysosomal pH, and cathepsin activity were assessed to reflect autophagy process. Proteins of interest were analyzed by Western blot. RESULTS DNLA pretreatment significantly inhibited axonal degeneration induced by Aβ25-35 peptide in vitro. Further studies revealed DNLA treatment increased autophagic flux through promoting formation and degradation of autophagosome in hippocampus neurons. Moreover, enhancement of autophagic flux was responsible for the protective effects of DNLA on axonal degeneration. CONCLUSIONS DNLA prevents Aβ25-35 -induced axonal degeneration via activation of autophagy process and could be a novel therapeutic target.
Collapse
Affiliation(s)
- Li-Sheng Li
- Department of Chemistry, Basic Medical Faculty, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yan-Liu Lu
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jing Nie
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yun-Yan Xu
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei Zhang
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wen-Jin Yang
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Hai Gong
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuan-Fu Lu
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yang Lu
- Department of Chemistry, Basic Medical Faculty, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Shan Shi
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
24
|
Berbusse GW, Woods LC, Vohra BPS, Naylor K. Mitochondrial Dynamics Decrease Prior to Axon Degeneration Induced by Vincristine and are Partially Rescued by Overexpressed cytNmnat1. Front Cell Neurosci 2016; 10:179. [PMID: 27486387 PMCID: PMC4949221 DOI: 10.3389/fncel.2016.00179] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/04/2016] [Indexed: 11/13/2022] Open
Abstract
Axon degeneration is a prominent feature of various neurodegenerative diseases, such as Parkinson's and Alzheimer's, and is often characterized by aberrant mitochondrial dynamics. Mitochondrial fission, fusion, and motility have been shown to be particularly important in progressive neurodegeneration. Thus we investigated these imperative dynamics, as well as mitochondrial fragmentation in vincristine induced axon degradation in cultured dorsal root ganglia (DRG) neurons. CytNmnat1 inhibits axon degeneration in various paradigms including vincristine toxicity. The mechanism of its protection is not yet fully understood; therefore, we also investigated the effect of cytNmnat1 on mitochondrial dynamics in vincristine treated neurons. We observed that vincristine treatment decreases the rate of mitochondrial fission, fusion and motility and induces mitochondrial fragmentation. These mitochondrial events precede visible axon degeneration. Overexpression of cytNmnat1 inhibits axon degeneration and preserves the normal mitochondrial dynamics and motility in vincristine treated neurons. We suggest the alterations in mitochondrial structure and dynamics are early events which lead to axon degeneration and cytNmnat1 blocks axon degeneration by halting the vincristine induced changes to mitochondrial structure and dynamics.
Collapse
Affiliation(s)
- Gregory W Berbusse
- Department of Cellular Physiology and Molecular Biophysics, University of Arkansas for Medical Sciences Little Rock, AR, USA
| | - Laken C Woods
- Department of Biology, University of Central Arkansas Conway, AR, USA
| | - Bhupinder P S Vohra
- Department of Neurology, Yale University School of Medicine New Haven, CT, USA
| | - Kari Naylor
- Department of Biology, University of Central Arkansas Conway, AR, USA
| |
Collapse
|
25
|
Tomasini MC, Borelli AC, Beggiato S, Ferraro L, Cassano T, Tanganelli S, Antonelli T. Differential Effects of Palmitoylethanolamide against Amyloid-β Induced Toxicity in Cortical Neuronal and Astrocytic Primary Cultures from Wild-Type and 3xTg-AD Mice. J Alzheimers Dis 2016; 46:407-21. [PMID: 25765918 DOI: 10.3233/jad-143039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Considering the heterogeneity of pathological changes occurring in Alzheimer's disease (AD), a therapeutic approach aimed both to neuroprotection and to neuroinflammation reduction may prove effective. Palmitoylethanolamide (PEA) has attracted attention for its anti-inflammatory/neuroprotective properties observed in AD animal models. OBJECTIVE AND METHODS We evaluated the protective role of PEA against amyloid-β₄₂ (Aβ₄₂) toxicity on cell viability and glutamatergic transmission in primary cultures of cerebral cortex neurons and astrocytes from the triple-transgenic murine model of AD (3xTg-AD) and their wild-type littermates (non-Tg) mice. RESULTS Aβ₄₂ (0.5 μM; 24 h) affects the cell viability in cultured cortical neurons and astrocytes from non-Tg mice, but not in those from 3xTg-AD mice. These effects were counteracted by the pretreatment with PEA (0.1 μM). Basal glutamate levels in cultured neurons and astrocytes from 3xTg-AD mice were lower than those observed in cultured cells from non-Tg mice. Aβ₄₂-exposure reduced and increased glutamate levels in non-Tg mouse cortical neurons and astrocytes, respectively. These effects were counteracted by the pretreatment with PEA. By itself, PEA did not affect cell viability and glutamate levels in cultured cortical neurons and astrocytes from non-Tg or 3xTg-AD mice. CONCLUSION The exposure to Aβ₄₂ induced toxic effects on cultured cortical neurons and astrocytes from non-Tg mice, but not in those from 3xTg-AD mice. Furthermore, PEA exerts differential effects against Aβ₄₂-induced toxicity in primary cultures of cortical neurons and astrocytes from non-Tg and 3xTg-AD mice. In particular, PEA displays protective properties in non-Tg but not in 3xTg-AD mouse neuronal cultured cells overexpressing Aβ.
Collapse
Affiliation(s)
- Maria Cristina Tomasini
- Department of Life Sciences and Biotechnology, University of Ferrara, Italy.,IRET Foundation, Ozzano Emilia, Bologna, Italy
| | | | - Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara, Italy.,IRET Foundation, Ozzano Emilia, Bologna, Italy
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Italy.,IRET Foundation, Ozzano Emilia, Bologna, Italy.,LTTA Centre, University of Ferrara, Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, Italy
| | - Sergio Tanganelli
- IRET Foundation, Ozzano Emilia, Bologna, Italy.,Department of Medical Sciences, University of Ferrara, Italy.,LTTA Centre, University of Ferrara, Italy
| | - Tiziana Antonelli
- IRET Foundation, Ozzano Emilia, Bologna, Italy.,Department of Medical Sciences, University of Ferrara, Italy.,LTTA Centre, University of Ferrara, Italy
| |
Collapse
|
26
|
Yang ZY, Kuboyama T, Kazuma K, Konno K, Tohda C. Active Constituents from Drynaria fortunei Rhizomes on the Attenuation of Aβ(25-35)-Induced Axonal Atrophy. JOURNAL OF NATURAL PRODUCTS 2015; 78:2297-300. [PMID: 26299900 DOI: 10.1021/acs.jnatprod.5b00290] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Axonal regeneration might contribute to the restoration of damaged neuronal networks and improvement of memory deficits in a murine Alzheimer's disease (AD) model. A search for axonal regenerative drugs was performed to discover novel therapeutic options for AD. In this study, an aqueous extract of Drynaria fortunei rhizomes reversed Aβ25-35-induced axonal atrophy in cultured cortical neurons of mice. Bioassay-guided fractionation of this extract led to the isolation and identification of compounds 1-5. Among them, (2S)-neoeriocitrin (2) and caffeic acid 4-O-glucoside (4) showed significant axonal elongation effects on Aβ25-35-induced atrophy.
Collapse
Affiliation(s)
- Zhi-You Yang
- Division of Neuromedical Science, Department of Bioscience, and ‡Division of Kampo Pharmaceutics, Department of Medicinal Resources, Institute of Natural Medicine, University of Toyama , 2630 Sugitani, Toyama 930-0194, Japan
| | - Tomoharu Kuboyama
- Division of Neuromedical Science, Department of Bioscience, and ‡Division of Kampo Pharmaceutics, Department of Medicinal Resources, Institute of Natural Medicine, University of Toyama , 2630 Sugitani, Toyama 930-0194, Japan
| | - Kohei Kazuma
- Division of Neuromedical Science, Department of Bioscience, and ‡Division of Kampo Pharmaceutics, Department of Medicinal Resources, Institute of Natural Medicine, University of Toyama , 2630 Sugitani, Toyama 930-0194, Japan
| | - Katsuhiro Konno
- Division of Neuromedical Science, Department of Bioscience, and ‡Division of Kampo Pharmaceutics, Department of Medicinal Resources, Institute of Natural Medicine, University of Toyama , 2630 Sugitani, Toyama 930-0194, Japan
| | - Chihiro Tohda
- Division of Neuromedical Science, Department of Bioscience, and ‡Division of Kampo Pharmaceutics, Department of Medicinal Resources, Institute of Natural Medicine, University of Toyama , 2630 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
27
|
Estacion M, Vohra BPS, Liu S, Hoeijmakers J, Faber CG, Merkies ISJ, Lauria G, Black JA, Waxman SG. Ca2+ toxicity due to reverse Na+/Ca2+ exchange contributes to degeneration of neurites of DRG neurons induced by a neuropathy-associated Nav1.7 mutation. J Neurophysiol 2015; 114:1554-64. [PMID: 26156380 DOI: 10.1152/jn.00195.2015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 07/06/2015] [Indexed: 12/19/2022] Open
Abstract
Gain-of-function missense mutations in voltage-gated sodium channel Nav1.7 have been linked to small-fiber neuropathy, which is characterized by burning pain, dysautonomia and a loss of intraepidermal nerve fibers. However, the mechanistic cascades linking Nav1.7 mutations to axonal degeneration are incompletely understood. The G856D mutation in Nav1.7 produces robust changes in channel biophysical properties, including hyperpolarized activation, depolarized inactivation, and enhanced ramp and persistent currents, which contribute to the hyperexcitability exhibited by neurons containing Nav1.8. We report here that cell bodies and neurites of dorsal root ganglion (DRG) neurons transfected with G856D display increased levels of intracellular Na(+) concentration ([Na(+)]) and intracellular [Ca(2+)] following stimulation with high [K(+)] compared with wild-type (WT) Nav1.7-expressing neurons. Blockade of reverse mode of the sodium/calcium exchanger (NCX) or of sodium channels attenuates [Ca(2+)] transients evoked by high [K(+)] in G856D-expressing DRG cell bodies and neurites. We also show that treatment of WT or G856D-expressing neurites with high [K(+)] or 2-deoxyglucose (2-DG) does not elicit degeneration of these neurites, but that high [K(+)] and 2-DG in combination evokes degeneration of G856D neurites but not WT neurites. Our results also demonstrate that 0 Ca(2+) or blockade of reverse mode of NCX protects G856D-expressing neurites from degeneration when exposed to high [K(+)] and 2-DG. These results point to [Na(+)] overload in DRG neurons expressing mutant G856D Nav1.7, which triggers reverse mode of NCX and contributes to Ca(2+) toxicity, and suggest subtype-specific blockade of Nav1.7 or inhibition of reverse NCX as strategies that might slow or prevent axon degeneration in small-fiber neuropathy.
Collapse
Affiliation(s)
- M Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - B P S Vohra
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - S Liu
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - J Hoeijmakers
- Department of Neurology, University Medical Center Maastricht, Maastricht, the Netherlands
| | - C G Faber
- Department of Neurology, University Medical Center Maastricht, Maastricht, the Netherlands
| | - I S J Merkies
- Department of Neurology, University Medical Center Maastricht, Maastricht, the Netherlands; Department of Neurology, Spaarne Hospital, Hoofddorp, the Netherlands; and
| | - G Lauria
- Neuroalgology Unit IRCCS Foundation "Carlo Besta" Neurological Institute, Milan, Italy
| | - J A Black
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - S G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut;
| |
Collapse
|
28
|
Ashok A, Rai NK, Tripathi S, Bandyopadhyay S. Exposure to As-, Cd-, and Pb-mixture induces Aβ, amyloidogenic APP processing and cognitive impairments via oxidative stress-dependent neuroinflammation in young rats. Toxicol Sci 2014; 143:64-80. [PMID: 25288670 DOI: 10.1093/toxsci/kfu208] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Environmental pollutants act as risk factors for Alzheimer's disease (AD), mainly affecting the aging population. We investigated early manifestations of AD-like pathology by a mixture of arsenic (As), cadmium (Cd), and lead (Pb), reported to impair neurodevelopment. We treated rats with As+Cd+Pb at their concentrations detected in groundwater of India, ie, 0.38, 0.098, and 0.22 ppm or 10 times of each, respectively, from gestation-05 to postnatal day-180. We identified dose-dependent increase in amyloid-beta (Aβ) in frontal cortex and hippocampus as early as post-weaning. The effect was strongly significant during early-adulthood, reaching levels comparable to an Aβ-infused AD-like rat model. The metals activated the proamyloidogenic pathway, mediated by increase in amyloid precursor protein (APP), and subsequent beta secretase (BACE) and presenilin (PS)-mediated APP-processing. Investigating the mechanism of Aβ-induction revealed an augmentation in oxidative stress-dependent neuroinflammation that stimulated APP expression through interleukin-responsive-APP-mRNA 5'-untranslated region. We then examined the effects of individual metals and binary mixtures in comparison with the tertiary. Among individual metals, Pb triggered maximum induction of Aβ, whereas individual As or Cd had a relatively non-significant effect on Aβ despite enhanced APP, owing to reduced induction of BACE and PS. Interestingly, when combined the metals demonstrated synergism, with a major contribution by As. The synergistic effect was significant and consistent in tertiary mixture, resulting in the augmentation of Aβ. Eventually, increase in Aβ culminated in cognitive impairments in the young rats. Together, our data demonstrate that exposure to As+Cd+Pb induces premature manifestation of AD-like pathology that is synergistic, and oxidative stress and inflammation dependent.
Collapse
Affiliation(s)
- Anushruti Ashok
- *Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow and Developmental Toxicology Division, CSIR-IITR Campus, Lucknow 226001, India *Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow and Developmental Toxicology Division, CSIR-IITR Campus, Lucknow 226001, India
| | - Nagendra Kumar Rai
- *Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow and Developmental Toxicology Division, CSIR-IITR Campus, Lucknow 226001, India *Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow and Developmental Toxicology Division, CSIR-IITR Campus, Lucknow 226001, India
| | - Sachin Tripathi
- *Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow and Developmental Toxicology Division, CSIR-IITR Campus, Lucknow 226001, India
| | - Sanghamitra Bandyopadhyay
- *Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow and Developmental Toxicology Division, CSIR-IITR Campus, Lucknow 226001, India *Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow and Developmental Toxicology Division, CSIR-IITR Campus, Lucknow 226001, India
| |
Collapse
|