1
|
Paitel ER, Pettigrew C, Moghekar A, Miller MI, Faria AV, Albert M, Soldan A. Alzheimer's disease cerebrospinal fluid biomarker levels and APOE genetic status are associated with hippocampal-cerebellar functional connectivity. Neurobiol Aging 2025; 151:107-116. [PMID: 40273528 DOI: 10.1016/j.neurobiolaging.2025.04.005] [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: 01/28/2025] [Revised: 03/20/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Recent research suggests that hippocampal-cerebellar (Hp-CB) functional connectivity may be altered early in the course of Alzheimer's disease (AD), given the early accumulation of AD pathology in the hippocampi and emerging evidence of cerebellar changes in early AD. This study analyzed the role of AD genetic risk (via APOE ε4 carrier status) and cerebrospinal fluid (CSF) biomarkers of AD pathology (ratio of phosphorylated tau (p-tau181) to amyloid beta (Aβ42/Aβ40)) on the relationship between age and functional Hp-CB resting state fMRI connectivity in 161 cognitively unimpaired older adults (M age =67.3; SD =9.0; 37 % APOE ε4 +). In multiple regression analyses with Hp-CB connectivity as the outcome, there were significant interactions between age and APOE ε4 status, and between age and CSF AD biomarkers. Older age was associated with greater Hp-CB connectivity in APOE ε4 non-carriers and participants with less abnormal CSF AD biomarkers. In contrast, Hp-CB connectivity was marginally lower with older age in ε4 carriers and those with more abnormal AD biomarkers. Furthermore, greater Hp-CB connectivity was associated with better episodic memory performance across all groups. These findings suggest that age-related increases in Hp-CB connectivity among APOE ε4 non-carriers and those with low AD biomarker levels reflect age-related changes that are largely unrelated to AD, while age-related decreases in Hp-CB connectivity in APOE ε4 carriers may reflect AD-related alterations. These findings also highlight the importance of cerebellar contributions to cognitive performance among older adults and suggest that Hp-CB connectivity may be altered in preclinical AD.
Collapse
Affiliation(s)
- Elizabeth R Paitel
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael I Miller
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andreia V Faria
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Antonioni A, Raho EM, Spampinato DA, Granieri E, Fadiga L, Di Lorenzo F, Koch G. The cerebellum in frontotemporal dementia: From neglected bystander to potential neuromodulatory target. A narrative review. Neurosci Biobehav Rev 2025; 174:106194. [PMID: 40324708 DOI: 10.1016/j.neubiorev.2025.106194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/29/2024] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Though cortical changes in frontotemporal dementia (FTD) are well-documented, the cerebellum's role, closely linked to these areas, remains unclear. OBJECTIVES To provide evidence on cerebellar involvement in FTD across clinical, genetic, imaging, neuropathological, and neurophysiological perspectives. Additionally, we sought evidence supporting the application of cerebellar non-invasive brain stimulation (NIBS) in FTD for both diagnostic and therapeutic purposes. METHODS We performed a literature review using MEDLINE (via PubMed), Scopus, and Web of Science databases. RESULTS We emphasized the involvement of specific cerebellar regions which differentiate each FTD subtypes and may account for some of the characteristic symptoms. Furthermore, we highlighted peculiarities in FTD genetic alterations. Finally, we outlined neurophysiological evidence supporting a role for the cerebellum in FTD pathogenesis. CONCLUSION The cerebellum is critically involved in the FTD spectrum. Moreover, it can be speculated that cerebellar modulation, as already shown in other neurodegenerative disorders, could restore the interneuronal intracortical circuits typically impaired in FTD patients, providing clinical improvements and fundamental outcome measures in clinical trials.
Collapse
Affiliation(s)
- Annibale Antonioni
- Doctoral Program in Translational Neurosciences and Neurotechnologies, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy.
| | - Emanuela Maria Raho
- University Unit of Neurology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Danny Adrian Spampinato
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, Rome 00179, Italy
| | - Enrico Granieri
- University Unit of Neurology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Luciano Fadiga
- Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, Ferrara 44121, Italy; Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Francesco Di Lorenzo
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, Rome 00179, Italy
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, Rome 00179, Italy; Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, Ferrara 44121, Italy; Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| |
Collapse
|
3
|
Xia J, Chan YH, Girish D, Rajapakse JC. Interpretable modality-specific and interactive graph convolutional network on brain functional and structural connectomes. Med Image Anal 2025; 102:103509. [PMID: 40020422 DOI: 10.1016/j.media.2025.103509] [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: 03/28/2024] [Revised: 01/24/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Both brain functional connectivity (FC) and structural connectivity (SC) provide distinct neural mechanisms for cognition and neurological disease. In addition, interactions between SC and FC within distributed association regions are related to alterations in cognition or neurological diseases, considering the inherent linkage between neural function and structure. However, there is a scarcity of existing learning-based methods that leverage both modality-specific characteristics and high-order interactions between the two modalities for regression or classification. Hence, this study proposes an interpretable modality-specific and interactive graph convolutional network (MS-Inter-GCN) that incorporates modality-specific information, reflecting the unique neural mechanism for each modality, and structure-function interactions, capturing the underlying foundation provided by white-matter fiber tracts for high-level brain function. In MS-Inter-GCN, we generate modality-specific task-relevant embeddings separately from both FC and SC using a graph convolutional encoder-decoder module. Subsequently, we learn the interactive weights between corresponding regions of FC and SC, reflecting the coupling strength, by employing an interactive module on the embeddings of both modalities. A novel graph structure is constructed, which uses modality-specific task-relevant embeddings and inserts the interactive weights as edges connecting corresponding regions of two modalities, and then is used for the regression or classification task. Finally, a post-hoc explainable technology - GNNExplainer- is used to identify salient regions and connections of each modality as well as salient interactions between FC and SC associated with tasks. We apply the proposed framework to fluid cognition prediction, Parkinson's disease (PD), Alzheimer's disease (AD), and schizophrenia (SZ) classification. Experimental results demonstrate that our method outperforms the other ten state-of-the-art methods on multi-modal brain features on all tasks. The GNNExplainer identifies salient structural and functional regions and connections for fluid cognition, PD, AD, and SZ. It confirms that strong structure-function coupling within the executive and control networks, combined with weak coupling within the motor network, is associated with fluid cognition. Moreover, structure-function decoupling in specific brain regions serves as a marker for different diseases: decoupling of the prefrontal, superior parietal, and superior occipital cortices is a marker of PD; decoupling of the middle frontal and lateral parietal cortices, temporal pole, and subcortical regions is indicative of AD; and decoupling of the prefrontal, parietal, and temporal cortices, as well as the cerebellum, contributes to SZ.
Collapse
Affiliation(s)
- Jing Xia
- College of Computing and Data Science, Nanyang Technological University, Singapore
| | - Yi Hao Chan
- College of Computing and Data Science, Nanyang Technological University, Singapore
| | - Deepank Girish
- College of Computing and Data Science, Nanyang Technological University, Singapore
| | - Jagath C Rajapakse
- College of Computing and Data Science, Nanyang Technological University, Singapore.
| |
Collapse
|
4
|
Yue L, Pan Y, Li W, Mao J, Hong B, Gu Z, Liu M, Shen D, Xiao S. Predicting cognitive decline: Deep-learning reveals subtle brain changes in pre-MCI stage. J Prev Alzheimers Dis 2025; 12:100079. [PMID: 39920001 DOI: 10.1016/j.tjpad.2025.100079] [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: 10/31/2024] [Revised: 12/22/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Mild cognitive impairment (MCI) and preclinical MCI (e.g., subjective cognitive decline, SCD) are considered risk states of dementia, such as Alzheimer's Disease (AD). However, it is challenging to accurately predict conversion from normal cognition (NC) to MCI, which is important for early detection and intervention. Since neuropathological changes may have occurred in the brain many years before clinical AD, we sought to detect the subtle brain changes in the pre-MCI stage using a deep-learning method based on structural Magnetic Resonance Imaging (MRI). OBJECTIVES To discover early structural neuroimaging changes that differentiate between stable and progressive cognitive status, and to establish a predictive model for MCI conversion. DESIGN, SETTING AND PARTICIPANTS We first created a unique deep-learning framework for pre-AD conversion prediction through the Alzheimer's Disease Neuroimaging Initiative-1 (ADNI-1) database (n = 845). Then, we tested the model on ADNI-2 (n = 321, followed 3 years) and our private study (n = 109), the China Longitudinal Aging Study (CLAS), to validate the rationality for pre-MCI conversion prediction. The CLAS is a 7-year community-based cohort study in Shanghai. Our framework consisted of two steps: 1) a single-ROI-based network (SRNet) for identifying informative regions in the brain, and 2) a multi-ROI-based network (MRNet) for pre-AD conversion prediction. We then utilized these "ROI-based deep learning" neural networks to create a composite score using advanced algorithm-building. We coined this score as the Progressive Index (PI), which serves as a metric for assessing the propensity of AD conversion. Ultimately, we employed the PI to gauge its predictive capability for MCI conversion in both ADNI-2 and CLAS datasets. MEASUREMENTS We primarily utilized baseline T1-weighted MRI scans to identify the most discriminative brain regions and subsequently developed the PI in both training and validation datasets. We compared the PI across different cognitive groups and conducted logistic regression models along with their AUCs, adjusting for education level, gender, neuropsychological test scores, and the presence of comorbid conditions. RESULTS We trained the SRNet and MRNet using 845 subjects from ADNI-1 with baseline MRI data, in which AD and progressive MCI (converting to AD within 3 years) patients were considered as positive samples, while NC and stable MCI (remaining stable for 3 years) subjects were considered as negative samples. The convolutional neural networks identified the top 10 regions of interest (ROIs) for distinguishing progressive from stable cases. These key brain regions included the hippocampus, amygdala, temporal lobe, insula, and anterior cerebellum. A total of 321 subjects from ADNI-2, including 209 NC (18 progressive NC (pNC), 113 stable NC (sNC), and 78 remaining NC (rNC)) and 112 SCD (11 pSCD, 5 sSCD, and 96 rSCD), as well as 109 subjects from CLAS, including 17 sNC, 16 pNC, 52 sSCD and 24 pSCD participated in the test set, separately. We found that the PI score effectively sorted all subjects by their stages (stable vs progressive). Furthermore, the PI score demonstrated excellent discrimination between the two outcomes in the CLAS data(p<0.001), even after controlling for age, gender, education level, depression symptoms, anxiety symptoms, somatic diseases, and baseline MoCA score. Better performance for prediction progression to MCI in CLAS was obtained when the PI score was combined with clinical measures (AUC=0.812; 95 %CI: 0.725-0.900). CONCLUSIONS This study effectively predicted the progression to MCI among order individuals at normal cognition state by deep learning algorithm with MRI scans. Exploring the key brain alterations during the very early stages, specifically the transition from NC to MCI, based on deep learning methods holds significant potential for further research and contributes to a deeper understanding of disease mechanisms.
Collapse
Affiliation(s)
- Ling Yue
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Yongsheng Pan
- School of Computer Science and Engineering, Northwestern Polytechnical University, 127 West Youyi Road, 710072, Xi'an, China
| | - Wei Li
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Junyan Mao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Bo Hong
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Zhen Gu
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Mingxia Liu
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, 130 Mason Farm Road, Chapel Hill, NC 27599, USA.
| | - Dinggang Shen
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai United Imaging Intelligence Co., Ltd., Shanghai 200232, China; Shanghai Clinical Research and Trial Center, Shanghai, 201210, China.
| | - Shifu Xiao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China.
| |
Collapse
|
5
|
Fan Y, Tian M, Chen Y, Qi X, Zhang Q, Yin K, Shi J, Xiao M. Cerebellar Crus II Regulates Recognition and Spatial Memory in Mice. Mol Neurobiol 2025:10.1007/s12035-025-04852-2. [PMID: 40198447 DOI: 10.1007/s12035-025-04852-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
The cerebellar Crus II is implicated in the late stages of Alzheimer's disease (AD), yet its specific roles in memory regulation and therapeutic potential remain unclear. Using in vivo fiber photometry, we observed robust activation of Crus II neurons in healthy mice during recognition memory tasks. Acute chemogenetic inhibition of Crus II neurons impaired recognition and spatial memory in mice. Polysynaptic circuit tracing revealed that Crus II neurons modulate neural activity in the contralateral prelimbic cortex (PrL) via the Crus II-cerebellar lateral nucleus (LN)-ventromedial thalamus/zona incerta (VM/ZI)-PrL pathway. In 5 × FAD mice, β-amyloid (Aβ) plaque deposition in Crus II exhibited age-dependent progression, occurring later and less severely compared to the prefrontal cortex. Chronic activation of Crus II neurons ameliorated recognition and spatial memory deficits in 5 × FAD mice. These findings highlight the cerebellar Crus II as a modulator of cognitive function and a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Yi Fan
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Minjie Tian
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Yan Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
| | - Xinyang Qi
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Qian Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
| | - Kuiying Yin
- Nanjing Research Institute of Electronic Technology, Nanjing, 210039, China
| | - Jingping Shi
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China.
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
6
|
Magalhães TNC, Maldonado T, Jackson TB, Hicks TH, Herrejon IA, Rezende TJR, Symm AC, Bernard JA. Cerebellar-hippocampal volume associations with behavioral outcomes following tDCS modulation. Brain Imaging Behav 2025; 19:384-394. [PMID: 39904871 DOI: 10.1007/s11682-025-00975-1] [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] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Here, we explore the relationship between transcranial direct current stimulation (tDCS) and brain-behavior interactions. We propose that tDCS perturbation allows for the investigation of relationships between brain volume and behavior. We focused on the hippocampus (HPC) and cerebellum (CB) regions that are implicated in our understanding of memory and motor skill acquisition. Seventy-four young adults (mean age: 22 ± 0.42 years, mean education: 14.7 ± 0.25 years) were randomly assigned to receive either anodal, cathodal, or sham stimulation. Following stimulation, participants completed computerized tasks assessing working memory and sequence learning in a magnetic resonance imaging (MRI) environment. We investigated the statistical interaction between CB and HPC volumes. Our findings showed that individuals with larger cerebellar volumes had shorter reaction times (RT) on a high-load working memory task in the sham stimulation group. In contrast, the anodal stimulation group exhibited faster RTs during the low-load working memory condition. These RT differences were associated with the cortical volumetric interaction between CB-HPC. Literature suggests that anodal stimulation down-regulates the CB and here, those with larger volumes perform more quickly, suggesting the potential need for additional cognitive resources to compensate for cerebellar downregulation or perturbation. This new insight suggests that tDCS can aid in revealing structure-function relationships, due to greater performance variability, especially in young adults. It may also reveal new targets of interest in the study of aging or in diseases where there is also greater behavioral variability.
Collapse
Affiliation(s)
- Thamires N C Magalhães
- Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX, 77840, United States of America.
| | - Ted Maldonado
- Department of Psychology, Indiana State University, Terre Haute, USA
| | | | - Tracey H Hicks
- Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX, 77840, United States of America
| | - Ivan A Herrejon
- Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX, 77840, United States of America
| | - Thiago J R Rezende
- Department of Neurology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Abigail C Symm
- Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX, 77840, United States of America
| | - Jessica A Bernard
- Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX, 77840, United States of America.
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
7
|
Qu H, Liu Y, Connolly JJ, Mentch FD, Kao C, Hakonarson H. Risk of Alzheimer's disease in Down syndrome: Insights gained by multi-omics. Alzheimers Dement 2025; 21:e14604. [PMID: 40207399 PMCID: PMC11982707 DOI: 10.1002/alz.14604] [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: 07/16/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 04/11/2025]
Abstract
Individuals with Down syndrome (DS) are highly susceptible to Alzheimer's disease (AD). The integration of genomics, transcriptomics, epigenomics, proteomics, and metabolomics enables unprecedented understanding of DS-AD, offering a detailed picture of this complex issue. The vast -omics data also present challenges that reflect the complexity of genetic information flow. These studies nonetheless reveal critical mechanisms behind AD risk, including unique observations in DS that differ from those seen in the general population and familial dominant AD. In addition, the correlations between the AD polygenic risk score and proteins related to female infertility and autoimmune thyroiditis corroborate clinical observations. Metabolomic data reveal disrupted metabolic networks, offering prospects for a dynamic score to create specialized nutritional interventions. By adopting a multidimensional perspective with integrated reductionism, the evolving landscape presents an opportunity to identify promising directions for developing precision strategies to mitigate the impact of AD in the DS population. HIGHLIGHTS: Individuals with Down syndrome (DS) are highly susceptible to Alzheimer's disease (AD). DS-AD is characterized by its polygenic nature, extending beyond chromosome 21 with significant contributions from various chromosomes. DS-AD also presents unique features that differ from those observed in the general population and familial dominant AD. Our review consolidates key findings from genomics, transcriptomics, epigenomics, proteomics, and metabolomics, providing a comprehensive view of the molecular mechanisms underlying DS-AD. We highlight promising research directions to further elucidate the pathogenesis of DS-AD.
Collapse
Affiliation(s)
- Hui‐Qi Qu
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Yichuan Liu
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - John J. Connolly
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Frank D. Mentch
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Charlly Kao
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Hakon Hakonarson
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Department of Pediatrics, The Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Division of Pulmonary MedicineChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| |
Collapse
|
8
|
Wang B, LeBel A, D'Mello AM. Ignoring the cerebellum is hindering progress in neuroscience. Trends Cogn Sci 2025; 29:318-330. [PMID: 39934082 DOI: 10.1016/j.tics.2025.01.004] [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: 09/16/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Traditionally considered a motor structure, the cerebellum has been shown to play a key role in several cognitive functions. However, for decades, the cerebellum has been largely overlooked and even deliberately excluded from 'whole-brain' neuroimaging studies. Here, we propose that the continued exclusion of the cerebellum has limited our understanding of whole-brain function. We describe reasons - both warranted and unwarranted - behind its historical exclusion from the neuroimaging literature, review literature describing the importance of the cerebellum and its unique role in brain function, and outline the potential unintended negative consequences of exclusion of the cerebellum for our comprehensive understanding of brain function and clinical disorders.
Collapse
Affiliation(s)
- Bangjie Wang
- Department of Psychology, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Amanda LeBel
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Anila M D'Mello
- Department of Psychology, University of Texas at Dallas, Richardson, TX 75080, USA; Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Peter O'Donnell Jr Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
9
|
Zhang H, Lu J, Zhang L, Hu J, Yue J, Ma Y, Yao Q, Jie P, Fan M, Fang J, Zhao J. Abnormal cerebellar activity and connectivity alterations of the cerebellar-limbic system in post-stroke cognitive impairment: a study based on resting state functional magnetic resonance imaging. Front Neurosci 2025; 19:1543760. [PMID: 40177371 PMCID: PMC11962788 DOI: 10.3389/fnins.2025.1543760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
Background Stroke is an important cause of cognitive impairment. Post-stroke cognitive impairment (PSCI) is a prevalent psychiatric disorder following stroke. However, the effects of PSCI on the cerebellum remain mostly unknown. Methods A total of 31 PSCI patients and 31 patients without cognitive impairment after stroke were included in this study. The Mini-Mental State Examination (MMSE) and the Montreal Cognitive Assessment (MoCA) were administered to all participants. Analyses of ALFF, fALFF, and ReHo were employed to investigate alterations in brain neuronal activity, while limbic connectivity analysis was utilized to reflect changes within the abnormal connections within brain regions. Results We found that ALFF values were increased in Cerebelum_7b_R, Cerebelum_Crus1_L. fALFF values were increased in Vermis_3. The ReHo values were increased in Cerebelum_8_R, Cerebelum_Crus2_R, Cerebelum_Crus1_L. The functional connection between Frontal_Mid_Orb_L and Cerebelum_Crus2_R brain regions was decreased. The functional connection between Hippocampus_L and Cerebelum_Crus2_R brain regions was decreased. The functional connection between Vermis_3 and Frontal_Med_Orb_L brain regions was decreased. Conclusion The severity of cognitive impairment may influence the extent of functional connectivity disruption between the cerebellum and the limbic system. Furthermore, atypical alterations in neuronal activity within cerebellar regions are associated with cognitive decline.
Collapse
Affiliation(s)
- Haiyi Zhang
- Department of Magnetic Resonance Imaging, The Affiliated Traditional Chinese Medicine Hospital, Luzhou, Sichuan, China
| | - Juan Lu
- Department of Magnetic Resonance Imaging, The Affiliated Traditional Chinese Medicine Hospital, Luzhou, Sichuan, China
| | - Lu Zhang
- Department of Acupuncture and Rehabilitation, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jidan Hu
- Department of Radiology, The Second People’s Hospital of Neijiang, Southwest Medical University, Neijiang, Sichuan, China
| | - Jiajun Yue
- Department of Magnetic Resonance Imaging, The Affiliated Traditional Chinese Medicine Hospital, Luzhou, Sichuan, China
| | - Yunhan Ma
- Department of Magnetic Resonance Imaging, The Affiliated Traditional Chinese Medicine Hospital, Luzhou, Sichuan, China
| | - Qi Yao
- Department of Magnetic Resonance Imaging, The Affiliated Traditional Chinese Medicine Hospital, Luzhou, Sichuan, China
| | - Pingping Jie
- Department of Magnetic Resonance Imaging, The Affiliated Traditional Chinese Medicine Hospital, Luzhou, Sichuan, China
| | - Min Fan
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Radiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jiliang Fang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhao
- Department of Magnetic Resonance Imaging, The Affiliated Traditional Chinese Medicine Hospital, Luzhou, Sichuan, China
| |
Collapse
|
10
|
Morell-Ortega S, Ruiz-Perez M, Gadea M, Vivo-Hernando R, Rubio G, Aparici F, Iglesia-Vaya MDL, Catheline G, Mansencal B, Coupé P, Manjón JV. DeepCERES: A deep learning method for cerebellar lobule segmentation using ultra-high resolution multimodal MRI. Neuroimage 2025; 308:121063. [PMID: 39922330 DOI: 10.1016/j.neuroimage.2025.121063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
This paper introduces a novel multimodal and high-resolution human brain cerebellum lobule segmentation method. Unlike current tools that operate at standard resolution (1 mm3) or using mono-modal data, the proposed method improves cerebellum lobule segmentation through the use of a multimodal and ultra-high resolution (0.125 mm3) training dataset. To develop the method, first, a database of semi-automatically labelled cerebellum lobules was created to train the proposed method with ultra-high resolution T1 and T2 MR images. Then, an ensemble of deep networks has been designed and developed, allowing the proposed method to excel in the complex cerebellum lobule segmentation task, improving precision while being memory efficient. Notably, our approach deviates from the traditional U-Net model by exploring alternative architectures. We have also integrated deep learning with classical machine learning methods incorporating a priori knowledge from multi-atlas segmentation which improved precision and robustness. Finally, a new online pipeline, named DeepCERES, has been developed to make available the proposed method to the scientific community requiring as input only a single T1 MR image at standard resolution.
Collapse
Affiliation(s)
- Sergio Morell-Ortega
- Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA), Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain.
| | - Marina Ruiz-Perez
- Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA), Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Marien Gadea
- Department of Psychobiology, Faculty of Psychology, Universitat de Valencia, Valencia, Spain
| | - Roberto Vivo-Hernando
- Instituto de Automática e Informática Industrial, Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Gregorio Rubio
- Departamento de matemática aplicada, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Fernando Aparici
- Área de Imagen Médica. Hospital Universitario y Politécnico La Fe. Valencia, Spain
| | - Maria de la Iglesia-Vaya
- Unidad Mixta de Imagen Biomédica FISABIO-CIPF. Fundación para el Fomento de la Investigación Sanitario y Biomédica de la Comunidad Valenciana - Valencia, Spain
| | - Gwenaelle Catheline
- Univ. Bordeaux, CNRS, UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Bordeaux, France
| | - Boris Mansencal
- CNRS, Univ. Bordeaux, Bordeaux INP, LABRI, UMR5800, in2brain, F-33400 Talence, France
| | - Pierrick Coupé
- CNRS, Univ. Bordeaux, Bordeaux INP, LABRI, UMR5800, in2brain, F-33400 Talence, France
| | - José V Manjón
- Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA), Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| |
Collapse
|
11
|
Suzumura S, Osawa A, Sugioka J, Kamiya M, Sano Y, Kandori A, Mizuguchi T, Uchida Y, Kagaya H, Kondo I. Differences in Finger Dexterity in Patients With Mild and Moderate Alzheimer's Disease-A Study of Cognitive Function by Disease Severity. Brain Behav 2025; 15:e70403. [PMID: 40059446 PMCID: PMC11891264 DOI: 10.1002/brb3.70403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/18/2024] [Accepted: 02/09/2025] [Indexed: 05/13/2025] Open
Abstract
AIM This study aimed to estimate the relationship between finger function and cognitive function in patients with Alzheimer's disease (AD). METHODS Patients diagnosed with AD at the Outpatient Center for Comprehensive Care and Research on Memory Disorder of the National Center for Geriatrics and Gerontology underwent a 15-s bimanual alternating tapping task to measure finger movements. After finger movement measurements, we classified the severity of AD into mild and moderate and compared the finger movements. The Mann-Whitney U test and effect size were used to compare parameter values between the two groups (mild and moderate AD), and the calculated p values were corrected using the Bonferroni method. The Spearman rank correlation coefficient was calculated to determine the association between finger parameters and cognitive function (Mini-Mental Examination [MMSE]). RESULTS Data from 163 patients with AD were analyzed. When comparing finger parameters between the mild AD (64 individuals) and moderate AD (99 individuals) groups, the moderate AD group demonstrated fewer taps (p = 0.005; r = 0.22) and a longer interval between taps with the thumb and index finger (p = 0.007; r = 0.21) than the mild AD group. The correlation between the MMSE score and finger function was weakly positive for the number of taps and weakly negative for the average of tapping interval. CONCLUSIONS These parameters reflect the decline in finger function associated with the advanced stages of dementia and may help assess the severity of AD. Additionally, these findings may have clinical utility in assessing the severity of AD, potentially enhancing diagnostic accuracy for differentiating stages of AD.
Collapse
Affiliation(s)
- Shota Suzumura
- Faculty of Rehabilitation, School of Health SciencesFujita Health UniversityToyoakeJapan
- Department of Rehabilitation MedicineNational Center for Geriatrics and GerontologyObuJapan
| | - Aiko Osawa
- Department of Rehabilitation MedicineNational Center for Geriatrics and GerontologyObuJapan
| | - Junpei Sugioka
- Department of Rehabilitation MedicineNational Center for Geriatrics and GerontologyObuJapan
| | - Masaki Kamiya
- Department of Rehabilitation MedicineNational Center for Geriatrics and GerontologyObuJapan
| | - Yuko Sano
- Research & Development Group, Healthcare Innovation CenterHitachi, Ltd.KokubunjiJapan
| | - Akihiko Kandori
- Research & Development Group, Center for Exploratory ResearchHitachi, LtdKokubunjiJapan
| | - Tomohiko Mizuguchi
- New Business Producing Division, Business Development Dept.Maxell, Ltd.YokohamaJapan
| | - Yoshiharu Uchida
- New Business Producing Division, Business Development Dept.Maxell, Ltd.YokohamaJapan
| | - Hitoshi Kagaya
- Department of Rehabilitation MedicineNational Center for Geriatrics and GerontologyObuJapan
| | - Izumi Kondo
- Department of Rehabilitation MedicineNational Center for Geriatrics and GerontologyObuJapan
| |
Collapse
|
12
|
Liang Y, Ma D, Li M, Wang Z, Hao C, Sun Y, Hao X, Zuo C, Li S, Feng Y, Qi S, Wang Y, Sun S, Xu YM, Andreassen OA, Shi C. Exome sequencing identifies novel genes associated with cerebellar volume and microstructure. Commun Biol 2025; 8:344. [PMID: 40025133 PMCID: PMC11873060 DOI: 10.1038/s42003-025-07797-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
Proteins encoded by exons are critical for cellular functions, and mutations in these genes often result in significant phenotypic effects. The cerebellum is linked to various heritable human disease phenotypes, yet genome-wide association studies have struggled to capture the effects of rare variants on cerebellar traits. This study conducts a large-scale exome association analysis using data from approximately 35,000 UK Biobank participants, examining seven cerebellar traits, including total cerebellar volume and white matter microstructure. We identify 90 genes associated with cerebellar traits, 60 of which were previously unreported in genome-wide association studies. Notable findings include the discovery of genes like PRKRA and TTK, as well as RASGRP3, linked to cerebellar volume and white matter microstructure. Gene enrichment analysis reveals associations with non-coding RNA processing, cognitive function, neurodegenerative diseases, and mental disorders, suggesting shared biological mechanisms between cerebellar phenotypes and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Yuanyuan Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dongrui Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zhiyun Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chenwei Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yuemeng Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chunyan Zuo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shuangjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yanmei Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shasha Qi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yunpeng Wang
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Shilei Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Ole A Andreassen
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
13
|
Hicks TH, Magalhães TNC, Bernard JA. The Human Cerebello-Hippocampal Circuit Across Adulthood. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638640. [PMID: 40027698 PMCID: PMC11870467 DOI: 10.1101/2025.02.17.638640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Direct communication between the hippocampus and cerebellum has been shown via coactivation and synchronized neuronal oscillations in animal models. Further, this novel cerebello-hippocampal circuit may be impacted by sex steroid hormones. The cerebellum and hippocampus are dense with estradiol and progesterone receptors relative to other brain regions. Females experience up to a 90% decrease in ovarian estradiol production after the menopausal transition. Postmenopausal women show lower cerebello-cortical and intracerebellar FC compared to reproductive aged females. Sex hormones are established modulators of both memory function and synaptic organization in the hippocampus in non-human animal studies. However, investigation of the cerebello-hippocampal (CB-HP) circuit has been limited to animal studies and small homogeneous samples of young adults as it relates to spatial navigation. Here, we investigate the CB-HP circuit in 138 adult humans (53% female) from 35-86 years of age, to define its FC patterns, and investigate its associations with behavior, hormone levels, and sex differences therein. We established robust FC patterns between the CB and HP in this sample. We predicted and found negative relationships between age and CB-HP FC. As expected, estradiol levels exhibited positive relationships with CB-HP. We found lower CB-HP FC with higher levels of progesterone. We provide the first characterization of the CB-HP circuit across middle and older adulthood and demonstrate that connectivity is sensitive to sex steroid hormone levels. This work provides the first clear CB-HP circuit mapping in the human brain and serves as a foundation for future work in neurological and psychiatric diseases.
Collapse
|
14
|
Yao C, Shan Y, Cui B, Chen Z, Bi S, Wang T, Yan S, Lu J. Hyperconnectivity and Connectome Gradient Dysfunction of Cerebello-Thalamo-Cortical Circuitry in Alzheimer's Disease Spectrum Disorders. CEREBELLUM (LONDON, ENGLAND) 2025; 24:43. [PMID: 39913059 DOI: 10.1007/s12311-025-01792-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 02/07/2025]
Abstract
Cerebellar functional connectivity changes have been reported in Alzheimer's disease (AD), but a comprehensive framework integrating these findings is lacking. This retrospective study investigates the cerebello-thalamo-cortical (CTC) circuit in AD, using functional gradient analysis to elucidate deficits and potential biomarkers. We analyzed data from 246 participants enrolled in the Alzheimer's Disease Neuroimaging Initiative (ADNI-3; NCT02854033), including 58 with AD, 103 with mild cognitive impairment (MCI), and 85 cognitively normal (CN) controls, matched for age and sex. All individuals underwent comprehensive neuropsychological assessments (MMSE, MoCA, ADAS-Cog) and MRI scans. We extracted mean time series for 270 brain regions (an extended Power atlas) and computed pairwise functional connectivity, focusing on CTC circuitry. Thalamic and cerebellar connectivity gradients were derived using voxel-wise correlation matrices and the BrainSpace toolbox, defining thalamic and cerebellar masks from the Melbourne subcortical atlas and AAL atlas, respectively. ANCOVA with post hoc analyses, controlling for age and sex, was conducted to assess abnormal CTC connectivity across AD, MCI, and CN groups. LASSO regression identified edges within the CTC circuitry that significantly differed between AD and CN, MCI and CN, AD and MCI, as well as was used to construct Logistic classification model. Pearson correlations were performed to examine relationships between mean CTC connectivity, individual edges, and cognitive scores (MMSE, MoCA, ADAS-Cog). To explore the hierarchical organization of the thalamus and cerebellum, global gradient distributions were compared across groups using two-sample Kolmogorov-Smirnov tests. Additionally, ANCOVA was applied to compare subfield- and functional-level gradients of the thalamus and cerebellum among AD, MCI, and CN. False discovery rate (FDR) corrections were used, setting the statistical significance threshold was set at P < 0.05. AD and MCI individuals exhibited increased CTC connectivity compared to CN (all P < 0.05). Average CTC connectivity did not correlate with cognitive scores (P > 0.05), but specific CTC edges were correlated. LASSO regression identified 20 discriminative edges, achieving high accuracy in AD-CN classification (AUC = 0.92 training, AUC = 0.80 test). Thalamic and cerebellar gradient distributions differed significantly across groups (all P < 0.05), with specific regions showing distinct gradient scores. Five cerebellar functional networks exhibited decreased gradient scores. Significant CTC hyperconnectivity in AD and MCI compared with CN suggests early thalamic and cerebellar dysregulation. Classification analyses effectively distinguished AD vs. CN but were moderate for MCI vs. CN and limited for MCI vs. AD. Gradient analyses revealed global- and subfield-level disruptions in AD, emphasizing the role of thalamic and cerebellar interactions in cognitive decline and offering potential diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Chenyang Yao
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Yi Shan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Bixiao Cui
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhigeng Chen
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Sheng Bi
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Tao Wang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Shaozhen Yan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.
| |
Collapse
|
15
|
Hyde VR, Zhou C, Fernandez JR, Chatterjee K, Ramakrishna P, Lin A, Fisher GW, Çeliker OT, Caldwell J, Bender O, Sauer PJ, Lugo-Martinez J, Bar DZ, D'Aiuto L, Shemesh OA. Anti-herpetic tau preserves neurons via the cGAS-STING-TBK1 pathway in Alzheimer's disease. Cell Rep 2025; 44:115109. [PMID: 39753133 DOI: 10.1016/j.celrep.2024.115109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on the presence of extracellular β-amyloid (Aβ) and intracellular hyperphosphorylated tau (p-tau). Emerging evidence suggests a potential link between AD pathologies and infectious agents, with herpes simplex virus 1 (HSV-1) being a leading candidate. Our investigation, using metagenomics, mass spectrometry, western blotting, and decrowding expansion pathology, detects HSV-1-associated proteins in human brain samples. Expression of the herpesvirus protein ICP27 increases with AD severity and strongly colocalizes with p-tau but not with Aβ. Modeling in human brain organoids shows that HSV-1 infection elevates tau phosphorylation. Notably, p-tau reduces ICP27 expression and markedly decreases post-infection neuronal death from 64% to 7%. This modeling prompts investigation into the cGAS-STING-TBK1 pathway products, nuclear factor (NF)-κB and IRF-3, which colocalizes with ICP27 and p-tau in AD. Furthermore, experimental activation of STING enhances tau phosphorylation, while TBK1 inhibition prevents it. Together, these findings suggest that tau phosphorylation acts as an innate immune response in AD, driven by cGAS-STING.
Collapse
Affiliation(s)
- Vanesa R Hyde
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoming Zhou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Juan R Fernandez
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krishnashis Chatterjee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pururav Ramakrishna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amanda Lin
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Gregory W Fisher
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Orhan Tunç Çeliker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jill Caldwell
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peter Joseph Sauer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Or A Shemesh
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
16
|
Parsaei M, Barahman G, Roumiani PH, Ranjbar E, Ansari S, Najafi A, Karimi H, Aarabi MH, Moghaddam HS. White matter correlates of cognition: A diffusion magnetic resonance imaging study. Behav Brain Res 2025; 476:115222. [PMID: 39216828 DOI: 10.1016/j.bbr.2024.115222] [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: 05/06/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Our comprehension of the interplay of cognition and the brain remains constrained. While functional imaging studies have identified cognitive brain regions, structural correlates of cognitive functions remain underexplored. Advanced methods like Diffusion Magnetic Resonance Imaging (DMRI) facilitate the exploration of brain connectivity and White Matter (WM) tract microstructure. Therefore, we conducted connectometry method on DMRI data, to reveal WM tracts associated with cognition. METHODS 125 healthy participants from the National Institute of Mental Health Intramural Healthy Volunteer Dataset were recruited. Multiple regression analyses were conducted between DMRI-derived Quantitative Anisotropy (QA) values within WM tracts and scores of participants in Flanker Inhibitory Control and Attention Test (attention), Dimensional Change Card Sort (executive function), Picture Sequence Memory Test (episodic memory), and List Sorting Working Memory Test (working memory) tasks from National Institute of Health toolbox. The significance level was set at False Discovery Rate (FDR)<0.05. RESULTS We identified significant positive correlations between the QA of WM tracts within the left cerebellum and bilateral fornix with attention, executive functioning, and episodic memory (FDR=0.018, 0.0002, and 0.0002, respectively), and a negative correlation between QA of WM tracts within bilateral cerebellum with attention (FDR=0.028). Working memory demonstrated positive correlations with QA of left inferior longitudinal and left inferior fronto-occipital fasciculi (FDR=0.0009), while it showed a negative correlation with QA of right cerebellar tracts (FDR=0.0005). CONCLUSION Our results underscore the intricate link between cognitive performance and WM integrity in frontal, temporal, and cerebellar regions, offering insights into early detection and targeted interventions for cognitive disorders.
Collapse
Affiliation(s)
- Mohammadamin Parsaei
- Maternal, Fetal & Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Gelayol Barahman
- School of Medicine, Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | | | - Ehsan Ranjbar
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Ansari
- Psychosomatic Medicine Research Center, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Anahita Najafi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanie Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Aarabi
- Department of Neuroscience, University of Padova, Padova, Italy; Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
| | - Hossein Sanjari Moghaddam
- Psychiatry and Psychology Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Young L, Richey LN, Law CA, Esagoff AI, Ismail Z, Senjem ML, Jack CR, Shrestha S, Gottesman RF, Moussawi K, Peters ME, Schneider ALC. Associations of Mild Behavioral Impairment Domains with Brain Volumes: Cross-sectional Analysis of Atherosclerosis Risk in Community (ARIC) Study. J Acad Consult Liaison Psychiatry 2025; 66:37-48. [PMID: 39603508 PMCID: PMC11903177 DOI: 10.1016/j.jaclp.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/03/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Mild behavioral impairment (MBI) has been associated with global brain atrophy, but the regional neural correlates of MBI symptoms are less clear, particularly among community-dwelling older individuals without dementia. OBJECTIVE Our objective was to examine the associations of MBI domains with gray matter (GM) volumes in a large population-based sample of older adults without dementia. METHODS We performed a cross-sectional study of 1445 community-dwelling older adults in the Atherosclerosis Risk in Communities Study who underwent detailed neurocognitive assessment and brain magnetic resonance imaging in 2011-2013. MBI domains were defined using an established algorithm that maps data collected from informants on the Neuropsychiatric Inventory Questionnaire to the 5 MBI domains of decreased motivation, affective dysregulation, impulse dyscontrol, social inappropriateness, and abnormal perception/thought content. We performed voxel-based morphometry analyses to investigate associations of any MBI domain symptoms with GM volumes. We additionally performed region-of-interest analyses using adjusted linear regression models to examine associations between individual MBI domains with a priori-hypothesized regional GM volumes. RESULTS Overall, the mean age of participants was 76.5 years; 59% were female, 21% were of Black race, and 26% had symptoms in at least one MBI domain. Participants with normal cognition comprised 60% of the population, and 40% had mild cognitive impairment. Compared to individuals without any MBI domain symptoms, voxel-based morphometry analyses showed that participants with symptoms in at least one MBI domain had consistently lower GM volumes in the cerebellum and bilateral temporal lobes, particularly involving the hippocampus. In adjusted region-of-interest models, affective dysregulation domain symptoms were associated with lower GM volume in the inferior temporal lobe (β = -0.34; 95% confidence interval = -0.64, -0.04), and impulse dyscontrol domain symptoms were associated with lower GM volume in the parahippocampal gyrus (β = -0.06; 95% confidence interval = -0.11, 0.00). CONCLUSIONS In this community-dwelling population of older adults without dementia, MBI symptoms were associated with lower GM volumes in regions commonly implicated in early Alzheimer's disease pathology. These findings lend support to the notion that MBI symptoms may be useful in identifying individuals at risk for future dementia.
Collapse
Affiliation(s)
- Lisa Young
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Lisa N Richey
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Connor A Law
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Aaron I Esagoff
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zahinoor Ismail
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada; NIHR Exeter Biomedical Research Centre, University of Exeter, Exeter, UK
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, MN; Mayo Clinic Department of Information Technology, Rochester, MN
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, MN
| | - Srishti Shrestha
- University of Mississippi Medical Center School of Medicine, The MIND Center and Department of Neurology, Oxford, MS
| | - Rebecca F Gottesman
- National Institutes of Health, National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD
| | - Khaled Moussawi
- Department of Neurology, University of California San Francisco, San Francisco, CA
| | - Matthew E Peters
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrea L C Schneider
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
18
|
Tao W, Lu X, Yuan S, Ye P, Zhang Z, Guan Q, Li H. Unstable functional brain states and reduced cerebro-cerebellar modularity in elderly individuals with subjective cognitive decline. Neuroimage 2025; 305:120969. [PMID: 39667538 DOI: 10.1016/j.neuroimage.2024.120969] [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: 05/21/2024] [Revised: 08/26/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024] Open
Abstract
The preclinical stage of Alzheimer's Disease (AD) holds great potential for intervention, therefore, it is crucial to elucidate the neural mechanisms underlying the progression of subjective cognitive decline (SCD). Previous studies have predominantly focused on the neural changes in the cerebrum associated with SCD, but have relatively neglected the cerebellum, and its functional relationship with the cerebrum. In the current study, we employed dynamic functional connectivity and large-scale brain network approaches to investigate the pathological characteristics of dynamic brain states and cerebro-cerebellar collaboration between SCD (n = 32) and the healthy elderly (n = 29) using resting-state fMRI. Two-way repeated measures ANOVA and permutation t-tests revealed significant group differences, with individuals with SCD exhibiting shorter state duration and more frequent transitions between states compared to the healthy elderly individuals. Additionally, individuals with SCD showed lower levels of intracerebellar functional connectivity, but higher levels of cerebellar-cerebral functional integration. Furthermore, the hub nodes of the functional networks in SCD shifted between the cerebellum and cerebrum across different brain states. These findings indicate that SCD exhibits greater state instability but may compensate for the negative effects of early disease by integrating cerebellar and cerebral networks, thereby maintaining cognitive performance. This study enhances our theoretical understanding of cerebellar-cerebral relationship changes in the early stages of AD and provides evidence for early interventions targeting the cerebellum.
Collapse
Affiliation(s)
- Wuhai Tao
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen 518060, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Xiaojie Lu
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Shuaike Yuan
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Peixuan Ye
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Qing Guan
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen 518060, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; University of Health and Rehabilitation Sciences,School of Social Development and Health Management, Qingdao, Shandong, 266113, China.
| | - Hehui Li
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen 518060, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
19
|
Gong L, Liu D, Zhang B, Yu S, Xi C. Sex-Specific Entorhinal Cortex Functional Connectivity in Cognitively Normal Older Adults with Amyloid-β Pathology. Mol Neurobiol 2025; 62:475-484. [PMID: 38867110 PMCID: PMC11711718 DOI: 10.1007/s12035-024-04243-z] [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: 08/01/2023] [Accepted: 05/06/2024] [Indexed: 06/14/2024]
Abstract
Sex and apolipoprotein E (APOE) genotype have been shown to influence the risk and progression of Alzheimer's disease (AD). However, the impact of these factors on the functional connectivity of the entorhinal cortex (ERC) in clinically unpaired older adults (CUOA) with amyloid-β (Aβ +) pathology remains unclear. A total of 1022 cognitively normal older adults with Aβ + (603 females and 586 APOE ε4 +) from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) study were included in this study. The 2 × 2 (gender, 2 APOE genotypes) analysis of covariance was performed to compare the demographic information, cognitive performance, and volumetric MRI data among these groups. Voxel-wise comparisons of bilateral ERC functional connectivity (FC) were conducted, and partial correlation analyses were used to explore the associations between cognitive performance and ERC-FC strength. We found that the APOE genotype influenced ERC functional connectivity mainly in the sensorimotor network (SMN). Males exhibited higher ERC-FC in the salience network (SN), while females displayed higher ERC-FC in the default mode network (DMN), executive control network (ECN), and reward network. The interplay of sex and APOE genotype on ERC-FC was observed in the SMN and cerebellar lobe. The ERC-FC was associated with executive function and memory performance in individuals with CUOA-Aβ + . Our findings provide evidence of sex-specific ERC functional connectivity compensation mechanism in cognitively normal older adults with Aβ + pathology. This study may contribute to a better understanding of the mechanisms underlying the early stages of AD and may help develop personalized interventions in preclinical AD.
Collapse
Affiliation(s)
- Liang Gong
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Duan Liu
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Bei Zhang
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Siyi Yu
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| | - Chunhua Xi
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Huaihe Road 390, Heifei, 230061, Anhui, China.
| |
Collapse
|
20
|
Chen Y, Qi Y, Hu Y, Qiu X, Qiu T, Li S, Liu M, Jia Q, Sun B, Liu C, Li T, Le W. Integrated cerebellar radiomic-network model for predicting mild cognitive impairment in Alzheimer's disease. Alzheimers Dement 2025; 21:e14361. [PMID: 39535490 PMCID: PMC11782160 DOI: 10.1002/alz.14361] [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: 08/22/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Pathological and neuroimaging alterations in the cerebellum of Alzheimer's disease (AD) patients have been documented. However, the role of cerebellum-derived radiomic and structural connectome modeling in the prediction of AD progression remains unclear. METHODS Radiomic features were extracted from magnetic resonance imaging (MRI) in the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset (n = 1319) and an in-house dataset (n = 308). Integrated machine learning models were developed to predict the conversion risk of normal cognition (NC) to mild cognitive impairment (MCI) over a 6-year follow-up. RESULTS The cerebellar models outperformed hippocampal models in distinguishing MCI from NC and in predicting transitions from NC to MCI across both cohorts. Key predictors included textural features in the right III and left I and II lobules, and network properties in Vermis I and II, which were associated with cognitive decline in AD. DISCUSSION Cerebellum-derived radiomic-network modeling shows promise as a tool for early identification and prediction of disease progression during the preclinical stage of AD. HIGHLIGHTS Altered cerebellar radiomic features and topological networks were identified in the subjects with mild cognitive impairment (MCI). The cerebellar radiomic-network integrated models outperformed hippocampal models in distinguishing MCI from normal cognition. The cerebellar radiomic model effectively predicts MCI risk and can stratify individuals into distinct risk categories. Specific cerebellar radiomic features are associated with cognitive impairment across various stages of amyloid beta and tau pathology.
Collapse
Affiliation(s)
- Yini Chen
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
- Department of RadiologyThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Yiwei Qi
- Department of RadiologyThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Yiying Hu
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
- Department of NeurologyThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Xinhui Qiu
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Tao Qiu
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Song Li
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Meichen Liu
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
- Department of NeurologyThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Qiqi Jia
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Bo Sun
- Department of RadiologyThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic ChemistryChinese Academy of SciencesShanghaiChina
| | - Tianbai Li
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Weidong Le
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated HospitalDalian Medical UniversityDalianChina
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu HospitalShanghaiChina
| | | |
Collapse
|
21
|
El Gazzar WB, Farag AA, Samir M, Bayoumi H, Youssef HS, Marei YM, Mohamed SK, Marei AM, Abdelfatah RM, Mahmoud MM, Aboelkomsan EAF, Khalfallah EKM, Anwer HM. Berberine chloride loaded nano-PEGylated liposomes attenuates imidacloprid-induced neurotoxicity by inhibiting NLRP3/Caspase-1/GSDMD-mediated pyroptosis. Biofactors 2025; 51:e2107. [PMID: 39074847 DOI: 10.1002/biof.2107] [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: 04/26/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024]
Abstract
Concerns have been expressed about imidacloprid (IMI), one of the most often used pesticides, and its potential neurotoxicity to non-target organisms. Chronic neuroinflammation is central to the pathology of several neurodegenerative disorders. Hence, exploring the molecular mechanism by which IMI would trigger neuroinflammation is particularly important. This study examined the neurotoxic effects of oral administration of IMI (45 mg/kg/day for 30 days) and the potential neuroprotective effect of berberine (Ber) chloride loaded nano-PEGylated liposomes (Ber-Lip) (10 mg/kg, intravenously every other day for 30 days) using laboratory rat. The histopathological changes, anti-oxidant and oxidative stress markers (GSH, SOD, and MDA), proinflammatory cytokines (IL1β and TNF-α), microglia phenotype markers (CD86 and iNOS for M1; CD163 for M2), the canonical pyroptotic pathway markers (NLRP3, caspase-1, GSDMD, and IL-18) and Alzheimer's disease markers (Neprilysin and beta amyloid [Aβ] deposits) were assessed. Oral administration of IMI resulted in apparent cerebellar histopathological alterations, oxidative stress, predominance of M1 microglia phenotype, significantly upregulated NLRP3, caspase-1, GSDMD, IL-18 and Aβ deposits and significantly decreased Neprilysin expression. Berberine reduced the IMI-induced aberrations in the measured parameters and improved the IMI-induced histopathological and ultrastructure alterations brought on by IMI. This study highlights the IMI neurotoxic effect and its potential contribution to the development of Alzheimer's disease and displayed the neuroprotective effect of Ber-Lip.
Collapse
Affiliation(s)
- Walaa Bayoumie El Gazzar
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
- Department of Medical Biochemistry and Molecular biology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| | - Amina A Farag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| | - Mohamed Samir
- Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharqia, Egypt
- School of Science, Faculty of Engineering and Science, University of Greenwich, Kent, UK
| | - Heba Bayoumi
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha City, Egypt
| | - Heba S Youssef
- Department of Physiology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| | - Yasmin Mohammed Marei
- Department of Medical Biochemistry and Molecular biology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| | - Shimaa K Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Azza M Marei
- Department of Zoology, Faculty of Science, Benha University, Benha City, Qalyubia, Egypt
| | - Reham M Abdelfatah
- Department of Pesticides, Faculty of Agriculture, Mansoura University, Mansoura, Egypt
| | | | | | - Eman Kamel M Khalfallah
- Department of Biochemistry, Toxicology and Feed Deficiency, Animal Health Research Institute (AHRI), Agricultural Research Center (ARC), Dokki, Giza, Egypt
| | - Hala Magdy Anwer
- Department of Physiology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| |
Collapse
|
22
|
Biljman K, Gozes I, Lam JCK, Li VOK. An experimental framework for conjoint measures of olfaction, navigation, and motion as pre-clinical biomarkers of Alzheimer's disease. J Alzheimers Dis Rep 2024; 8:1722-1744. [PMID: 40034341 PMCID: PMC11863766 DOI: 10.1177/25424823241307617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 11/19/2024] [Indexed: 03/05/2025] Open
Abstract
Elucidating Alzheimer's disease (AD) prodromal symptoms can resolve the outstanding challenge of early diagnosis. Based on intrinsically related substrates of olfaction and spatial navigation, we propose a novel experimental framework for their conjoint study. Artificial intelligence-driven multimodal study combining self-collected olfactory and motion data with available big clinical datasets can potentially promote high-precision early clinical screenings to facilitate timely interventions targeting neurodegenerative progression.
Collapse
Affiliation(s)
- Katarina Biljman
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Illana Gozes
- Elton Laboratory for Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, The Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Jacqueline CK Lam
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Victor OK Li
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
23
|
Winter S, Mahzarnia A, Anderson RJ, Han ZY, Tremblay J, Stout JA, Moon HS, Marcellino D, Dunson DB, Badea A. Brain network fingerprints of Alzheimer's disease risk factors in mouse models with humanized APOE alleles. Magn Reson Imaging 2024; 114:110251. [PMID: 39362319 PMCID: PMC11514054 DOI: 10.1016/j.mri.2024.110251] [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: 08/23/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
Alzheimer's disease (AD) presents complex challenges due to its multifactorial nature, poorly understood etiology, and late detection. The mechanisms through which genetic and modifiable risk factors influence disease susceptibility are under intense investigation, with APOE being the major genetic risk factor for late onset AD. Yet the impact of unique risk factors on brain networks is difficult to disentangle, and their interactions remain unclear. To model multiple risk factors, including APOE genotype, age, sex, diet, and immunity we used a cross sectional design, leveraging mice expressing human APOE and NOS2 genes, conferring a reduced immune response compared to mouse Nos2. We used network topological and GraphClass analyses of brain connectomes derived from accelerated diffusion-weighted MRI to assess the global and local impact of risk factors, in the absence of AD pathology. Aging and a high-fat diet impacted extensive networks comprising AD-vulnerable regions, including the temporal association cortex, amygdala, and the periaqueductal gray, involved in stress responses. Sex impacted networks including sexually dimorphic regions (thalamus, insula, hypothalamus) and key memory-processing areas (fimbria, septum). APOE genotypes modulated connectivity in memory, sensory, and motor regions, while diet and immunity both impacted the insula and hypothalamus. Notably, these risk factors converged on a circuit comprising 63 of 54,946 total connections (0.11% of the connectome), highlighting shared vulnerability amongst multiple AD risk factors in regions essential for sensory integration, emotional regulation, decision making, motor coordination, memory, homeostasis, and interoception. APOE genotype specific immune signatures support the design of interventions tailored to risk profiles. Sparse Canonical Correlation Analysis (CCA) including spatial memory as a risk factor resulted in a network comprising 80 edges, showing significant overlap with risk-associated networks from GraphClass. The largest overlaps were observed with networks impacted by diet (47 edges), immunity (39 edges), APOE3 vs 4 (26 edges), sex (23 edges), and age (19 edges), the resulting networks supporting the use of sensory cues in spatial memory retrieval. These network-based biomarkers hold translational value for distinguishing high-risk versus low-risk participants at preclinical AD stages, suggest circuits as potential therapeutic targets, and advance our understanding of network fingerprints associated with AD risk.
Collapse
Affiliation(s)
- Steven Winter
- Statistical Science, Trinity School, Duke University, Durham, NC 27710, USA
| | - Ali Mahzarnia
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert J Anderson
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zay Yar Han
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jessica Tremblay
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jacques A Stout
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA; Duke UNC Brain Imaging and Analysis Center, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hae Sol Moon
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Daniel Marcellino
- Department of Medical and Translational Biology, Umeå University, Umeå 901 87, Sweden; Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund 22184, Sweden
| | - David B Dunson
- Statistical Science, Trinity School, Duke University, Durham, NC 27710, USA
| | - Alexandra Badea
- Quantitative Imaging and Analysis Lab, Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA; Duke UNC Brain Imaging and Analysis Center, Duke University School of Medicine, Durham, NC 27710, USA; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA; Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
24
|
Liu G, Yang C, Wang X, Chen X, Cai H, Le W. Cerebellum in neurodegenerative diseases: Advances, challenges, and prospects. iScience 2024; 27:111194. [PMID: 39555407 PMCID: PMC11567929 DOI: 10.1016/j.isci.2024.111194] [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] [Indexed: 11/19/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a group of neurological disorders characterized by the progressive dysfunction of neurons and glial cells, leading to their structural and functional degradation in the central and/or peripheral nervous system. Historically, research on NDs has primarily focused on the brain, brain stem, or spinal cord associated with disease-related symptoms, often overlooking the role of the cerebellum. However, an increasing body of clinical and biological evidence suggests a significant connection between the cerebellum and NDs. In several NDs, cerebellar pathology and biochemical changes may start in the early disease stages. This article provides a comprehensive update on the involvement of the cerebellum in the clinical features and pathogenesis of multiple NDs, suggesting that the cerebellum is involved in the onset and progression of NDs through various mechanisms, including specific neurodegeneration, neuroinflammation, abnormal mitochondrial function, and altered metabolism. Additionally, this review highlights the significant therapeutic potential of cerebellum-related treatments for NDs.
Collapse
Affiliation(s)
- Guangdong Liu
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Cui Yang
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xin Wang
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xi Chen
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weidong Le
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 200237, China
| |
Collapse
|
25
|
Tasevski S, Kyung Nam H, Ghannam A, Moughni S, Atoui T, Mashal Y, Hatch N, Zhang Z. Tissue nonspecific alkaline phosphatase deficiency impairs Purkinje cell development and survival in a mouse model of infantile hypophosphatasia. Neuroscience 2024; 560:357-370. [PMID: 39369942 DOI: 10.1016/j.neuroscience.2024.10.005] [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: 06/15/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Loss-of-function mutations in the tissue-nonspecific alkaline phosphatase (TNAP) gene can result in hypophosphatasia (HPP), an inherited multi-systemic metabolic disorder that is well-known for skeletal and dental hypomineralization. However, emerging evidence shows that both adult and pediatric patients with HPP suffer from cognitive deficits, higher measures of depression and anxiety, and impaired sensorimotor skills. The cerebellum plays an important role in sensorimotor coordination, cognition, and emotion. To date, the impact of TNAP mutation on the cerebellar circuitry development and function remains poorly understood. The main objective of this study was to investigate the roles of TNAP in cerebellar development and function, with a particular focus on Purkinje cells, in a mouse model of infantile HPP. Male and female wild type (WT) and TNAP knockout (KO) mice underwent behavioral testing on postnatal day 13-14 and were euthanized after completion of behavioral tests. Cerebellar tissues were harvested for gene expression and immunohistochemistry analyses. We found that TNAP mutation resulted in significantly reduced body weight, shorter body length, and impaired sensorimotor functions in both male and female KO mice. These developmental and behavioral deficits were accompanied by abnormal Purkinje cell morphology and dysregulation of genes that regulates synaptic transmission, cellular growth, proliferation, and death. In conclusion, inactivation of TNAP via gene deletion causes developmental delays, sensorimotor impairment, and Purkinje cell maldevelopment. These results shed light on a new perspective of cerebellar dysfunction in HPP.
Collapse
Affiliation(s)
- Stefanie Tasevski
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Hwa Kyung Nam
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan-Ann Arbor, 1011 N University Ave, Ann Arbor, MI 48109, USA
| | - Amanda Ghannam
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Sara Moughni
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Tia Atoui
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Yara Mashal
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Nan Hatch
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan-Ann Arbor, 1011 N University Ave, Ann Arbor, MI 48109, USA
| | - Zhi Zhang
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA.
| |
Collapse
|
26
|
Sun J, Han JDJ, Chen W. Exploring the relationship among Alzheimer's disease, aging and cognitive scores through neuroimaging-based approach. Sci Rep 2024; 14:27472. [PMID: 39523370 PMCID: PMC11551169 DOI: 10.1038/s41598-024-78712-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder, with the Mini-Mental State Examination (MMSE) and Clinical Dementia Rating (CDR) serving significant roles in monitoring its progression. We hypothesize that while cognitive assessment scores can detect AD-related brain changes, the targeted brain regions may differ. Additionally, given AD's strong association with aging, we propose that specific brain regions are influenced by both AD pathology and aging, exhibiting strong correlations with both. To test these hypotheses, we developed a 3D convolutional network with a mixed-attention mechanism to recognize AD subjects from structural magnetic resonance imaging (sMRI) data and utilize 3D convolutional methods to pinpoint brain regions significantly correlated with the AD, MMSE, CDR and age. All models were trained and internally validated on 417 samples from the Alzheimer's Disease Neuroimaging Initiative (ADNI), and the classification model was externally validated on 382 samples from the Australian Imaging and Lifestyle flagship (AIBL). This approach provided robust support for using MMSE and CDR in assessing AD progression and visually illustrated the relationship between aging and AD. The analysis revealed correlations among the four identification tasks (AD, MMSE, CDR and age) and highlighted asymmetric brain lesions in both AD and aging. Notably, we found that AD can accelerate aging to some extent, and a significant correlation exists between the rate of aging and cognitive assessment scores. This offers new insights into the relationship between AD and aging.
Collapse
Affiliation(s)
- Jinhui Sun
- School of Cyber Science and Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, People's Republic of China.
| | - Weiyang Chen
- School of Cyber Science and Engineering, Qufu Normal University, Qufu, 273165, People's Republic of China.
| |
Collapse
|
27
|
Zhao Y. Mediation Analysis with Multiple Exposures and Multiple Mediators. Stat Med 2024; 43:4887-4898. [PMID: 39250913 PMCID: PMC11959452 DOI: 10.1002/sim.10215] [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: 01/10/2023] [Revised: 04/25/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024]
Abstract
A mediation analysis approach is proposed for multiple exposures, multiple mediators, and a continuous scalar outcome under the linear structural equation modeling framework. It assumes that there exist orthogonal components that demonstrate parallel mediation mechanisms on the outcome, and thus is named principal component mediation analysis (PCMA). Likelihood-based estimators are introduced for simultaneous estimation of the component projections and effect parameters. The asymptotic distribution of the estimators is derived for low-dimensional data. A bootstrap procedure is introduced for inference. Simulation studies illustrate the superior performance of the proposed approach. Applied to a proteomics-imaging dataset from the Alzheimer's disease neuroimaging initiative (ADNI), the proposed framework identifies protein deposition - brain atrophy - memory deficit mechanisms consistent with existing knowledge and suggests potential AD pathology by integrating data collected from different modalities.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
28
|
Wang X, Chen H, Tang T, Zhan X, Qin S, Hang T, Song M. Chronic Sleep Deprivation Altered the Expression of Memory-Related Genes and Caused Cognitive Memory Dysfunction in Mice. Int J Mol Sci 2024; 25:11634. [PMID: 39519186 PMCID: PMC11546330 DOI: 10.3390/ijms252111634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Lack of sleep, whether acute or chronic, is quite common and negatively affects an individual's memory and cognitive function. The question of whether chronic sleep deprivation (CSD) causes cognitive impairment to arise and progress is not well studied. To investigate the effects of CSD on memory and cognition, this study began by establishing a CSD mouse model. Behavioral experiments on animals revealed that CSD induced cognitive behavioral abnormalities reminiscent of Alzheimer's disease. Western blot experiments further demonstrated a considerable increase in amyloid-β (Aβ) expression in the mouse brain following CSD. Meanwhile, the hub gene Prkcg was searched for in the cerebellum using RNA-seq and bioinformatics analysis. PKCγ (Prkcg) expression was significantly reduced, as demonstrated by RT-qPCR and Western blot validations. Additionally, CSD was associated with downregulated CREB expression, decreased expression of the endothelin-converting enzyme (ECE1), and increased phosphorylation of ERK1/2 downstream of PKCγ. These findings suggested that CSD down-regulated PKCγ expression, decreased ECE1 expression, impaired Aβ degradation, and affected the PKCγ/ERK/CREB pathway and the synthesis of memory-related proteins. Overall, this study highlighted how CSD modulated PKCγ-related metabolism, impacting Aβ clearance and the production of memory-related proteins. Such insights are crucial for understanding and preventing sporadic Alzheimer's disease (sAD) associated with CSD.
Collapse
Affiliation(s)
| | | | | | | | | | - Taijun Hang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 211100, China; (X.W.); (H.C.); (T.T.); (X.Z.); (S.Q.)
| | - Min Song
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 211100, China; (X.W.); (H.C.); (T.T.); (X.Z.); (S.Q.)
| |
Collapse
|
29
|
Chen J, Han Z, Wang Z, Chen L, Wang S, Yao W, Xue Z. Identification of immune traits associated with neurodevelopmental disorders by two-sample Mendelian randomization analysis. BMC Psychiatry 2024; 24:728. [PMID: 39448971 PMCID: PMC11515564 DOI: 10.1186/s12888-024-06148-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND One of the main causes of health-related issues in children is neurodevelopmental disorders (NDDs), which include attention-deficit hyperactivity disorder (ADHD), autism spectrum disorder (ASD), and Tourette syndrome (TS). Nonetheless, there is relatively little prior research looking at the link between immunological inflammation and NDDs. Our work uses a two-sample Mendelian Randomization (MR) approach to provide a thorough evaluation of the causal effects of immune traits on ADHD, ASD, and TS. METHODS As exposures, 731 immunological traits' genetic associations were chosen, and the outcomes were genome-wide association data for ADHD, ASD, and TS. The inverse-variance weighted (IVW), weighted median (WM), and MR-Egger methods were used to conduct MR analysis. The results' robustness, heterogeneity, and horizontal pleiotropy were confirmed using extensive sensitivity analysis. RESULTS With single-nucleotide polymorphisms serving as instruments and false discovery rate (FDR) correction applied, the study found that significantly higher expression of CD62L on CD62L+ myeloid DC (IVW, OR: 0.926, 95% CI 0.896~0.958, P = 9.42 × 10-6, FDR = 0.007) and suggestively higher absolute cell count (AC) of CD28 + DN (CD4-CD8-) (IVW, OR: 0.852, 95% CI = 0.780 ∼ 0.932, P-value = 4.65 × 10-4, FDR = 0.170) was associated with a lower risk of ADHD. There was no pleiotropy, and the causal relationships were strong according to sensitivity, leave-one-out, and MR-Steiger directionality tests. For ASD and TS, no harmful or protective immune traits were observed. CONCLUSIONS The results of the study lend credence to the theory that deficiency in CD62L on CD62L+ myeloid DC and CD28 + DN (CD4-CD8) AC may contribute to the onset of ADHD.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pediatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, People's Republic of China
- Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Zhaopeng Han
- Department of Pediatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, People's Republic of China
| | - Zhuiyue Wang
- Department of Pediatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, People's Republic of China
| | - Lifei Chen
- Department of Pediatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, People's Republic of China
| | - Shuxia Wang
- Department of Pediatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, People's Republic of China
| | - Wenbo Yao
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, People's Republic of China.
| | - Zheng Xue
- Department of Pediatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, People's Republic of China.
| |
Collapse
|
30
|
Lin A, Chen Y, Chen Y, Ye Z, Luo W, Chen Y, Zhang Y, Wang W. MRI radiomics combined with machine learning for diagnosing mild cognitive impairment: a focus on the cerebellar gray and white matter. Front Aging Neurosci 2024; 16:1460293. [PMID: 39430972 PMCID: PMC11489926 DOI: 10.3389/fnagi.2024.1460293] [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: 07/05/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024] Open
Abstract
Objective Mild Cognitive Impairment (MCI) is a recognized precursor to Alzheimer's Disease (AD), presenting a significant risk of progression. Early detection and intervention in MCI can potentially slow disease advancement, offering substantial clinical benefits. This study employed radiomics and machine learning methodologies to distinguish between MCI and Normal Cognition (NC) groups. Methods The study included 172 MCI patients and 183 healthy controls from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, all of whom had 3D-T1 weighted MRI structural images. The cerebellar gray and white matter were segmented automatically using volBrain software, and radiomic features were extracted and screened through Pyradiomics. The screened features were then input into various machine learning models, including Random Forest (RF), Logistic Regression (LR), eXtreme Gradient Boosting (XGBoost), Support Vector Machines (SVM), K Nearest Neighbors (KNN), Extra Trees, Light Gradient Boosting Machine (LightGBM), and Multilayer Perceptron (MLP). Each model was optimized for penalty parameters through 5-fold cross-validation to construct radiomic models. The DeLong test was used to evaluate the performance of different models. Results The LightGBM model, which utilizes a combination of cerebellar gray and white matter features (comprising eight gray matter and eight white matter features), emerges as the most effective model for radiomics feature analysis. The model demonstrates an Area Under the Curve (AUC) of 0.863 for the training set and 0.776 for the test set. Conclusion Radiomic features based on the cerebellar gray and white matter, combined with machine learning, can objectively diagnose MCI, which provides significant clinical value for assisted diagnosis.
Collapse
Affiliation(s)
- Andong Lin
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Yini Chen
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yi Chen
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Zhinan Ye
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Weili Luo
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Ying Chen
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Yaping Zhang
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Wenjie Wang
- Department of Neurology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
31
|
Bernard JA. Cerebello-Hippocampal Interactions in the Human Brain: A New Pathway for Insights Into Aging. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2130-2141. [PMID: 38438826 PMCID: PMC11371944 DOI: 10.1007/s12311-024-01670-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 03/06/2024]
Abstract
The cerebellum is recognized as being important for optimal behavioral performance across task domains, including motor function, cognition, and affect. Decades of work have highlighted cerebello-thalamo-cortical circuits, from both structural and functional perspectives. However, these circuits of interest have been primarily (though not exclusively) focused on targets in the cerebral cortex. In addition to these cortical connections, the circuit linking the cerebellum and hippocampus is of particular interest. Recently, there has been an increased interest in this circuit, thanks in large part to novel findings in the animal literature demonstrating that neuronal firing in the cerebellum impacts that in the hippocampus. Work in the human brain has provided evidence for interactions between the cerebellum and hippocampus, though primarily this has been in the context of spatial navigation. Given the role of both regions in cognition and aging, and emerging evidence indicating that the cerebellum is impacted in age-related neurodegenerative disease such as Alzheimer's, I propose that further attention to this circuit is warranted. Here, I provide an overview of cerebello-hippocampal interactions in animal models and from human imaging and outline the possible utility of further investigations to improve our understanding of aging and age-related cognitive decline.
Collapse
Affiliation(s)
- Jessica A Bernard
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, 77843-4235, USA.
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, 77843-4235, USA.
| |
Collapse
|
32
|
Radosinska D, Gaal Kovalcikova A, Gardlik R, Chomova M, Snurikova D, Radosinska J, Vrbjar N. Oxidative Stress Markers and Na,K-ATPase Enzyme Kinetics Are Altered in the Cerebellum of Zucker Diabetic Fatty fa/fa Rats: A Comparison with Lean fa/+ and Wistar Rats. BIOLOGY 2024; 13:759. [PMID: 39452068 PMCID: PMC11505095 DOI: 10.3390/biology13100759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
Type 2 diabetes mellitus has been referred to as being closely related to oxidative stress, which may affect brain functions and brain glucose metabolism due to its high metabolic activity and lipid-rich content. Na,K-ATPase is an essential enzyme maintaining intracellular homeostasis, with properties that can sensitively mirror various pathophysiological conditions such as diabetes. The goal of this study was to determine oxidative stress markers as well as Na,K-ATPase activities in the cerebellum of Zucker diabetic fatty (ZDF) rats depending on diabetes severity. The following groups of male rats were used: Wistar, ZDF Lean (fa/+), and ZDF (fa/fa) rats, arbitrarily divided according to glycemia into ZDF obese (ZO, less severe diabetes) and ZDF diabetic (ZOD, advanced diabetes) groups. In addition to basic biometry and biochemistry, oxidative stress markers were assessed in plasma and cerebellar tissues. The Na, K-ATPase enzyme activity was measured at varying ATP substrate concentrations. The results indicate significant differences in basic biometric and biochemical parameters within all the studied groups. Furthermore, oxidative damage was greater in the cerebellum of both ZDF (fa/fa) groups compared with the controls. Interestingly, Na,K-ATPase enzyme activity was highest to lowest in the following order: ZOD > ZO > Wistar > ZDF lean rats. In conclusion, an increase in systemic oxidative stress resulting from diabetic conditions has a significant impact on the cerebellar tissue independently of diabetes severity. The increased cerebellar Na,K-ATPase activity may reflect compensatory mechanisms in aged ZDF (fa/fa) animals, rather than indicating cerebellar neurodegeneration: a phenomenon that warrants further investigation.
Collapse
Affiliation(s)
- Dominika Radosinska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Alexandra Gaal Kovalcikova
- Department of Pediatrics, National Institute of Children’s Diseases, Faculty of Medicine, Comenius University in Bratislava, 833 40 Bratislava, Slovakia;
| | - Roman Gardlik
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Maria Chomova
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 813 72 Bratislava, Slovakia;
| | - Denisa Snurikova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia; (D.S.); (N.V.)
| | - Jana Radosinska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 811 08 Bratislava, Slovakia
| | - Norbert Vrbjar
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia; (D.S.); (N.V.)
| |
Collapse
|
33
|
Ji J, Hou Z, He Y, Liu L, Xue F, Chen H, Yuan Z. Differential network knockoff filter with application to brain connectivity analysis. Stat Med 2024; 43:3830-3861. [PMID: 38922944 DOI: 10.1002/sim.10155] [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: 05/14/2023] [Revised: 04/30/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
The brain functional connectivity can typically be represented as a brain functional network, where nodes represent regions of interest (ROIs) and edges symbolize their connections. Studying group differences in brain functional connectivity can help identify brain regions and recover the brain functional network linked to neurodegenerative diseases. This process, known as differential network analysis focuses on the differences between estimated precision matrices for two groups. Current methods struggle with individual heterogeneity in measuring the brain connectivity, false discovery rate (FDR) control, and accounting for confounding factors, resulting in biased estimates and diminished power. To address these issues, we present a two-stage FDR-controlled feature selection method for differential network analysis using functional magnetic resonance imaging (fMRI) data. First, we create individual brain connectivity measures using a high-dimensional precision matrix estimation technique. Next, we devise a penalized logistic regression model that employs individual brain connectivity data and integrates a new knockoff filter for FDR control when detecting significant differential edges. Through extensive simulations, we showcase the superiority of our approach compared to other methods. Additionally, we apply our technique to fMRI data to identify differential edges between Alzheimer's disease and control groups. Our results are consistent with prior experimental studies, emphasizing the practical applicability of our method.
Collapse
Affiliation(s)
- Jiadong Ji
- Institute for Financial Studies, Shandong University, Jinan, Shandong, China
| | - Zhendong Hou
- Institute for Financial Studies, Shandong University, Jinan, Shandong, China
| | - Yong He
- Institute for Financial Studies, Shandong University, Jinan, Shandong, China
| | - Lei Liu
- Division of Biostatistics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hao Chen
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
34
|
Sajid M, Sharma R, Beheshti I, Tanveer M. Decoding cognitive health using machine learning: A comprehensive evaluation for diagnosis of significant memory concern. WIRES DATA MINING AND KNOWLEDGE DISCOVERY 2024; 14. [DOI: 10.1002/widm.1546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/29/2024] [Indexed: 01/03/2025]
Abstract
AbstractThe timely identification of significant memory concern (SMC) is crucial for proactive cognitive health management, especially in an aging population. Detecting SMC early enables timely intervention and personalized care, potentially slowing cognitive disorder progression. This study presents a state‐of‐the‐art review followed by a comprehensive evaluation of machine learning models within the randomized neural networks (RNNs) and hyperplane‐based classifiers (HbCs) family to investigate SMC diagnosis thoroughly. Utilizing the Alzheimer's Disease Neuroimaging Initiative 2 (ADNI2) dataset, 111 individuals with SMC and 111 healthy older adults are analyzed based on T1W magnetic resonance imaging (MRI) scans, extracting rich features. This analysis is based on baseline structural MRI (sMRI) scans, extracting rich features from gray matter (GM), white matter (WM), Jacobian determinant (JD), and cortical thickness (CT) measurements. In RNNs, deep random vector functional link (dRVFL) and ensemble dRVFL (edRVFL) emerge as the best classifiers in terms of performance metrics in the identification of SMC. In HbCs, Kernelized pinball general twin support vector machine (Pin‐GTSVM‐K) excels in CT and WM features, whereas Linear Pin‐GTSVM (Pin‐GTSVM‐L) and Linear intuitionistic fuzzy TSVM (IFTSVM‐L) performs well in the JD and GM features sets, respectively. This comprehensive evaluation emphasizes the critical role of feature selection, feature based‐interpretability and model choice in attaining an effective classifier for SMC diagnosis. The inclusion of statistical analyses further reinforces the credibility of the results, affirming the rigor of this analysis. The performance measures exhibit the suitability of this framework in aiding researchers with the automated and accurate assessment of SMC. The source codes of the algorithms and datasets used in this study are available at https://github.com/mtanveer1/SMC.This article is categorized under:
Technologies > Classification
Technologies > Machine Learning
Application Areas > Health Care
Collapse
Affiliation(s)
- M. Sajid
- Department of Mathematics Indian Institute of Technology Indore Indore India
| | - R. Sharma
- Department of Mathematics Indian Institute of Technology Indore Indore India
| | - I. Beheshti
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences University of Manitoba Winnipeg Manitoba Canada
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine Health Sciences Centre Winnipeg Manitoba Canada
| | - M. Tanveer
- Department of Mathematics Indian Institute of Technology Indore Indore India
| | | |
Collapse
|
35
|
Verpeut JL, Oostland M. The significance of cerebellar contributions in early-life through aging. Front Comput Neurosci 2024; 18:1449364. [PMID: 39258107 PMCID: PMC11384999 DOI: 10.3389/fncom.2024.1449364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Affiliation(s)
- Jessica L Verpeut
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Marlies Oostland
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
36
|
Lu Y, Xu K, Maydanchik N, Kang B, Pierce BL, Yang F, Chen LS. An integrative multi-context Mendelian randomization method for identifying risk genes across human tissues. Am J Hum Genet 2024; 111:1736-1749. [PMID: 39053459 PMCID: PMC11339623 DOI: 10.1016/j.ajhg.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Mendelian randomization (MR) provides valuable assessments of the causal effect of exposure on outcome, yet the application of conventional MR methods for mapping risk genes encounters new challenges. One of the issues is the limited availability of expression quantitative trait loci (eQTLs) as instrumental variables (IVs), hampering the estimation of sparse causal effects. Additionally, the often context- or tissue-specific eQTL effects challenge the MR assumption of consistent IV effects across eQTL and GWAS data. To address these challenges, we propose a multi-context multivariable integrative MR framework, mintMR, for mapping expression and molecular traits as joint exposures. It models the effects of molecular exposures across multiple tissues in each gene region, while simultaneously estimating across multiple gene regions. It uses eQTLs with consistent effects across more than one tissue type as IVs, improving IV consistency. A major innovation of mintMR involves employing multi-view learning methods to collectively model latent indicators of disease relevance across multiple tissues, molecular traits, and gene regions. The multi-view learning captures the major patterns of disease relevance and uses these patterns to update the estimated tissue relevance probabilities. The proposed mintMR iterates between performing a multi-tissue MR for each gene region and joint learning the disease-relevant tissue probabilities across gene regions, improving the estimation of sparse effects across genes. We apply mintMR to evaluate the causal effects of gene expression and DNA methylation for 35 complex traits using multi-tissue QTLs as IVs. The proposed mintMR controls genome-wide inflation and offers insights into disease mechanisms.
Collapse
Affiliation(s)
- Yihao Lu
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Ke Xu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, USA
| | - Nathaniel Maydanchik
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Bowei Kang
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Brandon L Pierce
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Fan Yang
- Yau Mathematical Sciences Center, Tsinghua University, Beijing, China; Yanqi Lake Beijing Institute of Mathematical Sciences and Applications, Beijing, China.
| | - Lin S Chen
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
37
|
Chen Y, Pan J, Lin A, Sun L, Li Y, Lin H, Pu R, Wang Y, Qi Y, Sun B. Cerebellar white and gray matter abnormalities in temporal lobe epilepsy: a voxel-based morphometry study. Front Neurosci 2024; 18:1417342. [PMID: 39156634 PMCID: PMC11328152 DOI: 10.3389/fnins.2024.1417342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
Background Previous structural neuroimaging studies linked cerebellar deficits to temporal lobe epilepsy (TLE). The functions of various cerebellar regions are increasingly being valued, and their changes in TLE patients warrant further in-depth investigation. In this study, we used the Spatially Unbiased Infratentorial (SUIT) toolbox with a new template to evaluate the cerebellar structural abnormalities in patients with TLE, and further explored the relationship between the changes of different cerebellar regions and cognition. Methods Thirty-two patients with TLE were compared with 39 healthy controls (HC) matched according to age, gender, handedness, and education level. All participants underwent a high-resolution T1-weighted MRI scan on a 3.0 Tesla scanner. We used a voxel-based morphometry (VBM) approach utilizing the SUIT toolbox to provide an optimized and fine-grained exploration of cerebellar structural alterations associated with TLE. Results Compared with HC, TLE patients showed a significant reduction in the volume of gray matter in the Left lobule VI and white matter in the Right Crus II. In the TLE patient group, we conducted partial correlation analysis between the volumes of different cerebellar regions and cognitive rating scale scores, such as MMSE and MoCA. The volume of the Left lobule VI (GM) exhibited a positive correlation with the MMSE score, but no significant correlation was found with the MoCA score. On the other hand, there was no significant correlation observed between the volume of the Right Crus II (WM) and the two cognitive scale scores mentioned above. Furthermore, it was observed that the MMSE was more effective than the MoCA in identifying epilepsy patients with cognitive impairment. Conclusion This study supported previous research indicating that temporal lobe epilepsy (TLE) is linked to structural changes in the cerebellum, specifically affecting the volume of both gray and white matter. These findings offer valuable insights into the neurobiology of TLE and hold potential to inform the development of enhanced diagnostic methods and more effective treatment approaches.
Collapse
Affiliation(s)
- Yini Chen
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingyu Pan
- Department of Neurology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Andong Lin
- Department of Neurology, Taizhou Municipal Hospital, Taizhou, China
| | - Lu Sun
- Department of Neurology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yufei Li
- Department of Neurology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Hongsen Lin
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Renwang Pu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Wang
- Department of Neurology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yiwei Qi
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bo Sun
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
38
|
Winter S, Mahzarnia A, Anderson RJ, Han ZY, Tremblay J, Stout J, Moon HS, Marcellino D, Dunson DB, Badea A. APOE, Immune Factors, Sex, and Diet Interact to Shape Brain Networks in Mouse Models of Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.04.560954. [PMID: 39005377 PMCID: PMC11244909 DOI: 10.1101/2023.10.04.560954] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Alzheimer's disease (AD) presents complex challenges due to its multifactorial nature, poorly understood etiology, and late detection. The mechanisms through which genetic, fixed and modifiable risk factors influence susceptibility to AD are under intense investigation, yet the impact of unique risk factors on brain networks is difficult to disentangle, and their interactions remain unclear. To model multiple risk factors including APOE genotype, age, sex, diet, and immunity we leveraged mice expressing the human APOE and NOS2 genes, conferring a reduced immune response compared to mouse Nos2. Employing graph analyses of brain connectomes derived from accelerated diffusion-weighted MRI, we assessed the global and local impact of risk factors in the absence of AD pathology. Aging and a high-fat diet impacted extensive networks comprising AD-vulnerable regions, including the temporal association cortex, amygdala, and the periaqueductal gray, involved in stress responses. Sex impacted networks including sexually dimorphic regions (thalamus, insula, hypothalamus) and key memory-processing areas (fimbria, septum). APOE genotypes modulated connectivity in memory, sensory, and motor regions, while diet and immunity both impacted the insula and hypothalamus. Notably, these risk factors converged on a circuit comprising 63 of 54,946 total connections (0.11% of the connectome), highlighting shared vulnerability amongst multiple AD risk factors in regions essential for sensory integration, emotional regulation, decision making, motor coordination, memory, homeostasis, and interoception. These network-based biomarkers hold translational value for distinguishing high-risk versus low-risk participants at preclinical AD stages, suggest circuits as potential therapeutic targets, and advance our understanding of network fingerprints associated with AD risk. Significance Statement Current interventions for Alzheimer's disease (AD) do not provide a cure, and are delivered years after neuropathological onset. Addressing the impact of risk factors on brain networks holds promises for early detection, prevention, and revealing putative therapeutic targets at preclinical stages. We utilized six mouse models to investigate the impact of factors, including APOE genotype, age, sex, immunity, and diet, on brain networks. Large structural connectomes were derived from high resolution compressed sensing diffusion MRI. A highly parallelized graph classification identified subnetworks associated with unique risk factors, revealing their network fingerprints, and a common network composed of 63 connections with shared vulnerability to all risk factors. APOE genotype specific immune signatures support the design of interventions tailored to risk profiles.
Collapse
Affiliation(s)
- Steven Winter
- Statistical Science, Trinity School, Duke University, Durham, NC, 27710 USA
| | - Ali Mahzarnia
- Department of Radiology, Duke University School of Medicine. Durham, NC, 27710. USA
| | - Robert J Anderson
- Department of Radiology, Duke University School of Medicine. Durham, NC, 27710. USA
| | - Zay Yar Han
- Department of Radiology, Duke University School of Medicine. Durham, NC, 27710. USA
| | - Jessica Tremblay
- Department of Radiology, Duke University School of Medicine. Durham, NC, 27710. USA
| | - Jacques Stout
- Duke UNC Brain Imaging and Analysis Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Hae Sol Moon
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Daniel Marcellino
- Department of Medical and Translational Biology, Umeå University, Umeå, 901 87, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, 22184, Sweden
| | - David B. Dunson
- Statistical Science, Trinity School, Duke University, Durham, NC, 27710 USA
| | - Alexandra Badea
- Department of Radiology, Duke University School of Medicine. Durham, NC, 27710. USA
- Duke UNC Brain Imaging and Analysis Center, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
- Department of Neurology, Duke University School of Medicine. Durham, NC, 27710, USA
| |
Collapse
|
39
|
Yang C, Liu G, Chen X, Le W. Cerebellum in Alzheimer's disease and other neurodegenerative diseases: an emerging research frontier. MedComm (Beijing) 2024; 5:e638. [PMID: 39006764 PMCID: PMC11245631 DOI: 10.1002/mco2.638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
The cerebellum is crucial for both motor and nonmotor functions. Alzheimer's disease (AD), alongside other dementias such as vascular dementia (VaD), Lewy body dementia (DLB), and frontotemporal dementia (FTD), as well as other neurodegenerative diseases (NDs) like Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and spinocerebellar ataxias (SCA), are characterized by specific and non-specific neurodegenerations in central nervous system. Previously, the cerebellum's significance in these conditions was underestimated. However, advancing research has elevated its profile as a critical node in disease pathology. We comprehensively review the existing evidence to elucidate the relationship between cerebellum and the aforementioned diseases. Our findings reveal a growing body of research unequivocally establishing a link between the cerebellum and AD, other forms of dementia, and other NDs, supported by clinical evidence, pathological and biochemical profiles, structural and functional neuroimaging data, and electrophysiological findings. By contrasting cerebellar observations with those from the cerebral cortex and hippocampus, we highlight the cerebellum's distinct role in the disease processes. Furthermore, we also explore the emerging therapeutic potential of targeting cerebellum for the treatment of these diseases. This review underscores the importance of the cerebellum in these diseases, offering new insights into the disease mechanisms and novel therapeutic strategies.
Collapse
Affiliation(s)
- Cui Yang
- Institute of Neurology Sichuan Provincial People's Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| | - Guangdong Liu
- Institute of Neurology Sichuan Provincial People's Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| | - Xi Chen
- Institute of Neurology Sichuan Provincial People's Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| | - Weidong Le
- Institute of Neurology Sichuan Provincial People's Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| |
Collapse
|
40
|
Guo X, Li J, Qi Y, Chen J, Jiang M, Zhu L, Liu Z, Wang H, Wang G, Wang X. Telomere length and micronuclei trajectories in APP/PS1 mouse model of Alzheimer's disease: Correlating with cognitive impairment and brain amyloidosis in a sexually dimorphic manner. Aging Cell 2024; 23:e14121. [PMID: 38450924 PMCID: PMC11113262 DOI: 10.1111/acel.14121] [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: 05/31/2023] [Revised: 12/31/2023] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Although studies have demonstrated that genome instability is accumulated in patients with Alzheimer's disease (AD), the specific types of genome instability linked to AD pathogenesis remain poorly understood. Here, we report the first characterization of the age- and sex-related trajectories of telomere length (TL) and micronuclei in APP/PS1 mice model and wild-type (WT) controls (C57BL/6). TL was measured in brain (prefrontal cortex, cerebellum, pituitary gland, and hippocampus), colon and skin, and MN was measured in bone marrow in 6- to 14-month-old mice. Variation in TL was attributable to tissue type, age, genotype and, to a lesser extent, sex. Compared to WT, APP/PS1 had a significantly shorter baseline TL across all examined tissues. TL was inversely associated with age in both genotypes and TL shortening was accelerated in brain of APP/PS1. Age-related increase of micronuclei was observed in both genotypes but was accelerated in APP/PS1. We integrated TL and micronuclei data with data on cognition performance and brain amyloidosis. TL and micronuclei were linearly correlated with cognition performance or Aβ40 and Aβ42 levels in both genotypes but to a greater extent in APP/PS1. These associations in APP/PS1 mice were dominantly driven by females. Together, our findings provide foundational knowledge to infer the TL and micronuclei trajectories in APP/PS1 mice during disease progression, and strongly support that TL attrition and micronucleation are tightly associated with AD pathogenesis in a female-biased manner.
Collapse
Affiliation(s)
- Xihan Guo
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Jianfei Li
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Yanmei Qi
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Juanlin Chen
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Minyan Jiang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Lina Zhu
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Zetong Liu
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Han Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Gongwu Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Xu Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
- Yeda Institute of Gene and Cell TherapyTaizhouZhejiangChina
| |
Collapse
|
41
|
Korte JA, Weakley A, Fernandez KD, Joiner WM, Fan AP. Neural Underpinnings of Learning in Dementia Populations: A Review of Motor Learning Studies Combined with Neuroimaging. J Cogn Neurosci 2024; 36:734-755. [PMID: 38285732 PMCID: PMC11934338 DOI: 10.1162/jocn_a_02116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
The intent of this review article is to serve as an overview of current research regarding the neural characteristics of motor learning in Alzheimer disease (AD) as well as prodromal phases of AD: at-risk populations, and mild cognitive impairment. This review seeks to provide a cognitive framework to compare various motor tasks. We will highlight the neural characteristics related to cognitive domains that, through imaging, display functional or structural changes because of AD progression. In turn, this motivates the use of motor learning paradigms as possible screening techniques for AD and will build upon our current understanding of learning abilities in AD populations.
Collapse
Affiliation(s)
- Jessica A. Korte
- Department of Biomedical Engineering, University of California, Davis
| | - Alyssa Weakley
- Department of Neurology, University of California, Davis
| | | | - Wilsaan M. Joiner
- Department of Biomedical Engineering, University of California, Davis
- Department of Neurology, University of California, Davis
- Department of Neurobiology, Physiology and Behavior, University of California, Davis
| | - Audrey P. Fan
- Department of Biomedical Engineering, University of California, Davis
- Department of Neurology, University of California, Davis
| |
Collapse
|
42
|
Biricioiu MR, Sarbu M, Ica R, Vukelić Ž, Clemmer DE, Zamfir AD. Human Cerebellum Gangliosides: A Comprehensive Analysis by Ion Mobility Tandem Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:683-695. [PMID: 38518248 DOI: 10.1021/jasms.3c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
The human cerebellum is an ultraspecialized region of the brain responsible for cognitive functions and movement coordination. The fine mechanisms through which the process of aging impacts such functions are not well understood; therefore, a rigorous exploration of this brain region at the molecular level is deemed necessary. Gangliosides, sialylated glycosphingolipids, highly and specifically expressed in the human central nervous system, represent possible molecular markers of cerebellum development and aging. In this context, for a comprehensive determination of development- and age-specific components, we have conducted here a comparative profiling and structural determination of the gangliosides expressed in fetal cerebellum in two intrauterine developmental stages and aged cerebellum by ion mobility separation (IMS) mass spectrometry (MS) and tandem MS (MS/MS). Due to the high sensitivity and efficiency of separation provided by IMS MS, no less than 551 chemically distinct species were identified, which represents 4.5 times more gangliosides than ever discovered in this brain region. The detailed assessment of fetal vs aged cerebellum gangliosidome showed marked discrepancies not only in the general number of the species expressed, but also in their sialylation patterns, the modifications of the glycan core, and the composition of the ceramides. All of these characteristics are potential markers of cerebellum development and aging. The structural analysis by collision-induced dissociation (CID) documented the occurrence of GD1b (d18:1/18:0) isomer in the fetal cerebellum in the second gestational trimester, with all probability of GQ1b (t18:1/18:0) in the near-term fetus and of GQ1b (d18:1/18:0) in aged cerebellum.
Collapse
Affiliation(s)
- Maria Roxana Biricioiu
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300224, Romania
- Department of Physics, West University of Timisoara, Timisoara 300223, Romania
| | - Mirela Sarbu
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300224, Romania
| | - Raluca Ica
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300224, Romania
| | - Željka Vukelić
- Department of Chemistry and Biochemistry, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - David E Clemmer
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Alina D Zamfir
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300224, Romania
- Institute for Research, Development and Innovation in Natural and Technical Sciences, Aurel Vlaicu University of Arad, Arad 310330, Romania
| |
Collapse
|
43
|
Magalhães TNC, Maldonado T, Jackson TB, Hicks TH, Herrejon IA, Rezende TJR, Symm AC, Bernard JA. Non-invasive neuromodulation of cerebello-hippocampal volume-behavior relationships. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587400. [PMID: 38617367 PMCID: PMC11014496 DOI: 10.1101/2024.03.29.587400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The study here explores the link between transcranial direct current stimulation (tDCS) and brain-behavior relationships. We propose that tDCS may indirectly influence the complex relationships between brain volume and behavior. We focused on the dynamics between the hippocampus (HPC) and cerebellum (CB) in cognitive processes, a relationship with significant implications for understanding memory and motor skills. Seventy-four young adults (mean age: 22±0.42 years, mean education: 14.7±0.25 years) were randomly assigned to receive either anodal, cathodal, or sham stimulation. Following stimulation, participants completed computerized tasks assessing working memory and sequence learning in a magnetic resonance imaging (MRI) environment. We investigated the statistical interaction between CB and HPC volumes. Our findings showed that individuals with larger cerebellar volumes had shorter reaction times (RT) on a high-load working memory task in the sham stimulation group. In contrast, the anodal stimulation group exhibited faster RTs during the low-load working memory condition. These RT differences were associated with the cortical volumetric interaction between CB-HPC. Literature suggests that anodal stimulation down-regulates the CB and here, those with larger volumes perform more quickly, suggesting the potential need for additional cognitive resources to compensate for cerebellar downregulation. This new insight suggests that tDCS can aid in revealing structure-function relationships, due to greater performance variability, especially in young adults. It may also reveal new targets of interest in the study of aging or in diseases where there is also greater behavioral variability.
Collapse
Affiliation(s)
- Thamires N. C. Magalhães
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Ted Maldonado
- Department of Psychology, Indiana State University, Terre Haute, United States of America
| | - T. Bryan Jackson
- Vanderbilt Memory & Alzheimer’s Center, Nashville, Tennessee, United States of America
| | - Tracey H. Hicks
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Ivan A. Herrejon
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Thiago J. R. Rezende
- Department of Neurology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Abigail C. Symm
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Jessica A. Bernard
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
44
|
Arleo A, Bareš M, Bernard JA, Bogoian HR, Bruchhage MMK, Bryant P, Carlson ES, Chan CCH, Chen LK, Chung CP, Dotson VM, Filip P, Guell X, Habas C, Jacobs HIL, Kakei S, Lee TMC, Leggio M, Misiura M, Mitoma H, Olivito G, Ramanoël S, Rezaee Z, Samstag CL, Schmahmann JD, Sekiyama K, Wong CHY, Yamashita M, Manto M. Consensus Paper: Cerebellum and Ageing. CEREBELLUM (LONDON, ENGLAND) 2024; 23:802-832. [PMID: 37428408 PMCID: PMC10776824 DOI: 10.1007/s12311-023-01577-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/08/2023] [Indexed: 07/11/2023]
Abstract
Given the key roles of the cerebellum in motor, cognitive, and affective operations and given the decline of brain functions with aging, cerebellar circuitry is attracting the attention of the scientific community. The cerebellum plays a key role in timing aspects of both motor and cognitive operations, including for complex tasks such as spatial navigation. Anatomically, the cerebellum is connected with the basal ganglia via disynaptic loops, and it receives inputs from nearly every region in the cerebral cortex. The current leading hypothesis is that the cerebellum builds internal models and facilitates automatic behaviors through multiple interactions with the cerebral cortex, basal ganglia and spinal cord. The cerebellum undergoes structural and functional changes with aging, being involved in mobility frailty and related cognitive impairment as observed in the physio-cognitive decline syndrome (PCDS) affecting older, functionally-preserved adults who show slowness and/or weakness. Reductions in cerebellar volume accompany aging and are at least correlated with cognitive decline. There is a strongly negative correlation between cerebellar volume and age in cross-sectional studies, often mirrored by a reduced performance in motor tasks. Still, predictive motor timing scores remain stable over various age groups despite marked cerebellar atrophy. The cerebello-frontal network could play a significant role in processing speed and impaired cerebellar function due to aging might be compensated by increasing frontal activity to optimize processing speed in the elderly. For cognitive operations, decreased functional connectivity of the default mode network (DMN) is correlated with lower performances. Neuroimaging studies highlight that the cerebellum might be involved in the cognitive decline occurring in Alzheimer's disease (AD), independently of contributions of the cerebral cortex. Grey matter volume loss in AD is distinct from that seen in normal aging, occurring initially in cerebellar posterior lobe regions, and is associated with neuronal, synaptic and beta-amyloid neuropathology. Regarding depression, structural imaging studies have identified a relationship between depressive symptoms and cerebellar gray matter volume. In particular, major depressive disorder (MDD) and higher depressive symptom burden are associated with smaller gray matter volumes in the total cerebellum as well as the posterior cerebellum, vermis, and posterior Crus I. From the genetic/epigenetic standpoint, prominent DNA methylation changes in the cerebellum with aging are both in the form of hypo- and hyper-methylation, and the presumably increased/decreased expression of certain genes might impact on motor coordination. Training influences motor skills and lifelong practice might contribute to structural maintenance of the cerebellum in old age, reducing loss of grey matter volume and therefore contributing to the maintenance of cerebellar reserve. Non-invasive cerebellar stimulation techniques are increasingly being applied to enhance cerebellar functions related to motor, cognitive, and affective operations. They might enhance cerebellar reserve in the elderly. In conclusion, macroscopic and microscopic changes occur in the cerebellum during the lifespan, with changes in structural and functional connectivity with both the cerebral cortex and basal ganglia. With the aging of the population and the impact of aging on quality of life, the panel of experts considers that there is a huge need to clarify how the effects of aging on the cerebellar circuitry modify specific motor, cognitive, and affective operations both in normal subjects and in brain disorders such as AD or MDD, with the goal of preventing symptoms or improving the motor, cognitive, and affective symptoms.
Collapse
Affiliation(s)
- Angelo Arleo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Martin Bareš
- First Department of Neurology, Faculty of Medicine, Masaryk University and St. Anne's Teaching Hospital, Brno, Czech Republic
- Department of Neurology, School of Medicine, University of Minnesota, Minneapolis, USA
| | - Jessica A Bernard
- Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX, 77843, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - Hannah R Bogoian
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| | - Muriel M K Bruchhage
- Department of Psychology, Stavanger University, Institute of Social Sciences, Kjell Arholms Gate 41, 4021, Stavanger, Norway
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Centre for Neuroimaging Sciences, Box 89, De Crespigny Park, London, PO, SE5 8AF, UK
- Rhode Island Hospital, Department for Diagnostic Imaging, 1 Hoppin St, Providence, RI, 02903, USA
- Department of Paediatrics, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Patrick Bryant
- Freie Universität Berlin, Fachbereich Mathematik und Informatik, Arnimallee 12, 14195, Berlin, Germany
| | - Erik S Carlson
- Department of Psychiatry and Behavioural Sciences, University of Washington, Seattle, WA, USA
- Geriatric Research, Education and Clinical Center, Veteran's Affairs Medical Center, Puget Sound, Seattle, WA, USA
| | - Chetwyn C H Chan
- Department of Psychology, The Education University of Hong Kong, New Territories, Tai Po, Hong Kong, China
| | - Liang-Kung Chen
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan
- Center for Geriatric and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan
- Taipei Municipal Gan-Dau Hospital (managed by Taipei Veterans General Hospital), Taipei, Taiwan
| | - Chih-Ping Chung
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Vonetta M Dotson
- Department of Psychology, Georgia State University, Atlanta, GA, USA
- Gerontology Institute, Georgia State University, Atlanta, GA, USA
| | - Pavel Filip
- Department of Neurology, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
- Center for Magnetic Resonance Research (CMRR), University of Minnesota, Minneapolis, MN, USA
| | - Xavier Guell
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Laboratory for Neuroanatomy and Cerebellar Neurobiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christophe Habas
- CHNO Des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, 75012, Paris, France
- Université Versailles St Quentin en Yvelines, Paris, France
| | - Heidi I L Jacobs
- School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, PO BOX 616, 6200, MD, Maastricht, The Netherlands
- Faculty of Psychology and Neuroscience, Department of Cognitive Neuroscience, Maastricht University, PO BOX 616, 6200, MD, Maastricht, The Netherlands
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Tatia M C Lee
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
- Laboratory of Neuropsychology and Human Neuroscience, Department of Psychology, The University of Hong Kong, Hong Kong, China
| | - Maria Leggio
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Ataxia Laboratory, I.R.C.C.S. Santa Lucia Foundation, Rome, Italy
| | - Maria Misiura
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| | - Hiroshi Mitoma
- Department of Medical Education, Tokyo Medical University, Tokyo, Japan
| | - Giusy Olivito
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Ataxia Laboratory, I.R.C.C.S. Santa Lucia Foundation, Rome, Italy
| | - Stephen Ramanoël
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
- Université Côte d'Azur, LAMHESS, Nice, France
| | - Zeynab Rezaee
- Noninvasive Neuromodulation Unit, Experimental Therapeutics & Pathophysiology Branch, National Institute of Mental Health, NIH, Bethesda, USA
| | - Colby L Samstag
- Department of Psychiatry and Behavioural Sciences, University of Washington, Seattle, WA, USA
- Geriatric Research, Education and Clinical Center, Veteran's Affairs Medical Center, Puget Sound, Seattle, WA, USA
| | - Jeremy D Schmahmann
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Laboratory for Neuroanatomy and Cerebellar Neurobiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ataxia Center, Cognitive Behavioural neurology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kaoru Sekiyama
- Graduate School of Advanced Integrated Studies in Human Survivability, Kyoto University, Kyoto, Japan
| | - Clive H Y Wong
- Department of Psychology, The Education University of Hong Kong, New Territories, Tai Po, Hong Kong, China
| | - Masatoshi Yamashita
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Osaka, Japan
| | - Mario Manto
- Service de Neurologie, Médiathèque Jean Jacquy, CHU-Charleroi, Charleroi, Belgium.
- Service des Neurosciences, University of Mons, Mons, Belgium.
| |
Collapse
|
45
|
Lyu W, Wu Y, Huynh KM, Ahmad S, Yap PT. A multimodal submillimeter MRI atlas of the human cerebellum. Sci Rep 2024; 14:5622. [PMID: 38453991 PMCID: PMC10920891 DOI: 10.1038/s41598-024-55412-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024] Open
Abstract
The human cerebellum is engaged in a broad array of tasks related to motor coordination, cognition, language, attention, memory, and emotional regulation. A detailed cerebellar atlas can facilitate the investigation of the structural and functional organization of the cerebellum. However, existing cerebellar atlases are typically limited to a single imaging modality with insufficient characterization of tissue properties. Here, we introduce a multifaceted cerebellar atlas based on high-resolution multimodal MRI, facilitating the understanding of the neurodevelopment and neurodegeneration of the cerebellum based on cortical morphology, tissue microstructure, and intra-cerebellar and cerebello-cerebral connectivity.
Collapse
Affiliation(s)
- Wenjiao Lyu
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Ye Wu
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Khoi Minh Huynh
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Sahar Ahmad
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Pew-Thian Yap
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA.
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
46
|
López-Ortiz S, Caruso G, Emanuele E, Menéndez H, Peñín-Grandes S, Guerrera CS, Caraci F, Nisticò R, Lucia A, Santos-Lozano A, Lista S. Digging into the intrinsic capacity concept: Can it be applied to Alzheimer's disease? Prog Neurobiol 2024; 234:102574. [PMID: 38266702 DOI: 10.1016/j.pneurobio.2024.102574] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Historically, aging research has largely centered on disease pathology rather than promoting healthy aging. The World Health Organization's (WHO) policy framework (2015-2030) underscores the significance of fostering the contributions of older individuals to their families, communities, and economies. The WHO has introduced the concept of intrinsic capacity (IC) as a key metric for healthy aging, encompassing five primary domains: locomotion, vitality, sensory, cognitive, and psychological. Past AD research, constrained by methodological limitations, has focused on single outcome measures, sidelining the complexity of the disease. Our current scientific milieu, however, is primed to adopt the IC concept. This is due to three critical considerations: (I) the decline in IC is linked to neurocognitive disorders, including AD, (II) cognition, a key component of IC, is deeply affected in AD, and (III) the cognitive decline associated with AD involves multiple factors and pathophysiological pathways. Our study explores the application of the IC concept to AD patients, offering a comprehensive model that could revolutionize the disease's diagnosis and prognosis. There is a dearth of information on the biological characteristics of IC, which are a result of complex interactions within biological systems. Employing a systems biology approach, integrating omics technologies, could aid in unraveling these interactions and understanding IC from a holistic viewpoint. This comprehensive analysis of IC could be leveraged in clinical settings, equipping healthcare providers to assess AD patients' health status more effectively and devise personalized therapeutic interventions in accordance with the precision medicine paradigm. We aimed to determine whether the IC concept could be extended from older individuals to patients with AD, thereby presenting a model that could significantly enhance the diagnosis and prognosis of this disease.
Collapse
Affiliation(s)
- Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | | | - Héctor Menéndez
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Saúl Peñín-Grandes
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Claudia Savia Guerrera
- Department of Educational Sciences, University of Catania, 95125 Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", 00133 Rome, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, 00143 Rome, Italy
| | - Alejandro Lucia
- Research Institute of the Hospital 12 de Octubre ('imas12'), 28041 Madrid, Spain; Faculty of Sport Sciences, European University of Madrid, 28670 Villaviciosa de Odón, Madrid, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), 28029 Madrid, Spain
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain; Research Institute of the Hospital 12 de Octubre ('imas12'), 28041 Madrid, Spain
| | - Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain.
| |
Collapse
|
47
|
Chen Y, Spina S, Callahan P, Grinberg LT, Seeley WW, Rosen HJ, Kramer JH, Miller BL, Rankin KP. Pathology-specific patterns of cerebellar atrophy in neurodegenerative disorders. Alzheimers Dement 2024; 20:1771-1783. [PMID: 38109286 PMCID: PMC10984510 DOI: 10.1002/alz.13551] [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: 06/27/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 12/20/2023]
Abstract
INTRODUCTION Associations of cerebellar atrophy with specific neuropathologies in Alzheimer's disease and related dementias (ADRD) have not been systematically analyzed. This study examined cerebellar gray matter volume across major pathological subtypes of ADRD. METHODS Cerebellar gray matter volume was examined using voxel-based morphometry in 309 autopsy-proven ADRD cases and 80 healthy controls. ADRD subtypes included AD, mixed Lewy body disease and AD (LBD-AD), and frontotemporal lobar degeneration (FTLD). Clinical function was assessed using the Clinical Dementia Rating (CDR) scale. RESULTS Distinct patterns of cerebellar atrophy were observed in all ADRD subtypes. Significant cerebellar gray matter changes appeared in the early stages of most subtypes and the very early stages of AD, LBD-AD, FTLD-TDP type A, and progressive supranuclear palsy. Cortical atrophy positively predicted cerebellar atrophy across all subtypes. DISCUSSION Our findings establish pathology-specific profiles of cerebellar atrophy in ADRD and propose cerebellar neuroimaging as a non-invasive biomarker for differential diagnosis and disease monitoring. HIGHLIGHTS Cerebellar atrophy was examined in 309 patients with autopsy-proven neurodegeneration. Distinct patterns of cerebellar atrophy are found in all pathological subtypes of Alzheimer's disease and related dementias (ADRD). Cerebellar atrophy is seen in early-stage (Clinical Dementia Rating [CDR] ≤1) AD, Lewy body dementia (LBD), frontotemporal lobar degeneration with tau-positive inclusion (FTLD-tau), and FTLD-transactive response DNA binding protein (FTLD-TDP). Cortical atrophy positively predicts cerebellar atrophy across all neuropathologies.
Collapse
Affiliation(s)
- Yu Chen
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Salvatore Spina
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Patrick Callahan
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lea T. Grinberg
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - William W. Seeley
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Howard J. Rosen
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Joel H. Kramer
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Bruce L. Miller
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Katherine P. Rankin
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
48
|
Bloch L, Friedrich CM. Systematic comparison of 3D Deep learning and classical machine learning explanations for Alzheimer's Disease detection. Comput Biol Med 2024; 170:108029. [PMID: 38308870 DOI: 10.1016/j.compbiomed.2024.108029] [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: 07/09/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Black-box deep learning (DL) models trained for the early detection of Alzheimer's Disease (AD) often lack systematic model interpretation. This work computes the activated brain regions during DL and compares those with classical Machine Learning (ML) explanations. The architectures used for DL were 3D DenseNets, EfficientNets, and Squeeze-and-Excitation (SE) networks. The classical models include Random Forests (RFs), Support Vector Machines (SVMs), eXtreme Gradient Boosting (XGBoost), Light Gradient Boosting (LightGBM), Decision Trees (DTs), and Logistic Regression (LR). For explanations, SHapley Additive exPlanations (SHAP) values, Local Interpretable Model-agnostic Explanations (LIME), Gradient-weighted Class Activation Mapping (GradCAM), GradCAM++ and permutation-based feature importance were implemented. During interpretation, correlated features were consolidated into aspects. All models were trained on the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset. The validation includes internal and external validation on the Australian Imaging and Lifestyle flagship study of Ageing (AIBL) and the Open Access Series of Imaging Studies (OASIS). DL and ML models reached similar classification performances. Regarding the brain regions, both types focus on different regions. The ML models focus on the inferior and middle temporal gyri, and the hippocampus, and amygdala regions previously associated with AD. The DL models focus on a wider range of regions including the optical chiasm, the entorhinal cortices, the left and right vessels, and the 4th ventricle which were partially associated with AD. One explanation for the differences is the input features (textures vs. volumes). Both types show reasonable similarity to a ground truth Voxel-Based Morphometry (VBM) analysis. Slightly higher similarities were measured for ML models.
Collapse
Affiliation(s)
- Louise Bloch
- Department of Computer Science, University of Applied Sciences and Arts Dortmund (FH Dortmund), Emil-Figge-Straße 42, Dortmund, 44227, North Rhine-Westphalia, Germany; Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, Hufelandstraße 55, Essen, 45122, North Rhine-Westphalia, Germany; Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen, Hufelandstraße 55, Essen, 45122, North Rhine-Westphalia, Germany.
| | - Christoph M Friedrich
- Department of Computer Science, University of Applied Sciences and Arts Dortmund (FH Dortmund), Emil-Figge-Straße 42, Dortmund, 44227, North Rhine-Westphalia, Germany; Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, Hufelandstraße 55, Essen, 45122, North Rhine-Westphalia, Germany.
| |
Collapse
|
49
|
Friese S, Ranzini G, Tuchtenhagen M, Lossow K, Hertel B, Pohl G, Ebert F, Bornhorst J, Kipp AP, Schwerdtle T. Long-term suboptimal dietary trace element supply does not affect trace element homeostasis in murine cerebellum. Metallomics 2024; 16:mfae003. [PMID: 38299785 PMCID: PMC10873500 DOI: 10.1093/mtomcs/mfae003] [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: 07/28/2023] [Accepted: 12/14/2023] [Indexed: 02/02/2024]
Abstract
The ageing process is associated with alterations of systemic trace element (TE) homeostasis increasing the risk, e.g. neurodegenerative diseases. Here, the impact of long-term modulation of dietary intake of copper, iron, selenium, and zinc was investigated in murine cerebellum. Four- and 40-wk-old mice of both sexes were supplied with different amounts of those TEs for 26 wk. In an adequate supply group, TE concentrations were in accordance with recommendations for laboratory mice while suboptimally supplied animals received only limited amounts of copper, iron, selenium, and zinc. An additional age-adjusted group was fed selenium and zinc in amounts exceeding recommendations. Cerebellar TE concentrations were measured by inductively coupled plasma-tandem mass spectrometry. Furthermore, the expression of genes involved in TE transport, DNA damage response, and DNA repair as well as selected markers of genomic stability [8-oxoguanine, incision efficiency toward 8-oxoguanine, 5-hydroxyuracil, and apurinic/apyrimidinic sites and global DNA (hydroxy)methylation] were analysed. Ageing resulted in a mild increase of iron and copper concentrations in the cerebellum, which was most pronounced in the suboptimally supplied groups. Thus, TE changes in the cerebellum were predominantly driven by age and less by nutritional intervention. Interestingly, deviation from adequate TE supply resulted in higher manganese concentrations of female mice even though the manganese supply itself was not modulated. Parameters of genomic stability were neither affected by age, sex, nor diet. Overall, this study revealed that suboptimal dietary TE supply does not substantially affect TE homeostasis in the murine cerebellum.
Collapse
Affiliation(s)
- Sharleen Friese
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Giovanna Ranzini
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Max Tuchtenhagen
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Kristina Lossow
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743 Jena, Germany
| | - Barbara Hertel
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Gabriele Pohl
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Franziska Ebert
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Julia Bornhorst
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstraße 20, 42119 Wuppertal, Germany
| | - Anna Patricia Kipp
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743 Jena, Germany
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| |
Collapse
|
50
|
Herrejon IA, Jackson TB, Hicks TH, Bernard JA. Functional Connectivity Differences in Distinct Dentato-Cortical Networks in Alzheimer's Disease and Mild Cognitive Impairment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578249. [PMID: 38352603 PMCID: PMC10862898 DOI: 10.1101/2024.02.02.578249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Recent research has implicated the cerebellum in Alzheimer's disease (AD), and cerebrocerebellar network connectivity is emerging as a possible contributor to symptom severity. The cerebellar dentate nucleus (DN) has parallel motor and non-motor sub-regions that project to motor and frontal regions of the cerebral cortex, respectively. These distinct dentato-cortical networks have been delineated in the non-human primate and human brain. Importantly, cerebellar regions prone to atrophy in AD are functionally connected to atrophied regions of the cerebral cortex, suggesting that dysfunction perhaps occurs at a network level. Investigating functional connectivity (FC) alterations of the DN is a crucial step in understanding the cerebellum in AD and in mild cognitive impairment (MCI). Inclusion of this latter group stands to provide insights into cerebellar contributions prior to diagnosis of AD. The present study investigated FC differences in dorsal (dDN) and ventral (vDN) DN networks in MCI and AD relative to cognitively normal participants (CN) and relationships between FC and behavior. Our results showed patterns indicating both higher and lower functional connectivity in both dDN and vDN in AD compared to CN. However, connectivity in the AD group was lower when compared to MCI. We argue that these findings suggest that the patterns of higher FC in AD may act as a compensatory mechanism. Additionally, we found associations between the individual networks and behavior. There were significant interactions between dDN connectivity and motor symptoms. However, both DN seeds were associated with cognitive task performance. Together, these results indicate that cerebellar DN networks are impacted in AD, and this may impact behavior. In concert with the growing body of literature implicating the cerebellum in AD, our work further underscores the importance of investigations of this region. We speculate that much like in psychiatric diseases such as schizophrenia, cerebellar dysfunction results in negative impacts on thought and the organization therein. Further, this is consistent with recent arguments that the cerebellum provides crucial scaffolding for cognitive function in aging. Together, our findings stand to inform future clinical work in the diagnosis and understanding of this disease.
Collapse
Affiliation(s)
- Ivan A. Herrejon
- Department of Psychological and Brain Sciences Texas A&M University
| | - T. Bryan Jackson
- Department of Psychological and Brain Sciences Texas A&M University
- Vanderbilt Memory and Alzheimer’s Center Vanderbilt University Medical Center
| | - Tracey H. Hicks
- Department of Psychological and Brain Sciences Texas A&M University
| | - Jessica A. Bernard
- Department of Psychological and Brain Sciences Texas A&M University
- Texas A&M Institute for Neuroscience Texas A&M University
| | | |
Collapse
|