1
|
Wu K, Gollo LL. Mapping and modeling age-related changes in intrinsic neural timescales. Commun Biol 2025; 8:167. [PMID: 39901043 PMCID: PMC11791184 DOI: 10.1038/s42003-025-07517-x] [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: 09/27/2024] [Accepted: 01/10/2025] [Indexed: 02/05/2025] Open
Abstract
Intrinsic timescales of brain regions exhibit heterogeneity, escalating with hierarchical levels, and are crucial for the temporal integration of external stimuli. Aging, often associated with cognitive decline, involves progressive neuronal and synaptic loss, reshaping brain structure and dynamics. However, the impact of these structural changes on temporal coding in the aging brain remains unclear. We mapped intrinsic timescales and gray matter volume (GMV) using magnetic resonance imaging (MRI) in young and elderly adults. We found shorter intrinsic timescales across multiple large-scale functional networks in the elderly cohort, and a significant positive association between intrinsic timescales and GMV. Additionally, age-related decline in performance on visual discrimination tasks was linked to a reduction in intrinsic timescales in the cuneus. To explain these age-related shifts, we developed an age-dependent spiking neuron network model. In younger subjects, brain regions were near a critical branching regime, while regions in elderly subjects had fewer neurons and synapses, pushing the dynamics toward a subcritical regime. The model accurately reproduced the empirical results, showing longer intrinsic timescales in young adults due to critical slowing down. Our findings reveal how age-related structural brain changes may drive alterations in brain dynamics, offering testable predictions and informing possible interventions targeting cognitive decline.
Collapse
Affiliation(s)
- Kaichao Wu
- Brain Networks and Modelling Laboratory and The Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
- Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia
| | - Leonardo L Gollo
- Brain Networks and Modelling Laboratory and The Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.
- Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia.
- Instituto de Física Interdisciplinary Sistemas Complejos, IFISC (UIB-CSIC), Campus Universitat de les Illes Balears, Palma de Mallorca, Spain.
| |
Collapse
|
2
|
Nicks R, Shah A, Stathas SA, Kirsch D, Horowitz SM, Saltiel N, Calderazzo SM, Butler MLMD, Cormier KA, Aytan N, Tu-Zahra F, Mathias R, Faheem F, Marcus S, Spurlock E, Fishbein L, Esnault CD, Boden A, Rosen G, Xia W, Daley S, Meng G, Martin BR, Daneshvar DH, Nowinski CJ, Alosco ML, Mez J, Tripodis Y, Huber BR, Alvarez VE, Cherry JD, McKee AC, Stein TD. Neurodegeneration in the cortical sulcus is a feature of chronic traumatic encephalopathy and associated with repetitive head impacts. Acta Neuropathol 2024; 148:79. [PMID: 39643767 PMCID: PMC11624223 DOI: 10.1007/s00401-024-02833-8] [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: 06/19/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024]
Abstract
Neurodegeneration is a seminal feature of many neurological disorders. Chronic traumatic encephalopathy (CTE) is caused by repetitive head impacts (RHI) and is characterized by sulcal tau pathology. However, quantitative assessments of regional neurodegeneration in CTE have not been described. In this study, we quantified three key neurodegenerative measures, including cortical thickness, neuronal density, and synaptic proteins, in contact sport athletes (n = 185) and non-athlete controls (n = 52) within the sulcal depth, middle, and gyral crest of the dorsolateral frontal cortex. Cortical thickness and neuronal density were decreased within the sulcus in CTE compared to controls (p's < 0.05). Measurements of synaptic proteins within the gyral crest showed a reduction of α-synuclein with CTE stage (p = 0.002) and variable changes in PSD-95 density. After adjusting for age, multiple linear regression models demonstrated a strong association between the duration of contact sports play and cortical thinning (p = 0.001) and neuronal loss (p = 0.032) within the sulcus. Additional regression models, adjusted for tau pathology, suggest that within the sulcus, the duration of play was associated with neuronal loss predominantly through tau pathology. In contrast, the association of duration of play with cortical thinning was minimally impacted by tau pathology. Overall, CTE is associated with cortical atrophy and a predominant sulcal neurodegeneration. Furthermore, the duration of contact sports play is associated with measures of neurodegeneration that are more severe in the cortical sulcus and may occur through tau-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Raymond Nicks
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Arsal Shah
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Spiro Anthony Stathas
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Daniel Kirsch
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Sarah M Horowitz
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Nicole Saltiel
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Samantha M Calderazzo
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Morgane L M D Butler
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kerry A Cormier
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Nurgul Aytan
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Fatima Tu-Zahra
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Rebecca Mathias
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Farwa Faheem
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | | | - Elizabeth Spurlock
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Lucas Fishbein
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Camille D Esnault
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Alexandra Boden
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Grace Rosen
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Weiming Xia
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Department of Biological Sciences, Kennedy College of Science, University of Massachusetts, Lowell, MA, USA
| | - Sarah Daley
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | | | - Brett R Martin
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Daniel H Daneshvar
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Massachusetts General Hospital, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Mass General Brigham-Spaulding Rehabilitation, Charlestown, MA, USA
| | - Christopher J Nowinski
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Concussion Legacy Foundation, Boston, MA, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Bertrand R Huber
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Victor E Alvarez
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Jonathan D Cherry
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ann C McKee
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA.
- VA Boston Healthcare System, Boston, MA, USA.
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- VA Bedford Healthcare System, Bedford, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
3
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
4
|
Yan Y, He X, Xu Y, Zhao F, Peng J, Shao Y. Pattern of heterogeneity in normal brain ageing: screening for mild cognitive impairment and its risk of progression with a radiomics model. Age Ageing 2024; 53:afae140. [PMID: 38984695 DOI: 10.1093/ageing/afae140] [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: 11/23/2023] [Revised: 05/01/2024] [Indexed: 07/11/2024] Open
Abstract
PURPOSE This study aimed to develop a normal brain ageing model based on magnetic resonance imaging and radiomics, therefore identifying radscore, an imaging indicator representing white matter heterogeneity and exploring the significance of radscore in detecting people's cognitive changes. METHODS Three hundred sixty cognitively normal (CN) subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database and 105 CN subjects from the Parkinson's Progression Markers Initiative database were used to develop the model. In ADNI, 230 mild cognitive impairment (MCI) subjects were matched with 230 CN old-aged subjects to evaluate their heterogeneity difference. One hundred four MCI subjects with 48 months of follow-up were divided into low and high heterogeneity groups. Kaplan-Meier survival curve analysis was used to observe the importance of heterogeneity results for predicting MCI progression. RESULTS The area under the receiver operating characteristic curve of the model in the training, internal test and external test sets was 0.7503, 0.7512 and 0.7514, respectively. There was a significantly positive correlation between age and radscore of CN subjects (r = 0.501; P < .001). The radscore of MCI subjects was significantly higher than that of matched CN subjects (P < .001). The median radscore ratios of MCI to CN from four age groups (66-70y, 71-75y, 76-80y and 81-85y) were 1.611, 1.760, 1.340 and 1.266, respectively. The probability to progression of low and high heterogeneity groups had a significant difference (P = .002). CONCLUSION When radscore is significantly higher than that of normal ageing, it is necessary to alert the possibility of cognitive impairment and deterioration.
Collapse
Affiliation(s)
- Yuting Yan
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Xiaodong He
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Yuyun Xu
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Fanfan Zhao
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Jiaxuan Peng
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Yuan Shao
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| |
Collapse
|
5
|
Park J, Wang J, Guan W, Gjesteby LA, Pollack D, Kamentsky L, Evans NB, Stirman J, Gu X, Zhao C, Marx S, Kim ME, Choi SW, Snyder M, Chavez D, Su-Arcaro C, Tian Y, Park CS, Zhang Q, Yun DH, Moukheiber M, Feng G, Yang XW, Keene CD, Hof PR, Ghosh SS, Frosch MP, Brattain LJ, Chung K. Integrated platform for multiscale molecular imaging and phenotyping of the human brain. Science 2024; 384:eadh9979. [PMID: 38870291 PMCID: PMC11830150 DOI: 10.1126/science.adh9979] [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] [Received: 03/28/2023] [Accepted: 04/22/2024] [Indexed: 06/15/2024]
Abstract
Understanding cellular architectures and their connectivity is essential for interrogating system function and dysfunction. However, we lack technologies for mapping the multiscale details of individual cells and their connectivity in the human organ-scale system. We developed a platform that simultaneously extracts spatial, molecular, morphological, and connectivity information of individual cells from the same human brain. The platform includes three core elements: a vibrating microtome for ultraprecision slicing of large-scale tissues without losing cellular connectivity (MEGAtome), a polymer hydrogel-based tissue processing technology for multiplexed multiscale imaging of human organ-scale tissues (mELAST), and a computational pipeline for reconstructing three-dimensional connectivity across multiple brain slabs (UNSLICE). We applied this platform for analyzing human Alzheimer's disease pathology at multiple scales and demonstrating scalable neural connectivity mapping in the human brain.
Collapse
Affiliation(s)
- Juhyuk Park
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
| | - Ji Wang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Webster Guan
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | | | | | - Lee Kamentsky
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Nicholas B. Evans
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Jeff Stirman
- LifeCanvas Technologies, Cambridge, MA 02141, USA
| | - Xinyi Gu
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA
| | - Chuanxi Zhao
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Slayton Marx
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Minyoung E. Kim
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Seo Woo Choi
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | | | - David Chavez
- MIT Lincoln Laboratory, Lexington, MA 02421, USA
| | - Clover Su-Arcaro
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Yuxuan Tian
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Chang Sin Park
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience, University of California, Los Angeles, CA 90024, USA
| | - Qiangge Zhang
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA
| | - Dae Hee Yun
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Mira Moukheiber
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Guoping Feng
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA
| | - X. William Yang
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience, University of California, Los Angeles, CA 90024, USA
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98115, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Center for Discovery and Innovation, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Satrajit S. Ghosh
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02114, USA
| | - Matthew P. Frosch
- C. S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Kwanghun Chung
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| |
Collapse
|
6
|
Ferrer I. Historical review: The golden age of the Golgi method in human neuropathology. J Neuropathol Exp Neurol 2024; 83:375-395. [PMID: 38622902 DOI: 10.1093/jnen/nlae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
Golgi methods were used to study human neuropathology in the 1970s, 1980s, and 1990s of the last century. Although a relatively small number of laboratories applied these methods, their impact was crucial by increasing knowledge about: (1) the morphology, orientation, and localization of neurons in human cerebral and cerebellar malformations and ganglionic tumors, and (2) the presence of abnormal structures including large and thin spines (spine dysgenesis) in several disorders linked to mental retardation, focal enlargements of the axon hillock and dendrites (meganeurites) in neuronal storage diseases, growth cone-like appendages in Alzheimer disease, as well as abnormal structures in other dementias. Although there were initial concerns about their reliability, reduced dendritic branches and dendritic spines were identified as common alterations in mental retardation, dementia, and other pathological conditions. Similar observations in appropriate experimental models have supported many abnormalities that were first identified using Golgi methods in human material. Moreover, electron microscopy, immunohistochemistry, fluorescent tracers, and combined methods have proven the accuracy of pioneering observations uniquely visualized as 3D images of fully stained individual neurons. Although Golgi methods had their golden age many years ago, these methods may still be useful complementary tools in human neuropathology.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de LLobregat, Spain
| |
Collapse
|
7
|
Sultana R, Butterfield DA. Protein Oxidation in Aging and Alzheimer's Disease Brain. Antioxidants (Basel) 2024; 13:574. [PMID: 38790679 PMCID: PMC11117785 DOI: 10.3390/antiox13050574] [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: 04/09/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Proteins are essential molecules that play crucial roles in maintaining cellular homeostasis and carrying out biological functions such as catalyzing biochemical reactions, structural proteins, immune response, etc. However, proteins also are highly susceptible to damage by reactive oxygen species (ROS) and reactive nitrogen species (RNS). In this review, we summarize the role of protein oxidation in normal aging and Alzheimer's disease (AD). The major emphasis of this review article is on the carbonylation and nitration of proteins in AD and mild cognitive impairment (MCI). The oxidatively modified proteins showed a strong correlation with the reported changes in brain structure, carbohydrate metabolism, synaptic transmission, cellular energetics, etc., of both MCI and AD brains compared to the controls. Some proteins were found to be common targets of oxidation and were observed during the early stages of AD, suggesting that those changes might be critical in the onset of symptoms and/or formation of the pathological hallmarks of AD. Further studies are required to fully elucidate the role of protein oxidation and nitration in the progression and pathogenesis of AD.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd., Richardson, TX 75080, USA;
| | - D. Allan Butterfield
- Department of Chemistry, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
8
|
Enduru N, Fernandes BS, Zhao Z. Dissecting the shared genetic architecture between Alzheimer's disease and frailty: a cross-trait meta-analyses of genome-wide association studies. Front Genet 2024; 15:1376050. [PMID: 38706793 PMCID: PMC11069310 DOI: 10.3389/fgene.2024.1376050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/04/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction: Frailty is the most common medical condition affecting the aging population, and its prevalence increases in the population aged 65 or more. Frailty is commonly diagnosed using the frailty index (FI) or frailty phenotype (FP) assessments. Observational studies have indicated the association of frailty with Alzheimer's disease (AD). However, the shared genetic and biological mechanism of these comorbidity has not been studied. Methods: To assess the genetic relationship between AD and frailty, we examined it at single nucleotide polymorphism (SNP), gene, and pathway levels. Results: Overall, 16 genome-wide significant loci (15 unique loci) (p meta-analysis < 5 × 10-8) and 22 genes (21 unique genes) were identified between AD and frailty using cross-trait meta-analysis. The 8 shared loci implicated 11 genes: CLRN1-AS1, CRHR1, FERMT2, GRK4, LINC01929, LRFN2, MADD, RP11-368P15.1, RP11-166N6.2, RNA5SP459, and ZNF652 between AD and FI, and 8 shared loci between AD and FFS implicated 11 genes: AFF3, C1QTNF4, CLEC16A, FAM180B, FBXL19, GRK4, LINC01104, MAD1L1, RGS12, ZDHHC5, and ZNF521. The loci 4p16.3 (GRK4) was identified in both meta-analyses. The colocalization analysis supported the results of our meta-analysis in these loci. The gene-based analysis revealed 80 genes between AD and frailty, and 4 genes were initially identified in our meta-analyses: C1QTNF4, CRHR1, MAD1L1, and RGS12. The pathway analysis showed enrichment for lipoprotein particle plasma, amyloid fibril formation, protein kinase regulator, and tau protein binding. Conclusion: Overall, our results provide new insights into the genetics of AD and frailty, suggesting the existence of non-causal shared genetic mechanisms between these conditions.
Collapse
Affiliation(s)
- Nitesh Enduru
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Brisa S. Fernandes
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
9
|
Morroni F, Caccamo A. Advances and Challenges in Gene Therapy for Alzheimer's Disease. J Alzheimers Dis 2024; 101:S417-S431. [PMID: 39422937 DOI: 10.3233/jad-230783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and behavioral impairments. Despite extensive research efforts, effective treatment options for AD remain limited. Recently, gene therapy has emerged as a promising avenue for targeted intervention in the pathogenesis of AD. This review will provide an overview of clinical and preclinical studies where gene therapy techniques have been utilized in the context of AD, highlighting their potential as novel therapeutic strategies. While challenges remain, ongoing research and technological advancement continue to enhance the potential of gene therapy as a targeted and personalized therapeutic approach for AD.
Collapse
Affiliation(s)
- Fabiana Morroni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
10
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
11
|
Zarifkar AH, Zarifkar A, Safaei S. Different paradigms of transcranial electrical stimulation induce structural changes in the CA1 region of the hippocampus in a rat model of Alzheimer's disease. Neurosci Lett 2024; 818:137570. [PMID: 38000774 DOI: 10.1016/j.neulet.2023.137570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023]
Abstract
One of the prominent sign of Alzheimer's disease (AD) is structural changes in the hippocampus. Recently, the new methods used to treat this disease is transcranial electrical stimulation (tES). This study evaluated the effect of four primary standards of tES, including tDCS, tACS, tRNS, and tPCS on beta-amyloid 25-35 (Aβ25-35)-induced structural changes in the CA1 region of hippocampus in male rats. For this purpose, rats weighing 250-275 g were selected, the cannula was embedded reciprocally into the hippocampi. Aβ25-35 (5 μg/ 2.5 ml/ day) was infused reciprocally for four continuous days.Then, animals were then given tES for 6 days.Subsequently, structural changes in the hippocampal CA1 were evaluated using the stereological method. Aβ25-35 resulted in loss of neurons (P < 0.01) and decreased hippocampal volume (P < 0.05). However, the administration of tES paradigms prevented these changes. The results proposed that through the improvement of hippocampal cell number and volume, tES paradigms can retain efficiency in remediating structural impairments in AD. From this, it can be concluded that other tES paradigms besides tDCS can also be considered for the treatment of AD.
Collapse
Affiliation(s)
- Amir Hossein Zarifkar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Biology Research Center, Larestan University of Medical Sciences, Larestan, Iran.
| | - Asadollah Zarifkar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Safaei
- Gerash Amir-al-Momenin Medical and Educational Center, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
12
|
Lu K, Li C, Liu J, Wang J, Li Y, He B, Li J, Zhang X, Wei M, Tian Y, Zhang R, Zhang C, Zhang Y. Impairments in endogenous AMPA receptor dynamics correlates with learning deficits in Alzheimer's disease model mice. Proc Natl Acad Sci U S A 2023; 120:e2303878120. [PMID: 37748061 PMCID: PMC10556575 DOI: 10.1073/pnas.2303878120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023] Open
Abstract
AMPA receptors (AMPARs) play a critical role in synaptic plasticity and learning and memory, and dysfunction or dysregulation of AMPARs could lead to various neurological and psychiatric disorders, such as Alzheimer's disease (AD). However, the dynamics and/or longitudinal changes of AMPARs in vivo during AD pathogenesis remain elusive. Here, employing 5xFAD SEP-GluA1 KI mice, we investigated endogenous AMPA receptor dynamics in a whisker deflection-associated Go/No-go learning paradigm. We found a significant increase in synaptosomal AMPA receptor subunits GluA1 in WT mice after learning, while no such changes were detected in 7-mo-old 5xFAD mice. Daily training led to an increase in endogenous spine surface GluA1 in Control mice, while this increase was absent in 5xFAD-KI mice which correlates with its learning defects in Go/No-go paradigm. Furthermore, we demonstrated that the onset of abnormal AMPAR dynamics corresponds temporally with microglia and astrocyte overactivation. Our results have shown that impairments in endogenous AMPA receptor dynamics play an important role in learning deficits in 5xFAD mice and AD pathogenesis.
Collapse
Affiliation(s)
- Kongjie Lu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Chenyang Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing100083, China
| | - Jinpeng Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Yongfeng Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Bin He
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Junzhao Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Xiaochen Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin300072, China
| | - Mengping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing100069, China
| | - Yonglu Tian
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
- School of Psychological and Cognitive Sciences, Peking University, Beijing100871, China
| | - Rong Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing100069, China
| | - Yong Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| |
Collapse
|
13
|
Falaki A, Cuadra C, Lewis MM, Prado-Rico JM, Huang X, Latash ML. Multi-muscle synergies in preparation for gait initiation in Parkinson's disease. Clin Neurophysiol 2023; 154:12-24. [PMID: 37524005 DOI: 10.1016/j.clinph.2023.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/20/2023] [Accepted: 06/25/2023] [Indexed: 08/02/2023]
Abstract
OBJECTIVE We investigated changes in indices of muscle synergies prior to gait initiation and the effects of gaze shift in patients with Parkinson's disease (PD). A long-term objective of the study is to develop a method for quantitative assessment of gait-initiation problems in PD. METHODS PD patients without clinical signs of postural instability and two control groups (age-matched and young) performed a gait initiation task in a self-paced manner, with and without a quick prior gaze shift produced by turning the head. Muscle groups with parallel scaling of activation levels (muscle modes) were identified as factors in the muscle activation space. Synergy index stabilizing center of pressure trajectory in the anterior-posterior and medio-lateral directions (indices of stability) was quantified in the muscle mode space. A drop in the synergy index in preparation to gait initiation (anticipatory synergy adjustment, ASA) was quantified. RESULTS Compared to the control groups, PD patients showed significantly smaller synergy indices and ASA for both directions of the center of pressure shift. Both PD and age-matched controls, but not younger controls, showed detrimental effects of the prior gaze shift on the ASA indices. CONCLUSIONS PD patients without clinically significant posture or gait disorders show impaired stability of the center of pressure and its diminished adjustment during gait initiation. SIGNIFICANCE The indices of stability and ASA may be useful to monitor pre-clinical gait disorders, and lower ASA may be relevant to emergence of freezing of gait in PD.
Collapse
Affiliation(s)
- Ali Falaki
- Department of Neurosciences, University of Montreal, Montreal, Quebec, Canada
| | - Cristian Cuadra
- Department of Physical Therapy, Emory University, Atlanta, GA, USA; Exercise and Rehabilitation Sciences Laboratory, School of Physical Therapy, Faculty of Rehabilitation Sciences, Universidad Andres Bello, 7591538 Santiago, Chile
| | - Mechelle M Lewis
- Department of Neurology, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, USA; Department of Pharmacology, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, USA
| | - Janina M Prado-Rico
- Department of Neurology, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, USA
| | - Xuemei Huang
- Department of Neurology, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, USA; Department of Pharmacology, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, USA; Department of Radiology, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, USA; Department of Neurosurgery, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, USA; Department of Kinesiology, The Pennsylvania State University, University Park, PA, USA
| | - Mark L Latash
- Department of Kinesiology, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
14
|
Eugenín J, Eugenín-von Bernhardi L, von Bernhardi R. Age-dependent changes on fractalkine forms and their contribution to neurodegenerative diseases. Front Mol Neurosci 2023; 16:1249320. [PMID: 37818457 PMCID: PMC10561274 DOI: 10.3389/fnmol.2023.1249320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
The chemokine fractalkine (FKN, CX3CL1), a member of the CX3C subfamily, contributes to neuron-glia interaction and the regulation of microglial cell activation. Fractalkine is expressed by neurons as a membrane-bound protein (mCX3CL1) that can be cleaved by extracellular proteases generating several sCX3CL1 forms. sCX3CL1, containing the chemokine domain, and mCX3CL1 have high affinity by their unique receptor (CX3CR1) which, physiologically, is only found in microglia, a resident immune cell of the CNS. The activation of CX3CR1contributes to survival and maturation of the neural network during development, glutamatergic synaptic transmission, synaptic plasticity, cognition, neuropathic pain, and inflammatory regulation in the adult brain. Indeed, the various CX3CL1 forms appear in some cases to serve an anti-inflammatory role of microglia, whereas in others, they have a pro-inflammatory role, aggravating neurological disorders. In the last decade, evidence points to the fact that sCX3CL1 and mCX3CL1 exhibit selective and differential effects on their targets. Thus, the balance in their level and activity will impact on neuron-microglia interaction. This review is focused on the description of factors determining the emergence of distinct fractalkine forms, their age-dependent changes, and how they contribute to neuroinflammation and neurodegenerative diseases. Changes in the balance among various fractalkine forms may be one of the mechanisms on which converge aging, chronic CNS inflammation, and neurodegeneration.
Collapse
Affiliation(s)
- Jaime Eugenín
- Facultad de Química y Biología, Departamento de Biología, Universidad de Santiago de Chile, USACH, Santiago, Chile
| | | | - Rommy von Bernhardi
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
15
|
Wang S, Wang L, Bu Q, Wei Q, Jiang L, Dai Y, Zhang N, Kuang W, Zhao Y, Cen X. Methamphetamine exposure drives cell cycle exit and aberrant differentiation in rat hippocampal-derived neurospheres. Front Pharmacol 2023; 14:1242109. [PMID: 37795025 PMCID: PMC10546213 DOI: 10.3389/fphar.2023.1242109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
Introduction: Methamphetamine (METH) abuse by pregnant drug addicts causes toxic effects on fetal neurodevelopment; however, the mechanism underlying such effect of METH is poorly understood. Methods: In the present study, we applied three-dimensional (3D) neurospheres derived from the embryonic rat hippocampal tissue to investigate the effect of METH on neurodevelopment. Through the combination of whole genome transcriptional analyses, the involved cell signalings were identified and investigated. Results: We found that METH treatment for 24 h significantly and concentration-dependently reduced the size of neurospheres. Analyses of genome-wide transcriptomic profiles found that those down-regulated differentially expressed genes (DEGs) upon METH exposure were remarkably enriched in the cell cycle progression. By measuring the cell cycle and the expression of cell cycle-related checkpoint proteins, we found that METH exposure significantly elevated the percentage of G0/G1 phase and decreased the levels of the proteins involved in the G1/S transition, indicating G0/G1 cell cycle arrest. Furthermore, during the early neurodevelopment stage of neurospheres, METH caused aberrant cell differentiation both in the neurons and astrocytes, and attenuated migration ability of neurospheres accompanied by increased oxidative stress and apoptosis. Conclusion: Our findings reveal that METH induces an aberrant cell cycle arrest and neuronal differentiation, impairing the coordination of migration and differentiation of neurospheres.
Collapse
Affiliation(s)
- Shaomin Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Wei
- Cell and Immunology Laboratory, Chengdu West China Frontier Pharmatech Co., Ltd., Chengdu, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ni Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weihong Kuang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Gonzalez‐Rodriguez M, Villar‐Conde S, Astillero‐Lopez V, Villanueva‐Anguita P, Ubeda‐Banon I, Flores‐Cuadrado A, Martinez‐Marcos A, Saiz‐Sanchez D. Human amygdala involvement in Alzheimer's disease revealed by stereological and dia-PASEF analysis. Brain Pathol 2023; 33:e13180. [PMID: 37331354 PMCID: PMC10467039 DOI: 10.1111/bpa.13180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/06/2023] [Indexed: 06/20/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of pathological amyloid-β (Aβ) and Tau proteins. According to the prion-like hypothesis, both proteins can seed and disseminate through brain regions through neural connections and glial cells. The amygdaloid complex (AC) is involved early in the disease, and its widespread connections with other brain regions indicate that it is a hub for propagating pathology. To characterize changes in the AC as well as the involvement of neuronal and glial cells in AD, a combined stereological and proteomic analysis was performed in non-Alzheimer's disease and AD human samples. The synaptic alterations identified by proteomic data analysis could be related to the volume reduction observed in AD by the Cavalieri probe without neuronal loss. The pathological markers appeared in a gradient pattern with the medial region (cortical nucleus, Co) being more affected than lateral regions, suggesting the relevance of connections in the distribution of the pathology among different brain regions. Generalized astrogliosis was observed in every AC nucleus, likely related to deposits of pathological proteins. Astrocytes might mediate phagocytic microglial activation, whereas microglia might play a dual role since protective and toxic phenotypes have been described. These results highlight the potential participation of the amygdala in the disease spreading from/to olfactory areas, the temporal lobe and beyond. Proteomic data are available via ProteomeXchange with identifier PXD038322.
Collapse
Affiliation(s)
- Melania Gonzalez‐Rodriguez
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Sandra Villar‐Conde
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Veronica Astillero‐Lopez
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Patricia Villanueva‐Anguita
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Isabel Ubeda‐Banon
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Alicia Flores‐Cuadrado
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Alino Martinez‐Marcos
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Daniel Saiz‐Sanchez
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| |
Collapse
|
17
|
Tribble JR, Hui F, Quintero H, El Hajji S, Bell K, Di Polo A, Williams PA. Neuroprotection in glaucoma: Mechanisms beyond intraocular pressure lowering. Mol Aspects Med 2023; 92:101193. [PMID: 37331129 DOI: 10.1016/j.mam.2023.101193] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/25/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
Glaucoma is a common, complex, multifactorial neurodegenerative disease characterized by progressive dysfunction and then loss of retinal ganglion cells, the output neurons of the retina. Glaucoma is the most common cause of irreversible blindness and affects ∼80 million people worldwide with many more undiagnosed. The major risk factors for glaucoma are genetics, age, and elevated intraocular pressure. Current strategies only target intraocular pressure management and do not directly target the neurodegenerative processes occurring at the level of the retinal ganglion cell. Despite strategies to manage intraocular pressure, as many as 40% of glaucoma patients progress to blindness in at least one eye during their lifetime. As such, neuroprotective strategies that target the retinal ganglion cell and these neurodegenerative processes directly are of great therapeutic need. This review will cover the recent advances from basic biology to on-going clinical trials for neuroprotection in glaucoma covering degenerative mechanisms, metabolism, insulin signaling, mTOR, axon transport, apoptosis, autophagy, and neuroinflammation. With an increased understanding of both the basic and clinical mechanisms of the disease, we are closer than ever to a neuroprotective strategy for glaucoma.
Collapse
Affiliation(s)
- James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flora Hui
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia; Department of Optometry & Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Heberto Quintero
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Sana El Hajji
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Katharina Bell
- NHMRC Clinical Trials Centre, University of Sydney, Australia; Eye ACP Duke-NUS, Singapore
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
18
|
Kim EJ, Kim JJ. Neurocognitive effects of stress: a metaparadigm perspective. Mol Psychiatry 2023; 28:2750-2763. [PMID: 36759545 PMCID: PMC9909677 DOI: 10.1038/s41380-023-01986-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/11/2023]
Abstract
Stressful experiences, both physical and psychological, that are overwhelming (i.e., inescapable and unpredictable), can measurably affect subsequent neuronal properties and cognitive functioning of the hippocampus. At the cellular level, stress has been shown to alter hippocampal synaptic plasticity, spike and local field potential activity, dendritic morphology, neurogenesis, and neurodegeneration. At the behavioral level, stress has been found to impair learning and memory for declarative (or explicit) tasks that are based on cognition, such as verbal recall memory in humans and spatial memory in rodents, while facilitating those that are based on emotion, such as differential fear conditioning in humans and contextual fear conditioning in rodents. These vertically related alterations in the hippocampus, procedurally observed after subjects have undergone stress, are generally believed to be mediated by recurrently elevated circulating hypothalamic-pituitary-adrenal (HPA) axis effector hormones, glucocorticoids, directly acting on hippocampal neurons densely populated with corticosteroid receptors. The main purposes of this review are to (i) provide a synopsis of the neurocognitive effects of stress in a historical context that led to the contemporary HPA axis dogma of basic and translational stress research, (ii) critically reappraise the necessity and sufficiency of the glucocorticoid hypothesis of stress, and (iii) suggest an alternative metaparadigm approach to monitor and manipulate the progression of stress effects at the neural coding level. Real-time analyses can reveal neural activity markers of stress in the hippocampus that can be used to extrapolate neurocognitive effects across a range of stress paradigms (i.e., resolve scaling and dichotomous memory effects issues) and understand individual differences, thereby providing a novel neurophysiological scaffold for advancing future stress research.
Collapse
Affiliation(s)
- Eun Joo Kim
- Department of Psychology, University of Washington, Seattle, WA, 98195, USA
- School of Psychology, Korea University, Seoul, 02841, Republic of Korea
| | - Jeansok J Kim
- Department of Psychology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
19
|
Telser J, Grossmann K, Wohlwend N, Risch L, Saely CH, Werner P. Phosphorylated tau in Alzheimer's disease. Adv Clin Chem 2023; 116:31-111. [PMID: 37852722 DOI: 10.1016/bs.acc.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
There is a need for blood biomarkers to detect individuals at different Alzheimer's disease (AD) stages because obtaining cerebrospinal fluid-based biomarkers is invasive and costly. Plasma phosphorylated tau proteins (p-tau) have shown potential as such biomarkers. This systematic review was conducted according to the PRISMA guidelines and aimed to determine whether quantification of plasma tau phosphorylated at threonine 181 (p-tau181), threonine 217 (p-tau217) and threonine 231 (p-tau231) is informative in the diagnosis of AD. All p-tau isoforms increase as a function of Aβ-accumulation and discriminate healthy individuals from those at preclinical AD stages with high accuracy. P-tau231 increases earliest, followed by p-tau181 and p-tau217. In advanced stages, all p-tau isoforms are associated with the clinical classification of AD and increase with disease severity, with the greatest increase seen for p-tau217. This is also reflected by a better correlation of p-tau217 with Aβ scans, whereas both, p-tau217 and p-tau181 correlated equally with tau scans. However, at the very advanced stages, p-tau181 begins to plateau, which may mirror the trajectory of the Aβ pathology and indicate an association with a more intermediate risk of AD. Across the AD continuum, the incremental increase in all biomarkers is associated with structural changes in widespread brain regions and underlying cognitive decline. Furthermore, all isoforms differentiate AD from non-AD neurodegenerative disorders, making them specific for AD. Incorporating p-tau181, p-tau217 and p-tau231 in clinical use requires further studies to examine ideal cut-points and harmonize assays.
Collapse
Affiliation(s)
- Julia Telser
- Faculty of Medical Science, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Laboratory Dr. Risch, Vaduz, Liechtenstein
| | - Kirsten Grossmann
- Faculty of Medical Science, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Laboratory Dr. Risch, Vaduz, Liechtenstein
| | - Niklas Wohlwend
- Laboratory Dr. Risch, Vaduz, Liechtenstein; Department of Internal Medicine Spital Grabs, Spitalregion Rheintal Werdenberg Sarganserland, Grabs, Switzerland
| | - Lorenz Risch
- Faculty of Medical Science, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Laboratory Dr. Risch, Vaduz, Liechtenstein; University Institute of Clinical Chemistry, University Hospital and University of Bern, Inselspital, Bern, Switzerland
| | - Christoph H Saely
- Faculty of Medical Science, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Philipp Werner
- Department of Neurology, State Hospital of Rankweil, Academic Teaching Hospital, Rankweil, Austria.
| |
Collapse
|
20
|
Reekes TH, Ledbetter CR, Alexander JS, Stokes KY, Pardue S, Bhuiyan MAN, Patterson JC, Lofton KT, Kevil CG, Disbrow EA. Elevated plasma sulfides are associated with cognitive dysfunction and brain atrophy in human Alzheimer's disease and related dementias. Redox Biol 2023; 62:102633. [PMID: 36924684 PMCID: PMC10026043 DOI: 10.1016/j.redox.2023.102633] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023] Open
Abstract
Emerging evidence indicates that vascular stress is an important contributor to the pathophysiology of Alzheimer's disease and related dementias (ADRD). Hydrogen sulfide (H2S) and its metabolites (acid-labile (e.g., iron-sulfur clusters) and bound (e.g., per-, poly-) sulfides) have been shown to modulate both vascular and neuronal homeostasis. We recently reported that elevated plasma sulfides were associated with cognitive dysfunction and measures of microvascular disease in ADRD. Here we extend our previous work to show associations between elevated sulfides and magnetic resonance-based metrics of brain atrophy and white matter integrity. Elevated bound sulfides were associated with decreased grey matter volume, while increased acid labile sulfides were associated with decreased white matter integrity and greater ventricular volume. These findings are consistent with alterations in sulfide metabolism in ADRD which may represent maladaptive responses to oxidative stress.
Collapse
Affiliation(s)
- Tyler H Reekes
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Shreveport, United States; Center for Brain Health, LSU Health Shreveport, United States
| | - Christina R Ledbetter
- Center for Brain Health, LSU Health Shreveport, United States; Department of Neurosurgery, LSU Health Shreveport, United States
| | - J Steven Alexander
- Center for Brain Health, LSU Health Shreveport, United States; Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Neurology, LSU Health Shreveport, United States; Department of Molecular and Cellular Physiology, LSU Health Shreveport, United States
| | - Karen Y Stokes
- Center for Brain Health, LSU Health Shreveport, United States; Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Molecular and Cellular Physiology, LSU Health Shreveport, United States
| | - Sibile Pardue
- Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Pathology and Translational Pathobiology, LSU Health Shreveport, United States
| | | | - James C Patterson
- Center for Brain Health, LSU Health Shreveport, United States; Department of Psychiatry and Behavioral Medicine, LSU Health Shreveport, United States
| | - Katelyn T Lofton
- Center for Brain Health, LSU Health Shreveport, United States; Department of Neurology, LSU Health Shreveport, United States; Department of Psychiatry and Behavioral Medicine, LSU Health Shreveport, United States
| | - Christopher G Kevil
- Center for Brain Health, LSU Health Shreveport, United States; Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Pathology and Translational Pathobiology, LSU Health Shreveport, United States.
| | - Elizabeth A Disbrow
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Shreveport, United States; Center for Brain Health, LSU Health Shreveport, United States; Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Neurology, LSU Health Shreveport, United States.
| |
Collapse
|
21
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
22
|
The times they are a-changin': a proposal on how brain flexibility goes beyond the obvious to include the concepts of "upward" and "downward" to neuroplasticity. Mol Psychiatry 2023; 28:977-992. [PMID: 36575306 PMCID: PMC10005965 DOI: 10.1038/s41380-022-01931-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022]
Abstract
Since the brain was found to be somehow flexible, plastic, researchers worldwide have been trying to comprehend its fundamentals to better understand the brain itself, make predictions, disentangle the neurobiology of brain diseases, and finally propose up-to-date treatments. Neuroplasticity is simple as a concept, but extremely complex when it comes to its mechanisms. This review aims to bring to light an aspect about neuroplasticity that is often not given enough attention as it should, the fact that the brain's ability to change would include its ability to disconnect synapses. So, neuronal shrinkage, decrease in spine density or dendritic complexity should be included within the concept of neuroplasticity as part of its mechanisms, not as an impairment of it. To that end, we extensively describe a variety of studies involving topics such as neurodevelopment, aging, stress, memory and homeostatic plasticity to highlight how the weakening and disconnection of synapses organically permeate the brain in so many ways as a good practice of its intrinsic physiology. Therefore, we propose to break down neuroplasticity into two sub-concepts, "upward neuroplasticity" for changes related to synaptic construction and "downward neuroplasticity" for changes related to synaptic deconstruction. With these sub-concepts, neuroplasticity could be better understood from a bigger landscape as a vector in which both directions could be taken for the brain to flexibly adapt to certain demands. Such a paradigm shift would allow a better understanding of the concept of neuroplasticity to avoid any data interpretation bias, once it makes clear that there is no morality with regard to the organic and physiological changes that involve dynamic biological systems as seen in the brain.
Collapse
|
23
|
Zeng F, Parker K, Zhan Y, Miller M, Zhu MY. Upregulated DNA Damage-Linked Biomarkers in Parkinson's Disease Model Mice. ASN Neuro 2023; 15:17590914231152099. [PMID: 36683340 PMCID: PMC9880594 DOI: 10.1177/17590914231152099] [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: 08/05/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023] Open
Abstract
SUMMARY STATEMENT The present study examined expression of DNA damage markers in VMAT2 Lo PD model mice. The results demonstrate there is a significant increase in these DNA damage markers mostly in the brain regions of 18- and 23-month-old model mice, indicating oxidative stress-induced DNA lesion is an important pathologic feature of this mouse model.
Collapse
Affiliation(s)
- Fei Zeng
- Department of Neurology, Renmin Hospital of the Wuhan University,
Wuhan, China
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| | - Karsten Parker
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| | - Yanqiang Zhan
- Department of Neurology, Renmin Hospital of the Wuhan University,
Wuhan, China
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| | - Matthew Miller
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| | - Meng-Yang Zhu
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| |
Collapse
|
24
|
Gernone F, Uva A, Cavalera MA, Zatelli A. Neurogenic Bladder in Dogs, Cats and Humans: A Comparative Review of Neurological Diseases. Animals (Basel) 2022; 12:3233. [PMID: 36496754 PMCID: PMC9739254 DOI: 10.3390/ani12233233] [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: 10/09/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Lower urinary tract disease (LUTD) includes abnormalities in the structure and function of the bladder and the urethra. LUTD caused by neurological disease is defined neurogenic bladder (NB). The integrity of the central nervous system (CNS) and peripheral nervous system (PNS) is required to explicate normal micturition, maintaining the proper function of bladder and urethra. The location and type of neurological lesions influence the pattern of clinical manifestations, potential treatment, and prognosis. Though, in dogs and cats, spinal cord injury is considered mainly responsible for bladder and/or urethra incompetence, other disorders, congenital or acquired, involving CNS or PNS, could play a role in NB. In veterinary medicine, the information about the epidemiology, prevalence, etiopathogenesis, diagnosis and treatment of NB are scattered. The aim of this study is to provide an overview of the epidemiology, prevalence, clinical findings, diagnosis and prognosis for NB in dogs and cats compared with humans.
Collapse
Affiliation(s)
- Floriana Gernone
- Department of Veterinary Medicine, University of Bari, 70010 Valenzano, Italy
| | | | | | | |
Collapse
|
25
|
Denk A, Müller K, Schlosser S, Heissner K, Gülow K, Müller M, Schmid S. Liver diseases as a novel risk factor for delirium in the ICU-Delirium and hepatic encephalopathy are two distinct entities. PLoS One 2022; 17:e0276914. [PMID: 36413529 PMCID: PMC9681112 DOI: 10.1371/journal.pone.0276914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/15/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Delirium prevalence is high in critical care settings. We examined the incidence, risk factors, and outcome of delirium in a medical intensive care unit (MICU) with a particular focus on liver diseases. We analyzed this patient population in terms of delirium risk prediction and differentiation between delirium and hepatic encephalopathy. METHODS We conducted an observational study and included 164 consecutive patients admitted to an MICU of a university hospital. Patients were assessed for delirium using the Confusion Assessment Method for ICUs and the Richmond Agitation-Sedation Scale (RASS). On admission and at the onset of delirium Sequential Organ Failure Assessment (SOFA) score was determined. A population of patients with liver disease was compared to a population with gastrointestinal diseases. In the population with liver diseases, hepatic encephalopathy was graded according to the West Haven classification. We analyzed the incidence, subtype, predisposing, precipitating, and health-care setting-related factors, treatment, outcome of delirium and the association between delirium and hepatic encephalopathy in patients with liver diseases. RESULTS The incidence of delirium was 32.5% (n = 53). Univariable binary regression analyses adjusted by the Holm-Bonferroni method showed that the development of delirium was significantly determined by 10 risk factors: Alcohol abuse (p = 0.016), severity of disease (Simplified Acute Physiology Score (SAPS) II, p = 0.016), liver diseases (p = 0.030) and sepsis (p = 0.016) compared to the control group (gastrointestinal (GI) diseases and others), increased sodium (p = 0.016), creatinine (p = 0.030), urea (p = 0.032) or bilirubin (p = 0.042), decreased hemoglobin (p = 0.016), and mechanical ventilation (p = 0.016). Of note, we identified liver diseases as a novel and relevant risk factor for delirium. Hepatic encephalopathy was not a risk factor for delirium. Delirium and hepatic encephalopathy are both life-threatening but clearly distinct conditions. The median SOFA score for patients with delirium at delirium onset was significantly higher than the SOFA score of all patients at admission (p = 0.008). Patients with delirium had five times longer ICU stays (p = 0.004) and three times higher in-hospital mortality (p = 0.036). Patients with delirium were five times more likely to be transferred to an intensive medical rehabilitation unit for post-intensive care (p = 0.020). Treatment costs per case were more than five times higher in patients with delirium than in patients without delirium (p = 0.004). CONCLUSIONS The 10 risk factors identified in this study should be assessed upon admission to ICU for effective detection, prevention, and treatment of delirium. Liver diseases are a novel risk factor for delirium with a level of significance comparable to sepsis as an established risk factor. Of note, in patients with liver diseases delirium and hepatic encephalopathy should be recognized as distinct entities to initiate appropriate treatment. Therefore, we propose a new algorithm for efficient diagnosis, characterization, and treatment of altered mental status in the ICU. This algorithm integrates the 10 risk factor prediction-model for delirium and prompts grading of the severity of hepatic encephalopathy using the West Haven classification if liver disease is present or newly diagnosed.
Collapse
Affiliation(s)
- Alexander Denk
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Germany
| | - Karolina Müller
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Sophie Schlosser
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Germany
| | - Klaus Heissner
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Germany
| | - Karsten Gülow
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Germany
| | - Martina Müller
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Germany
| | - Stephan Schmid
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
26
|
Wang M, Song WM, Ming C, Wang Q, Zhou X, Xu P, Krek A, Yoon Y, Ho L, Orr ME, Yuan GC, Zhang B. Guidelines for bioinformatics of single-cell sequencing data analysis in Alzheimer's disease: review, recommendation, implementation and application. Mol Neurodegener 2022; 17:17. [PMID: 35236372 PMCID: PMC8889402 DOI: 10.1186/s13024-022-00517-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive cognitive impairment and neurodegeneration. Extensive clinical and genomic studies have revealed biomarkers, risk factors, pathways, and targets of AD in the past decade. However, the exact molecular basis of AD development and progression remains elusive. The emerging single-cell sequencing technology can potentially provide cell-level insights into the disease. Here we systematically review the state-of-the-art bioinformatics approaches to analyze single-cell sequencing data and their applications to AD in 14 major directions, including 1) quality control and normalization, 2) dimension reduction and feature extraction, 3) cell clustering analysis, 4) cell type inference and annotation, 5) differential expression, 6) trajectory inference, 7) copy number variation analysis, 8) integration of single-cell multi-omics, 9) epigenomic analysis, 10) gene network inference, 11) prioritization of cell subpopulations, 12) integrative analysis of human and mouse sc-RNA-seq data, 13) spatial transcriptomics, and 14) comparison of single cell AD mouse model studies and single cell human AD studies. We also address challenges in using human postmortem and mouse tissues and outline future developments in single cell sequencing data analysis. Importantly, we have implemented our recommended workflow for each major analytic direction and applied them to a large single nucleus RNA-sequencing (snRNA-seq) dataset in AD. Key analytic results are reported while the scripts and the data are shared with the research community through GitHub. In summary, this comprehensive review provides insights into various approaches to analyze single cell sequencing data and offers specific guidelines for study design and a variety of analytic directions. The review and the accompanied software tools will serve as a valuable resource for studying cellular and molecular mechanisms of AD, other diseases, or biological systems at the single cell level.
Collapse
Affiliation(s)
- Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Won-min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Chen Ming
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Qian Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Peng Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Yonejung Yoon
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Lap Ho
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Miranda E. Orr
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| |
Collapse
|
27
|
Bartley SC, Proctor MT, Xia H, Ho E, Kang DS, Schuster K, Bicca MA, Seckler HS, Viola KL, Patrie SM, Kelleher NL, De Mello FG, Klein WL. An Essential Role for Alzheimer’s-Linked Amyloid Beta Oligomers in Neurodevelopment: Transient Expression of Multiple Proteoforms during Retina Histogenesis. Int J Mol Sci 2022; 23:ijms23042208. [PMID: 35216328 PMCID: PMC8875314 DOI: 10.3390/ijms23042208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
Human amyloid beta peptide (Aβ) is a brain catabolite that at nanomolar concentrations can form neurotoxic oligomers (AβOs), which are known to accumulate in Alzheimer’s disease. Because a predisposition to form neurotoxins seems surprising, we have investigated whether circumstances might exist where AβO accumulation may in fact be beneficial. Our investigation focused on the embryonic chick retina, which expresses the same Aβ as humans. Using conformation-selective antibodies, immunoblots, mass spectrometry, and fluorescence microscopy, we discovered that AβOs are indeed present in the developing retina, where multiple proteoforms are expressed in a highly regulated cell-specific manner. The expression of the AβO proteoforms was selectively associated with transiently expressed phosphorylated Tau (pTau) proteoforms that, like AβOs, are linked to Alzheimer’s disease (AD). To test whether the AβOs were functional in development, embryos were cultured ex ovo and then injected intravitreally with either a beta-site APP-cleaving enzyme 1 (BACE-1) inhibitor or an AβO-selective antibody to prematurely lower the levels of AβOs. The consequence was disrupted histogenesis resulting in dysplasia resembling that seen in various retina pathologies. We suggest the hypothesis that embryonic AβOs are a new type of short-lived peptidergic hormone with a role in neural development. Such a role could help explain why a peptide that manifests deleterious gain-of-function activity when it oligomerizes in the aging brain has been evolutionarily conserved.
Collapse
Affiliation(s)
- Samuel C. Bartley
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Madison T. Proctor
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Hongjie Xia
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Evelyn Ho
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Dong S. Kang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Kristen Schuster
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Maíra A. Bicca
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Henrique S. Seckler
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; (H.S.S.); (S.M.P.)
| | - Kirsten L. Viola
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Steven M. Patrie
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; (H.S.S.); (S.M.P.)
| | - Neil L. Kelleher
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA;
| | - Fernando G. De Mello
- Instituto de Biofísica Carlos Chagas Filho (IBCCF), Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - William L. Klein
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-847-591-5510
| |
Collapse
|
28
|
Rengel KF, Boncyk CS, Hughes CG. Postoperative Delirium Prevention and Novel Cognitive Therapy Interventions. CURRENT ANESTHESIOLOGY REPORTS 2022. [DOI: 10.1007/s40140-021-00501-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Wu BS, Zhang YR, Li HQ, Kuo K, Chen SD, Dong Q, Liu Y, Yu JT. Cortical structure and the risk for Alzheimer's disease: a bidirectional Mendelian randomization study. Transl Psychiatry 2021; 11:476. [PMID: 34526483 PMCID: PMC8443658 DOI: 10.1038/s41398-021-01599-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/16/2021] [Accepted: 08/27/2021] [Indexed: 02/08/2023] Open
Abstract
Progressive loss of neurons in a specific brain area is one of the manifestations of Alzheimer's disease (AD). Much effort has been devoted to investigating brain atrophy and AD. However, the causal relationship between cortical structure and AD is not clear. We conducted a bidirectional two-sample Mendelian randomization analysis to investigate the causal relationship between cortical structure (surface area and thickness of the whole cortex and 34 cortical regions) and AD risk. Genetic variants used as instruments came from a large genome-wide association meta-analysis of cortical structure (33,992 participants of European ancestry) and AD (AD and AD-by-proxy, 71,880 cases, 383,378 controls). We found suggestive associations of the decreased surface area of the temporal pole (OR (95% CI): 0.95 (0.9, 0.997), p = 0.04), and decreased thickness of cuneus (OR (95% CI): 0.93 (0.89, 0.98), p = 0.006) with higher AD risk. We also found a suggestive association of vulnerability to AD with the decreased surface area of precentral (β (SE): -43.4 (21.3), p = 0.042) and isthmus cingulate (β (SE): -18.5 (7.3), p = 0.011). However, none of the Bonferroni-corrected p values of the causal relationship between cortical structure and AD met the threshold. We show suggestive evidence of an association of the atrophy of the temporal pole and cuneus with higher AD risk. In the other direction, there was a suggestive causal relationship between vulnerability to AD and the decreased surface area of the precentral and isthmus cingulate. Our findings shed light on the associations of cortical structure with the occurrence of AD.
Collapse
Affiliation(s)
- Bang-Sheng Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Qi Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kevin Kuo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Geula C, Dunlop SR, Ayala I, Kawles AS, Flanagan ME, Gefen T, Mesulam MM. Basal forebrain cholinergic system in the dementias: Vulnerability, resilience, and resistance. J Neurochem 2021; 158:1394-1411. [PMID: 34272732 PMCID: PMC8458251 DOI: 10.1111/jnc.15471] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/15/2023]
Abstract
The basal forebrain cholinergic neurons (BFCN) provide the primary source of cholinergic innervation of the human cerebral cortex. They are involved in the cognitive processes of learning, memory, and attention. These neurons are differentially vulnerable in various neuropathologic entities that cause dementia. This review summarizes the relevance to BFCN of neuropathologic markers associated with dementias, including the plaques and tangles of Alzheimer's disease (AD), the Lewy bodies of diffuse Lewy body disease, the tauopathy of frontotemporal lobar degeneration (FTLD-TAU) and the TDP-43 proteinopathy of FTLD-TDP. Each of these proteinopathies has a different relationship to BFCN and their corticofugal axons. Available evidence points to early and substantial degeneration of the BFCN in AD and diffuse Lewy body disease. In AD, the major neurodegenerative correlate is accumulation of phosphotau in neurofibrillary tangles. However, these neurons are less vulnerable to the tauopathy of FTLD. An intriguing finding is that the intracellular tau of AD causes destruction of the BFCN, whereas that of FTLD does not. This observation has profound implications for exploring the impact of different species of tauopathy on neuronal survival. The proteinopathy of FTLD-TDP shows virtually no abnormal inclusions within the BFCN. Thus, the BFCN are highly vulnerable to the neurodegenerative effects of tauopathy in AD, resilient to the neurodegenerative effect of tauopathy in FTLD and apparently resistant to the emergence of proteinopathy in FTLD-TDP and perhaps also in Pick's disease. Investigations are beginning to shed light on the potential mechanisms of this differential vulnerability and their implications for therapeutic intervention.
Collapse
Affiliation(s)
- Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Sara R Dunlop
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Ivan Ayala
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Allegra S Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Marek-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
31
|
Śliwińska MA, Cały A, Borczyk M, Ziółkowska M, Skonieczna E, Chilimoniuk M, Bernaś T, Giese KP, Radwanska K. Long-term Memory Upscales Volume of Postsynaptic Densities in the Process that Requires Autophosphorylation of αCaMKII. Cereb Cortex 2021; 30:2573-2585. [PMID: 31800021 DOI: 10.1093/cercor/bhz261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
It is generally accepted that formation and storage of memory relies on alterations of the structure and function of brain circuits. However, the structural data, which show learning-induced and long-lasting remodeling of synapses, are still very sparse. Here, we reconstruct 1927 dendritic spines and their postsynaptic densities (PSDs), representing a postsynaptic part of the glutamatergic synapse, in the hippocampal area CA1 of the mice that underwent spatial training. We observe that in young adult (5 months), mice volume of PSDs, but not the volume of the spines, is increased 26 h after the training. The training-induced growth of PSDs is specific for the dendritic spines that lack smooth endoplasmic reticulum and spine apparatuses, and requires autophosphorylation of αCaMKII. Interestingly, aging alters training-induced ultrastructural remodeling of dendritic spines. In old mice, both the median volumes of dendritic spines and PSDs shift after training toward bigger values. Overall, our data support the hypothesis that formation of memory leaves long-lasting footprint on the ultrastructure of brain circuits; however, the form of circuit remodeling changes with age.
Collapse
Affiliation(s)
- Małgorzata Alicja Śliwińska
- Laboratory of Molecular Basis of Behavior, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland.,Laboratory of Imaging Tissue Structure and Function, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Anna Cały
- Laboratory of Molecular Basis of Behavior, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Malgorzata Borczyk
- Laboratory of Molecular Basis of Behavior, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Magdalena Ziółkowska
- Laboratory of Molecular Basis of Behavior, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Edyta Skonieczna
- Laboratory of Molecular Basis of Behavior, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Magdalena Chilimoniuk
- Laboratory of Molecular Basis of Behavior, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Tytus Bernaś
- Laboratory of Imaging Tissue Structure and Function, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland.,Department of Anatomy and Neurology, VCU School of Medicine, Richmond, VA 23298, USA
| | - K Peter Giese
- Department of Basic and Clinical Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, The Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw 02-093, Poland
| |
Collapse
|
32
|
Gasiorowska A, Wydrych M, Drapich P, Zadrozny M, Steczkowska M, Niewiadomski W, Niewiadomska G. The Biology and Pathobiology of Glutamatergic, Cholinergic, and Dopaminergic Signaling in the Aging Brain. Front Aging Neurosci 2021; 13:654931. [PMID: 34326765 PMCID: PMC8315271 DOI: 10.3389/fnagi.2021.654931] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
The elderly population is growing worldwide, with important health and socioeconomic implications. Clinical and experimental studies on aging have uncovered numerous changes in the brain, such as decreased neurogenesis, increased synaptic defects, greater metabolic stress, and enhanced inflammation. These changes are associated with cognitive decline and neurobehavioral deficits. Although aging is not a disease, it is a significant risk factor for functional worsening, affective impairment, disease exaggeration, dementia, and general disease susceptibility. Conversely, life events related to mental stress and trauma can also lead to accelerated age-associated disorders and dementia. Here, we review human studies and studies on mice and rats, such as those modeling human neurodegenerative diseases, that have helped elucidate (1) the dynamics and mechanisms underlying the biological and pathological aging of the main projecting systems in the brain (glutamatergic, cholinergic, and dopaminergic) and (2) the effect of defective glutamatergic, cholinergic, and dopaminergic projection on disabilities associated with aging and neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. Detailed knowledge of the mechanisms of age-related diseases can be an important element in the development of effective ways of treatment. In this context, we briefly analyze which adverse changes associated with neurodegenerative diseases in the cholinergic, glutaminergic and dopaminergic systems could be targeted by therapeutic strategies developed as a result of our better understanding of these damaging mechanisms.
Collapse
Affiliation(s)
- Anna Gasiorowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Wydrych
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Patrycja Drapich
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Zadrozny
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Steczkowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Wiktor Niewiadomski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Grazyna Niewiadomska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
33
|
van der Thiel MM, Freeze WM, Verheggen ICM, Wong SM, de Jong JJA, Postma AA, Hoff EI, Gronenschild EHBM, Verhey FR, Jacobs HIL, Ramakers IHGB, Backes WH, Jansen JFA. Associations of increased interstitial fluid with vascular and neurodegenerative abnormalities in a memory clinic sample. Neurobiol Aging 2021; 106:257-267. [PMID: 34320463 DOI: 10.1016/j.neurobiolaging.2021.06.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 12/21/2022]
Abstract
The vascular and neurodegenerative processes related to clinical dementia cause cell loss which induces, amongst others, an increase in interstitial fluid (ISF). We assessed microvascular, parenchymal integrity, and a proxy of ISF volume alterations with intravoxel incoherent motion imaging in 21 healthy controls and 53 memory clinic patients - mainly affected by neurodegeneration (mild cognitive impairment, Alzheimer's disease dementia), vascular pathology (vascular cognitive impairment), and presumed to be without significant pathology (subjective cognitive decline). The microstructural components were quantified with spectral analysis using a non-negative least squares method. Linear regression was employed to investigate associations of these components with hippocampal and white matter hyperintensity (WMH) volumes. In the normal appearing white matter, a large fint (a proxy of ISF volume) was associated with a large WMH volume and low hippocampal volume. Likewise, a large fint value was associated with a lower hippocampal volume in the hippocampi. Large ISF volume (fint) was shown to be a prominent factor associated with both WMHs and neurodegenerative abnormalities in memory clinic patients and is argued to play a potential role in impaired glymphatic functioning.
Collapse
Affiliation(s)
- Merel M van der Thiel
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Whitney M Freeze
- Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Inge C M Verheggen
- Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Sau May Wong
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Joost J A de Jong
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Alida A Postma
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Erik I Hoff
- Department of Neurology, Zuyderland Medical Center Heerlen, Heerlen, the Netherlands
| | - Ed H B M Gronenschild
- Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Frans R Verhey
- Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Heidi I L Jacobs
- Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Inez H G B Ramakers
- Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands; School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
34
|
Kovács-Valasek A, Pöstyéni E, Dénes V, Mester A, Sétáló G, Gábriel R. Age-Related Alterations of Proteins in Albino Wistar Rat Retina. Cells Tissues Organs 2021; 210:135-150. [PMID: 34218223 DOI: 10.1159/000515447] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/04/2021] [Indexed: 01/05/2023] Open
Abstract
Imbalance of homeostasis causes permanent changes in the body with time. The central nervous system is especially prone to these changes since it possesses limited regenerative capacity. In the retina, neurons are damaged during the aging process, and this eventually leads to deterioration of vision. In our 2-year-long study, we examined genetically closely related rat individuals to disclose the hidden retinal causes of age-associated visual dysfunction. Morphometric analysis showed significant reduction of the retina thickness with aging, particularly that of the inner plexiform layer. To reveal changes between the age groups, we used immunohistochemistry against vesicular glutamate transporter 1 protein for photoreceptor and bipolar cell terminals, Brn3a for ganglion cells, calbindin 28 kDa for horizontal cells, parvalbumin for AII amacrines, protein kinase Cα for rod bipolar cells, tyrosine hydroxylase for dopaminergic cells, glial fibrillary acidic protein for glial cells, and peanut-agglutinin labeling for cones. The most significant decrease was observed in the density of photoreceptor and the ganglion cells in the aging process. By using immunocytochemistry and western blot technique, we observed that calbindin and vesicular glutamate transporter 1 protein staining do not change much with aging; tyrosine hydroxylase, parvalbumin and calretinin showed the highest immunoreactivity during the midlife period. Most interestingly, the level of glial fibrillary acidic protein also changes similarly to the previously named markers. Our results provide further evidence that protein content is modified at least in some cell populations of the rat retina, and the number of retinal cells declined with aging. We conclude that senescence alone may cause structural and functional damage in the retinal tissue.
Collapse
Affiliation(s)
- Andrea Kovács-Valasek
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Etelka Pöstyéni
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Viktória Dénes
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Adrienn Mester
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - György Sétáló
- Department of Medical Biology, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Róbert Gábriel
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
35
|
Pereira JB, Janelidze S, Stomrud E, Palmqvist S, van Westen D, Dage JL, Mattsson-Carlgren N, Hansson O. Plasma markers predict changes in amyloid, tau, atrophy and cognition in non-demented subjects. Brain 2021; 144:2826-2836. [PMID: 34077494 PMCID: PMC8557344 DOI: 10.1093/brain/awab163] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/26/2021] [Accepted: 04/02/2021] [Indexed: 11/13/2022] Open
Abstract
It is currently unclear whether plasma biomarkers can be used as independent prognostic tools to predict changes associated with early Alzheimer's disease (AD). In this study we sought to address this question by assessing whether plasma biomarkers can predict changes in amyloid load, tau accumulation, brain atrophy and cognition in non-demented individuals. To achieve this, plasma amyloid-β 42/40 (Aβ42/40), phosphorylated-tau181 (P-tau181), phosphorylated-tau217 (P-tau217) and neurofilament light (NfL) were determined in 159 non-demented individuals, 123 patients with AD dementia and 35 patients with a non-AD dementia from the Swedish BioFINDER-2 study, who underwent longitudinal amyloid (18 F-flutemetamol) and tau (18 F-RO948) positron emission tomography (PET), structural magnetic resonance imaging (T1-weighted) and cognitive testing. Our univariate linear mixed effect models showed there were several significant associations between the plasma biomarkers with imaging and cognitive measures. However, when all biomarkers were included in the same multivariate linear mixed effect models, we found that increased longitudinal amyloid-PET signals were independently predicted by low baseline plasma Aβ42/40 (p = 0.012), whereas increased tau-PET signals, brain atrophy and worse cognition were independently predicted by high plasma P-tau217 (p < 0.004). These biomarkers performed equally well or better than the corresponding biomarkers measured in the cerebrospinal fluid. In addition, they showed a similar performance to binary plasma biomarker values defined using the Youden index, which can be more easily implemented in the clinic. In addition, plasma Aβ42/40 and P-tau217 did not predict longitudinal changes in patients with a non-AD neurodegenerative disorder. In conclusion, our findings indicate that plasma Aβ42/40 and P-tau217 could be useful in clinical practice, research and drug development as prognostic markers of future AD pathology.
Collapse
Affiliation(s)
- Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, 141 83 Huddinge, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Memory Clinic, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Memory Clinic, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Danielle van Westen
- Diagnostic Radiology, Department of Clinical Sciences Lund, Lund University, 221 85 Lund, Sweden.,Image and Function, Skåne University Hospital, Malmö 205 02, Sweden
| | | | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, 221 84 Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Memory Clinic, Skåne University Hospital, 214 28 Malmö, Sweden
| |
Collapse
|
36
|
Abstract
An aging-related reduction in the brain's functional reserve may explain why delirium is more frequent in the elderly than in younger people insofar as the reserve becomes inadequate to cover the metabolic requirements that are critically increased by stressors. The aim of this paper is to review the normal aging-related changes that theoretically compromise complex mental activities, neuronal and synaptic densities, and the neurocomputational flexibility of the functional reserve. A pivotal factor is diminished connectivity, which is substantially due to the loss of synapses and should specifically affect association systems and cholinergic fibres in delirious patients. However, micro-angiopathy with impaired blood flow autoregulation, increased blood/brain barrier permeability, changes in cerebrospinal fluid dynamics, weakened mitochondrial performance, and a pro-inflammatory involution of the immune system may also jointly affect neurons and their synaptic assets, and even cause the progression of delirium to dementia regardless of the presence of co-existing plaques, tangles, or other pathological markers. On the other hand, the developmental growth in functional reserve during childhood and adolescence makes the brain increasingly resistant to delirium, and residual reserve can allow the elderly to recover. These data support the view that functional reserve is the variable that confronts stressors and governs the risk and intensity of and recovery from delirium. Although people of any age are at risk of delirium, the elderly are at greater risk because aging and age-dependent structural changes inevitably affect the brain's functional reserve.
Collapse
|
37
|
Suri S, Bulte D, Chiesa ST, Ebmeier KP, Jezzard P, Rieger SW, Pitt JE, Griffanti L, Okell TW, Craig M, Chappell MA, Blockley NP, Kivimäki M, Singh-Manoux A, Khir AW, Hughes AD, Deanfield JE, Jensen DEA, Green SF, Sigutova V, Jansen MG, Zsoldos E, Mackay CE. Study Protocol: The Heart and Brain Study. Front Physiol 2021; 12:643725. [PMID: 33868011 PMCID: PMC8046163 DOI: 10.3389/fphys.2021.643725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/03/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND It is well-established that what is good for the heart is good for the brain. Vascular factors such as hypertension, diabetes, and high cholesterol, and genetic factors such as the apolipoprotein E4 allele increase the risk of developing both cardiovascular disease and dementia. However, the mechanisms underlying the heart-brain association remain unclear. Recent evidence suggests that impairments in vascular phenotypes and cerebrovascular reactivity (CVR) may play an important role in cognitive decline. The Heart and Brain Study combines state-of-the-art vascular ultrasound, cerebrovascular magnetic resonance imaging (MRI) and cognitive testing in participants of the long-running Whitehall II Imaging cohort to examine these processes together. This paper describes the study protocol, data pre-processing and overarching objectives. METHODS AND DESIGN The 775 participants of the Whitehall II Imaging cohort, aged 65 years or older in 2019, have received clinical and vascular risk assessments at 5-year-intervals since 1985, as well as a 3T brain MRI scan and neuropsychological tests between 2012 and 2016 (Whitehall II Wave MRI-1). Approximately 25% of this cohort are selected for the Heart and Brain Study, which involves a single testing session at the University of Oxford (Wave MRI-2). Between 2019 and 2023, participants will undergo ultrasound scans of the ascending aorta and common carotid arteries, measures of central and peripheral blood pressure, and 3T MRI scans to measure CVR in response to 5% carbon dioxide in air, vessel-selective cerebral blood flow (CBF), and cerebrovascular lesions. The structural and diffusion MRI scans and neuropsychological battery conducted at Wave MRI-1 will also be repeated. Using this extensive life-course data, the Heart and Brain Study will examine how 30-year trajectories of vascular risk throughout midlife (40-70 years) affect vascular phenotypes, cerebrovascular health, longitudinal brain atrophy and cognitive decline at older ages. DISCUSSION The study will generate one of the most comprehensive datasets to examine the longitudinal determinants of the heart-brain association. It will evaluate novel physiological processes in order to describe the optimal window for managing vascular risk in order to delay cognitive decline. Ultimately, the Heart and Brain Study will inform strategies to identify at-risk individuals for targeted interventions to prevent or delay dementia.
Collapse
Affiliation(s)
- Sana Suri
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Daniel Bulte
- Oxford Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Scott T. Chiesa
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Klaus P. Ebmeier
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | - Peter Jezzard
- FMRIB Centre, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Sebastian W. Rieger
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
- FMRIB Centre, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Jemma E. Pitt
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Ludovica Griffanti
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
- FMRIB Centre, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Thomas W. Okell
- FMRIB Centre, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Martin Craig
- Radiological Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nottingham Biomedical Research Centre, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Michael A. Chappell
- Radiological Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nottingham Biomedical Research Centre, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | | | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Archana Singh-Manoux
- Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Ashraf W. Khir
- Mechanical Engineering, Brunel University London, Uxbridge, United Kingdom
| | - Alun D. Hughes
- MRC Unit for Lifelong Health and Ageing, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - John E. Deanfield
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Daria E. A. Jensen
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Sebastian F. Green
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | - Veronika Sigutova
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | - Michelle G. Jansen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Enikő Zsoldos
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Clare E. Mackay
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
38
|
Koch SC, Nelson A, Hartenstein V. Structural aspects of the aging invertebrate brain. Cell Tissue Res 2021; 383:931-947. [PMID: 33409654 PMCID: PMC7965346 DOI: 10.1007/s00441-020-03314-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/28/2020] [Indexed: 11/26/2022]
Abstract
Aging is characterized by a decline in neuronal function in all animal species investigated so far. Functional changes are accompanied by and may be in part caused by, structurally visible degenerative changes in neurons. In the mammalian brain, normal aging shows abnormalities in dendrites and axons, as well as ultrastructural changes in synapses, rather than global neuron loss. The analysis of the structural features of aging neurons, as well as their causal link to molecular mechanisms on the one hand, and the functional decline on the other hand is crucial in order to understand the aging process in the brain. Invertebrate model organisms like Drosophila and C. elegans offer the opportunity to apply a forward genetic approach to the analysis of aging. In the present review, we aim to summarize findings concerning abnormalities in morphology and ultrastructure in invertebrate brains during normal aging and compare them to what is known for the mammalian brain. It becomes clear that despite of their considerably shorter life span, invertebrates display several age-related changes very similar to the mammalian condition, including the retraction of dendritic and axonal branches at specific locations, changes in synaptic density and increased accumulation of presynaptic protein complexes. We anticipate that continued research efforts in invertebrate systems will significantly contribute to reveal (and possibly manipulate) the molecular/cellular pathways leading to neuronal aging in the mammalian brain.
Collapse
Affiliation(s)
- Sandra C Koch
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Annie Nelson
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Volker Hartenstein
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA.
| |
Collapse
|
39
|
Ohm DT, Fought AJ, Martersteck A, Coventry C, Sridhar J, Gefen T, Weintraub S, Bigio E, Mesulam M, Rogalski E, Geula C. Accumulation of neurofibrillary tangles and activated microglia is associated with lower neuron densities in the aphasic variant of Alzheimer's disease. Brain Pathol 2021; 31:189-204. [PMID: 33010092 PMCID: PMC7855834 DOI: 10.1111/bpa.12902] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/27/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
The neurofibrillary tangles (NFT) and amyloid-ß plaques (AP) that comprise Alzheimer's disease (AD) neuropathology are associated with neurodegeneration and microglial activation. Activated microglia exist on a dynamic spectrum of morphologic subtypes that include resting, surveillant microglia capable of converting to activated, hypertrophic microglia closely linked to neuroinflammatory processes and AD neuropathology in amnestic AD. However, quantitative analyses of microglial subtypes and neurons are lacking in non-amnestic clinical AD variants, including primary progressive aphasia (PPA-AD). PPA-AD is a language disorder characterized by cortical atrophy and NFT densities concentrated to the language-dominant hemisphere. Here, a stereologic investigation of five PPA-AD participants determined the densities and distributions of neurons and microglial subtypes to examine how cellular changes relate to AD neuropathology and may contribute to cortical atrophy. Adjacent series of sections were immunostained for neurons (NeuN) and microglia (HLA-DR) from bilateral language and non-language regions where in vivo cortical atrophy and Thioflavin-S-positive APs and NFTs were previously quantified. NeuN-positive neurons and morphologic subtypes of HLA-DR-positive microglia (i.e., resting [ramified] microglia and activated [hypertrophic] microglia) were quantified using unbiased stereology. Relationships between neurons, microglia, AD neuropathology, and cortical atrophy were determined using linear mixed models. NFT densities were positively associated with hypertrophic microglia densities (P < 0.01) and inversely related to neuron densities (P = 0.01). Hypertrophic microglia densities were inversely related to densities of neurons (P < 0.01) and ramified microglia (P < 0.01). Ramified microglia densities were positively associated with neuron densities (P = 0.02) and inversely related to cortical atrophy (P = 0.03). Our findings provide converging evidence of divergent roles for microglial subtypes in patterns of neurodegeneration, which includes hypertrophic microglia likely driving a neuroinflammatory response more sensitive to NFTs than APs in PPA-AD. Moreover, the accumulation of both NFTs and activated hypertrophic microglia in association with low neuron densities suggest they may collectively contribute to focal neurodegeneration characteristic of PPA-AD.
Collapse
Affiliation(s)
- Daniel T. Ohm
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Angela J. Fought
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIL
| | - Adam Martersteck
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Jaiashre Sridhar
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Eileen Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIL
| | - M.‐Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIL
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| |
Collapse
|
40
|
Huang L, Zhou H, Chen K, Chen X, Yang G. Learning-Dependent Dendritic Spine Plasticity Is Reduced in the Aged Mouse Cortex. Front Neural Circuits 2020; 14:581435. [PMID: 33324172 PMCID: PMC7726160 DOI: 10.3389/fncir.2020.581435] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/05/2020] [Indexed: 11/13/2022] Open
Abstract
Aging is accompanied by a progressive decrease in learning and memory function. Synaptic loss, one of the hallmarks of normal aging, likely plays an important role in age-related cognitive decline. But little is known about the impact of advanced age on synaptic plasticity and neuronal function in vivo. In this study, we examined the structural dynamics of postsynaptic dendritic spines as well as calcium activity of layer 5 pyramidal neurons in the cerebral cortex of young and old mice. Using transcranial two-photon microscopy, we found that in both sensory and motor cortices, the elimination rates of dendritic spines were comparable between young (3-5 months) and mature adults (8-10 months), but seemed higher in old mice (>20 months), contributing to a reduction of total spine number in the old brain. During the process of motor learning, old mice compared to young mice had fewer new spines formed in the primary motor cortex. Motor training-evoked somatic calcium activity in layer 5 pyramidal neurons of the motor cortex was also lower in old than young mice, which was associated with the decline of motor learning ability during aging. Together, these results demonstrate the effects of aging on learning-dependent synapse remodeling and neuronal activity in the living cortex and suggest that synaptic deficits may contribute to age-related learning impairment.
Collapse
Affiliation(s)
- Lianyan Huang
- Department of Anesthesiology, New York University School of Medicine, New York, NY, United States.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Hang Zhou
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, United States
| | - Kai Chen
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, United States
| | - Xiao Chen
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Guang Yang
- Department of Anesthesiology, New York University School of Medicine, New York, NY, United States.,Department of Anesthesiology, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
41
|
Kirch C, Gollo LL. Spatially resolved dendritic integration: towards a functional classification of neurons. PeerJ 2020; 8:e10250. [PMID: 33282551 PMCID: PMC7694565 DOI: 10.7717/peerj.10250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/06/2020] [Indexed: 01/19/2023] Open
Abstract
The vast tree-like dendritic structure of neurons allows them to receive and integrate input from many neurons. A wide variety of neuronal morphologies exist, however, their role in dendritic integration, and how it shapes the response of the neuron, is not yet fully understood. Here, we study the evolution and interactions of dendritic spikes in excitable neurons with complex real branch structures. We focus on dozens of digitally reconstructed illustrative neurons from the online repository NeuroMorpho.org, which contains over 130,000 neurons. Yet, our methods can be promptly extended to any other neuron. This approach allows us to estimate and map specific and heterogeneous patterns of activity observed across extensive dendritic trees with thousands of compartments. We propose a classification of neurons based on the location of the soma (centrality) and the number of branches connected to the soma. These are key topological factors in determining the neuron's energy consumption, firing rate, and the dynamic range, which quantifies the range in synaptic input rate that can be reliably encoded by the neuron's firing rate. Moreover, we find that bifurcations, the structural building blocks of complex dendrites, play a major role in increasing the dynamic range of neurons. Our results provide a better understanding of the effects of neuronal morphology in the diversity of neuronal dynamics and function.
Collapse
Affiliation(s)
- Christoph Kirch
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Queensland University of Technology, Brisbane, QLD, Australia
| | - Leonardo L. Gollo
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Queensland University of Technology, Brisbane, QLD, Australia
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
42
|
Neuron Loss in Alzheimer's Disease: Translation in Transgenic Mouse Models. Int J Mol Sci 2020; 21:ijms21218144. [PMID: 33143374 PMCID: PMC7663280 DOI: 10.3390/ijms21218144] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Transgenic mouse models represent an essential tool for the exploration of Alzheimer’s disease (AD) pathological mechanisms and the development of novel treatments, which at present provide only symptomatic and transient effects. While a variety of mouse models successfully reflects the main neuropathological hallmarks of AD, such as extracellular amyloid-β (Aβ) deposits, intracellular accumulation of Tau protein, the development of micro- and astrogliosis, as well as behavioral deficits, substantial neuron loss, as a key feature of the disease, seems to be more difficult to achieve. In this review, we summarize information on classic and more recent transgenic mouse models for AD, focusing in particular on loss of pyramidal, inter-, and cholinergic neurons. Although the cause of neuron loss in AD is still a matter of scientific debate, it seems to be linked to intraneuronal Aβ accumulation in several transgenic mouse models, especially in pyramidal neurons.
Collapse
|
43
|
Delirium Superimposed on Dementia in Perioperative Period and Intensive Care. J Clin Med 2020; 9:jcm9103279. [PMID: 33066174 PMCID: PMC7601948 DOI: 10.3390/jcm9103279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/28/2020] [Accepted: 10/06/2020] [Indexed: 11/30/2022] Open
Abstract
Delirium is a life-threatening condition, the causes of which are still not fully understood. It may develop in patients with pre-existing dementia. Delirium superimposed on dementia (DSD) can go completely unnoticed with routine examination. It may happen in the perioperative period and in the critical care setting, especially in the ageing population. Difficulties in diagnosing and lack of specific pharmacological and non-pharmacological treatment make DSD a seriously growing problem. Patient-oriented, multidirectional preventive measures should be applied to reduce the risk of DSD. For this reason, anesthesiologists and intensive care specialists should be aware of this interesting condition in their everyday clinical practice.
Collapse
|
44
|
Poon CH, Wang Y, Fung ML, Zhang C, Lim LW. Rodent Models of Amyloid-Beta Feature of Alzheimer's Disease: Development and Potential Treatment Implications. Aging Dis 2020; 11:1235-1259. [PMID: 33014535 PMCID: PMC7505263 DOI: 10.14336/ad.2019.1026] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide and causes severe financial and social burdens. Despite much research on the pathogenesis of AD, the neuropathological mechanisms remain obscure and current treatments have proven ineffective. In the past decades, transgenic rodent models have been used to try to unravel this disease, which is crucial for early diagnosis and the assessment of disease-modifying compounds. In this review, we focus on transgenic rodent models used to study amyloid-beta pathology in AD. We also discuss their possible use as promising tools for AD research. There is still no effective treatment for AD and the development of potent therapeutics are urgently needed. Many molecular pathways are susceptible to AD, ranging from neuroinflammation, immune response, and neuroplasticity to neurotrophic factors. Studying these pathways may shed light on AD pathophysiology as well as provide potential targets for the development of more effective treatments. This review discusses the advantages and limitations of these models and their potential therapeutic implications for AD.
Collapse
Affiliation(s)
- Chi Him Poon
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingyi Wang
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Lung Fung
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chengfei Zhang
- 2Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
45
|
Rizzo A, Gambino G, Sardo P, Rizzo V. Being in the Past and Perform the Future in a Virtual World: VR Applications to Assess and Enhance Episodic and Prospective Memory in Normal and Pathological Aging. Front Hum Neurosci 2020; 14:297. [PMID: 32848672 PMCID: PMC7417675 DOI: 10.3389/fnhum.2020.00297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 07/03/2020] [Indexed: 11/20/2022] Open
Abstract
The process of aging commonly features a gradual deterioration in cognitive performance and, in particular, the decline of memory. Despite the increased longevity of the world’s population, the prevalence of neurodegenerative conditions, such as dementia, continues to be a major burden on public health, and consequently, the latest research has been focused on memory and aging. Currently, the failure of episodic and Prospective memory (PM) is one of the main complaints in the elderly, considered among the early symptoms of dementia. It is therefore increasingly important to define more clearly the boundaries between normal and pathological aging. Recently, researchers have begun to build and apply Virtual Environments (VE) to the explicit purpose of better understanding the performance of episodic and PM in complex and realistic contexts, with the perspective of further developing effective training procedures that depend on reliable cognitive assessment methods. Virtual technology offers higher levels of realism than “pen and paper” testing and at the same time more experimental control than naturalistic settings. In this mini-review article, we examine the outcomes of recently available studies on virtual reality technology applications developed for the assessment and improvement of episodic and/or PM. To consider the latest technology, we selected 29 articles that have been published in the last 10 years. These documents show that VR-based technologies can provide a valid basis for screening and treatment and, through increased sensory stimulation and enriched environments reproducing the scenarios of everyday life, could represent effective stimulating experiences even in pathological aging.
Collapse
Affiliation(s)
- Azzurra Rizzo
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, Università Degli Studi di Palermo, Palermo, Italy
| | - Giuditta Gambino
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, Università Degli Studi di Palermo, Palermo, Italy
| | - Pierangelo Sardo
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, Università Degli Studi di Palermo, Palermo, Italy
| | - Valerio Rizzo
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, Università Degli Studi di Palermo, Palermo, Italy
| |
Collapse
|
46
|
CircGRIA1 shows an age-related increase in male macaque brain and regulates synaptic plasticity and synaptogenesis. Nat Commun 2020; 11:3594. [PMID: 32681011 PMCID: PMC7367861 DOI: 10.1038/s41467-020-17435-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/01/2020] [Indexed: 01/11/2023] Open
Abstract
Circular RNAs (circRNAs) are abundant in mammalian brain and some show age-dependent expression patterns. Here, we report that circGRIA1, a conserved circRNA isoform derived from the genomic loci of α-mino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor subunit Gria1, shows an age-related and male-specific increase in expression in the rhesus macaque prefrontal cortex and hippocampus. We show circGRIA1 is predominantly localized to the nucleus, and find an age-related increase in its association with the promoter region of Gria1 gene, suggesting it has a regulatory role in Gria1 transcription. In vitro and in vivo manipulation of circGRIA1 negatively regulates Gria1 mRNA and protein levels. Knockdown of circGRIA1 results in an age-related improvement of synaptogenesis, and GluR1 activity-dependent synaptic plasticity in the hippocampal neurons in males. Our findings underscore the importance of circRNA regulation and offer an insight into the biology of brain aging.
Collapse
|
47
|
Adult-Born Hippocampal Neurons Undergo Extended Development and Are Morphologically Distinct from Neonatally-Born Neurons. J Neurosci 2020; 40:5740-5756. [PMID: 32571837 DOI: 10.1523/jneurosci.1665-19.2020] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 02/28/2020] [Accepted: 05/04/2020] [Indexed: 11/21/2022] Open
Abstract
During immature stages, adult-born neurons pass through critical periods for survival and plasticity. It is generally assumed that by 2 months of age adult-born neurons are mature and equivalent to the broader neuronal population, raising questions of how they might contribute to hippocampal function in old age when neurogenesis has declined. However, few have examined adult-born neurons beyond the critical period or directly compared them to neurons born in infancy. Here, we used a retrovirus to visualize functionally relevant morphological features of 2- to 24-week-old adult-born neurons in male rats. From 2 to 7 weeks, neurons grew and attained a relatively mature phenotype. However, several features of 7-week-old neurons suggested a later wave of growth: these neurons had larger nuclei, thicker dendrites, and more dendritic filopodia than all other groups. Indeed, between 7 and 24 weeks, adult-born neurons gained additional dendritic branches, formed a second primary dendrite, acquired more mushroom spines, and had enlarged mossy fiber presynaptic terminals. Compared with neonatal-born neurons, old adult-born neurons had greater spine density, larger presynaptic terminals, and more putative efferent filopodial contacts onto inhibitory neurons. By integrating rates of cell birth and growth across the life span, we estimate that adult neurogenesis ultimately produces half of the cells and the majority of spines in the dentate gyrus. Critically, protracted development contributes to the plasticity of the hippocampus through to the end of life, even after cell production declines. Persistent differences from neonatal-born neurons may additionally endow adult-born neurons with unique functions even after they have matured.SIGNIFICANCE STATEMENT Neurogenesis occurs in the hippocampus throughout adult life and contributes to memory and emotion. It is generally assumed that new neurons have the greatest impact on behavior when they are immature and plastic. However, since neurogenesis declines dramatically with age, it is unclear how they might contribute to behavior later in life when cell proliferation has slowed. Here we find that newborn neurons mature over many months in rats and may end up with distinct morphological features compared with neurons born in infancy. Using a mathematical model, we estimate that a large fraction of neurons is added in adulthood. Moreover, their extended growth produces a reserve of plasticity that persists even after neurogenesis has declined to low rates.
Collapse
|
48
|
Gul Z, Demircan C, Bagdas D, Buyukuysal RL. Aging protects rat cortical slices against to oxygen-glucose deprivation induced damage. Int J Neurosci 2020; 130:1183-1191. [PMID: 32064981 DOI: 10.1080/00207454.2020.1730830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Objective: In present study, we aimed to clarify effect of aging on the susceptibility of brain tissue to neurodegeneration induced by ischemia.Methods: Damage induced by oxygen-glucose deprivation (OGD) followed by reoxygenation (REO) were compared in cortical slices prepared from young (3 months of age) and aged (22-24 months of age) male Sprague Dawley rats.Results: After incubation of the slices in an oxygen and glucose containing control condition, 2,3,5-triphenyl tetrazolium chloride (TTC) staining intensity was found significantly high in aged cortical slices. Although thirty minutes incubation of the slices in OGD medium followed by REO (OGD-REO) caused similar decline in TTC staining in young and aged cortical slices, staining intensity was still significantly higher in the slices prepared from aged animals. Thirty minutes of OGD-REO, on the other hand, also caused more increase in lactate dehydrogenase (LDH) leakage from young slices. While water contents of the slices were almost equal under control condition, it was significantly high in young cortical slices after OGD-REO incubations. In contrary to these findings, OGD and REO caused more increases in S100B output from aged rat cortical slices. S100B levels in brain regions including the cerebral cortex were also found higher in aged rats.Conclusion: All these results indicate that, cortical slices prepared from aged male rats are significantly less responsive to in vitro OGD-REO induced alterations. Since protein S100B outputs were almost doubled from aged cortical slices, a possible involvement of this enhanced S100B output seems to be likely.
Collapse
Affiliation(s)
- Zulfiye Gul
- Faculty of Medicine, Department of Medical Pharmacology, Bahcesehir University, Istanbul, Turkey
| | - Celaleddin Demircan
- Faculty of Medicine, Department of Internal Medicine, Uludag University, Bursa, Turkey
| | - Deniz Bagdas
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | | |
Collapse
|
49
|
Sharma A, Kazim SF, Larson CS, Ramakrishnan A, Gray JD, McEwen BS, Rosenberg PA, Shen L, Pereira AC. Divergent roles of astrocytic versus neuronal EAAT2 deficiency on cognition and overlap with aging and Alzheimer's molecular signatures. Proc Natl Acad Sci U S A 2019; 116:21800-21811. [PMID: 31591195 PMCID: PMC6815169 DOI: 10.1073/pnas.1903566116] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The excitatory amino acid transporter 2 (EAAT2) is the major glutamate transporter in the brain expressed predominantly in astrocytes and at low levels in neurons and axonal terminals. EAAT2 expression is reduced in aging and sporadic Alzheimer's disease (AD) patients' brains. The role EAAT2 plays in cognitive aging and its associated mechanisms remains largely unknown. Here, we show that conditional deletion of astrocytic and neuronal EAAT2 results in age-related cognitive deficits. Astrocytic, but not neuronal EAAT2, deletion leads to early deficits in short-term memory and in spatial reference learning and long-term memory. Neuronal EAAT2 loss results in late-onset spatial reference long-term memory deficit. Neuronal EAAT2 deletion leads to dysregulation of the kynurenine pathway, and astrocytic EAAT2 deficiency results in dysfunction of innate and adaptive immune pathways, which correlate with cognitive decline. Astrocytic EAAT2 deficiency also shows transcriptomic overlaps with human aging and AD. Overall, the present study shows that in addition to the widely recognized astrocytic EAAT2, neuronal EAAT2 plays a role in hippocampus-dependent memory. Furthermore, the gene expression profiles associated with astrocytic and neuronal EAAT2 deletion are substantially different, with the former associated with inflammation and synaptic function similar to changes observed in human AD and gene expression changes associated with inflammation similar to the aging human brain.
Collapse
Affiliation(s)
- Abhijeet Sharma
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Syed Faraz Kazim
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Chloe S Larson
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065
| | - Aarthi Ramakrishnan
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jason D Gray
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065
| | - Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065;
| | - Paul A Rosenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115
| | - Li Shen
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ana C Pereira
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029;
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
50
|
Temido-Ferreira M, Coelho JE, Pousinha PA, Lopes LV. Novel Players in the Aging Synapse: Impact on Cognition. J Caffeine Adenosine Res 2019; 9:104-127. [PMID: 31559391 PMCID: PMC6761599 DOI: 10.1089/caff.2019.0013] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
While neuronal loss has long been considered as the main contributor to age-related cognitive decline, these alterations are currently attributed to gradual synaptic dysfunction driven by calcium dyshomeostasis and alterations in ionotropic/metabotropic receptors. Given the key role of the hippocampus in encoding, storage, and retrieval of memory, the morpho- and electrophysiological alterations that occur in the major synapse of this network-the glutamatergic-deserve special attention. We guide you through the hippocampal anatomy, circuitry, and function in physiological context and focus on alterations in neuronal morphology, calcium dynamics, and plasticity induced by aging and Alzheimer's disease (AD). We provide state-of-the art knowledge on glutamatergic transmission and discuss implications of these novel players for intervention. A link between regular consumption of caffeine-an adenosine receptor blocker-to decreased risk of AD in humans is well established, while the mechanisms responsible have only now been uncovered. We review compelling evidence from humans and animal models that implicate adenosine A2A receptors (A2AR) upsurge as a crucial mediator of age-related synaptic dysfunction. The relevance of this mechanism in patients was very recently demonstrated in the form of a significant association of the A2AR-encoding gene with hippocampal volume (synaptic loss) in mild cognitive impairment and AD. Novel pathways implicate A2AR in the control of mGluR5-dependent NMDAR activation and subsequent Ca2+ dysfunction upon aging. The nature of this receptor makes it particularly suited for long-term therapies, as an alternative for regulating aberrant mGluR5/NMDAR signaling in aging and disease, without disrupting their crucial constitutive activity.
Collapse
Affiliation(s)
- Mariana Temido-Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana E. Coelho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Paula A. Pousinha
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), CNRS UMR7275, Université Côte d'Azur, Valbonne, France
| | - Luísa V. Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|