1
|
Chang Y, Liu J, Xu X, Sun S, Zhang J, Zhang X, Lu G, Xiao S, Cao Y, Wu R, Wu J, Liu R, Wang R. Subcortical tau deposition and plasma glial fibrillary acidic protein as predictors of cognitive decline in mild cognitive impairment and Alzheimer's disease. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-07016-x. [PMID: 39690275 DOI: 10.1007/s00259-024-07016-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024]
Abstract
PURPOSE This study aimed to investigate the correlation between subcortical tau-positron emission tomography (Tau-PET) and plasma glial fibrillary acidic protein (GFAP) levels and cognitive function in participants with cognitively unimpaired (CU), mild cognitive impairment (MCI) and Alzheimer's disease (AD) conditions. METHODS 105 participants with amyloid (Aβ) PET and Tau-PET scans were enrolled. Region of interest (ROI) level and voxel-wise comparisons were performed between those three groups. Correlations between standardized uptake value ratio (SUVR) and cognitive performance were analyzed. The diagnostic performance of Tau-PET, Aβ-PET, and plasma GFAP, both individually and combined, was evaluated by calculating the area under the curve (AUC) from receiver operating characteristic (ROC) analyses. RESULTS Plasma GFAP levels in the AD and MCI groups were higher than those in the CU group. The AD and MCI groups showed higher Tau-PET load at the amygdala, accumbens, putamen, pallidum, hippocampus, para-hippocampus and olfactory tubercle than the CU group (p < 0.05). In the MCI group, the mean tau SUVR in the combined subcortical ROI negatively correlated with cognitive scores (r = -0.38, p = 0.02). The combination of Tau-PET, Aβ-PET and plasma GFAP provided optimal diagnostic accuracy for classifying AD from MCI, with an AUC of 0.82, a sensitivity of 0.69 and a specificity of 0.81. CONCLUSIONS Subcortical tau deposition and increased plasma GFAP levels are associated with cognitive impairment in MCI patients.
Collapse
Affiliation(s)
- Yan Chang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurology, International Headache Center, The First Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Jiajin Liu
- Department of Nuclear Medicine, The First Medical Centre, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xiaodan Xu
- Department of Nuclear Medicine, The First Medical Centre, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shuwei Sun
- Department of Nuclear Medicine, The First Medical Centre, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Jinming Zhang
- Department of Nuclear Medicine, The First Medical Centre, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, The First Medical Centre, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Guangshuang Lu
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurology, International Headache Center, The First Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- Department of Pediatrics, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, 237000, China
| | - Shaobo Xiao
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurology, International Headache Center, The First Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yuanyan Cao
- Central Research Institute, Beijing United Imaging Research Institute of Intelligent Imaging, Beijing, 100089, China
| | - Runze Wu
- Central Research Institute, Beijing United Imaging Research Institute of Intelligent Imaging, Beijing, 100089, China
| | - Jun Wu
- R&D Center, Beijing Kaixianghongkang Company, Beijing, 100029, China
| | - Ruozhuo Liu
- Department of Neurology, International Headache Center, The First Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Ruimin Wang
- Department of Nuclear Medicine, The First Medical Centre, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
2
|
Kolling LJ, Chimenti MS, Marcinkiewcz CA. Spatial differences in gene expression across the dorsal raphe nucleus in a model of early Alzheimer's disease. J Alzheimers Dis 2024:13872877241299119. [PMID: 39584353 DOI: 10.1177/13872877241299119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
BACKGROUND Persons with Alzheimer's disease (AD) present with changes in mood, sleep, and arousal that may precede the clinical manifestation of cognitive decline. These early symptoms can be driven by changes in the serotonergic (5-HT) nuclei of the brainstem, particularly the dorsal raphe nucleus (DRN). It is unclear why all 5-HT neurons do not simultaneously develop AD pathology that progresses at the same rate. OBJECTIVE We sought to identify any underlying genetic components associated with susceptibility or resistance of 5-HT neurons to AD pathology. METHODS The Visium Spatial Gene Expression platform was used to identify transcriptomic changes across the DRN in a preclinical model of early AD, human tau-overexpressing mice (htau mice). We further used RNAscope and immunohistochemical assessment to validate findings of primary interest. RESULTS We find that the DRN of htau mice differentially expresses AD-related genes, including those related to kinase binding, ion channel activity, ligand-receptor interactions, and regulation of serine/threonine kinases. We further find that computational sub-clustering of the DRN is consistent with previous circuitry-driven characterizations, allowing for spatial bounding of distinct subregions within the DRN. Of these, we find the dorsolateral DRN is preferentially impacted by 5-HT neuron loss and development of tau pathology, which coincides with increased expression of the long noncoding RNA Map2k3os. CONCLUSIONS Map2k3os may serve regulatory roles relevant for tau phosphorylation and warrants further investigation to characterize its interactions. Overall, this report demonstrates the power of large-scale spatial transcriptomics technologies, while underscoring the need for convergent-data validation to overcome their limitations.
Collapse
Affiliation(s)
- Louis J Kolling
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Michael S Chimenti
- Iowa Institute of Human Genetics Bioinformatics Division, University of Iowa, IA City, IA, USA
| | | |
Collapse
|
3
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [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: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
4
|
Olchanyi MD, Augustinack J, Haynes RL, Lewis LD, Cicero N, Li J, Destrieux C, Folkerth RD, Kinney HC, Fischl B, Brown EN, Iglesias JE, Edlow BL. Histology-guided MRI segmentation of brainstem nuclei critical to consciousness. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.26.24314117. [PMID: 39399006 PMCID: PMC11469455 DOI: 10.1101/2024.09.26.24314117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
While substantial progress has been made in mapping the connectivity of cortical networks responsible for conscious awareness, neuroimaging analysis of subcortical arousal networks that modulate arousal (i.e., wakefulness) has been limited by a lack of a robust segmentation procedures for brainstem arousal nuclei. Automated segmentation of brainstem arousal nuclei is an essential step toward elucidating the physiology of arousal in human consciousness and the pathophysiology of disorders of consciousness. We created a probabilistic atlas of brainstem arousal nuclei built on diffusion MRI scans of five ex vivo human brain specimens scanned at 750 μm isotropic resolution. Labels of arousal nuclei used to generate the probabilistic atlas were manually annotated with reference to nucleus-specific immunostaining in two of the five brain specimens. We then developed a Bayesian segmentation algorithm that utilizes the probabilistic atlas as a generative model and automatically identifies brainstem arousal nuclei in a resolution- and contrast-agnostic manner. The segmentation method displayed high accuracy in both healthy and lesioned in vivo T1 MRI scans and high test-retest reliability across both T1 and T2 MRI contrasts. Finally, we show that the segmentation algorithm can detect volumetric changes and differences in magnetic susceptibility within brainstem arousal nuclei in Alzheimer's disease and traumatic coma, respectively. We release the probabilistic atlas and Bayesian segmentation tool in FreeSurfer to advance the study of human consciousness and its disorders.
Collapse
|
5
|
Xu Q, Chen Y, Miller S, Bajaj K, Santana J, Badawy M, Lyu H, Liu Y, He N, Yan F, Haacke EM. In Vivo visualization of white matter fiber tracts in the brainstem using low flip angle double echo 3D gradient echo imaging at 3T. Neuroimage 2024; 300:120857. [PMID: 39299660 DOI: 10.1016/j.neuroimage.2024.120857] [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: 07/15/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND White matter (WM) fiber tracts in the brainstem communicate with various regions in the cerebrum, cerebellum, and spinal cord. Clinically, small lesions, malformations, or histopathological changes in the brainstem can cause severe neurological disorders. A direct and non-invasive assessment approach could bring valuable information about the intricate anatomical variations of the white matter fiber tracts and nuclei. Although tractography from diffusion tensor imaging has been commonly used to map the WM fiber tracts connectivity, it is difficult to differentiate the complex WM tracts anatomically. Both high field MRI methods and ultrahigh-field MRI methods at 7T and 11.7 T have been used to enhance the contrast of WM fiber tracts. Despite their promising results, it is still challenging to achieve wide clinical adoption at 3T. In this study, we explored a clinically feasible method using a proton density weighted (PDW) 3D gradient echo (GRE) sequence to directly image the WM fiber tracts in the brainstem at 3T in vivo. METHODS We optimized a 3D high resolution, double echo, short TR, PDW GRE sequence on 5 healthy volunteers using a clinical 3T scanner to visualize the complicated anatomy of WM fiber tracts in the brain stem. Tissue properties including T1, proton density and T2* from in vivo quantitative MRI data were used for simulations to determine the optimal flip angle for the sequence. The visualization of multiple WM fiber tracts in the brainstem was assessed qualitatively and quantitatively using relative contrast and contrast-to-noise ratio (CNR). To improve the CNR, the final images were created by averaging over all echoes from two consecutive scans at the optimal flip angle. The results were compared to anatomical atlases and histology sections to identify the major fiber tracts. All the identified major fiber tracts were labeled on axial, sagittal and coronal slices. RESULTS The WM fiber tracts were found to have distinct hypointense signal throughout the brainstem and most of the major WM fiber tracts, such as the corticospinal tract, medial lemniscus, medial longitudinal fasciculus, and central tegmental tract, in the brainstem up to and including the thalamus were identified in all subjects. Both qualitative and quantitative evaluations showed that the 3° scan offered the best contrast for WM fiber tracts for a TR of 20 ms. The average over the first two echo times and two consecutive 3° scans gave a CNR of 47.8 ± 6.2 for the pyramidal tracts in particular and CNRs values greater than 6.5 ± 2.4 for the rest of the fiber tracts. CONCLUSIONS All the major fiber tracts in the brainstem could be visualized. Given the reasonably short scan time of 10 min at 3T, double echo PDW GRE sequence is a very practical approach for clinical adoption.
Collapse
Affiliation(s)
- Qiuyun Xu
- Department of Radiology, Wayne State University, 3990 John R, 4201 St Antoine, Detroit Receiving Hospital 3L-8, Detroit, MI, 48201, USA
| | - Yongsheng Chen
- Department of Neurology, Detroit Medical Center, Wayne State University, University Health Center-8th floor, 4201 St Antoine, Detroit, MI, 48201, USA
| | - Stephan Miller
- Department of Radiology, Detroit Medical Center, Wayne State University School of Medicine, 3901 Beaubien Boulevard, Detroit, MI, 48201, USA
| | - Kunal Bajaj
- Department of Radiology, Detroit Medical Center, Wayne State University School of Medicine, 3901 Beaubien Boulevard, Detroit, MI, 48201, USA
| | - Jairo Santana
- Department of Radiology, Detroit Medical Center, Wayne State University School of Medicine, 3901 Beaubien Boulevard, Detroit, MI, 48201, USA
| | - Mohamed Badawy
- Department of Radiology, Detroit Medical Center, Wayne State University School of Medicine, 3901 Beaubien Boulevard, Detroit, MI, 48201, USA
| | - Haiying Lyu
- Department of Radiology, Ruijin Hospital, No. 197 Ruijin Er Road, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Liu
- Department of Radiology, Ruijin Hospital, No. 197 Ruijin Er Road, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Naying He
- Department of Radiology, Ruijin Hospital, No. 197 Ruijin Er Road, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, No. 197 Ruijin Er Road, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - E Mark Haacke
- Department of Radiology, Wayne State University, 3990 John R, 4201 St Antoine, Detroit Receiving Hospital 3L-8, Detroit, MI, 48201, USA; Department of Neurology, Detroit Medical Center, Wayne State University, University Health Center-8th floor, 4201 St Antoine, Detroit, MI, 48201, USA; Department of Radiology, Ruijin Hospital, No. 197 Ruijin Er Road, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Biomedical Engineering, Wayne State University, 3990 John R, 4201 St Antoine, Detroit Receiving Hospital 3L-8, Detroit, MI, 48201, USA.
| |
Collapse
|
6
|
Pierson SR, Fiock KL, Wang R, Balasubramanian N, Reinhardt J, Khan KM, James TD, Hunter ML, Cooper BJ, Williamsen HR, Betters R, Deniz K, Lee G, Aldridge G, Hefti MM, Marcinkiewcz CA. Tau pathology in the dorsal raphe may be a prodromal indicator of Alzheimer's disease. Mol Psychiatry 2024:10.1038/s41380-024-02664-9. [PMID: 39143322 DOI: 10.1038/s41380-024-02664-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 06/22/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024]
Abstract
Protein aggregation in brainstem nuclei is thought to occur in the early stages of Alzheimer's disease (AD), but its specific role in driving prodromal symptoms and disease progression is largely unknown. The dorsal raphe nucleus (DRN) contains a large population of serotonin (5-hydroxytryptamine; 5-HT) neurons that regulate mood, reward-related behavior, and sleep, which are all disrupted in AD. We report here that tau pathology is present in the DRN of individuals 25-80 years old without a known history of dementia, and its prevalence was comparable to the locus coeruleus (LC). By comparison, fewer cases were positive for other pathological proteins including α-synuclein, β-amyloid, and TDP-43. To evaluate how early tau pathology impacts behavior, we overexpressed human P301L-tau in the DRN of mice and observed depressive-like behaviors and hyperactivity without deficits in spatial memory. Tau pathology was predominantly found in neurons relative to glia and colocalized with a significant proportion of Tph2-expressing neurons in the DRN. 5-HT neurons were also hyperexcitable in P301L-tauDRN mice, and there was an increase in the amplitude of excitatory post-synaptic currents (EPSCs). Moreover, astrocytic density was elevated in the DRN and accompanied by an increase in IL-1α and Frk expression, which suggests increased inflammatory signaling. Additionally, tau pathology was detected in axonal processes in the thalamus, hypothalamus, amygdala, and caudate putamen. A significant proportion of this tau pathology colocalized with the serotonin reuptake transporter (SERT), suggesting that tau may spread in an anterograde manner to regions outside the DRN. Together these results indicate that tau pathology accumulates in the DRN in a subset of individuals over 50 years and may lead to behavioral dysregulation, 5-HT neuronal dysfunction, and activation of local astrocytes which may be prodromal indicators of AD.
Collapse
Affiliation(s)
- Samantha R Pierson
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kimberly L Fiock
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Ruixiang Wang
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Jessica Reinhardt
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kanza M Khan
- Psychological Sciences Department, Daemen University, Amherst, NY, 14226, USA
| | - Thomas D James
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Mikayla L Hunter
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Benjamin J Cooper
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Ryan Betters
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kaancan Deniz
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
| | - Gloria Lee
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Georgina Aldridge
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Marco M Hefti
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Catherine A Marcinkiewcz
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
7
|
Agostinho D, Simões M, Castelo-Branco M. Predicting conversion from mild cognitive impairment to Alzheimer's disease: a multimodal approach. Brain Commun 2024; 6:fcae208. [PMID: 38961871 PMCID: PMC11220508 DOI: 10.1093/braincomms/fcae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/09/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024] Open
Abstract
Successively predicting whether mild cognitive impairment patients will progress to Alzheimer's disease is of significant clinical relevance. This ability may provide information that can be leveraged by emerging intervention approaches and thus mitigate some of the negative effects of the disease. Neuroimaging biomarkers have gained some attention in recent years and may be useful in predicting the conversion of mild cognitive impairment to Alzheimer's disease. We implemented a novel multi-modal approach that allowed us to evaluate the potential of different imaging modalities, both alone and in different degrees of combinations, in predicting the conversion to Alzheimer's disease of mild cognitive impairment patients. We applied this approach to the imaging data from the Alzheimer's Disease Neuroimaging Initiative that is a multi-modal imaging dataset comprised of MRI, Fluorodeoxyglucose PET, Florbetapir PET and diffusion tensor imaging. We included a total of 480 mild cognitive impairment patients that were split into two groups: converted and stable. Imaging data were segmented into atlas-based regions of interest, from which relevant features were extracted for the different imaging modalities and used to construct machine-learning models to classify mild cognitive impairment patients into converted or stable, using each of the different imaging modalities independently. The models were then combined, using a simple weight fusion ensemble strategy, to evaluate the complementarity of different imaging modalities and their contribution to the prediction accuracy of the models. The single-modality findings revealed that the model, utilizing features extracted from Florbetapir PET, demonstrated the highest performance with a balanced accuracy of 83.51%. Concerning multi-modality models, not all combinations enhanced mild cognitive impairment conversion prediction. Notably, the combination of MRI with Fluorodeoxyglucose PET emerged as the most promising, exhibiting an overall improvement in predictive capabilities, achieving a balanced accuracy of 78.43%. This indicates synergy and complementarity between the two imaging modalities in predicting mild cognitive impairment conversion. These findings suggest that β-amyloid accumulation provides robust predictive capabilities, while the combination of multiple imaging modalities has the potential to surpass certain single-modality approaches. Exploring modality-specific biomarkers, we identified the brainstem as a sensitive biomarker for both MRI and Fluorodeoxyglucose PET modalities, implicating its involvement in early Alzheimer's pathology. Notably, the corpus callosum and adjacent cortical regions emerged as potential biomarkers, warranting further study into their role in the early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Daniel Agostinho
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), ICNAS, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Science and Technology, Centre for Informatics and Systems of the University of Coimbra (CISUC), 3030-790 Coimbra, Portugal
- Intelligent Systems Associate Laboratory (LASI), 4800-058 Guimarães, Portugal
| | - Marco Simões
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), ICNAS, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Science and Technology, Centre for Informatics and Systems of the University of Coimbra (CISUC), 3030-790 Coimbra, Portugal
- Intelligent Systems Associate Laboratory (LASI), 4800-058 Guimarães, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), ICNAS, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Intelligent Systems Associate Laboratory (LASI), 4800-058 Guimarães, Portugal
| |
Collapse
|
8
|
Ono M, Ito T, Yamaki S, Hori Y, Zhou Q, Zhao X, Muramoto S, Yamamoto R, Furuyama T, Sakata-Haga H, Hatta T, Hamaguchi T, Kato N. Spatiotemporal development of the neuronal accumulation of amyloid precursor protein and the amyloid plaque formation in the brain of 3xTg-AD mice. Heliyon 2024; 10:e28821. [PMID: 38596059 PMCID: PMC11002285 DOI: 10.1016/j.heliyon.2024.e28821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024] Open
Abstract
The amyloid plaque is a hallmark of Alzheimer's disease. The accumulation of the amyloid precursor protein (APP) in the neuronal structure is assumed to lead to amyloid plaque formation through the excessive production of β-amyloid protein. To study the relationship between the neuronal accumulation of APP and amyloid plaque formation, we histologically analyzed their development in the different brain regions in 3xTg-AD mice, which express Swedish mutated APP (APPSWE) in the neurons. Observation throughout the brain revealed APPSWE-positive somata in the broad regions. Quantitative model analysis showed that the somatic accumulation of APPSWE developed firstly in the hippocampus from a very early age (<1 month) and proceeded slower in the isocortex. In line with this, the hippocampus was the first region to form amyloid plaques at the age of 9-12 months, while amyloid plaques were rarely observed in the isocortex. Females had more APPSWE-positive somata and plaques than males. Furthermore, amyloid plaques were observed in the lateral septum and pontine grey, which did not contain APPSWE-positive somata but only the APPSWE-positive fibers. These results suggested that neuronal accumulation of APPSWE, both in somatodendritic and axonal domains, is closely related to the formation of amyloid plaques.
Collapse
Affiliation(s)
- Munenori Ono
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Tetsufumi Ito
- Systems Function and Morphology, University of Toyama, Toyama, 930-0194, Japan
| | - Sachiko Yamaki
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Yoshie Hori
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Qing Zhou
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Xirun Zhao
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Shinji Muramoto
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Takafumi Furuyama
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Hiromi Sakata-Haga
- Department of Anatomy, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Tsuyoshi Hamaguchi
- Department of Neurology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| |
Collapse
|
9
|
Zhan Q, Kong F, Shao S, Zhang B, Huang S. Pathogenesis of Depression in Alzheimer's Disease. Neurochem Res 2024; 49:548-556. [PMID: 38015411 DOI: 10.1007/s11064-023-04061-0] [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: 08/31/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023]
Abstract
Depression is a prevalent occurrence among Alzheimer's disease (AD) patients, yet its underlying mechanism remains unclear. Recent investigations have revealed that several pathophysiological changes associated with Alzheimer's disease can lead to mood disorders. These alterations include irregularities in monoamine neurotransmitters, disruptions in glutamatergic synaptic transmission, neuro-inflammation, dysfunction within the hypothalamic-pituitary-adrenocortical (HPA) axis, diminished levels of brain-derived neurotrophic factor (BDNF), and hippocampal atrophy. This review consolidates research findings from pertinent fields to elucidate the mechanisms underlying depression in Alzheimer's disease, aiming to provide valuable insights for the study of its mechanisms and clinical treatment.
Collapse
Affiliation(s)
- Qingyang Zhan
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Fanyi Kong
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Shuai Shao
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Bo Zhang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Shuming Huang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| |
Collapse
|
10
|
Wrzesień A, Andrzejewski K, Jampolska M, Kaczyńska K. Respiratory Dysfunction in Alzheimer's Disease-Consequence or Underlying Cause? Applying Animal Models to the Study of Respiratory Malfunctions. Int J Mol Sci 2024; 25:2327. [PMID: 38397004 PMCID: PMC10888758 DOI: 10.3390/ijms25042327] [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: 01/17/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative brain disease that is the most common cause of dementia among the elderly. In addition to dementia, which is the loss of cognitive function, including thinking, remembering, and reasoning, and behavioral abilities, AD patients also experience respiratory disturbances. The most common respiratory problems observed in AD patients are pneumonia, shortness of breath, respiratory muscle weakness, and obstructive sleep apnea (OSA). The latter is considered an outcome of Alzheimer's disease and is suggested to be a causative factor. While this narrative review addresses the bidirectional relationship between obstructive sleep apnea and Alzheimer's disease and reports on existing studies describing the most common respiratory disorders found in patients with Alzheimer's disease, its main purpose is to review all currently available studies using animal models of Alzheimer's disease to study respiratory impairments. These studies on animal models of AD are few in number but are crucial for establishing mechanisms, causation, implementing potential therapies for respiratory disorders, and ultimately applying these findings to clinical practice. This review summarizes what is already known in the context of research on respiratory disorders in animal models, while pointing out directions for future research.
Collapse
Affiliation(s)
| | | | | | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.W.); (K.A.); (M.J.)
| |
Collapse
|
11
|
Saternos H, Hamlett ED, Guzman S, Head E, Granholm AC, Ledreux A. Unique Pathology in the Locus Coeruleus of Individuals with Down Syndrome. J Alzheimers Dis 2024; 101:541-561. [PMID: 39213062 DOI: 10.3233/jad-240043] [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: 09/04/2024]
Abstract
Background Down syndrome (DS) is one of the most commonly occurring chromosomal conditions. Most individuals with DS develop Alzheimer's disease (AD) by 50 years of age. Recent evidence suggests that AD pathology in the locus coeruleus (LC) is an early event in sporadic AD. It is likely that the widespread axonal network of LC neurons contributes to the spread of tau pathology in the AD brain, although this has not been investigated in DS-AD. Objective The main purpose of this study was to profile AD pathology and neuroinflammation in the LC, comparing AD and DS-AD in postmortem human tissues. Methods We utilized immunofluorescence and semi-quantitative analyses of pTau (4 different forms), amyloid-β (Aβ), glial, and neuronal markers in the LC across 36 cases (control, DS-AD, and AD) to compare the different pathological profiles. Results Oligomeric tau was highly elevated in DS-AD cases compared to LOAD or EOAD cases. The distribution of staining for pT231 was elevated in DS-AD and EOAD compared to the LOAD group. The DS-AD group exhibited increased Aβ immunostaining compared to AD cases. The number of tau-bearing neurons was also significantly different between the EOAD and DS-AD cases compared to the LOAD cases. Conclusions While inflammation, pTau, and Aβ are all involved in AD pathology, their contribution to disease progression may differ depending on the diagnosis. Our results suggest that DS-AD and EOAD may be more similar in pathology than LOAD. Our study highlights unique avenues to further our understanding of the mechanisms governing AD neuropathology.
Collapse
Affiliation(s)
- Hannah Saternos
- Department of Neurosurgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric D Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Samuel Guzman
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California at Irvine, Irvine, CA, USA
| | - Ann-Charlotte Granholm
- Department of Neurosurgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aurélie Ledreux
- Department of Neurosurgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
12
|
Frank B, Walsh M, Hurley L, Groh J, Blennow K, Zetterberg H, Tripodis Y, Budson AE, O'Connor MK, Martin B, Weller J, McKee A, Qiu W, Stein TD, Stern RA, Mez J, Henson R, Long J, Aschenbrenner AJ, Babulal GM, Morris JC, Schindler S, Alosco ML. Cognition Mediates the Association Between Cerebrospinal Fluid Biomarkers of Amyloid and P-Tau and Neuropsychiatric Symptoms. J Alzheimers Dis 2024; 100:1055-1073. [PMID: 38995786 DOI: 10.3233/jad-240125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Background Neuropsychiatric symptoms (NPS) can be an early manifestation of Alzheimer's disease (AD). However, the associations among NPS, cognition, and AD biomarkers across the disease spectrum are unclear. Objective We analyzed cross-sectional mediation pathways between cerebrospinal fluid (CSF) biomarkers of AD (Aβ1-42, p-tau181), cognitive function, and NPS. Methods Primary models included 781 participants from the National Alzheimer's Coordinating Center (NACC) data set who had CSF analyzed for AD biomarkers using Lumipulse. NPS were assessed with the Neuropsychiatric Inventory Questionnaire (NPI-Q). We assessed cognition with the harmonized MMSE/MoCA, as well as neuropsychological tests sensitive to AD pathology: story recall, naming, animal fluency, and Trails B. The Clinical Dementia Rating (CDR®) scale assessed dementia severity. Mediation models were estimated with Kemeny metric covariance in a structural equation model framework, controlling for age, education, sex, and APOEɛ4. Results The sample was older adults (M = 73.85, SD = 6.68; 49.9% male, 390; 27.9% dementia, 218) who were predominantly white (n = 688, 88.1%). Higher p-tau181/Aβ1-42 ratio predicted higher NPI-Q, which was partially mediated by the MMSE/MoCA and, in a second model, story recall. No other pathway was statistically significant. Both the MMSE/MoCA and NPI-Q independently mediated the association between p-tau181/Aβ1-42 ratio and CDR global impairment. With dementia excluded, p-tau181/Aβ1-42 ratio was no longer associated with the NPI-Q. Conclusions NPS may be secondary to cognitive impairment and AD pathology through direct and indirect pathways. NPS independently predict dementia severity in AD. However, AD pathology likely plays less of a role in NPS in samples without dementia.
Collapse
Affiliation(s)
- Brandon Frank
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Michael Walsh
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Landon Hurley
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
| | - Jenna Groh
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Andrew E Budson
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Maureen K O'Connor
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, U.S. Department of Veteran Affairs, Bedford, MA, USA
| | - Brett Martin
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Jason Weller
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Ann McKee
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Wendy Qiu
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Robert A Stern
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Framingham Heart Study, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Rachel Henson
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Justin Long
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Ganesh M Babulal
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Suzanne Schindler
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
13
|
Tian J, Du E, Guo L. Mitochondrial Interaction with Serotonin in Neurobiology and Its Implication in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:1165-1177. [PMID: 38025801 PMCID: PMC10657725 DOI: 10.3233/adr-230070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/16/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is a lethal neurodegenerative disorder characterized by severe brain pathologies and progressive cognitive decline. While the exact cause of this disease remains unknown, emerging evidence suggests that dysregulation of neurotransmitters contributes to the development of AD pathology and symptoms. Serotonin, a critical neurotransmitter in the brain, plays a pivotal role in regulating various brain processes and is implicated in neurological and psychiatric disorders, including AD. Recent studies have shed light on the interplay between mitochondrial function and serotonin regulation in brain physiology. In AD, there is a deficiency of serotonin, along with impairments in mitochondrial function, particularly in serotoninergic neurons. Additionally, altered activity of mitochondrial enzymes, such as monoamine oxidase, may contribute to serotonin dysregulation in AD. Understanding the intricate relationship between mitochondria and serotonin provides valuable insights into the underlying mechanisms of AD and identifies potential therapeutic targets to restore serotonin homeostasis and alleviate AD symptoms. This review summarizes the recent advancements in unraveling the connection between brain mitochondria and serotonin, emphasizing their significance in AD pathogenesis and underscoring the importance of further research in this area. Elucidating the role of mitochondria in serotonin dysfunction will promote the development of therapeutic strategies for the treatment and prevention of this neurodegenerative disorder.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Eric Du
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
- Blue Valley West High School, Overland Park, KS, USA
| | - Lan Guo
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
14
|
Ranjan A, Biswas S, Mallick BN. Rapid eye movement sleep loss associated cytomorphometric changes and neurodegeneration. Sleep Med 2023; 110:25-34. [PMID: 37524037 DOI: 10.1016/j.sleep.2023.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/10/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023]
Abstract
Rapid eye movement sleep (REMS) is essential for leading normal healthy living at least in higher-order mammals, including humans. In this review, we briefly survey the available literature for evidence linking cytomorphometric changes in the brain due to loss of REMS. As a mechanism of action, we add evidence that REMS loss elevates noradrenaline (NA) levels in the brain, which affects neuronal cytomorphology. These changes may be a compensatory mechanism as the changes return to normal after the subjects recover from the loss of REMS or if during REMS deprivation, the subjects are treated with NA-adrenoceptor antagonist prazosin (PRZ). We had proposed earlier that one of the fundamental functions of REMS is to maintain the level of NA in the brain. We elaborate on this idea to propose that if REMS loss continues without recovery, the sustained level of NA breaks down neurophysiologically active compensatory mechanism/s starting with changes in the neuronal cytomorphology, followed by their degeneration, leading to acute and chronic pathological conditions. Identification of neuronal cytomorphological changes could prove to be of significance for predicting future neuronal (brain) damage as well as an indicator for REMS health. Although current brain imaging techniques may not enable us to visualize changes in neuronal cytomorphology, given the rapid technological progress including use of artificial intelligence, we are optimistic that it may be a reality soon. Finally, we propose that maintenance of optimum REMS must be considered a criterion for leading a healthy life.
Collapse
Affiliation(s)
- Amit Ranjan
- Department of Zoology, Mahatma Gandhi Central University, Motihari, East Champaran, Bihar, 845401, India.
| | - Sudipta Biswas
- Math, Science, Engineering Department, South Mountain Community College, 7050 S 24th St, Phoenix, AZ, 85042, USA
| | - Birendra Nath Mallick
- Amity Institute of Neuropsychology & Neurosciences, Amity University Campus, Sector 125, Gautam Budh Nagar, Noida, 201313, Uttar Pradesh, India
| |
Collapse
|
15
|
Nageeb Hasan SM, Clarke CL, McManamon Strand TP, Bambico FR. Putative pathological mechanisms of late-life depression and Alzheimer's Disease. Brain Res 2023:148423. [PMID: 37244602 DOI: 10.1016/j.brainres.2023.148423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by progressive impairment in cognition and memory. AD is accompanied by several neuropsychiatric symptoms, with depression being the most prominent. Although depression has long been known to be associated with AD, controversial findings from preclinical and clinical studies have obscured the precise nature of this association. However recent evidence suggests that depression could be a prodrome or harbinger of AD. Evidence indicates that the major central serotonergic nucleus-the dorsal raphe nucleus (DRN)-shows very early AD pathology: neurofibrillary tangles made of hyperphosphorylated tau protein and degenerated neurites. AD and depression share common pathophysiologies, including functional deficits of the serotonin (5-HT) system. 5-HT receptors have modulatory effects on the progression of AD pathology i.e., reduction in Aβ load, increased hyper-phosphorylation of tau, decreased oxidative stress etc. Moreover, preclinical models show a role for specific channelopathies that result in abnormal regional activational and neuroplasticity patterns. One of these concerns the pathological upregulation of the small conductance calcium-activated potassium (SK) channel in corticolimbic structure. This has also been observed in the DRN in both diseases. The SKC is a key regulator of cell excitability and long-term potentiation (LTP). SKC over-expression is positively correlated with aging and cognitive decline, and is evident in AD. Pharmacological blockade of SKCs has been reported to reverse symptoms of depression and AD. Thus, aberrant SKC functioning could be related to depression pathophysiology and diverts its late-life progression towards the development of AD. We summarize findings from preclinical and clinical studies suggesting a molecular linkage between depression and AD pathology. We also provide a rationale for considering SKCs as a novel pharmacological target for the treatment of AD-associated symptoms.
Collapse
Affiliation(s)
- S M Nageeb Hasan
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada.
| | - Courtney Leigh Clarke
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada
| | | | - Francis Rodriguez Bambico
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada; Behavioural Neurobiology Laboratory, Centre for Addiction and Mental Health, Toronto, ON, M5T1R8, Canada
| |
Collapse
|
16
|
Klingenberg M, Stark D, Eitel F, Budding C, Habes M, Ritter K. Higher performance for women than men in MRI-based Alzheimer's disease detection. Alzheimers Res Ther 2023; 15:84. [PMID: 37081528 PMCID: PMC10116672 DOI: 10.1186/s13195-023-01225-6] [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: 08/15/2022] [Accepted: 04/03/2023] [Indexed: 04/22/2023]
Abstract
INTRODUCTION Although machine learning classifiers have been frequently used to detect Alzheimer's disease (AD) based on structural brain MRI data, potential bias with respect to sex and age has not yet been addressed. Here, we examine a state-of-the-art AD classifier for potential sex and age bias even in the case of balanced training data. METHODS Based on an age- and sex-balanced cohort of 432 subjects (306 healthy controls, 126 subjects with AD) extracted from the ADNI data base, we trained a convolutional neural network to detect AD in MRI brain scans and performed ten different random training-validation-test splits to increase robustness of the results. Classifier decisions for single subjects were explained using layer-wise relevance propagation. RESULTS The classifier performed significantly better for women (balanced accuracy [Formula: see text]) than for men ([Formula: see text]). No significant differences were found in clinical AD scores, ruling out a disparity in disease severity as a cause for the performance difference. Analysis of the explanations revealed a larger variance in regional brain areas for male subjects compared to female subjects. DISCUSSION The identified sex differences cannot be attributed to an imbalanced training dataset and therefore point to the importance of examining and reporting classifier performance across population subgroups to increase transparency and algorithmic fairness. Collecting more data especially among underrepresented subgroups and balancing the dataset are important but do not always guarantee a fair outcome.
Collapse
Affiliation(s)
- Malte Klingenberg
- Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Neurosciences, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Didem Stark
- Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Neurosciences, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Fabian Eitel
- Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Neurosciences, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Céline Budding
- Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kerstin Ritter
- Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Neurosciences, Berlin, Germany.
- Bernstein Center for Computational Neuroscience, Berlin, Germany.
| |
Collapse
|
17
|
Langer Horvat L, Španić Popovački E, Babić Leko M, Zubčić K, Horvat L, Mustapić M, Hof PR, Šimić G. Anterograde and Retrograde Propagation of Inoculated Human Tau Fibrils and Tau Oligomers in a Non-Transgenic Rat Tauopathy Model. Biomedicines 2023; 11:1004. [PMID: 37189622 PMCID: PMC10135744 DOI: 10.3390/biomedicines11041004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 05/17/2023] Open
Abstract
The tauopathy of Alzheimer's disease (AD) is first observed in the brainstem and entorhinal cortex, spreading trans-synaptically along specific pathways to other brain regions with recognizable patterns. Tau propagation occurs retrogradely and anterogradely (trans-synaptically) along a given pathway and through exosomes and microglial cells. Some aspects of in vivo tau spreading have been replicated in transgenic mice models expressing a mutated human MAPT (tau) gene and in wild-type mice. In this study, we aimed to characterize the propagation of different forms of tau species in non-transgenic 3-4 months old wild-type rats after a single unilateral injection of human tau oligomers and tau fibrils into the medial entorhinal cortex (mEC). We determined whether different variants of the inoculated human tau protein, tau fibrils, and tau oligomers, would induce similar neurofibrillary changes and propagate in an AD-related pattern, and how tau-related pathological changes would correlate with presumed cognitive impairment. We injected human tau fibrils and tau oligomers stereotaxically into the mEC and examined the distribution of tau-related changes at 3 days and 4, 8, and 11 months post-injection using antibodies AT8 and MC1, which reveal early phosphorylation and aberrant conformation of tau, respectively, HT7, anti-synaptophysin, and the Gallyas silver staining method. Human tau oligomers and tau fibrils exhibited some similarities and some differences in their ability to seed and propagate tau-related changes. Both human tau fibrils and tau oligomers rapidly propagated from the mEC anterogradely into the hippocampus and various parts of the neocortex. However, using a human tau-specific HT7 antibody, 3 days post-injection we found inoculated human tau oligomers in the red nucleus, primary motor, and primary somatosensory cortex, a finding not seen in animals inoculated with human tau fibrils. In animals inoculated with human tau fibrils, 3 days post-injection the HT7 antibody showed fibrils in the pontine reticular nucleus, a finding explained only by uptake of human tau fibrils by incoming presynaptic fibers to the mEC and retrograde transport of inoculated human tau fibrils to the brainstem. Rats inoculated with human tau fibrils showed as early as 4 months after inoculation a spread of phosphorylated tau protein at the AT8 epitopes throughout the brain, dramatically faster propagation of neurofibrillary changes than with human tau oligomers. The overall severity of tau protein changes 4, 8, and 11 months after inoculation of human tau oligomers and tau fibrils correlated well with spatial working memory and cognition impairments, as measured by the T-maze spontaneous alternation, novel object recognition, and object location tests. We concluded that this non-trangenic rat model of tauopathy, especially when using human tau fibrils, demonstrates rapidly developing pathologic alterations in neurons, synapses, and identifiable pathways together with cognitive and behavioral changes, through the anterograde and retrograde spreading of neurofibrillary degeneration. Therefore, it represents a promising model for future experimental studies of primary and secondary tauopathies, especially AD.
Collapse
Affiliation(s)
- Lea Langer Horvat
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Ena Španić Popovački
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Klara Zubčić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Luka Horvat
- Department of Molecular Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Maja Mustapić
- Laboratory of Clinical Investigation, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
18
|
El-Latif AAA, Chelloug SA, Alabdulhafith M, Hammad M. Accurate Detection of Alzheimer's Disease Using Lightweight Deep Learning Model on MRI Data. Diagnostics (Basel) 2023; 13:diagnostics13071216. [PMID: 37046434 PMCID: PMC10093003 DOI: 10.3390/diagnostics13071216] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 04/14/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment and aberrant protein deposition in the brain. Therefore, the early detection of AD is crucial for the development of effective treatments and interventions, as the disease is more responsive to treatment in its early stages. It is worth mentioning that deep learning techniques have been successfully applied in recent years to a wide range of medical imaging tasks, including the detection of AD. These techniques have the ability to automatically learn and extract features from large datasets, making them well suited for the analysis of complex medical images. In this paper, we propose an improved lightweight deep learning model for the accurate detection of AD from magnetic resonance imaging (MRI) images. Our proposed model achieves high detection performance without the need for deeper layers and eliminates the use of traditional methods such as feature extraction and classification by combining them all into one stage. Furthermore, our proposed method consists of only seven layers, making the system less complex than other previous deep models and less time-consuming to process. We evaluate our proposed model using a publicly available Kaggle dataset, which contains a large number of records in a small dataset size of only 36 Megabytes. Our model achieved an overall accuracy of 99.22% for binary classification and 95.93% for multi-classification tasks, which outperformed other previous models. Our study is the first to combine all methods used in the publicly available Kaggle dataset for AD detection, enabling researchers to work on a dataset with new challenges. Our findings show the effectiveness of our lightweight deep learning framework to achieve high accuracy in the classification of AD.
Collapse
Affiliation(s)
- Ahmed A Abd El-Latif
- EIAS Data Science Lab, College of Computer and Information Sciences, Prince Sultan University, P.O. Box 66833, Riyadh 11586, Saudi Arabia
- Department of Mathematics and Computer Science, Faculty of Science, Menoufia University, Shibin El Kom 32511, Egypt
| | - Samia Allaoua Chelloug
- Department of Information Technology, College of Computer and Information Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Maali Alabdulhafith
- Department of Information Technology, College of Computer and Information Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mohamed Hammad
- EIAS Data Science Lab, College of Computer and Information Sciences, Prince Sultan University, P.O. Box 66833, Riyadh 11586, Saudi Arabia
- Department of Information Technology, Faculty of Computers and Information, Menoufia University, Shibin El Kom 32511, Egypt
| |
Collapse
|
19
|
Humphrey CM, Hooker JW, Thapa M, Wilcox MJ, Ostrowski D, Ostrowski TD. Synaptic loss and gliosis in the nucleus tractus solitarii with streptozotocin-induced Alzheimer's disease. Brain Res 2023; 1801:148202. [PMID: 36521513 PMCID: PMC9840699 DOI: 10.1016/j.brainres.2022.148202] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/21/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Obstructive sleep apnea is highly prevalent in Alzheimer's disease (AD). However, brainstem centers controlling respiration have received little attention in AD research, and mechanisms behind respiratory dysfunction in AD are not understood. The nucleus tractus solitarii (nTS) is an important brainstem center for respiratory control and chemoreflex function. Alterations of nTS integrity, like those shown in AD patients, likely affect neuronal processing and adequate control of breathing. We used the streptozotocin-induced rat model of AD (STZ-AD) to analyze cellular changes in the nTS that corroborate previously documented respiratory dysfunction. We used 2 common dosages of STZ (2 and 3 mg/kg STZ) for model induction and evaluated the early impact on cell populations in the nTS. The hippocampus served as control region to identify site-specific effects of STZ. There was significant atrophy in the caudal nTS of the 3 mg/kg STZ-AD group only, an area known to integrate chemoafferent information. Also, the hippocampus had significant atrophy with the highest STZ dosage tested. Both STZ-AD groups showed respiratory dysfunction along with multiple indices for astroglial and microglial activation. These changes were primarily located in the caudal and intermediate nTS. While there was no change of astrocytes in the hippocampus, microglial activation was accompanied by a reduction in synaptic density. Together, our data demonstrate that STZ-AD induces site-specific effects on all major cell types, primarily in the caudal/intermediate nTS. Both STZ dosages used in this study produced a similar outcome and can be used for future studies examining the initial symptoms of STZ-AD.
Collapse
Affiliation(s)
- Chuma M Humphrey
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University, 800 W. Jefferson St., Kirksville, MO, USA
| | - John W Hooker
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University, 800 W. Jefferson St., Kirksville, MO, USA
| | - Mahima Thapa
- Department of Biology, Truman State University, 100 E. Normal Ave., Kirksville, MO, USA
| | - Mason J Wilcox
- Department of Biology, Truman State University, 100 E. Normal Ave., Kirksville, MO, USA
| | - Daniela Ostrowski
- Department of Biology, Truman State University, 100 E. Normal Ave., Kirksville, MO, USA
| | - Tim D Ostrowski
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University, 800 W. Jefferson St., Kirksville, MO, USA.
| |
Collapse
|
20
|
Zeng X, Wang Z, Tan W, Petersen E, Cao X, LaBella A, Boccia A, Franceschi D, de Leon M, Chiang GCY, Qi J, Biegon A, Zhao W, Goldan AH. A conformal TOF-DOI Prism-PET prototype scanner for high-resolution quantitative neuroimaging. Med Phys 2023; 50:10.1002/mp.16223. [PMID: 36651630 PMCID: PMC11025680 DOI: 10.1002/mp.16223] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Positron emission tomography (PET) has had a transformative impact on oncological and neurological applications. However, still much of PET's potential remains untapped with limitations primarily driven by low spatial resolution, which severely hampers accurate quantitative PET imaging via the partial volume effect (PVE). PURPOSE We present experimental results of a practical and cost-effective ultra-high resolution brain-dedicated PET scanner, using our depth-encoding Prism-PET detectors arranged along a compact and conformal gantry, showing substantial reduction in PVE and accurate radiotracer uptake quantification in small regions. METHODS The decagon-shaped prototype scanner has a long diameter of 38.5 cm, a short diameter of 29.1 cm, and an axial field-of-view (FOV) of 25.5 mm with a single ring of 40 Prism-PET detector modules. Each module comprises a 16 × 16 array of 1.5 × 1.5 × 20-mm3 lutetium yttrium oxyorthosillicate (LYSO) scintillator crystals coupled 4-to-1 to an 8 × 8 array of silicon photomultiplier (SiPM) pixels on one end and to a prismatoid light guide array on the opposite end. The scanner's performance was evaluated by measuring depth-of-interaction (DOI) resolution, energy resolution, timing resolution, spatial resolution, sensitivity, and image quality of ultra-micro Derenzo and three-dimensional (3D) Hoffman brain phantoms. RESULTS The full width at half maximum (FWHM) DOI, energy, and timing resolutions of the scanner are 2.85 mm, 12.6%, and 271 ps, respectively. Not considering artifacts due to mechanical misalignment of detector blocks, the intrinsic spatial resolution is 0.89-mm FWHM. Point source images reconstructed with 3D filtered back-projection (FBP) show an average spatial resolution of 1.53-mm FWHM across the entire FOV. The peak absolute sensitivity is 1.2% for an energy window of 400-650 keV. The ultra-micro Derenzo phantom study demonstrates the highest reported spatial resolution performance for a human brain PET scanner with perfect reconstruction of 1.00-mm diameter hot-rods. Reconstructed images of customized Hoffman brain phantoms prove that Prism-PET enables accurate radiotracer uptake quantification in small brain regions (2-3 mm). CONCLUSIONS Prism-PET will substantially strengthen the utility of quantitative PET in neurology for early diagnosis of neurodegenerative diseases, and in neuro-oncology for improved management of both primary and metastatic brain tumors.
Collapse
Affiliation(s)
- Xinjie Zeng
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
- Department of Electrical and Computer Engineering, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, US
| | - Zipai Wang
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, US
| | - Wanbin Tan
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, US
| | - Eric Petersen
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, US
| | - Xinjie Cao
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
- Department of Electrical and Computer Engineering, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, US
| | - Andy LaBella
- Department of Radiology, Boston children’s Hospital, Boston, MA, US
| | - Anthony Boccia
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
| | - Dinko Franceschi
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
| | - Mony de Leon
- Department of Radiology, Weill Cornell Medical College, Cornell University, New York, NY, US
| | - Gloria Chia-Yi Chiang
- Department of Radiology, Weill Cornell Medical College, Cornell University, New York, NY, US
| | - Jinyi Qi
- Department of Biomedical Engineering, University of California, Davis, CA, US
| | - Anat Biegon
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
| | - Wei Zhao
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
| | - Amir H. Goldan
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, US
| |
Collapse
|
21
|
Smith GS, Protas H, Kuwabara H, Savonenko A, Nassery N, Gould NF, Kraut M, Avramopoulos D, Holt D, Dannals RF, Nandi A, Su Y, Reiman EM, Chen K. Molecular imaging of the association between serotonin degeneration and beta-amyloid deposition in mild cognitive impairment. Neuroimage Clin 2023; 37:103322. [PMID: 36680976 PMCID: PMC9869478 DOI: 10.1016/j.nicl.2023.103322] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/28/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
BACKGROUND Degeneration of the serotonin system has been observed in Alzheimer's disease (AD) and in mild cognitive impairment (MCI). In transgenic amyloid mouse models, serotonin degeneration is detected prior to widespread cortical beta-amyloid (Aβ) deposition, also suggesting that serotonin degeneration may be observed in preclinical AD. METHODS The differences in the distribution of serotonin degeneration (reflected by the loss of the serotonin transporter, 5-HTT) relative to Aβ deposition was measured with positron emission tomography in a group of individuals with MCI and a group of healthy older adults. A multi-modal partial least squares (mmPLS) algorithm was applied to identify the spatial covariance pattern between 5-HTT availability and Aβ deposition. RESULTS Forty-five individuals with MCI and 35 healthy older adults were studied, 22 and 27 of whom were included in the analyses who were "amyloid positive" and "amyloid negative", respectively. A pattern of lower cortical, subcortical and limbic 5-HTT availability and higher cortical Aβ deposition distinguished the MCI from the healthy older control participants. Greater expression of this pattern was correlated with greater deficits in memory and executive function in the MCI group, not in the control group. CONCLUSION A spatial covariance pattern of lower 5-HTT availability and Aβ deposition was observed to a greater extent in an MCI group relative to a control group and was associated with cognitive impairment in the MCI group. The results support the application of mmPLS to understand the neurochemical changes associated with Aβ deposition in the course of preclinical AD.
Collapse
Affiliation(s)
- Gwenn S Smith
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | - Hiroto Kuwabara
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alena Savonenko
- Department of Pathology (Neuropathology), Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Najlla Nassery
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Neda F Gould
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Kraut
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitri Avramopoulos
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel Holt
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert F Dannals
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ayon Nandi
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | | | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| |
Collapse
|
22
|
Tian J, Stucky CS, Wang T, Muma NA, Johnson M, Du H. Mitochondrial Dysfunction Links to Impaired Hippocampal Serotonin Release in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2023; 93:605-619. [PMID: 37066917 PMCID: PMC10416312 DOI: 10.3233/jad-230072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND Deprivation of extracellular serotonin has been linked to cognitive decline and neuropsychiatric disturbances in Alzheimer's disease (AD). However, despite degeneration of serotonin-producing neurons, whether serotonin release is affected in AD-sensitive brain regions is unknown. OBJECTIVE This study investigated the impact of mitochondrial dysfunction in decreased hippocampal serotonin release in AD amyloidosis mouse model 5xFAD mice. METHODS Electrochemical assays were applied to examine hippocampal serotonin release. We also employed multidisciplinary techniques to determine the role of oligomeric amyloid-β (Aβ) in hippocampal mitochondrial deficits and serotonin release deficiency. RESULTS 5xFAD mice exhibited serotonin release decrease and relatively moderate downregulation of serotonergic fiber density as well as serotonin content in the hippocampal region. Further experiments showed an inhibitory effect of oligomeric amyloid-β (Aβ) on hippocampal serotonin release without affecting the density of serotonergic fibers. Pharmaceutical uncoupling of mitochondrial oxidative phosphorylation (OXPHOS) disrupted hippocampal serotonin release in an ex vivo setting. This echoes the mitochondrial defects in serotonergic fibers in 5xFAD mice and oligomeric Aβ-challenged primary serotonergic neuron cultures and implicates a link between mitochondrial dysfunction and serotonin transmission defects in AD-relevant pathological settings. CONCLUSION The most parsimonious interpretation of our findings is that mitochondrial dysfunction is a phenotypic change of serotonergic neurons, which potentially plays a role in the development of serotonergic failure in AD-related conditions.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | | | - Tienju Wang
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Nancy A. Muma
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Michael Johnson
- Department of Chemistry, University of Kansas, Lawrence, KS, USA
| | - Heng Du
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
- Higuchi Biosciences Center, University of Kansas, Lawrence, KS, USA
- Alzheimer’s Disease Center, University of Kansas Medical Center, Lawrence, KS, USA
| |
Collapse
|
23
|
Severity estimation of brainstem in dementia MR images using moth flame optimized segmentation and fused deep feature selection. Neural Comput Appl 2022. [DOI: 10.1007/s00521-022-08167-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
24
|
Babić Leko M, Nikolac Perković M, Španić E, Švob Štrac D, Pleić N, Vogrinc Ž, Gunjača I, Bežovan D, Nedić Erjavec G, Klepac N, Borovečki F, Zemunik T, Pivac N, Hof PR, Šimić G. Serotonin Receptor Gene Polymorphisms Are Associated with Cerebrospinal Fluid, Genetic, and Neuropsychological Biomarkers of Alzheimer's Disease. Biomedicines 2022; 10:3118. [PMID: 36551873 PMCID: PMC9775360 DOI: 10.3390/biomedicines10123118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/26/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
A decrease in serotonergic transmission throughout the brain is among the earliest pathological changes in Alzheimer's disease (AD). Serotonergic receptors are also affected in AD. Polymorphisms in genes of serotonin (5HT) receptors have been mostly associated with behavioral and psychological symptoms of dementia (BPSD). In this study, we examined if AD patients carrying different genotypes in 5HTR1B rs13212041, 5HTR2A rs6313 (T102C), 5HTR2C rs3813929 (-759C/T), and 5HTR6 rs1805054 (C267T) polymorphisms have a higher risk of faster disease progression (assessed by neuropsychological testing), are more prone to develop AD-related pathology (reflected by levels of cerebrospinal fluid [CSF] AD biomarkers), or have an association with an apolipoprotein E (APOE) haplotype. This study included 115 patients with AD, 53 patients with mild cognitive impairment (MCI), and 2701 healthy controls. AD biomarkers were determined in the CSF of AD and MCI patients using enzyme-linked immunosorbent assays (ELISA), while polymorphisms were determined using either TaqMan SNP Genotyping Assays or Illumina genotyping platforms. We detected a significant decrease in the CSF amyloid β1-42 (Aβ1-42) and an increase in p-tau181/Aβ1-42 ratio in carriers of the T allele in the 5HTR2C rs3813929 (-759C/T) polymorphism. A significantly higher number of APOE ε4 allele carriers was observed among individuals carrying a TT genotype within the 5HTR2A T102C polymorphism, a C allele within the 5HTR1B rs13212041 polymorphism, and a T allele within the 5HTR6 rs1805054 (C267T) polymorphism. Additionally, individuals carrying the C allele within the 5HTR1B rs13212041 polymorphism were significantly more represented among AD patients and had poorer performances on the Rey-Osterrieth test. Carriers of the T allele within the 5HTR6 rs1805054 had poorer performances on the MMSE and ADAS-Cog. As all four analyzed polymorphisms of serotonin receptor genes showed an association with either genetic, CSF, or neuropsychological biomarkers of AD, they deserve further investigation as potential early genetic biomarkers of AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, 10000 Zagreb, Croatia
- Department of Medical Biology, School of Medicine, University of Split, 21000 Split, Croatia
| | | | - Ena Španić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, 10000 Zagreb, Croatia
| | - Dubravka Švob Štrac
- Department of Molecular Medicine, Institute Ruđer Bošković, 10000 Zagreb, Croatia
| | - Nikolina Pleić
- Department of Medical Biology, School of Medicine, University of Split, 21000 Split, Croatia
| | - Željka Vogrinc
- Laboratory for Neurobiochemistry, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Ivana Gunjača
- Department of Medical Biology, School of Medicine, University of Split, 21000 Split, Croatia
| | | | | | - Nataša Klepac
- Department of Neurology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Tatijana Zemunik
- Department of Medical Biology, School of Medicine, University of Split, 21000 Split, Croatia
| | - Nela Pivac
- Department of Molecular Medicine, Institute Ruđer Bošković, 10000 Zagreb, Croatia
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, 10000 Zagreb, Croatia
| |
Collapse
|
25
|
Caligiore D, Giocondo F, Silvetti M. The Neurodegenerative Elderly Syndrome (NES) hypothesis: Alzheimer and Parkinson are two faces of the same disease. IBRO Neurosci Rep 2022; 13:330-343. [PMID: 36247524 PMCID: PMC9554826 DOI: 10.1016/j.ibneur.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/07/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022] Open
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) and Parkinson's disease (PD) share monoamine and alpha-synuclein (αSyn) dysfunctions, often beginning years before clinical manifestations onset. The triggers for these impairments and the causes leading these early neurodegenerative processes to become AD or PD remain unclear. We address these issues by proposing a radically new perspective to frame AD and PD: they are different manifestations of one only disease we call "Neurodegenerative Elderly Syndrome (NES)". NES goes through three phases. The seeding stage, which starts years before clinical signs, and where the part of the brain-body affected by the initial αSyn and monoamine dysfunctions, influences the future possible progression of NES towards PD or AD. The compensatory stage, where the clinical symptoms are still silent thanks to compensatory mechanisms keeping monoamine concentrations homeostasis. The bifurcation stage, where NES becomes AD or PD. We present recent literature supporting NES and discuss how this hypothesis could radically change the comprehension of AD and PD comorbidities and the design of novel system-level diagnostic and therapeutic actions.
Collapse
Affiliation(s)
- Daniele Caligiore
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
- AI2Life s.r.l., Innovative Start-Up, ISTC-CNR Spin-Off, Via Sebino 32, Rome 00199, Italy
| | - Flora Giocondo
- Laboratory of Embodied Natural and Artificial Intelligence, Institute of Cognitive Sciences and Technologies, National Research Council (LENAI-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
| | - Massimo Silvetti
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
| |
Collapse
|
26
|
Takahashi M, Akamatsu G, Iwao Y, Tashima H, Yoshida E, Yamaya T. Small nuclei identification with a hemispherical brain PET. EJNMMI Phys 2022; 9:69. [PMID: 36209191 PMCID: PMC9547762 DOI: 10.1186/s40658-022-00498-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To confirm the performance of the first hemispherical positron emission tomography (PET) for the brain (Vrain) that we developed to visualise the small nuclei in the deep brain area, we compared 18F-fluorodeoxyglucose (FDG) brain images with whole-body PET images. METHODS Ten healthy male volunteers (aged 22-45 years) underwent a representative clinical whole-body PET, followed by Vrain each for 10 min. These two scans were initiated 30 min and 45 min after FDG injection (4.1 ± 0.5 MBq/kg), respectively. First, we visually identified the small nuclei and then compared their standardised uptake values (SUVs) with the participants' age. Next, the SUVs of each brain region, which were determined by applying a volume-of-interest template for anatomically normalised PET images, were compared between the brain images with the Vrain and those with the whole-body PET images. RESULTS Small nuclei, such as the inferior colliculus, red nucleus, and substantia nigra, were more clearly visualised in Vrain than in whole-body PET. The anterior nucleus and dorsomedial nucleus in the thalamus and raphe nucleus in the brainstem were identified in Vrain but not in whole-body PET. The SUVs of the inferior colliculus and dentate gyrus in the cerebellum positively correlated with age (Spearman's correlation coefficient r = 0.811, p = 0.004; r = 0.738, p = 0.015, respectively). The SUVs of Vrain were slightly higher in the mesial temporal and medial parietal lobes than those in whole-body PET. CONCLUSIONS This was the first time that the raphe nuclei, anterior nuclei, and dorsomedial nuclei were successfully visualised using the first hemispherical brain PET. TRIAL REGISTRATION : Japan Registry of Clinical Trials, jRCTs032210086, Registered 13 May 2021, https://jrct.niph.go.jp/latest-detail/jRCTs032210086 .
Collapse
Affiliation(s)
- Miwako Takahashi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| | - Go Akamatsu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Yuma Iwao
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Hideaki Tashima
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Eiji Yoshida
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Taiga Yamaya
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| |
Collapse
|
27
|
A Review on Characteristics of Experimental Research on Acupuncture Treatment for Alzheimer's Disease: Study Design. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8243704. [PMID: 35855822 PMCID: PMC9288276 DOI: 10.1155/2022/8243704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/31/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
Abstract
Background This review aims to systematically summarize and analyze recent high-quality animal research results about the use of acupuncture in Alzheimer's disease (AD) patients. This information will be useful in providing a reference for future experimental research and an experimental basis for the clinical use of acupuncture in the treatment of AD. Methods We utilized and referenced various electronic libraries from their inception to November 2021. Relevant information was reviewed and information such as the journal names, publication records, animal model selections and preparations, intervention measures, acupoint selections, detection methods, and detection indicators was extracted. Results. A total of 75 eligible studies were selected for additional review. Male SAMP8 mice, APP/PS1 double transgenic mice, Sprague-Dawley (SD) rats, and Wistar rats were the four commonly used animal strains in the experiments. The animals were categorized as transgenic and surgical mouse models. Experimental interventions included manual acupuncture (MA), Electro-acupuncture (EA), Moxibustion, and EA combined with Moxibustion. A retention time of 20 min was the optimal timing for experimental studies, with 14 sessions chosen as the most common treatment time. EA was the most prescribed acupuncture treatment type with continuous wave, 2 Hz frequency, and 1 mA electric current selected as frequently used parameters. A total of 78 acupoint prescriptions were analyzed involving 21 acupoints. The top 3 combinations of common acupoints were GV20 ⟶ EX-HN3, GV20 ⟶ BL23, and GV20 ⟶ GV26. A total of 39 articles had positive drug control groups, sham acupuncture, and/or nonacupoint control groups. Furthermore, 10 types of behavioral tests, 29 detection methods, 178 evaluation indicators, and 18 tissue samples were included in the analysis. Conclusions By collating these high-quality research studies systematically and comprehensively, acupuncture was found to be a viable and effective treatment in AD animal models. In addition, when designing experiments, researchers could refer to the detailed data provided here to make better schemes and maybe conduct more investigations in unresearched areas.
Collapse
|
28
|
Šimić G, Krsnik Ž, Knezović V, Kelović Z, Mathiasen ML, Junaković A, Radoš M, Mulc D, Španić E, Quattrocolo G, Hall VJ, Zaborszky L, Vukšić M, Olucha Bordonau F, Kostović I, Witter MP, Hof PR. Prenatal development of the human entorhinal cortex. J Comp Neurol 2022; 530:2711-2748. [DOI: 10.1002/cne.25344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Goran Šimić
- Department of Neuroscience Croatian Institute for Brain Research University of Zagreb Medical School, Zagreb, HR Croatia
| | - Željka Krsnik
- Department of Neuroscience Croatian Institute for Brain Research University of Zagreb Medical School, Zagreb, HR Croatia
| | - Vinka Knezović
- Department of Neuroscience Croatian Institute for Brain Research University of Zagreb Medical School, Zagreb, HR Croatia
| | - Zlatko Kelović
- Department of Anatomy University of Zagreb Medical School, Zagreb, HR Croatia
| | - Mathias Lysholt Mathiasen
- Department of Veterinary and Animal Sciences Faculty of Health Sciences University of Copenhagen, Frederiksberg C, DK Denmark
| | - Alisa Junaković
- Department of Neuroscience Croatian Institute for Brain Research University of Zagreb Medical School, Zagreb, HR Croatia
| | - Milan Radoš
- Department of Neuroscience Croatian Institute for Brain Research University of Zagreb Medical School, Zagreb, HR Croatia
| | - Damir Mulc
- Psychiatric Hospital Vrapče University of Zagreb Medical School, Zagreb, HR Croatia
| | - Ena Španić
- Department of Neuroscience Croatian Institute for Brain Research University of Zagreb Medical School, Zagreb, HR Croatia
| | - Giulia Quattrocolo
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation Norwegian University of Science and Technology Trondheim NO Norway
| | - Vanessa Jane Hall
- Department of Veterinary and Animal Sciences Faculty of Health Sciences University of Copenhagen, Frederiksberg C, DK Denmark
| | - Laszlo Zaborszky
- Center for Molecular and Behavioral Neuroscience Rutgers, The State University of New Jersey Newark New Jersey USA
| | - Mario Vukšić
- Department of Neuroscience Croatian Institute for Brain Research University of Zagreb Medical School, Zagreb, HR Croatia
| | - Francisco Olucha Bordonau
- Department of Medicine School of Medical Sciences Universitat Jaume I Castellón de la Plana ES Spain
| | - Ivica Kostović
- Department of Neuroscience Croatian Institute for Brain Research University of Zagreb Medical School, Zagreb, HR Croatia
| | - Menno P. Witter
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation Norwegian University of Science and Technology Trondheim NO Norway
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute Icahn School of Medicine at Mount Sinai New York New York USA
| |
Collapse
|
29
|
Latimer CS, Stair JG, Hincks JC, Currey HN, Bird TD, Keene CD, Kraemer BC, Liachko NF. TDP-43 promotes tau accumulation and selective neurotoxicity in bigenic Caenorhabditis elegans. Dis Model Mech 2022; 15:275149. [PMID: 35178571 PMCID: PMC9066518 DOI: 10.1242/dmm.049323] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/11/2022] [Indexed: 11/20/2022] Open
Abstract
Although amyloid β (Aβ) and tau aggregates define the neuropathology of Alzheimer's disease (AD), TDP-43 has recently emerged as a co-morbid pathology in more than half of patients with AD. Individuals with concomitant Aβ, tau and TDP-43 pathology experience accelerated cognitive decline and worsened brain atrophy, but the molecular mechanisms of TDP-43 neurotoxicity in AD are unknown. Synergistic interactions among Aβ, tau and TDP-43 may be responsible for worsened disease outcomes. To study the biology underlying this process, we have developed new models of protein co-morbidity using the simple animal Caenorhabditis elegans. We demonstrate that TDP-43 specifically enhances tau but not Aβ neurotoxicity, resulting in neuronal dysfunction, pathological tau accumulation and selective neurodegeneration. Furthermore, we find that synergism between tau and TDP-43 is rescued by loss-of-function of the robust tau modifier sut-2. Our results implicate enhanced tau neurotoxicity as the primary driver underlying worsened clinical and neuropathological phenotypes in AD with TDP-43 pathology, and identify cell-type specific sensitivities to co-morbid tau and TDP-43. Determining the relationship between co-morbid TDP-43 and tau is crucial to understand, and ultimately treat, mixed pathology AD.
Collapse
Affiliation(s)
- Caitlin S. Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Jade G. Stair
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Joshua C. Hincks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Heather N. Currey
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Thomas D. Bird
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Department of Neurology, University of Washington, Seattle, WA 98104, USA,Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Brian C. Kraemer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA,Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Nicole F. Liachko
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA,Author for correspondence ()
| |
Collapse
|
30
|
Guerrero-Gonzalez J, Surgent O, Adluru N, Kirk GR, Dean III DC, Kecskemeti SR, Alexander AL, Travers BG. Improving Imaging of the Brainstem and Cerebellum in Autistic Children: Transformation-Based High-Resolution Diffusion MRI (TiDi-Fused) in the Human Brainstem. Front Integr Neurosci 2022; 16:804743. [PMID: 35310466 PMCID: PMC8928227 DOI: 10.3389/fnint.2022.804743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Diffusion-weighted magnetic resonance imaging (dMRI) of the brainstem is technically challenging, especially in young autistic children as nearby tissue-air interfaces and motion (voluntary and physiological) can lead to artifacts. This limits the availability of high-resolution images, which are desirable for improving the ability to study brainstem structures. Furthermore, inherently low signal-to-noise ratios, geometric distortions, and sensitivity to motion not related to molecular diffusion have resulted in limited techniques for high-resolution data acquisition compared to other modalities such as T1-weighted imaging. Here, we implement a method for achieving increased apparent spatial resolution in pediatric dMRI that hinges on accurate geometric distortion correction and on high fidelity within subject image registration between dMRI and magnetization prepared rapid acquisition gradient echo (MPnRAGE) images. We call this post-processing pipeline T1 weighted-diffusion fused, or "TiDi-Fused". Data used in this work consists of dMRI data (2.4 mm resolution, corrected using FSL's Topup) and T1-weighted (T1w) MPnRAGE anatomical data (1 mm resolution) acquired from 128 autistic and non-autistic children (ages 6-10 years old). Accurate correction of geometric distortion permitted for a further increase in apparent resolution of the dMRI scan via boundary-based registration to the MPnRAGE T1w. Estimation of fiber orientation distributions and further analyses were carried out in the T1w space. Data processed with the TiDi-Fused method were qualitatively and quantitatively compared to data processed with conventional dMRI processing methods. Results show the advantages of the TiDi-Fused pipeline including sharper brainstem gray-white matter tissue contrast, improved inter-subject spatial alignment for group analyses of dMRI based measures, accurate spatial alignment with histology-based imaging of the brainstem, reduced variability in brainstem-cerebellar white matter tracts, and more robust biologically plausible relationships between age and brainstem-cerebellar white matter tracts. Overall, this work identifies a promising pipeline for achieving high-resolution imaging of brainstem structures in pediatric and clinical populations who may not be able to endure long scan times. This pipeline may serve as a gateway for feasibly elucidating brainstem contributions to autism and other conditions.
Collapse
Affiliation(s)
- Jose Guerrero-Gonzalez
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| | - Olivia Surgent
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Nagesh Adluru
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, United States
| | - Gregory R. Kirk
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Douglas C. Dean III
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Andrew L. Alexander
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| | - Brittany G. Travers
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Occupational Therapy Program in the Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
31
|
Chlamydia pneumoniae can infect the central nervous system via the olfactory and trigeminal nerves and contributes to Alzheimer's disease risk. Sci Rep 2022; 12:2759. [PMID: 35177758 PMCID: PMC8854390 DOI: 10.1038/s41598-022-06749-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/07/2022] [Indexed: 02/07/2023] Open
Abstract
Chlamydia pneumoniae is a respiratory tract pathogen but can also infect the central nervous system (CNS). Recently, the link between C. pneumoniae CNS infection and late-onset dementia has become increasingly evident. In mice, CNS infection has been shown to occur weeks to months after intranasal inoculation. By isolating live C. pneumoniae from tissues and using immunohistochemistry, we show that C. pneumoniae can infect the olfactory and trigeminal nerves, olfactory bulb and brain within 72 h in mice. C. pneumoniae infection also resulted in dysregulation of key pathways involved in Alzheimer’s disease pathogenesis at 7 and 28 days after inoculation. Interestingly, amyloid beta accumulations were also detected adjacent to the C. pneumoniae inclusions in the olfactory system. Furthermore, injury to the nasal epithelium resulted in increased peripheral nerve and olfactory bulb infection, but did not alter general CNS infection. In vitro, C. pneumoniae was able to infect peripheral nerve and CNS glia. In summary, the nerves extending between the nasal cavity and the brain constitute invasion paths by which C. pneumoniae can rapidly invade the CNS likely by surviving in glia and leading to Aβ deposition.
Collapse
|
32
|
Monoranu CM, Hartmann T, Strobel S, Heinsen H, Riederer P, Distel L, Bohnert S. Is There Any Evidence of Monocytes Involvement in Alzheimer's Disease? A Pilot Study on Human Postmortem Brain. J Alzheimers Dis Rep 2022; 5:887-897. [PMID: 35088038 PMCID: PMC8764630 DOI: 10.3233/adr-210052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/23/2021] [Indexed: 12/19/2022] Open
Abstract
Background The role of neuroinflammation has become more evident in the pathogenesis of neurodegenerative diseases. Increased expression of microglial markers is widely reported in Alzheimer's disease (AD), but much less is known about the role of monocytes in AD pathogenesis. In AD animal models, bone marrow-derived monocytes appear to infiltrate the parenchyma and contribute to the phagocytosis of amyloid-β depositions, but this infiltration has not been established in systematic studies of the human brain postmortem. Objective In addition to assessing the distribution of different subtypes of microglia by immunostaining for CD68, HLA-DR, CD163, and CD206, we focused on the involvement of C-chemokine receptor type2 (CCR2) positive monocytes during the AD course. Methods We used formalin-fixed and paraffin-embedded tissue from four vulnerable brain regions (hippocampus, occipital lobe, brainstem, and cerebellum) from neuropathologically characterized AD cases at different Braak stages and age-matched controls. Results Only singular migrated CCR2-positive cells were found in all brain regions and stages. The brainstem showed the highest number of positive cells overall, followed by the hippocampus. This mechanism of recruitment seems to work less efficiently in the human brain at an advanced age, and the ingress of monocytes obviously takes place in much reduced numbers or not at all. Conclusion In contrast to studies on animal models, we observed only a quite low level of myeloid monocytes associated with AD pathology. Furthermore, we provide evidence associating early microglial reactions carried out in particular by pro-inflammatory cells with early effects on tangle- and plaque-positive vulnerable brain regions.
Collapse
Affiliation(s)
- Camelia-Maria Monoranu
- Institute of Pathology, Department of Neuropathology, Julius-Maximilian-University of Wuerzburg, Wuerzburg, Germany
| | - Tim Hartmann
- Institute of Pathology, Department of Neuropathology, Julius-Maximilian-University of Wuerzburg, Wuerzburg, Germany
| | - Sabrina Strobel
- Institute of Pathology, Julius-Maximilian-University of Wuerzburg, Wuerzburg, Germany
| | - Helmut Heinsen
- Department of Psychiatry, Morphological Brain Research Unit, University of Wuerzburg, Wuerzburg, Germany.,Department of Pathology, University of São Paulo, São Paulo, Brazil
| | - Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, University of Wuerzburg, Wuerzburg, Germany.,Department of Psychiatry, University of South Denmark, Odense, Denmark
| | - Luitpold Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Bohnert
- Institute of Forensic Medicine, Julius-Maximilian-University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
33
|
Yu ZW, Wang Y, Li X, Fu YH, Yuan Y, Li HY, Gao XY. Brainstem auditory evoked potential in cognitive impairment in patients with type 2 diabetes mellitus. Exp Gerontol 2022; 159:111684. [PMID: 34995724 DOI: 10.1016/j.exger.2021.111684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 12/12/2021] [Accepted: 12/25/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Type 2 diabetes mellitus (T2DM) can cause mild cognitive impairment (MCI) which threatens the health of patients. So the diagnosis of MCI is particularly important. It is reported that brainstem auditory evoked potential (BAEP) is a sensitive tool to detect the brainstem function in patients with T2DM. This study aimed to investigate the relationship between BAEP and MCI in patients with T2DM. METHODS A total of 244 T2DM patients with normal hearing, including 117 normal cognition patients and 127 MCI patients, were recruited in this cross-sectional study. Each subject underwent the BAEP examination. The diagnosis of MCI was based on the diagnostic guideline developed by the National Institute on Aging-Alzheimer's Association workgroups. The Montreal Cognitive Assessment (MoCA) was used to assess the cognitive function of the subjects. RESULTS Compared with the normal cognition group, the patients in the MCI group had longer latencies of waves III and V and interpeak latencies (IPL) I-V in both ears (P < 0.05). The significant negative correlations were found between the latencies of waves III, V, IPL I-V, and MoCA score in both ears (P < 0.05). Logistic regression showed that the prolongations of latunits of waves III and V and IPL I-V in both ears were still associated with MCI after adjustment for mixed factors (P < 0.05). CONCLUSION These results indicate abnormal auditory pathway in brainstem of T2DM patients with MCI. BAEP may contribute to the clinical diagnosis of MCI in patients with T2DM.
Collapse
Affiliation(s)
- Zi-Wei Yu
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ying Wang
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xin Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yu-Hong Fu
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yue Yuan
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hui-Yao Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xin-Yuan Gao
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
34
|
Homolak J, Babic Perhoc A, Knezovic A, Osmanovic Barilar J, Koc F, Stanton C, Ross RP, Salkovic-Petrisic M. Disbalance of the Duodenal Epithelial Cell Turnover and Apoptosis Accompanies Insensitivity of Intestinal Redox Homeostasis to Inhibition of the Brain Glucose-Dependent Insulinotropic Polypeptide Receptors in a Rat Model of Sporadic Alzheimer's Disease. Neuroendocrinology 2022; 112:744-762. [PMID: 34607331 DOI: 10.1159/000519988] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/29/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Gastrointestinal dyshomeostasis is investigated in the context of metabolic dysfunction, systemic, and neuroinflammation in Alzheimer's disease. Dysfunctional gastrointestinal redox homeostasis and the brain-gut incretin axis have been reported in the rat model of insulin-resistant brain state-driven neurodegeneration induced by intracerebroventricular streptozotocin (STZ-icv). We aimed to assess whether (i) the structural epithelial changes accompany duodenal oxidative stress; (ii) the brain glucose-dependent insulinotropic polypeptide receptor (GIP-R) regulates redox homeostasis of the duodenum; and (iii) the STZ-icv brain-gut axis is resistant to pharmacological inhibition of the brain GIP-R. METHODS GIP-R inhibitor [Pro3]-GIP (85 μg/kg) was administered intracerebroventricularly to the control and the STZ-icv rats 1 month after model induction. Thiobarbituric acid reactive substances (TBARSs) were measured in the plasma and duodenum, and the sections were analyzed morphometrically. Caspase-3 expression and activation were assessed by Western blot and multiplex fluorescent signal amplification. RESULTS Intracerebroventricular [Pro3]-GIP decreased plasma TBARSs in the control and STZ-icv animals and increased duodenal TBARSs in the controls. In the controls, inhibition of brain GIP-R affected duodenal epithelial cells, but not villus structure, while all morphometric parameters were altered in the STZ-icv-treated animals. Morphometric changes in the STZ-icv animals were accompanied by reduced levels of caspase-3. Suppression of brain GIP-R inhibited duodenal caspase-3 activation. CONCLUSION Brain GIP-R seems to be involved in the regulation of duodenal redox homeostasis and epithelial cell turnover. Resistance of the brain-gut GIP axis and morphological changes indicative of abnormal epithelial cell turnover accompany duodenal oxidative stress in the STZ-icv rats.
Collapse
Affiliation(s)
- Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ana Babic Perhoc
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Jelena Osmanovic Barilar
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Fatma Koc
- APC Microbiome Ireland, Cork, Ireland
- Teagasc Food Research Centre, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Cork, Ireland
- Teagasc Food Research Centre, Cork, Ireland
| | | | - Melita Salkovic-Petrisic
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
35
|
Jellinger KA. Recent update on the heterogeneity of the Alzheimer’s disease spectrum. J Neural Transm (Vienna) 2021; 129:1-24. [DOI: 10.1007/s00702-021-02449-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/25/2021] [Indexed: 02/03/2023]
|
36
|
von Linstow CU, Waider J, Bergh MSS, Anzalone M, Madsen C, Nicolau AB, Wirenfeldt M, Lesch KP, Finsen B. The Combined Effects of Amyloidosis and Serotonin Deficiency by Tryptophan Hydroxylase-2 Knockout Impacts Viability of the APP/PS1 Mouse Model of Alzheimer’s Disease. J Alzheimers Dis 2021; 85:1283-1300. [DOI: 10.3233/jad-210581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: A decline of brain serotonin (5-HT) is held responsible for the changes in mood that can be observed in Alzheimer’s disease (AD). However, 5-HT’ergic signaling is also suggested to reduce the production of pathogenic amyloid-4β (Aβ). Objective: To investigate the effect of targeted inactivation of tryptophan hydroxylase-2 (Tph2), which is essential for neuronal 5-HT synthesis, on amyloidosis in amyloid precursor protein (APP)swe/presenilin 1 (PS1) ΔE9 transgenic mice. Methods: Triple-transgenic (3xTg) APP/PS1 mice with partial (+/-) or complete Tph2 knockout (–/–) were allowed to survive until 6 months old with APP/PS1, Tph2–/–, and wildtype mice. Survival and weight were recorded. Levels of Aβ 42/40/38, soluble APPα (sAβPPα) and sAβPPβ, and cytokines were analyzed by mesoscale, neurotransmitters by mass spectrometry, and gene expression by quantitative PCR. Tph2, microglia, and Aβ were visualized histologically. Results: Tph2 inactivation in APP/PS1 mice significantly reduced viability, without impacting soluble and insoluble Aβ 42 and Aβ 40 in neocortex and hippocampus, and with only mild changes of soluble Aβ 42/Aβ 40. However, sAβPPα and sAβPPβ in hippocampus and Aβ 38 and Aβ 40 in cerebrospinal fluid were reduced. 3xTg–/–mice were devoid of Tph2 immunopositive fibers and 5-HT. Cytokines were unaffected by genotype, as were neocortical TNF, HTR2a and HTR2b mRNA levels in Tph2–/– mice. Microglia clustered around Aβ plaques regardless of genotype. Conclusion: The results suggest that Tph2 inactivation influences AβPP processing, at least in the hippocampus, although levels of Aβ are unchanged. The reduced viability of 3xTg–/–mice could indicate that 5-HT protects against the seizures that can impact the viability of APP/PS1 mice.
Collapse
Affiliation(s)
- Christian Ulrich von Linstow
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Würzburg, Germany
| | - Marianne Skov-Skov Bergh
- Section for Drug Abuse Research, Department of Forensic Sciences, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Marco Anzalone
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Cecilie Madsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Aina Battle Nicolau
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Martin Wirenfeldt
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
- Department of Pathology, Institute of Clinical Science, Odense University Hospital, Denmark
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
37
|
Ali AA, Khalil MG, Abd El-Latif DM, Okda T, Abdelaziz AI, Abu-Elfotuh K, Kamal MM, Wahid A. The influence of vinpocetine alone or in combination with Epigallocatechin-3-gallate, Coenzyme COQ10, Vitamin E and Selenium as a potential neuroprotective combination against aluminium-induced Alzheimer's disease in Wistar Albino Rats. Arch Gerontol Geriatr 2021; 98:104557. [PMID: 34706318 DOI: 10.1016/j.archger.2021.104557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/09/2021] [Accepted: 10/10/2021] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is one of such diseases that represent the most prominent cause of dementia in elderly people. To explore the possible neuroprotective effect as well as mechanism of action of Vinpocetine either alone or in combination with EGCG, CoQ10, or VE & Se in ameliorating aluminum chloride-induced AD in rats. Rats were received AlCl3 (70 mg/kg) intraperitoneal daily dose for 30 days along with EGCG (10 mg/kg, I.P), CoQ10 (200 mg/kg, P.O), VE (100 mg/kg, P.O) & Se (1 mg/kg, P.O) as well as Vinpocetine (20 mg/kg, P.O) either alone or in combination. Results revealed that the combination of Vinpocetine with EGCG showed the best neuroprotection. This protection in the brain was indicated by the significant decrease in Aβ and ACHE. The same pattern of results were shown in the levels of monoamines and BDNF. In addition, the combination of Vinpocetine with EGCG showed more pronounced anti-inflammatory (TNF-α, IL-1β) and antioxidant (MDA, SOD, TAC) effects in comparison to other combinations. These results were confirmed using histopathological examinations as well as DNA fragmentation assays. Vinpocetine with EGCG showed pronounced protection on neurons against AD induced by AlCl3 in rats.
Collapse
Affiliation(s)
- Azza A Ali
- Department of Pharmacology and Toxicology; Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mona G Khalil
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Doaa M Abd El-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Tarek Okda
- Department of Biochemistry, Faculty of pharmacy, Damanhour University, Egypt
| | - Aya I Abdelaziz
- Medical Research Center, Faculty of pharmacy, Heliopolis University, Egypt
| | - Karema Abu-Elfotuh
- Department of Pharmacology and Toxicology; Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mona M Kamal
- Department of Pharmacology and Toxicology; Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ahmed Wahid
- Department of Pharmaceutical Biochemistry, Faculty of pharmacy, Alexandria University, Egypt.
| |
Collapse
|
38
|
Anti-Oxidative, Anti-Inflammatory and Anti-Apoptotic Effects of Flavonols: Targeting Nrf2, NF-κB and p53 Pathways in Neurodegeneration. Antioxidants (Basel) 2021; 10:antiox10101628. [PMID: 34679762 PMCID: PMC8533072 DOI: 10.3390/antiox10101628] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Neurodegenerative diseases are one of the leading causes of disability and death worldwide. Intracellular transduction pathways that end in the activation of specific transcription factors are highly implicated in the onset and progression of pathological changes related to neurodegeneration, of which those related to oxidative stress (OS) and neuroinflammation are particularly important. Here, we provide a brief overview of the key concepts related to OS- and neuroinflammation-mediated neuropathological changes in neurodegeneration, together with the role of transcription factors nuclear factor erythroid 2-related factor 2 (Nrf2) and nuclear factor-κB (NF-κB). This review is focused on the transcription factor p53 that coordinates the cellular response to diverse genotoxic stimuli, determining neuronal death or survival. As current pharmacological options in the treatment of neurodegenerative disease are only symptomatic, many research efforts are aimed at uncovering efficient disease-modifying agents. Natural polyphenolic compounds demonstrate powerful anti-oxidative, anti-inflammatory and anti-apoptotic effects, partially acting as modulators of signaling pathways. Herein, we review the current understanding of the therapeutic potential and limitations of flavonols in neuroprotection, with emphasis on their anti-oxidative, anti-inflammatory and anti-apoptotic effects along the Nrf2, NF-κB and p53 pathways. A better understanding of cellular and molecular mechanisms of their action may pave the way toward new treatments.
Collapse
|
39
|
Llano DA, Kwok SS, Devanarayan V. Reported Hearing Loss in Alzheimer's Disease Is Associated With Loss of Brainstem and Cerebellar Volume. Front Hum Neurosci 2021; 15:739754. [PMID: 34630060 PMCID: PMC8498578 DOI: 10.3389/fnhum.2021.739754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/03/2021] [Indexed: 11/13/2022] Open
Abstract
Multiple epidemiological studies have revealed an association between presbycusis and Alzheimer’s Disease (AD). Unfortunately, the neurobiological underpinnings of this relationship are not clear. It is possible that the two disorders share a common, as yet unidentified, risk factor, or that hearing loss may independently accelerate AD pathology. Here, we examined the relationship between reported hearing loss and brain volumes in normal, mild cognitive impairment (MCI) and AD subjects using a publicly available database. We found that among subjects with AD, individuals that reported hearing loss had smaller brainstem and cerebellar volumes in both hemispheres than individuals without hearing loss. In addition, we found that these brain volumes diminish in size more rapidly among normal subjects with reported hearing loss and that there was a significant interaction between cognitive diagnosis and the relationship between reported hearing loss and these brain volumes. These data suggest that hearing loss is linked to brainstem and cerebellar pathology, but only in the context of the pathological state of AD. We hypothesize that the presence of AD-related pathology in both the brainstem and cerebellum creates vulnerabilities in these brain regions to auditory deafferentation-related atrophy. These data have implications for our understanding of the potential neural substrates for interactions between hearing loss and AD.
Collapse
Affiliation(s)
- Daniel A Llano
- Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Carle Neuroscience Institute, Urbana, IL, United States.,Carle Illinois College of Medicine, Urbana, IL, United States.,Beckman Institute for Advanced Science and Technology, Urbana, IL, United States
| | - Susanna S Kwok
- Carle Illinois College of Medicine, Urbana, IL, United States
| | - Viswanath Devanarayan
- Eisai Inc., Woodcliff Lake, NJ, United States.,Department of Mathematics, Statistics and Computer Science, University of Illinois at Chicago, Chicago, IL, United States
| | | |
Collapse
|
40
|
Plini ERG, O’Hanlon E, Boyle R, Sibilia F, Rikhye G, Kenney J, Whelan R, Melnychuk MC, Robertson IH, Dockree PM. Examining the Role of the Noradrenergic Locus Coeruleus for Predicting Attention and Brain Maintenance in Healthy Old Age and Disease: An MRI Structural Study for the Alzheimer's Disease Neuroimaging Initiative. Cells 2021; 10:1829. [PMID: 34359997 PMCID: PMC8306442 DOI: 10.3390/cells10071829] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/08/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
The noradrenergic theory of Cognitive Reserve (Robertson, 2013-2014) postulates that the upregulation of the locus coeruleus-noradrenergic system (LC-NA) originating in the brainstem might facilitate cortical networks involved in attention, and protracted activation of this system throughout the lifespan may enhance cognitive stimulation contributing to reserve. To test the above-mentioned theory, a study was conducted on a sample of 686 participants (395 controls, 156 mild cognitive impairment, 135 Alzheimer's disease) investigating the relationship between LC volume, attentional performance and a biological index of brain maintenance (BrainPAD-an objective measure, which compares an individual's structural brain health, reflected by their voxel-wise grey matter density, to the state typically expected at that individual's age). Further analyses were carried out on reserve indices including education and occupational attainment. Volumetric variation across groups was also explored along with gender differences. Control analyses on the serotoninergic (5-HT), dopaminergic (DA) and cholinergic (Ach) systems were contrasted with the noradrenergic (NA) hypothesis. The antithetic relationships were also tested across the neuromodulatory subcortical systems. Results supported by Bayesian modelling showed that LC volume disproportionately predicted higher attentional performance as well as biological brain maintenance across the three groups. These findings lend support to the role of the noradrenergic system as a key mediator underpinning the neuropsychology of reserve, and they suggest that early prevention strategies focused on the noradrenergic system (e.g., cognitive-attentive training, physical exercise, pharmacological and dietary interventions) may yield important clinical benefits to mitigate cognitive impairment with age and disease.
Collapse
Affiliation(s)
- Emanuele R. G. Plini
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Erik O’Hanlon
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
- Department of Psychiatry, Royal College of Surgeons in Ireland, Hospital Rd, Beaumont, 9QRH+4F Dublin, Ireland
- Department of Psychiatry, School of Medicine Dublin, Trinity College Dublin, 152-160 Pearse St, 8QV3+99 Dublin, Ireland;
| | - Rory Boyle
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Francesca Sibilia
- Department of Psychiatry, School of Medicine Dublin, Trinity College Dublin, 152-160 Pearse St, 8QV3+99 Dublin, Ireland;
| | - Gaia Rikhye
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Joanne Kenney
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Robert Whelan
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland;
| | - Michael C. Melnychuk
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Ian H. Robertson
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland;
| | - Paul M. Dockree
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| |
Collapse
|
41
|
Gallo A, Pillet LE, Verpillot R. New frontiers in Alzheimer's disease diagnostic: Monoamines and their derivatives in biological fluids. Exp Gerontol 2021; 152:111452. [PMID: 34182050 DOI: 10.1016/j.exger.2021.111452] [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] [Received: 11/10/2020] [Revised: 04/29/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
Current diagnosis of Alzheimer's disease (AD) relies on a combination of neuropsychological evaluations, biomarker measurements and brain imaging. Nevertheless, these approaches are either expensive, invasive or lack sensitivity to early AD stages. The main challenge of ongoing research is therefore to identify early non-invasive biomarkers to diagnose AD at preclinical stage. Accumulating evidence support the hypothesis that initial degeneration of profound monoaminergic nuclei may trigger a transneuronal spread of AD pathology towards hippocampus and cortex. These studies aroused great interest on monoamines, i.e. noradrenaline (NA), dopamine (D) ad serotonin (5-HT), as early hallmarks of AD pathology. The present work reviews current literature on the potential role of monoamines and related metabolites as biomarkers of AD. First, morphological changes in the monoaminergic systems during AD are briefly described. Second, we focus on concentration changes of these molecules and their derivatives in biological fluids, including cerebrospinal fluid, obtained by lumbar puncture, and blood or urine, sampled via less invasive procedures. Starting from initial observations, we then discuss recent insights on metabolomics-based analysis, highlighting the promising clinical utility of monoamines for the identification of a molecular AD signature, aimed at improving early diagnosis and discrimination from other dementia.
Collapse
|
42
|
Bouhrara M, Cortina LE, Khattar N, Rejimon AC, Ajamu S, Cezayirli DS, Spencer RG. Maturation and degeneration of the human brainstem across the adult lifespan. Aging (Albany NY) 2021; 13:14862-14891. [PMID: 34115614 PMCID: PMC8221341 DOI: 10.18632/aging.203183] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/20/2021] [Indexed: 04/12/2023]
Abstract
Brainstem tissue microstructural properties change across the adult lifespan. However, studies elucidating the biological processes that govern brainstem maturation and degeneration in-vivo are lacking. In the present work, conducted on a large cohort of 140 cognitively unimpaired subjects spanning a wide age range of 21 to 94 years, we implemented a multi-parameter approach to characterize the sex- and age differences. In addition, we examined regional correlations between myelin water fraction (MWF), a direct measure of myelin content, and diffusion tensor imaging indices, and transverse and longitudinal relaxation rates to evaluate whether these metrics provide information complementary to MWF. We observed region-dependent differences in myelin content and axonal density with age and found that both exhibit an inverted U-shape association with age in several brainstem substructures. We emphasize that the microstructural differences captured by our distinct MRI metrics, along with their weak associations with MWF, strongly indicate the potential of using these outcome measures in a multi-parametric approach. Furthermore, our results support the gain-predicts-loss hypothesis of tissue maturation and degeneration in the brainstem. Indeed, our results indicate that myelination follows a temporally symmetric time course across the adult life span, while axons appear to degenerate significantly more rapidly than they mature.
Collapse
Affiliation(s)
- Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luis E. Cortina
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Nikkita Khattar
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Abinand C. Rejimon
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Samuel Ajamu
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Defne S. Cezayirli
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Richard G. Spencer
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
43
|
Jové M, Mota-Martorell N, Torres P, Ayala V, Portero-Otin M, Ferrer I, Pamplona R. The Causal Role of Lipoxidative Damage in Mitochondrial Bioenergetic Dysfunction Linked to Alzheimer's Disease Pathology. Life (Basel) 2021; 11:life11050388. [PMID: 33923074 PMCID: PMC8147054 DOI: 10.3390/life11050388] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 01/18/2023] Open
Abstract
Current shreds of evidence point to the entorhinal cortex (EC) as the origin of the Alzheimer’s disease (AD) pathology in the cerebrum. Compared with other cortical areas, the neurons from this brain region possess an inherent selective vulnerability derived from particular oxidative stress conditions that favor increased mitochondrial molecular damage with early bioenergetic involvement. This alteration of energy metabolism is the starting point for subsequent changes in a multitude of cell mechanisms, leading to neuronal dysfunction and, ultimately, cell death. These events are induced by changes that come with age, creating the substrate for the alteration of several neuronal pathways that will evolve toward neurodegeneration and, consequently, the development of AD pathology. In this context, the present review will focus on description of the biological mechanisms that confer vulnerability specifically to neurons of the entorhinal cortex, the changes induced by the aging process in this brain region, and the alterations at the mitochondrial level as the earliest mechanism for the development of AD pathology. Current findings allow us to propose the existence of an altered allostatic mechanism at the entorhinal cortex whose core is made up of mitochondrial oxidative stress, lipid metabolism, and energy production, and which, in a positive loop, evolves to neurodegeneration, laying the basis for the onset and progression of AD pathology.
Collapse
Affiliation(s)
- Mariona Jové
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Natàlia Mota-Martorell
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Pascual Torres
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Victoria Ayala
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Manuel Portero-Otin
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Bellvitge University Hospital/Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain
- Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), ISCIII, 28220 Madrid, Spain
- Correspondence: (I.F.); (R.P.)
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
- Correspondence: (I.F.); (R.P.)
| |
Collapse
|
44
|
Babić Leko M, Nikolac Perković M, Klepac N, Švob Štrac D, Borovečki F, Pivac N, Hof PR, Šimić G. Relationships of Cerebrospinal Fluid Alzheimer's Disease Biomarkers and COMT, DBH, and MAOB Single Nucleotide Polymorphisms. J Alzheimers Dis 2021; 73:135-145. [PMID: 31771069 PMCID: PMC7029364 DOI: 10.3233/jad-190991] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The noradrenergic and dopaminergic systems are affected in Alzheimer’s disease (AD). Polymorphisms in genes encoding enzymes and proteins that are components of these systems can affect products of transcription and translation and lead to altered enzymatic activity and alterations in overall dopamine and noradrenaline levels. Catechol-O-methyltransferase (COMT) and monoamine oxidase B (MAOB) are the enzymes that regulate degradation of dopamine, while dopamine β-hydroxylase (DBH) is involved in synthesis of noradrenaline. COMT Val158Met (rs4680), DBH rs1611115 (also called –1021C/T or –970C/T), and MAOB rs1799836 (also called A644G) polymorphisms have been previously associated with AD. We assessed whether these polymorphisms are associated with cerebrospinal fluid (CSF) AD biomarkers including total tau (t-tau), phosphorylated tau proteins (p-tau181, p-tau199, and p-tau231), amyloid-β42 (Aβ42), and visinin-like protein 1 (VILIP-1) to test possible relationships of specific genotypes and pathological levels of CSF AD biomarkers. The study included 233 subjects: 115 AD, 53 mild cognitive impairment, 54 subjects with other primary causes of dementia, and 11 healthy controls. Significant decrease in Aβ42 levels was found in patients with GG compared to AG COMT Val158Met genotype, while t-tau and p-tau181 levels were increased in patients with AA compared to AG COMT Val158Met genotype. Aβ42 levels were also decreased in carriers of A allele in MAO-B rs1799836 polymorphism, while p-tau181 levels were increased in carriers of T allele in DBH rs1611115 polymorphism. These results indicate that COMT Val158Met, DBH rs1611115, and MAOB rs1799836 polymorphisms deserve further investigation as genetic markers of AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | | | - Nataša Klepac
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nela Pivac
- Department of Molecular Medicine, Institute Ruđer Bošković, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
45
|
Patthy Á, Murai J, Hanics J, Pintér A, Zahola P, Hökfelt TGM, Harkany T, Alpár A. Neuropathology of the Brainstem to Mechanistically Understand and to Treat Alzheimer's Disease. J Clin Med 2021; 10:jcm10081555. [PMID: 33917176 PMCID: PMC8067882 DOI: 10.3390/jcm10081555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder as yet without effective therapy. Symptoms of this disorder typically reflect cortical malfunction with local neurohistopathology, which biased investigators to search for focal triggers and molecular mechanisms. Cortex, however, receives massive afferents from caudal brain structures, which do not only convey specific information but powerfully tune ensemble activity. Moreover, there is evidence that the start of AD is subcortical. The brainstem harbors monoamine systems, which establish a dense innervation in both allo- and neocortex. Monoaminergic synapses can co-release neuropeptides either by precisely terminating on cortical neurons or, when being “en passant”, can instigate local volume transmission. Especially due to its early damage, malfunction of the ascending monoaminergic system emerges as an early sign and possible trigger of AD. This review summarizes the involvement and cascaded impairment of brainstem monoaminergic neurons in AD and discusses cellular mechanisms that lead to their dysfunction. We highlight the significance and therapeutic challenges of transmitter co-release in ascending activating system, describe the role and changes of local connections and distant afferents of brainstem nuclei in AD, and summon the rapidly increasing diagnostic window during the last few years.
Collapse
Affiliation(s)
- Ágoston Patthy
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Murai
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Hanics
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
| | - Anna Pintér
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Péter Zahola
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Tomas G. M. Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
| | - Tibor Harkany
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
- Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, 1090 Vienna, Austria
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
- Correspondence:
| |
Collapse
|
46
|
Steinbusch HWM, Dolatkhah MA, Hopkins DA. Anatomical and neurochemical organization of the serotonergic system in the mammalian brain and in particular the involvement of the dorsal raphe nucleus in relation to neurological diseases. PROGRESS IN BRAIN RESEARCH 2021; 261:41-81. [PMID: 33785137 DOI: 10.1016/bs.pbr.2021.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The brainstem is a neglected brain area in neurodegenerative diseases, including Alzheimer's and Parkinson's disease, frontotemporal lobar degeneration and autonomic dysfunction. In Depression, several observations have been made in relation to changes in one particular the Dorsal Raphe Nucleus (DRN) which also points toward as key area in various age-related and neurodevelopmental diseases. The DRN is further thought to be related to stress regulated processes and cognitive events. It is involved in neurodegeneration, e.g., amyloid plaques, neurofibrillary tangles, and impaired synaptic transmission in Alzheimer's disease as shown in our autopsy findings. The DRN is a phylogenetically old brain area, with projections that reach out to a large number of regions and nuclei of the central nervous system, particularly in the forebrain. These ascending projections contain multiple neurotransmitters. One of the main reasons for the past and current interest in the DRN is its involvement in depression, and its main transmitter serotonin. The DRN also points toward the increased importance and focus of the brainstem as key area in various age-related and neurodevelopmental diseases. This review describes the morphology, ascending projections and the complex neurotransmitter nature of the DRN, stressing its role as a key research target into the neural bases of depression.
Collapse
Affiliation(s)
- Harry W M Steinbusch
- Department of Cellular Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands; Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology-DGIST, Daegu, South Korea.
| | | | - David A Hopkins
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
47
|
Babić Leko M, Hof PR, Šimić G. Alterations and interactions of subcortical modulatory systems in Alzheimer's disease. PROGRESS IN BRAIN RESEARCH 2021; 261:379-421. [PMID: 33785136 DOI: 10.1016/bs.pbr.2020.07.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) is not fully understood. Here we summarize current knowledge on the involvement of the serotonergic, noradrenergic, dopaminergic, cholinergic, and opioid systems in AD, emphasizing the importance of interactions between the serotonergic and the other subcortical modulatory systems during the progression of AD. In physiological conditions, all neurotransmitter systems function in concert and are interdependent at both the neuroanatomical and molecular levels. Through their early involvement in AD, cognitive and behavioral abilities that rely on their interactions also become disrupted. Considering that serotonin (5HT) regulates the release of noradrenaline (NA), dopamine (DA) and acetylcholine (ACh), any alteration in 5HT levels leads to disturbance of NA, DA, and ACh homeostasis in the brain. One of the earliest pathological changes during the prodromal phase of AD is a decrease of serotonergic transmission throughout the brain, with serotonergic receptors being also affected. Additionally, serotonergic and noradrenergic as well as serotonergic and dopaminergic nuclei are reciprocally interconnected. As the serotonergic dorsal raphe nucleus (DRN) is affected by pathological changes early in AD, and the noradrenergic locus coeruleus (LC) and dopaminergic ventral tegmental area (VTA) exhibit AD-related pathological changes, their connectivity also becomes altered in AD. Such disrupted interactions among neurotransmitter systems in AD can be used in the development of multi-target drugs. Some of the potential AD therapeutics (such as ASS234, RS67333, tropisetron) target multiple neurotransmitter systems to achieve the best possible improvement of cognitive and behavioral deficits observed in AD. Here, we review how serotonergic system interacts with other subcortical modulatory systems (noradrenergic, dopaminergic, cholinergic, and opioid systems) during AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia.
| |
Collapse
|
48
|
Toljan K, Homolak J. Circadian changes in Alzheimer's disease: Neurobiology, clinical problems, and therapeutic opportunities. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:285-300. [PMID: 34225969 DOI: 10.1016/b978-0-12-819975-6.00018-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The understanding of Alzheimer's disease (AD) pathophysiology is an active area of research, and the traditional focus on hippocampus, amyloid and tau protein, and memory impairment has been expanded with components like neuroinflammation, insulin resistance, and circadian rhythm alterations. The bidirectional vicious cycle of neuroinflammation and neurodegeneration on a molecular level may cause functional deficits already long before the appearance of overt clinical symptoms. Located at the crossroads of metabolic, circadian, and hormonal signaling, the hypothalamus has been identified as another brain region affected by AD pathophysiology. Current findings on hypothalamic dysfunction open a broader horizon for studying AD pathogenesis and offer new opportunities for diagnosis and therapy. While treatments with cholinomimetics and memantine form a first line of pharmacological treatment, additional innovative research is pursued toward the development of antiinflammatory, growth factor, or antidiabetic types of medication. Following recent epidemiological data showing associations of AD incidence with modern societal and "life-style"-related risk factors, also nonpharmacological interventions, including sleep optimization, are being developed and some have been shown to be beneficial. Circadian aspects in AD are relevant from a pathophysiological standpoint, but they can also have an important role in pharmacologic and nonpharmacologic interventions, and appropriate timing of sleep, meals, and medication may boost therapeutic efficacy.
Collapse
Affiliation(s)
- Karlo Toljan
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States.
| | - Jan Homolak
- Department of Pharmacology, and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
49
|
Yu CC, He C, Du YJ, Gao S, Lin YF, Wang SQ, Wang L, Wang J, Wang XS, Jiang T, Kong LH. Preventive electroacupuncture reduces cognitive deficits in a rat model of D-galactose-induced aging. Neural Regen Res 2021; 16:916-923. [PMID: 33229729 PMCID: PMC8178792 DOI: 10.4103/1673-5374.297090] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Acupuncture can reduce cognitive deficits in Alzheimer’s disease. However, whether electroacupuncture can prevent or alleviate the cognitive deficits in animal models of aging remains poorly understood. Studies have shown that disordered epigenetic modifications play a critical role in age-related cognitive decline. Therefore, we hypothesized that preventive electroacupuncture might improve cognitive functions during aging by regulating epigenetic modifications. A rat model of aging was produced by intraperitoneal injection of 120 mg/kg D-galactose for 8 weeks. Baihui and Shenshu acupoints were stimulated by electroacupuncture for 8 weeks from the first day of D-galactose administration. Preventive electroacupuncture alleviated memory impairment, decreased tau hyperphosphorylation, and reduced glycogen synthase kinase-3β protein and mRNA expression levels in the brainstem dorsal raphe nucleus, where intracellular neurofibrillary tangle lesions first occur. In addition, the DNA methylation level in the promoter region of the glycogen synthase kinase-3β gene was increased. The effects of preventive electroacupuncture were stronger than those of preventive acupuncture. Intraperitoneal injection of 0.4 mg/kg 5-aza-2′-deoxycytidine, an inhibitor of DNA methyltransferase that blocks epigenetic modifications, antagonized the effects of preventive electroacupuncture. Our results suggest that preventive electroacupuncture treatment alleviates cognitive impairment in aging rats probably by affecting the epigenetic modification of the glycogen synthase kinase-3β gene in the dorsal raphe nucleus. This study was approved by the Animal Ethics Committee of Hubei University of Chinese Medicine, China (approval No. HUCMS201712001) on November 28, 2017.
Collapse
Affiliation(s)
- Chao-Chao Yu
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital; The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Chuan He
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Shan Gao
- Department of Acupuncture & Moxibustion, Wuhan Hospital of Integrated Chinese & Western Medicine, Tongji Medical College of Huazhong University of Science & Technology, Wuhan, Hubei Province, China
| | - Yuan-Fang Lin
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital; The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Shu-Qin Wang
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital; The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Li Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Jia Wang
- Department of Acupuncture & Moxibustion, Wuhan Hospital of Integrated Chinese & Western Medicine, Tongji Medical College of Huazhong University of Science & Technology, Wuhan, Hubei Province, China
| | - Xue-Song Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Tao Jiang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Li-Hong Kong
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| |
Collapse
|
50
|
Zubčić K, Hof PR, Šimić G, Jazvinšćak Jembrek M. The Role of Copper in Tau-Related Pathology in Alzheimer's Disease. Front Mol Neurosci 2020; 13:572308. [PMID: 33071757 PMCID: PMC7533614 DOI: 10.3389/fnmol.2020.572308] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
All tauopathies, including Alzheimer's disease (AD), are characterized by the intracellular accumulation of abnormal forms of tau protein in neurons and glial cells, which negatively affect microtubule stability. Under physiological conditions, tubulin-associated unit (Tau) protein is intrinsically disordered, almost without secondary structure, and is not prone to aggregation. In AD, it assembles, and forms paired helical filaments (PHFs) that further build-up neurofibrillary tangles (NFTs). Aggregates are composed of hyperphosphorylated tau protein that is more prone to aggregation. The pathology of AD is also linked to disturbed copper homeostasis, which promotes oxidative stress (OS). Copper imbalance is widely observed in AD patients. Deregulated copper ions may initiate and exacerbate tau hyperphosphorylation and formation of β-sheet-rich tau fibrils that ultimately contribute to synaptic failure, neuronal death, and cognitive decline observed in AD patients. The present review summarizes factors affecting the process of tau aggregation, conformational changes of small peptide sequences in the microtubule-binding domain required for these motifs to act as seeding sites in aggregation, and the role of copper in OS induction, tau hyperphosphorylation and tau assembly. A better understanding of the various factors that affect tau aggregation under OS conditions may reveal new targets and novel pharmacological approaches for the therapy of AD.
Collapse
Affiliation(s)
- Klara Zubčić
- Laboratory for Developmental Neuropathology, Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Laboratory for Developmental Neuropathology, Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Maja Jazvinšćak Jembrek
- Laboratory for Protein Dynamics, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia.,Department of Psychology, Catholic University of Croatia, Zagreb, Croatia
| |
Collapse
|