1
|
Kamen Y, Chapman TW, Piedra ET, Ciolkowski ME, Hill RA. Transient upregulation of procaspase-3 during oligodendrocyte fate decisions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623446. [PMID: 39605489 PMCID: PMC11601457 DOI: 10.1101/2024.11.13.623446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Oligodendrocytes are generated throughout life and in neurodegenerative conditions from brain resident oligodendrocyte precursor cells (OPCs). The transition from OPC to oligodendrocyte involves a complex cascade of molecular and morphological states that position the cell to make a fate decision to integrate as a myelinating oligodendrocyte or die through apoptosis. Oligodendrocyte maturation impacts the cell death mechanisms that occur in degenerative conditions, but it is unclear if and how the cell death machinery changes as OPCs transition into oligodendrocytes. Here, we discovered that differentiating oligodendrocytes transiently upregulate the zymogen procaspase-3, equipping these cells to make a survival decision during differentiation. Pharmacological inhibition of caspase-3 decreases oligodendrocyte density, indicating that procaspase-3 upregulation promotes differentiation. Moreover, using procaspase-3 as a marker, we show that oligodendrocyte differentiation continues in the aging cortex and white matter. Taken together, our data establish procaspase-3 as a differentiating oligodendrocyte marker and provide insight into the underlying mechanisms occurring during the decision to integrate or die.
Collapse
Affiliation(s)
- Yasmine Kamen
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Timothy W. Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Enrique T. Piedra
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | | | - Robert A. Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
2
|
Raz N, Daugherty A, Khatib D, Dahle C, Rajan U, Zajac‐Benitez C, Stanley J. Mapping Age Differences in Brain Energy Metabolites and Metabolic Markers of Cellular Membrane Production and Degradation With 31P Magnetic Resonance Spectroscopy. Hum Brain Mapp 2024; 45:e70039. [PMID: 39391993 PMCID: PMC11467684 DOI: 10.1002/hbm.70039] [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: 05/29/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Using Phosphorus Magnetic Resonance Spectroscopy (31P MRS), we examined five metabolites associated with brain energy cycle, and cellular membrane production and degradation in 11 brain regions of 48 children (age 6-15), and 80 middle-aged and older adults (age 52-87). Levels of phosphomonoesters (PMEs) and phosphodiesters (PDEs), gamma plus alpha adenosine triphosphate (γαATP), phosphocreatine (PCr) and inorganic phosphate (Pi), were residualized on the total amplitude value. PMEs were greater in children compared to adults, whereas PDEs showed the opposite age difference. Higher γαATP and lower Pi were found in children compared to adults. The age group differences were particularly salient in the association cortices and anterior white matter. Among children, age correlated negatively with PMEs and positively with PDEs in association cortices. Compared to children, adults had lower intracellular pH. The results suggest reduction in membrane synthesis and increase in membrane degradation in adolescents and to a greater degree in adults compared to younger children. Concomitant reduction in γαATP and increase in Pi are consistent with reduced energy consumption in adolescents and further drop in middle-aged and older adults, although it is impossible to distinguish declines in energy supply from reduced demand due to shrinking neuropil, without longitudinal studies.
Collapse
Affiliation(s)
- Naftali Raz
- Department of PsychologyStony Brook UniversityStony BrookNew YorkUSA
- Center for Lifespan PsychologyMax Planck Institute for Human DevelopmentBerlinGermany
| | - Ana M. Daugherty
- Department of Psychology, and the Institute of GerontologyWayne State UniversityDetroitMichiganUSA
| | - Dalal Khatib
- Department of Psychiatry and Behavioral NeurosciencesWayne State UniversityDetroitMichiganUSA
| | - Cheryl L. Dahle
- Institute of GerontologyWayne State UniversityDetroitMichiganUSA
| | - Usha Rajan
- Department of Psychiatry and Behavioral NeurosciencesWayne State UniversityDetroitMichiganUSA
| | - Caroline Zajac‐Benitez
- Department of Psychiatry and Behavioral NeurosciencesWayne State UniversityDetroitMichiganUSA
| | - Jeffrey A. Stanley
- Department of Psychiatry and Behavioral NeurosciencesWayne State UniversityDetroitMichiganUSA
| |
Collapse
|
3
|
Kumar A, Pal A, Singh P, Rani I, Tondolo V, Rongioletti M, Squitti R. Might Diet, APOE-APOA1 Axis, and Iron Metabolism Provide Clues About the Discrepancy in Alzheimer's Disease Occurrence Between Humans and Chimpanzees? A Bioinformatics-Based Re-Analysis of Gene Expression Data on Mice Fed with Human and Chimpanzee Diets. Biol Trace Elem Res 2024; 202:3750-3759. [PMID: 37938458 DOI: 10.1007/s12011-023-03932-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
The emergence of conflicting reports on the natural occurrence of Alzheimer's disease (AD) in non-human primates has prompted research on the comparison of the role of diet-associated changes in gene expression between humans and non-human primates. This article analyzes the effects of different human and chimpanzee diets and their link with apolipoproteins, lipid, and iron (Fe) metabolism, starting from available data, to find out any gap in the existing knowledge. By using a system biology approach, we have re-analyzed the liver and brain RNA seq data of mice fed with either human or chimpanzee diet for 2 weeks to look for genetic differences that may explain the differences in AD occurrence between those two classes. In liver samples of mice fed with the chimpanzee diet in comparison to the human diet, apolipoprotein A-1, ceruloplasmin, and 10 other genes were upregulated while 21 genes were downregulated. However, brain apolipoprotein E4 gene expression was not changed upon diet. Genetic, structural, and functional differences in apolipoprotein E protein, along with differences in Fe metabolisms and a longer lifespan of humans during evolution may account for the observed disparity.
Collapse
Affiliation(s)
- Ashok Kumar
- Centre for Systems Biology and Bioinformatics, Panjab University, Chandigarh, India
| | - Amit Pal
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Kalyani, West Bengal, 741245, India.
| | - Parminder Singh
- Centre for Systems Biology and Bioinformatics, Panjab University, Chandigarh, India
| | - Isha Rani
- Department of Biochemistry, Maharishi Markandeshwar College of Medical Sciences and Research (MMCMSR), Sadopur, Ambala, India
| | - Vincenzo Tondolo
- Digestive and Colorectal Surgery, Ospedale Isola Tiberina, Gemelli Isola, 00186, Rome, Italy
- Digestive Surgery Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy
| | - Mauro Rongioletti
- Department of Laboratory Science, Ospedale Isola Tiberina, Gemelli Isola, 00186, Rome, Italy
| | - Rosanna Squitti
- Department of Laboratory Science, Ospedale Isola Tiberina, Gemelli Isola, 00186, Rome, Italy
| |
Collapse
|
4
|
Ibañez S, Sengupta N, Luebke JI, Wimmer K, Weaver CM. Myelin dystrophy impairs signal transmission and working memory in a multiscale model of the aging prefrontal cortex. eLife 2024; 12:RP90964. [PMID: 39028036 PMCID: PMC11259433 DOI: 10.7554/elife.90964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
Abstract
Normal aging leads to myelin alterations in the rhesus monkey dorsolateral prefrontal cortex (dlPFC), which are positively correlated with degree of cognitive impairment. It is hypothesized that remyelination with shorter and thinner myelin sheaths partially compensates for myelin degradation, but computational modeling has not yet explored these two phenomena together systematically. Here, we used a two-pronged modeling approach to determine how age-related myelin changes affect a core cognitive function: spatial working memory. First, we built a multicompartment pyramidal neuron model fit to monkey dlPFC empirical data, with an axon including myelinated segments having paranodes, juxtaparanodes, internodes, and tight junctions. This model was used to quantify conduction velocity (CV) changes and action potential (AP) failures after demyelination and subsequent remyelination. Next, we incorporated the single neuron results into a spiking neural network model of working memory. While complete remyelination nearly recovered axonal transmission and network function to unperturbed levels, our models predict that biologically plausible levels of myelin dystrophy, if uncompensated by other factors, can account for substantial working memory impairment with aging. The present computational study unites empirical data from ultrastructure up to behavior during normal aging, and has broader implications for many demyelinating conditions, such as multiple sclerosis or schizophrenia.
Collapse
Affiliation(s)
- Sara Ibañez
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of MedicineBostonUnited States
- Centre de Recerca Matemàtica, Edifici C, Campus BellaterraBellaterraSpain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Edifici CBellaterraSpain
| | - Nilapratim Sengupta
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of MedicineBostonUnited States
- Department of Mathematics, Franklin and Marshall CollegeLancasterUnited States
| | - Jennifer I Luebke
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of MedicineBostonUnited States
| | - Klaus Wimmer
- Centre de Recerca Matemàtica, Edifici C, Campus BellaterraBellaterraSpain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Edifici CBellaterraSpain
| | - Christina M Weaver
- Department of Mathematics, Franklin and Marshall CollegeLancasterUnited States
| |
Collapse
|
5
|
Chen J, Li J, Wang X, Fu X, Ke J, Li J, Wen J, Cheng K, Li S, Shi Z. Heme Oxygenase-1 Gene (GT)n Polymorphism Linked to Deep White Matter Hyperintensities, Not Periventricular Hyperintensities. J Am Heart Assoc 2024; 13:e033981. [PMID: 38818928 PMCID: PMC11255616 DOI: 10.1161/jaha.123.033981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Oxidative stress plays a principal role in the pathogenesis of white matter hyperintensities (WMHs). The induction of heme oxygenase-1 (HO-1) gene in the brain represents 1 of the pivotal mechanisms to counteract the noxious effects of reactive oxygen species, and the transcriptional modulation of HO-1 induction depends on the length of a GT-repeat (GT)n in the promoter region. We investigated whether the HO-1 gene (GT)n polymorphism is associated with the risk of WMHs. METHODS AND RESULTS A total of 849 subjects from the memory clinic were consecutively enrolled, and the HO-1 (GT)n genotype was determined. WMHs were assessed with the Fazekas scale and further divided into periventricular WMHs and deep WMHs (DWMHs). Allelic HO-1 (GT)n polymorphisms were classified as short (≤24 (GT)n), median (25≤[GT]n<31), or long (31≤[GT]n). Multivariate logistic regression analysis was used to evaluate the effect of the HO-1 (GT)n variants on WMHs. The number of repetitions of the HO-1 gene (GT)n ranged from 15 to 39 with a bimodal distribution at lengths 23 and 30. The proportion of S/S genotypes was higher for moderate/severe DWMHs than none/mild DWMHs (22.22% versus 12.44%; P=0.001), but the association for periventricular WMHs was not statistically significant. Logistic regression suggested that the S/S genotype was significantly associated with moderate/severe DWMHs (S/S versus non-S/S: odds ratio, 2.001 [95% CI, 1.323-3.027]; P<0.001). The HO-1 gene (GT)n S/S genotype and aging synergistically contributed to the progression of DWMHs (relative excess risk attributable to interaction, 6.032 [95% CI, 0.149-11.915]). CONCLUSIONS Short (GT)n variants in the HO-1 gene may confer susceptibility to rather than protection from DWMHs, but not periventricular WMHs. REGISTRATION URL: https://www.chictr.org.cn; Unique identifier: ChiCTR2100045869.
Collapse
Affiliation(s)
- Junting Chen
- Department of Neurology and Memory CenterThe 10th Affiliate Hospital, Southern Medical UniversityDongguanChina
- Postgraduate SchoolGuangdong Medical UniversityZhanjiangGuangdongChina
| | - Jinrui Li
- Department of Neurology and Memory CenterThe 10th Affiliate Hospital, Southern Medical UniversityDongguanChina
- The 1st Clinical Medical SchoolSouthern Medical UniversityDongguanChina
| | - Xiaomian Wang
- Postgraduate SchoolGuangdong Medical UniversityZhanjiangGuangdongChina
| | - Xiaoli Fu
- Department of Neurology and Memory CenterThe 10th Affiliate Hospital, Southern Medical UniversityDongguanChina
| | - Jianxia Ke
- The 1st Clinical Medical SchoolSouthern Medical UniversityDongguanChina
| | - Jintao Li
- The 1st Clinical Medical SchoolSouthern Medical UniversityDongguanChina
| | - Jia Wen
- Postgraduate SchoolGuangdong Medical UniversityZhanjiangGuangdongChina
| | - Kailin Cheng
- Postgraduate SchoolGuangdong Medical UniversityZhanjiangGuangdongChina
| | - Shuen Li
- Department of Neurology and Memory CenterThe 10th Affiliate Hospital, Southern Medical UniversityDongguanChina
| | - Zhu Shi
- Department of Neurology and Memory CenterThe 10th Affiliate Hospital, Southern Medical UniversityDongguanChina
- Postgraduate SchoolGuangdong Medical UniversityZhanjiangGuangdongChina
- The 1st Clinical Medical SchoolSouthern Medical UniversityDongguanChina
| |
Collapse
|
6
|
Chapman TW, Kamen Y, Piedra ET, Hill RA. Oligodendrocyte Maturation Alters the Cell Death Mechanisms That Cause Demyelination. J Neurosci 2024; 44:e1794232024. [PMID: 38395617 PMCID: PMC10977033 DOI: 10.1523/jneurosci.1794-23.2024] [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: 09/21/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Myelinating oligodendrocytes die in human disease and early in aging. Despite this, the mechanisms that underly oligodendrocyte death are not resolved and it is also not clear whether these mechanisms change as oligodendrocyte lineage cells are undergoing differentiation and maturation. Here, we used a combination of intravital imaging, single-cell ablation, and cuprizone-mediated demyelination, in both female and male mice, to discover that oligodendrocyte maturation dictates the dynamics and mechanisms of cell death. After single-cell phototoxic damage, oligodendrocyte precursor cells underwent programmed cell death within hours, differentiating oligodendrocytes died over several days, while mature oligodendrocytes took weeks to die. Importantly cells at each maturation stage all eventually died but did so with drastically different temporal dynamics and morphological features. Consistent with this, cuprizone treatment initiated a caspase-3-dependent form of rapid cell death in differentiating oligodendrocytes, while mature oligodendrocytes never activated this executioner caspase. Instead, mature oligodendrocytes exhibited delayed cell death which was marked by DNA damage and disruption in poly-ADP-ribose subcellular localization. Thus, oligodendrocyte maturation plays a key role in determining the mechanism of death a cell undergoes in response to the same insult. This means that oligodendrocyte maturation is important to consider when designing strategies for preventing cell death and preserving myelin while also enhancing the survival of new oligodendrocytes in demyelinating conditions.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Yasmine Kamen
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Enrique T Piedra
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| |
Collapse
|
7
|
McElroy CL, Wang B, Zhang H, Jin K. Cerebellum and Aging: Update and Challenges. Aging Dis 2024; 15:2345-2360. [PMID: 38502583 PMCID: PMC11567260 DOI: 10.14336/ad.2024.0220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
The cerebellum plays a vital role in the aging process. With the aging of the cerebellum, there is a decline in balance and motor function, particularly fine motor skills, and an increased risk of falling. However, in recent years, numerous studies have revealed that the cerebellum has several roles besides balance and fine motor skills, such as cognitive function and memory. It also plays a role in many neurodegenerative diseases. Interestingly, the cerebellum ages more rapidly than other brain regions, including the hippocampus. With increasing studies reporting that the cerebellum has a more prominent and interconnected role in the brain, it is essential to understand why aging affects it more, leading to solutions to help curb the accelerated decline. Here, we summarize the cerebellum's function and look at how it ages at the cellular, molecular, and functional levels. Additionally, we explore the the effects of alcoholism on the aging cerebellum as well as the role of the cerebellum in diseases such as Alzheimer's, Parkinson's, and Multiple Sclerosis.
Collapse
Affiliation(s)
| | | | | | - Kunlin Jin
- Department of Pharmacology and Neuroscience, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
8
|
Guan Y, Cheng CH, Bellomo LI, Narain S, Bigornia SJ, Garelnabi MO, Scott T, Ordovás JM, Tucker KL, Bhadelia R, Koo BB. APOE4 allele-specific associations between diet, multimodal biomarkers, and cognition among Puerto Rican adults in Massachusetts. Front Aging Neurosci 2023; 15:1285333. [PMID: 38035273 PMCID: PMC10684694 DOI: 10.3389/fnagi.2023.1285333] [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: 08/29/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Background Apolipoprotein E (APOE) is the strongest genetic risk factor for sporadic Alzheimer's Disease (AD), and the ε4 allele (APOE4) may interact with lifestyle factors that relate to brain structural changes, underlying the increased risk of AD. However, the exact role of APOE4 in mediating interactions between the peripheral circulatory system and the central nervous system, and how it may link to brain and cognitive aging requires further elucidation. In this analysis, we investigated the association between APOE4 carrier status and multimodal biomarkers (diet, blood markers, clinical diagnosis, brain structure, and cognition) in the context of gene-environment interactions. Methods Participants were older adults from a longitudinal observational study, the Boston Puerto Rican Health Study (BPRHS), who self-identified as of Puerto Rican descent. Demographics, APOE genotype, diet, blood, and clinical data were collected at baseline and at approximately 12th year, with the addition of multimodal brain magnetic resonance imaging (MRI) (T1-weighted and diffusion) and cognitive testing acquired at 12-year. Measures were compared between APOE4 carriers and non-carriers, and associations between multimodal variables were examined using correlation and multivariate network analyses within each group. Results A total of 156 BPRHS participants (mean age at imaging = 68 years, 77% female, mean follow-up 12.7 years) with complete multimodal data were included in the current analysis. APOE4 carriers (n = 43) showed reduced medial temporal lobe (MTL) white matter (WM) microstructural integrity and lower mini-mental state examination (MMSE) score than non-carriers (n = 113). This pattern was consistent with an independent sample from the Alzheimer's Disease Neuroimaging Initiative (ADNI) of n = 283 non-Hispanic White adults without dementia (mean age = 75, 40% female). Within BPRHS, carriers showed distinct connectivity patterns between multimodal biomarkers, characterized by stronger direct network connections between baseline diet/blood markers with 12-year blood/clinical measures, and between blood markers (especially lipids and cytokines) and WM. Cardiovascular burden (i.e., hypertension and diabetes status) was associated with WM integrity for both carriers and non-carriers. Conclusion APOE4 carrier status affects interactions between dietary factors, multimodal blood biomarkers, and MTL WM integrity across ~12 years of follow-up, which may reflect increased peripheral-central systems crosstalk following blood-brain barrier breakdown in carriers.
Collapse
Affiliation(s)
- Yi Guan
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Chia Hsin Cheng
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Luis I. Bellomo
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Sriman Narain
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Sherman J. Bigornia
- Department of Agriculture, Nutrition, and Food Systems, College of Life Sciences and Agriculture, University of New Hampshire, Durham, NH, United States
| | - Mahdi O. Garelnabi
- Department of Public Health, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, United States
| | - Tammy Scott
- School of Medicine, Tufts University, Boston, MA, United States
| | - José M. Ordovás
- Nutrition and Genomics Laboratory, J.M.-US Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
- IMDEA Alimentacion, Madrid, Spain
- CIBER Fisiopatologia de la Obesidad y la Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Katherine L. Tucker
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, United States
- Center for Population Health, University of Massachusetts Lowell, Lowell, MA, United States
| | - Rafeeque Bhadelia
- Neuroradiology Section, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Bang-Bon Koo
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
9
|
He S, Guan Y, Cheng CH, Moore TL, Luebke JI, Killiany RJ, Rosene DL, Koo BB, Ou Y. Human-to-monkey transfer learning identifies the frontal white matter as a key determinant for predicting monkey brain age. Front Aging Neurosci 2023; 15:1249415. [PMID: 38020785 PMCID: PMC10646581 DOI: 10.3389/fnagi.2023.1249415] [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: 06/28/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The application of artificial intelligence (AI) to summarize a whole-brain magnetic resonance image (MRI) into an effective "brain age" metric can provide a holistic, individualized, and objective view of how the brain interacts with various factors (e.g., genetics and lifestyle) during aging. Brain age predictions using deep learning (DL) have been widely used to quantify the developmental status of human brains, but their wider application to serve biomedical purposes is under criticism for requiring large samples and complicated interpretability. Animal models, i.e., rhesus monkeys, have offered a unique lens to understand the human brain - being a species in which aging patterns are similar, for which environmental and lifestyle factors are more readily controlled. However, applying DL methods in animal models suffers from data insufficiency as the availability of animal brain MRIs is limited compared to many thousands of human MRIs. We showed that transfer learning can mitigate the sample size problem, where transferring the pre-trained AI models from 8,859 human brain MRIs improved monkey brain age estimation accuracy and stability. The highest accuracy and stability occurred when transferring the 3D ResNet [mean absolute error (MAE) = 1.83 years] and the 2D global-local transformer (MAE = 1.92 years) models. Our models identified the frontal white matter as the most important feature for monkey brain age predictions, which is consistent with previous histological findings. This first DL-based, anatomically interpretable, and adaptive brain age estimator could broaden the application of AI techniques to various animal or disease samples and widen opportunities for research in non-human primate brains across the lifespan.
Collapse
Affiliation(s)
- Sheng He
- Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Yi Guan
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Chia Hsin Cheng
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Tara L. Moore
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jennifer I. Luebke
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Ronald J. Killiany
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Douglas L. Rosene
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Bang-Bon Koo
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Yangming Ou
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
10
|
Ibañez S, Sengupta N, Luebke JI, Wimmer K, Weaver CM. Myelin dystrophy in the aging prefrontal cortex leads to impaired signal transmission and working memory decline: a multiscale computational study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555476. [PMID: 37693412 PMCID: PMC10491254 DOI: 10.1101/2023.08.30.555476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Normal aging leads to myelin alternations in the rhesus monkey dorsolateral prefrontal cortex (dlPFC), which are often correlated with cognitive impairment. It is hypothesized that remyelination with shorter and thinner myelin sheaths partially compensates for myelin degradation, but computational modeling has not yet explored these two phenomena together systematically. Here, we used a two-pronged modeling approach to determine how age-related myelin changes affect a core cognitive function: spatial working memory. First we built a multicompartment pyramidal neuron model fit to monkey dlPFC data, with axon including myelinated segments having paranodes, juxtaparanodes, internodes, and tight junctions, to quantify conduction velocity (CV) changes and action potential (AP) failures after demyelination and subsequent remyelination in a population of neurons. Lasso regression identified distinctive parameter sets likely to modulate an axon's susceptibility to CV changes following demyelination versus remyelination. Next we incorporated the single neuron results into a spiking neural network model of working memory. While complete remyelination nearly recovered axonal transmission and network function to unperturbed levels, our models predict that biologically plausible levels of myelin dystrophy, if uncompensated by other factors, can account for substantial working memory impairment with aging. The present computational study unites empirical data from electron microscopy up to behavior on aging, and has broader implications for many demyelinating conditions, such as multiple sclerosis or schizophrenia.
Collapse
Affiliation(s)
- Sara Ibañez
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA 02118
- Centre de Recerca Matemàtica, Edifici C, Campus Bellaterra, 08193 Bellaterra, Spain
| | - Nilapratim Sengupta
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA 02118
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, USA 17604
| | - Jennifer I Luebke
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA 02118
| | - Klaus Wimmer
- Centre de Recerca Matemàtica, Edifici C, Campus Bellaterra, 08193 Bellaterra, Spain
| | - Christina M Weaver
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, USA 17604
| |
Collapse
|
11
|
Thompson González N, Machanda Z, Emery Thompson M. Age-related social selectivity: An adaptive lens on a later life social phenotype. Neurosci Biobehav Rev 2023; 152:105294. [PMID: 37380041 PMCID: PMC10529433 DOI: 10.1016/j.neubiorev.2023.105294] [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: 01/31/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
Age-related social selectivity is a process in which older humans reduce their number of social partners to a subset of positive and emotionally fulfilling relationships. Although selectivity has been attributed to humans' unique perceptions of time horizons, recent evidence demonstrates that these social patterns and processes occur in other non-human primates, suggesting an evolutionarily wider phenomenon. Here, we develop the hypothesis that selective social behavior is an adaptive strategy that allows social animals to balance the costs and benefits of navigating social environments in the face of age-related functional declines. We first aim to distinguish social selectivity from the non-adaptive social consequences of aging. We then outline multiple mechanisms by which social selectivity in old age may enhance fitness and healthspan. Our goal is to lay out a research agenda to identify selective strategies and their potential benefits. Given the importance of social support for health across primates, understanding why aging individuals lose social connections and how they can remain resilient has vital applications to public health research.
Collapse
Affiliation(s)
- Nicole Thompson González
- Integrative Anthropological Sciences, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Zarin Machanda
- Department of Anthropology, Tufts University, Medford, MA 02155, USA
| | | |
Collapse
|
12
|
Nguyen JN, Chauhan A. Bystanders or not? Microglia and lymphocytes in aging and stroke. Neural Regen Res 2023; 18:1397-1403. [PMID: 36571333 PMCID: PMC10075112 DOI: 10.4103/1673-5374.360345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
As the average age of the world population increases, more people will face debilitating aging-associated conditions, including dementia and stroke. Not only does the incidence of these conditions increase with age, but the recovery afterward is often worse in older patients. Researchers and health professionals must unveil and understand the factors behind age-associated diseases to develop a therapy for older patients. Aging causes profound changes in the immune system including the activation of microglia in the brain. Activated microglia promote T lymphocyte transmigration leading to an increase in neuroinflammation, white matter damage, and cognitive impairment in both older humans and rodents. The presence of T and B lymphocytes is observed in the aged brain and correlates with worse stroke outcomes. Preclinical strategies in stroke target either microglia or the lymphocytes or the communications between them to promote functional recovery in aged subjects. In this review, we examine the role of the microglia and T and B lymphocytes in aging and how they contribute to cognitive impairment. Additionally, we provide an important update on the contribution of these cells and their interactions in preclinical aged stroke.
Collapse
Affiliation(s)
- Justin N. Nguyen
- University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| |
Collapse
|
13
|
Moore TL, Medalla M, Ibañez S, Wimmer K, Mojica CA, Killiany RJ, Moss MB, Luebke JI, Rosene DL. Neuronal properties of pyramidal cells in lateral prefrontal cortex of the aging rhesus monkey brain are associated with performance deficits on spatial working memory but not executive function. GeroScience 2023:10.1007/s11357-023-00798-2. [PMID: 37106282 PMCID: PMC10400510 DOI: 10.1007/s11357-023-00798-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Age-related declines in cognitive abilities occur as early as middle-age in humans and rhesus monkeys. Specifically, performance by aged individuals on tasks of executive function (EF) and working memory (WM) is characterized by greater frequency of errors, shorter memory spans, increased frequency of perseverative responses, impaired use of feedback and reduced speed of processing. However, how aging precisely differentially impacts specific aspects of these cognitive functions and the distinct brain areas mediating cognition are not well understood. The prefrontal cortex (PFC) is known to mediate EF and WM and is an area that shows a vulnerability to age-related alterations in neuronal morphology. In the current study, we show that performance on EF and WM tasks exhibited significant changes with age, and these impairments correlate with changes in biophysical properties of layer 3 (L3) pyramidal neurons in lateral LPFC (LPFC). Specifically, there was a significant age-related increase in excitability of L3 LPFC pyramidal neurons, consistent with previous studies. Further, this age-related hyperexcitability of LPFC neurons was significantly correlated with age-related decline on a task of WM, but not an EF task. The current study characterizes age-related performance on tasks of WM and EF and provides insight into the neural substrates that may underlie changes in both WM and EF with age.
Collapse
Affiliation(s)
- Tara L Moore
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 700 Albany Street, W701, MA, 02118, Boston, USA.
- Center for Systems Neuroscience, Boston University, MA, 02115, Boston, USA.
| | - Maria Medalla
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 700 Albany Street, W701, MA, 02118, Boston, USA
- Center for Systems Neuroscience, Boston University, MA, 02115, Boston, USA
| | - Sara Ibañez
- Centre de Recerca Matemàtica, Edifici C, Campus Bellaterra, 08193, Bellaterra, Spain
| | - Klaus Wimmer
- Centre de Recerca Matemàtica, Edifici C, Campus Bellaterra, 08193, Bellaterra, Spain
| | - Chromewell A Mojica
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 700 Albany Street, W701, MA, 02118, Boston, USA
| | - Ronald J Killiany
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 700 Albany Street, W701, MA, 02118, Boston, USA
- Center for Systems Neuroscience, Boston University, MA, 02115, Boston, USA
| | - Mark B Moss
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 700 Albany Street, W701, MA, 02118, Boston, USA
- Center for Systems Neuroscience, Boston University, MA, 02115, Boston, USA
| | - Jennifer I Luebke
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 700 Albany Street, W701, MA, 02118, Boston, USA
- Center for Systems Neuroscience, Boston University, MA, 02115, Boston, USA
| | - Douglas L Rosene
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 700 Albany Street, W701, MA, 02118, Boston, USA
- Center for Systems Neuroscience, Boston University, MA, 02115, Boston, USA
| |
Collapse
|
14
|
Chapman TW, Olveda GE, Bame X, Pereira E, Hill RA. Oligodendrocyte death initiates synchronous remyelination to restore cortical myelin patterns in mice. Nat Neurosci 2023; 26:555-569. [PMID: 36928635 PMCID: PMC10208560 DOI: 10.1038/s41593-023-01271-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/09/2023] [Indexed: 03/18/2023]
Abstract
Myelin degeneration occurs in neurodegenerative diseases and aging. In these conditions, resident oligodendrocyte progenitor cells (OPCs) differentiate into oligodendrocytes that carry out myelin repair. To investigate the cellular dynamics underlying these events, we developed a noninflammatory demyelination model that combines intravital two-photon imaging with a single-cell ablation technique called two-photon apoptotic targeted ablation (2Phatal). Oligodendrocyte 2Phatal in both sexes results in a myelin degeneration cascade that triggers rapid forms of synchronous remyelination on defined axons. This remyelination is driven by oligodendrocytes differentiated from a subset of morphologically distinct, highly branched OPCs. Moreover, remyelination efficiency depends on the initial myelin patterns, as well as the age of the organism. In summary, using 2Phatal, we show a form of rapid synchronous remyelination, mediated by a distinct subset of OPCs, capable of restoring the original myelin patterning in adulthood but not aging.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Genaro E Olveda
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Xhoela Bame
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Elizabeth Pereira
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
15
|
Moore TL, Medalla M, Iba Ez S, Wimmer K, Mojica CA, Killiany RJ, Moss MB, Luebke JI, Rosene DL. Neuronal properties of pyramidal cells in lateral prefrontal cortex of the aging rhesus monkey brain are associated with performance deficits on spatial working memory but not executive function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527321. [PMID: 36798388 PMCID: PMC9934587 DOI: 10.1101/2023.02.07.527321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Age-related declines in cognitive abilities occur as early as middle-age in humans and rhesus monkeys. Specifically, performance by aged individuals on tasks of executive function (EF) and working memory (WM) is characterized by greater frequency of errors, shorter memory spans, increased frequency of perseverative responses, impaired use of feedback and reduced speed of processing. However, how aging precisely differentially impacts specific aspects of these cognitive functions and the distinct brain areas mediating cognition are not well understood. The prefrontal cortex (PFC) is known to mediate EF and WM and is an area that shows a vulnerability to age-related alterations in neuronal morphology. In the current study, we show that performance on EF and WM tasks exhibited significant changes with age and these impairments correlate with changes in biophysical properties of L3 pyramidal neurons in lateral LPFC (LPFC). Specifically, there was a significant age-related increase in excitability of Layer 3 LPFC pyramidal neurons, consistent with previous studies. Further, this age-related hyperexcitability of LPFC neurons was significantly correlated with age-related decline on a task of WM, but not an EF task. The current study characterizes age-related performance on tasks of WM and EF and provides insight into the neural substrates that may underlie changes in both WM and EF with age.
Collapse
|
16
|
Bao S, Liao C, Xu N, Deng A, Luo Y, Ouyang Z, Guo X, Liu Y, Ke T, Yang J. Prediction of brain age using quantitative parameters of synthetic magnetic resonance imaging. Front Aging Neurosci 2022; 14:963668. [PMID: 36457759 PMCID: PMC9705592 DOI: 10.3389/fnagi.2022.963668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Brain tissue changes dynamically during aging. The purpose of this study was to use synthetic magnetic resonance imaging (syMRI) to evaluate the changes in relaxation values in different brain regions during brain aging and to construct a brain age prediction model. Materials and methods Quantitative MRI was performed on 1,000 healthy people (≥ 18 years old) from September 2020 to October 2021. T1, T2 and proton density (PD) values were simultaneously measured in 17 regions of interest (the cerebellar hemispheric cortex, pons, amygdala, hippocampal head, hippocampal tail, temporal lobe, occipital lobe, frontal lobe, caudate nucleus, lentiform nucleus, dorsal thalamus, centrum semiovale, parietal lobe, precentral gyrus, postcentral gyrus, substantia nigra, and red nucleus). The relationship between the relaxation values and age was investigated. In addition, we analyzed the relationship between brain tissue values and sex. Finally, the participants were divided into two age groups: < 60 years old and ≥ 60 years old. Logistic regression analysis was carried out on the two groups of data. According to the weight of related factors, a brain age prediction model was established and verified. Results We obtained the specific reference value range of different brain regions of individuals in different age groups and found that there were differences in relaxation values in brain tissue between different sexes in the same age group. Moreover, the relaxation values of most brain regions in males were slightly higher than those in females. In the study of age and brain relaxation, it was found that brain relaxation values were correlated with age. The T1 values of the centrum semiovale increased with age, the PD values of the centrum semiovale increased with age, while the T2 values of the caudate nucleus and lentiform nucleus decreased with age. Seven brain age prediction models were constructed with high sensitivity and specificity, among which the combined T1, T2 and PD values showed the best prediction efficiency. In the training set, the area under the curve (AUC), specificity and sensitivity were 0.959 [95% confidence interval (CI): 0.945–0.974], 91.51% and 89.36%, respectively. In the test cohort, the above indicators were 0.916 (95% CI: 0.882–0.951), 89.24% and 80.33%, respectively. Conclusion Our study provides specific reference ranges of T1, T2, and PD values in different brain regions from healthy adults of different ages. In addition, there are differences in brain relaxation values in some brain regions between different sexes, which help to provide new ideas for brain diseases that differ according to sex. The brain age model based on synthetic MRI is helpful to determine brain age.
Collapse
|
17
|
Gullstrand J, Claidière N, Fagot J. Age effect in expert cognitive flexibility in Guinea baboons (Papio papio). Behav Brain Res 2022; 434:114043. [PMID: 35933047 DOI: 10.1016/j.bbr.2022.114043] [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: 01/19/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/02/2022]
Abstract
Cognitive flexibility in non-human primates is traditionally measured with the conceptual set shifting task (CSST). In our laboratory, Guinea baboons (N = 24) were continuously tested with a CSST task during approximately 10 years. Our task involved the presentation of three stimuli on a touch screen all made from 3 possible colours and 3 shapes. The subjects had to touch the stimulus containing the stimulus dimension (e.g., green) that was constantly rewarded until the stimulus dimension changed. Analysis of perseveration responses, scores and response times collected during the last two years of testing (approximately 1.6 million trials) indicate (1) that the baboons have developed an "expert" form of cognitive flexibility and (2) that their performance was age-dependent, it was at a developing stage in juveniles, optimal in adults, declining in middle-aged, and strongly impaired in the oldest age group. A direct comparison with the data collected by Bonté , Flemming & Fagot (2011) on some of the same baboons and same task as in the current study indicates that (3) the performance of all age groups has improved after 10 years of training, even for the now old individuals. All these data validate the use of non-human primates as models of human cognitive flexibility and suggest that cognitive flexibility in humans has a long evolutionary history.
Collapse
Affiliation(s)
- Julie Gullstrand
- Laboratory of Cognitive Psychology, CNRS, Aix-Marseille University, and Primatology Station of the CNRS-Celphedia, France.
| | - Nicolas Claidière
- Laboratory of Cognitive Psychology, CNRS, Aix-Marseille University, and Primatology Station of the CNRS-Celphedia, France
| | - Joel Fagot
- Laboratory of Cognitive Psychology, CNRS, Aix-Marseille University, and Primatology Station of the CNRS-Celphedia, France.
| |
Collapse
|
18
|
Benitez A, Jensen JH, Thorn K, Dhiman S, Fountain-Zaragoza S, Rieter WJ, Spampinato MV, Hamlett ED, Nietert PJ, Falangola MDF, Helpern JA. Greater diffusion restriction in white matter in Preclinical Alzheimer's disease. Ann Neurol 2022; 91:864-877. [PMID: 35285067 DOI: 10.1002/ana.26353] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/14/2022] [Accepted: 03/07/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVE The Alzheimer's Continuum is biologically defined by beta-amyloid deposition which, at the earliest stages, is superimposed upon white matter degeneration in aging. However, the extent to which these co-occurring changes are characterized is relatively under-explored. The goal of this study was to use Diffusional Kurtosis Imaging (DKI) and biophysical modeling to detect and describe amyloid-related white matter changes in preclinical Alzheimer's disease (AD). METHODS Cognitively unimpaired participants ages 45-85 completed brain MRI, amyloid PET (florbetapir), neuropsychological testing, and other clinical measures at baseline in a cohort study. We tested whether beta amyloid-negative (AB-) and -positive (AB+) participants differed on DKI-based conventional (i.e. Fractional Anisotropy [FA], Mean Diffusivity [MD], Mean Kurtosis [MK]) and modeling (i.e. Axonal Water Fraction [AWF], extra-axonal radial diffusivity [De,⊥ ]) metrics, and whether these metrics were associated with other biomarkers. RESULTS We found significantly greater diffusion restriction (higher FA/AWF, lower MD/ De,⊥ ) in white matter in AB+ than AB- (partial η2 = 0.08-0.19), more notably in the extra-axonal space within primarily late-myelinating tracts. Diffusion metrics predicted amyloid status incrementally over age (AUC=0.84) with modest yet selective associations, where AWF (a marker of axonal density) correlated with speed/executive functions and neurodegeneration, whereas De,⊥ (a marker of gliosis/myelin repair) correlated with amyloid deposition and white matter hyperintensity volume. INTERPRETATION These results support prior evidence of a non-monotonic change in diffusion behavior, where an early increase in diffusion restriction is hypothesized to reflect inflammation and myelin repair prior to an ensuing decrease in diffusion restriction, indicating glial and neuronal degeneration. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Andreana Benitez
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA
- Department of Radiology and Radiological Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Jens H Jensen
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Kathryn Thorn
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Siddhartha Dhiman
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Stephanie Fountain-Zaragoza
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA
| | - William J Rieter
- Department of Radiology and Radiological Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Maria Vittoria Spampinato
- Department of Radiology and Radiological Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Eric D Hamlett
- Department of Pathology and Laboratory Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Paul J Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Maria de Fatima Falangola
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Joseph A Helpern
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
19
|
Ahn K, Lee SJ, Mook-Jung I. White matter-associated microglia: New players in brain aging and neurodegenerative diseases. Ageing Res Rev 2022; 75:101574. [PMID: 35093614 DOI: 10.1016/j.arr.2022.101574] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 12/20/2022]
Abstract
There has been growing interest in brain aging and rejuvenation. It is well known that brain aging is one of the leading causes of neurodegenerative diseases, such as Alzheimer's disease, but brain aging alone can cause cognitive decline. Microglia are thought to act as 'conductors' of white matter aging by modulating diverse glial cells and phagocytosing white matter-derived myelin debris. A recent study identified a specific subpopulation of microglia in the white matter of aged mice, termed white matter-associated microglia (WAM). Additionally, senescent microglia show impaired phagocytic function and altered lipid metabolism, which cause accumulation of lipid metabolites and eventually lead to myelin sheath degeneration. These results suggest that senescent WAM could be pivotal players in axonal loss during brain aging. The aim of this review is to assess the current state of knowledge on brain aging, with an emphasis on the roles of the white matter and microglia, and suggest potential approaches for rejuvenating the aged brain.
Collapse
Affiliation(s)
- Kyusik Ahn
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SNU Dementia Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Inhee Mook-Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SNU Dementia Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
20
|
Kim T, Aizenstein HJ, Snitz BE, Cheng Y, Chang YF, Roush RE, Huppert TJ, Cohen A, Doman J, Becker JT. Tract Specific White Matter Lesion Load Affects White Matter Microstructure and Their Relationships With Functional Connectivity and Cognitive Decline. Front Aging Neurosci 2022; 13:760663. [PMID: 35185514 PMCID: PMC8848259 DOI: 10.3389/fnagi.2021.760663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022] Open
Abstract
White matter hyperintensities (WMHs) are associated with cognitive decline. Assessing the effect of WMH on WM microstructural changes and its relationships with structural and functional connectivity to multiple cognitive domains are helpful to better understand the pathophysiological processes of cognitive impairment. 65 participants (49 normal and 16 MCI subjects, age: 67.4 ± 8.3 years, 44 females) were studied at 3T. The WMHs and fifty fiber tracts were automatically segmented from the T1/T2-weighted images and diffusion-weighted images, respectively. Tract-profiles of WMH were compared with those of apparent fiber density (AFD). The relationship between AFD and tract connectivity (TC) was assessed. Functional connectivity (FC) between tract ends obtained from resting-state functional MRI was examined in relation to TC. Tract-specific relationships of WMH, TC and FC with a multi-domain neuropsychological test battery and Montreal Cognitive Assessment (MoCA) were also separately assessed by lasso linear regression. Indirect pathways of TC and FC between WMH and multiple cognitive measures were tested using the mediation analysis. Higher WMH loads in WM tracts were locally matched with the reduced AFD, which was related to decrease in TC. However, no direct relationship was found between TC and FC. Tract-specific changes on WMH, TC and FC for each cognitive performance may explain that macro- and microstructural and functional changes are associated differently with each cognitive domain in a fiber specific manner. In these identified tracts, the differences between normal and MCI for WMH and TC were increased, and the relationships of WMH, TC and FC with cognitive outcomes were more significant, compared to the results from all tracts. Indirect pathways of two-step (TC-FC) between WMH and all cognitive domains were significant (p < 0.0083 with Bonferroni correction), while the separated indirect pathways through TC and through FC were different depending on cognitive domain. Deterioration in specific cognitive domains may be affected by alterations in a set of different tracts that are differently associated with macrostructural, microstructural, and function changes. Thus, assessments of WMH and its associated changes on specific tracts help for better understanding of the interrelationships of multiple changes in cognitive impairment.
Collapse
Affiliation(s)
- Tae Kim
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- *Correspondence: Tae Kim,
| | - Howard J. Aizenstein
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Beth E. Snitz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yu Cheng
- Departments of Statistics and Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yue-Fang Chang
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rebecca E. Roush
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Theodore J. Huppert
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Deparement of Electrical and Computer Engineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Annie Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jack Doman
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - James T. Becker
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
21
|
Chang W, Weaver CM, Medalla M, Moore TL, Luebke JI. Age-related alterations to working memory and to pyramidal neurons in the prefrontal cortex of rhesus monkeys begin in early middle-age and are partially ameliorated by dietary curcumin. Neurobiol Aging 2022; 109:113-124. [PMID: 34715442 PMCID: PMC8671373 DOI: 10.1016/j.neurobiolaging.2021.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/18/2021] [Accepted: 09/08/2021] [Indexed: 01/03/2023]
Abstract
Layer 3 (L3) pyramidal neurons in aged rhesus monkey lateral prefrontal cortex (LPFC) exhibit significantly elevated excitability in vitro and reduced spine density compared to neurons in young subjects. The time-course of these alterations, and whether they can be ameliorated in middle age by the powerful anti-oxidant curcumin is unknown. We compared the properties of L3 pyramidal neurons from the LPFC of behaviorally characterized rhesus monkeys over the adult lifespan using whole-cell patch clamp recordings and neuronal reconstructions. Working memory (WM) impairment, neuronal hyperexcitability, and spine loss began in middle age. There was no significant relationship between neuronal properties and WM performance. Middle-aged subjects given curcumin exhibited better WM performance and less neuronal excitability compared to control subjects. These findings suggest that the appropriate time frame for intervention for age-related cognitive changes is early middle age, and points to the efficacy of curcumin in delaying WM decline. Because there was no relationship between excitability and behavior, the effects of curcumin on these measures appear to be independent.
Collapse
Affiliation(s)
- W Chang
- Department of Anatomy & Neurobiology, Boston University School of Medicine 650 Albany St., Boston MA 02118 USA
| | - C M Weaver
- Department of Mathematics, Franklin and Marshall College, Lancaster PA 17604 USA
| | - M Medalla
- Department of Anatomy & Neurobiology, Boston University School of Medicine 650 Albany St., Boston MA 02118 USA
| | - T L Moore
- Department of Anatomy & Neurobiology, Boston University School of Medicine 650 Albany St., Boston MA 02118 USA
| | - J I Luebke
- Department of Anatomy & Neurobiology, Boston University School of Medicine 650 Albany St., Boston MA 02118 USA.
| |
Collapse
|
22
|
Freire-Cobo C, Edler MK, Varghese M, Munger E, Laffey J, Raia S, In SS, Wicinski B, Medalla M, Perez SE, Mufson EJ, Erwin JM, Guevara EE, Sherwood CC, Luebke JI, Lacreuse A, Raghanti MA, Hof PR. Comparative neuropathology in aging primates: A perspective. Am J Primatol 2021; 83:e23299. [PMID: 34255875 PMCID: PMC8551009 DOI: 10.1002/ajp.23299] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/24/2021] [Accepted: 06/06/2021] [Indexed: 12/27/2022]
Abstract
While humans exhibit a significant degree of neuropathological changes associated with deficits in cognitive and memory functions during aging, non-human primates (NHP) present with more variable expressions of pathological alterations among individuals and species. As such, NHP with long life expectancy in captivity offer an opportunity to study brain senescence in the absence of the typical cellular pathology caused by age-related neurodegenerative illnesses commonly seen in humans. Age-related changes at neuronal population, single cell, and synaptic levels have been well documented in macaques and marmosets, while age-related and Alzheimer's disease-like neuropathology has been characterized in additional species including lemurs as well as great apes. We present a comparative overview of existing neuropathologic observations across the primate order, including classic age-related changes such as cell loss, amyloid deposition, amyloid angiopathy, and tau accumulation. We also review existing cellular and ultrastructural data on neuronal changes, such as dendritic attrition and spine alterations, synaptic loss and pathology, and axonal and myelin pathology, and discuss their repercussions on cellular and systems function and cognition.
Collapse
Affiliation(s)
- Carmen Freire-Cobo
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Melissa K Edler
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Merina Varghese
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emily Munger
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Jessie Laffey
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sophia Raia
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Selena S In
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bridget Wicinski
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Maria Medalla
- Department of Anatomy and Neurobiology, Center for Systems Neuroscience, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sylvia E Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
- Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Joseph M Erwin
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
| | - Elaine E Guevara
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
- Department of Evolutionary Anthropology, Duke University, Durham, North Carolina, USA
| | - Chet C Sherwood
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
| | - Jennifer I Luebke
- Department of Anatomy and Neurobiology, Center for Systems Neuroscience, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Agnès Lacreuse
- Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Mary A Raghanti
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
23
|
Lomidze N, Zhvania MG, Tizabi Y, Japaridze N, Pochkhidze N, Rzayev F, Lordkipanidze T. Aging affects cognition and hippocampal ultrastructure in male Wistar rats. Dev Neurobiol 2021; 81:833-846. [PMID: 34047044 DOI: 10.1002/dneu.22839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/11/2021] [Accepted: 05/16/2021] [Indexed: 12/18/2022]
Abstract
It is now well established that aging is associated with emotional and cognitive changes. Although the basis of such changes is not fully understood, ultrastructural alterations in key brain areas are likely contributing factors. Recently, we reported that aging-related anxiety in male Wistar rats is associated with ultrastructural changes in the central nucleus of amygdala, an area that plays important role in emotional regulation. In this study, we evaluated the cognitive performance of adolescent, adult, and aged male Wistar rats in multi-branch maze (MBM) as well as in Morris water maze (MWM). We also performed ultrastructural analysis of the CA1 region of the hippocampus, an area intimately involved in cognitive function. The behavioral data indicate significant impairments in few indices of cognitive functions in both tests in aged rats compared to the other two age groups. Concomitantly, a total number of presynaptic vesicles as well as vesicles in the resting pool were significantly lower, whereas postsynaptic mitochondrial area was significantly higher in aged rats compared to the other age groups. No significant differences in presynaptic terminal area or postsynaptic mitochondrial number were detected between the three age groups. These results indicate that selective ultrastructural changes in specific hippocampal region may accompany cognitive decline in aging rats.
Collapse
Affiliation(s)
- Nino Lomidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| | - Mzia G Zhvania
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashviloi Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Yousef Tizabi
- Department of Pharmacology Howard, University College of Medicine, Washington, District of Columbia, USA
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashviloi Center of Experimental Biomedicine, Tbilisi, Georgia.,Medical School, New Vision University, Tbilisi, Georgia
| | - Nino Pochkhidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashviloi Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Fuad Rzayev
- Department of Histology, Embryology and Cytology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Tamar Lordkipanidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| |
Collapse
|
24
|
Corpus callosum morphology across the lifespan in baboons (Papio anubis): A cross-sectional study of relative mid-sagittal surface area and thickness. Neurosci Res 2021; 171:19-26. [PMID: 33744333 DOI: 10.1016/j.neures.2021.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 11/22/2022]
Abstract
The corpus callosum enables integration and coordination of cognitive processing between the cerebral hemispheres. In the aging human brain, these functions are affected by progressive axon and myelin deteriorations, reflected as atrophy of the midsagittal corpus callosum in old age. In non-human primates, these degenerative processes are less pronounced as previous morphometric studies on capuchin monkey, rhesus monkeys, and chimpanzees do not find old-age callosal atrophy. In the present study, we extend these previous findings by studying callosal development of the olive baboon (Papio anubis) across the lifespan and compare it to chimpanzee and human data. For this purpose, total relative (to forebrain volume) midsagittal area, subsectional area, and regional thickness of the corpus callosum were assessed in 91 male and female baboons using non-invasive MRI-based morphometry. The studied age range was 2.5-26.6 years and lifespan trajectories were fitted using general additive modelling. Relative area of the total and anterior corpus callosum showed a positive linear trajectory. That is, both measures increased slowly but continuously from childhood into old age, and no decline was observed in old age. Thus, comparable with all other non-human primates studied to-date, baboons do not show callosal atrophy in old age. This observation lends supports to the notion that atrophy of the corpus callosum is a unique characteristic of human brain aging.
Collapse
|
25
|
Batterman KV, Cabrera PE, Moore TL, Rosene DL. T Cells Actively Infiltrate the White Matter of the Aging Monkey Brain in Relation to Increased Microglial Reactivity and Cognitive Decline. Front Immunol 2021; 12:607691. [PMID: 33664743 PMCID: PMC7920950 DOI: 10.3389/fimmu.2021.607691] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Normal aging is characterized by declines in processing speed, learning, memory, and executive function even in the absence of neurodegenerative diseases such as Alzheimer's Disease (AD). In normal aging monkeys and humans, neuronal loss does not account for cognitive impairment. Instead, loss of white matter volume and an accumulation of myelin sheath pathology begins in middle age and is associated with cognitive decline. It is unknown what causes this myelin pathology, but it likely involves increased neuroinflammation in white matter and failures in oligodendrocyte function (maturation and repair). In frontal white matter tracts vulnerable to myelin damage, microglia become chronically reactive and secrete harmful pro-inflammatory cytokines. Despite being in a phagocytic state, these microglia are ineffective at phagocytosing accruing myelin debris, which directly inhibits myelin sheath repair. Here, we asked whether reported age-related increases in pro-inflammatory markers were accompanied by an adaptive immune response involving T cells. We quantified T cells with immunohistochemistry in the brains of 34 cognitively characterized monkeys and found an age-related increase in perivascular T cells that surround CNS vasculature. We found a surprising age-related increase in T cells that infiltrate the white matter parenchyma. In the cingulum bundle the percentage of these parenchymal T cells increased with age relative to those in the perivascular space. In contrast, infiltrating T cells were rarely found in surrounding gray matter regions. We assessed whether T cell infiltration correlated with fibrinogen extravasation from the vasculature as a measure of BBB leakiness and found no correlation, suggesting that T cell infiltration is not a result of passive extravasation. Importantly, the density of T cells in the cingulum bundle correlated with microglial reactivity and with cognitive impairment. This is the first demonstration that T cell infiltration of white matter is associated with cognitive decline in the normal aging monkey.
Collapse
Affiliation(s)
- Katelyn V Batterman
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Payton E Cabrera
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Tara L Moore
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States.,Laboratory of Interventions for Cortical Injury and Cognitive Decline, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States.,Center for Systems Neuroscience, Boston University, Boston, MA, United States
| | - Douglas L Rosene
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States.,Center for Systems Neuroscience, Boston University, Boston, MA, United States
| |
Collapse
|
26
|
Stonebarger GA, Urbanski HF, Woltjer RL, Vaughan KL, Ingram DK, Schultz PL, Calderazzo SM, Siedeman JA, Mattison JA, Rosene DL, Kohama SG. Amyloidosis increase is not attenuated by long-term calorie restriction or related to neuron density in the prefrontal cortex of extremely aged rhesus macaques. GeroScience 2020; 42:1733-1749. [PMID: 32876855 PMCID: PMC7732935 DOI: 10.1007/s11357-020-00259-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/24/2020] [Indexed: 01/30/2023] Open
Abstract
As human lifespan increases and the population ages, diseases of aging such as Alzheimer's disease (AD) are a major cause for concern. Although calorie restriction (CR) as an intervention has been shown to increase healthspan in many species, few studies have examined the effects of CR on brain aging in primates. Using postmortem tissue from a cohort of extremely aged rhesus monkeys (22-44 years old, average age 31.8 years) from a longitudinal CR study, we measured immunohistochemically labeled amyloid beta plaques in Brodmann areas 32 and 46 of the prefrontal cortex, areas that play key roles in cognitive processing, are sensitive to aging and, in humans, are also susceptible to AD pathogenesis. We also evaluated these areas for cortical neuron loss, which has not been observed in younger cohorts of aged monkeys. We found a significant increase in plaque density with age, but this was unaffected by diet. Moreover, there was no change in neuron density with age or treatment. These data suggest that even in the oldest-old rhesus macaques, amyloid beta plaques do not lead to overt neuron loss. Hence, the rhesus macaque serves as a pragmatic animal model for normative human aging but is not a complete model of the neurodegeneration of AD. This model of aging may instead prove most useful for determining how even the oldest monkeys are protected from AD, and this information may therefore yield valuable information for clinical AD treatments.
Collapse
Affiliation(s)
- G A Stonebarger
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - H F Urbanski
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - R L Woltjer
- Department of Pathology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - K L Vaughan
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, NIH, Dickerson, MD, 20842, USA
- Charles River, Wilmington, MA, 01867, USA
| | - D K Ingram
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - P L Schultz
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MD, 02218, USA
| | - S M Calderazzo
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MD, 02218, USA
| | - J A Siedeman
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MD, 02218, USA
| | - J A Mattison
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, NIH, Dickerson, MD, 20842, USA
| | - D L Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MD, 02218, USA
| | - S G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR, 97006, USA.
| |
Collapse
|
27
|
Datta D, Enwright JF, Arion D, Paspalas CD, Morozov YM, Lewis DA, Arnsten AFT. Mapping Phosphodiesterase 4D (PDE4D) in Macaque Dorsolateral Prefrontal Cortex: Postsynaptic Compartmentalization in Layer III Pyramidal Cell Circuits. Front Neuroanat 2020; 14:578483. [PMID: 33328902 PMCID: PMC7714912 DOI: 10.3389/fnana.2020.578483] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022] Open
Abstract
cAMP signaling has powerful, negative effects on cognitive functions of the primate dorsolateral prefrontal cortex (dlPFC), opening potassium channels to reduce firing and impair working memory, and increasing tau phosphorylation in aging neurons. This contrasts with cAMP actions in classic circuits, where it enhances plasticity and transmitter release. PDE4 isozymes regulate cAMP actions, and thus have been a focus of research and drug discovery. Previous work has focused on the localization of PDE4A and PDE4B in dlPFC, but PDE4D is also of great interest, as it is the predominant PDE4 isoform in primate association cortex, and PDE4D expression decreases with aging in human dlPFC. Here we used laser-capture microdissection transcriptomics and found that PDE4D message is enriched in pyramidal cells compared to GABAergic PV-interneurons in layer III of the human dlPFC. A parallel study in rhesus macaques using high-spatial resolution immunoelectron microscopy revealed the ultrastructural locations of PDE4D in primate dlPFC with clarity not possible in human post-mortem tissue. PDE4D was especially prominent in dendrites associated with microtubules, mitochondria, and likely smooth endoplasmic reticulum (SER). There was substantial postsynaptic labeling in dendritic spines, associated with the SER spine-apparatus near glutamatergic-like axospinous synapses, but sparse labeling in axon terminals. We also observed dense PDE4D labeling perisynaptically in astroglial leaflets ensheathing glutamatergic connections. These data suggest that PDE4D is strategically positioned to regulate cAMP signaling in dlPFC glutamatergic synapses and circuits, especially in postsynaptic compartments where it is localized to influence cAMP actions on intracellular trafficking, mitochondrial physiology, and internal calcium release.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - John F. Enwright
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dominique Arion
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Constantinos D. Paspalas
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Yury M. Morozov
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - David A. Lewis
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Amy F. T. Arnsten
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
28
|
Köhncke Y, Düzel S, Sander MC, Lindenberger U, Kühn S, Brandmaier AM. Hippocampal and Parahippocampal Gray Matter Structural Integrity Assessed by Multimodal Imaging Is Associated with Episodic Memory in Old Age. Cereb Cortex 2020; 31:1464-1477. [PMID: 33150357 PMCID: PMC7869080 DOI: 10.1093/cercor/bhaa287] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/29/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023] Open
Abstract
Maintained structural integrity of hippocampal and cortical gray matter may explain why some older adults show rather preserved episodic memory. However, viable measurement models for estimating individual differences in gray matter structural integrity are lacking; instead, findings rely on fallible single indicators of integrity. Here, we introduce multitrait–multimethod methodology to capture individual differences in gray matter integrity, based on multimodal structural imaging in a large sample of 1522 healthy adults aged 60–88 years from the Berlin Aging Study II, including 333 participants who underwent magnetic resonance imaging. Structural integrity factors expressed the common variance of voxel-based morphometry, mean diffusivity, and magnetization transfer ratio for each of four regions of interest: hippocampus, parahippocampal gyrus, prefrontal cortex, and precuneus. Except for precuneus, the integrity factors correlated with episodic memory. Associations with hippocampal and parahippocampal integrity persisted after controlling for age, sex, and education. Our results support the proposition that episodic memory ability in old age benefits from maintained structural integrity of hippocampus and parahippocampal gyrus. Exploratory follow-up analyses on sex differences showed that this effect is restricted to men. Multimodal factors of structural brain integrity might help to improve our biological understanding of human memory aging.
Collapse
Affiliation(s)
- Ylva Köhncke
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| | - Sandra Düzel
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| | - Myriam C Sander
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| | - Ulman Lindenberger
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany.,Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| | - Simone Kühn
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.,Lise Meitner Group for Environmental Neuroscience, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| | - Andreas M Brandmaier
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany.,Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| |
Collapse
|
29
|
Chiou KL, Montague MJ, Goldman EA, Watowich MM, Sams SN, Song J, Horvath JE, Sterner KN, Ruiz-Lambides AV, Martínez MI, Higham JP, Brent LJN, Platt ML, Snyder-Mackler N. Rhesus macaques as a tractable physiological model of human ageing. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190612. [PMID: 32951555 DOI: 10.1098/rstb.2019.0612] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Research in the basic biology of ageing is increasingly identifying mechanisms and modifiers of ageing in short-lived organisms such as worms and mice. The ultimate goal of such work is to improve human health, particularly in the growing segment of the population surviving into old age. Thus far, few interventions have robustly transcended species boundaries in the laboratory, suggesting that changes in approach are needed to avoid costly failures in translational human research. In this review, we discuss both well-established and alternative model organisms for ageing research and outline how research in nonhuman primates is sorely needed, first, to translate findings from short-lived organisms to humans, and second, to understand key aspects of ageing that are unique to primate biology. We focus on rhesus macaques as a particularly promising model organism for ageing research owing to their social and physiological similarity to humans as well as the existence of key resources that have been developed for this species. As a case study, we compare gene regulatory signatures of ageing in the peripheral immune system between humans and rhesus macaques from a free-ranging study population in Cayo Santiago. We show that both mRNA expression and DNA methylation signatures of immune ageing are broadly shared between macaques and humans, indicating strong conservation of the trajectory of ageing in the immune system. We conclude with a review of key issues in the biology of ageing for which macaques and other nonhuman primates may uniquely contribute valuable insights, including the effects of social gradients on health and ageing. We anticipate that continuing research in rhesus macaques and other nonhuman primates will play a critical role in conjunction with the model organism and human biodemographic research in ultimately improving translational outcomes and extending health and longevity in our ageing population. This article is part of the theme issue 'Evolution of the primate ageing process'.
Collapse
Affiliation(s)
- Kenneth L Chiou
- Department of Psychology, University of Washington, Seattle, WA 98195, USA.,Department of Pathology, Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Washington, Seattle, WA 98195, USA.,Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85281, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Marina M Watowich
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Sierra N Sams
- Department of Psychology, University of Washington, Seattle, WA 98195, USA
| | - Jeff Song
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
| | - Julie E Horvath
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA.,Research and Collections Section, North Carolina Museum of Natural Sciences, Raleigh, NC 27601, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,Department of Evolutionary Anthropology, Duke University, Durham, NC 27708, USA
| | - Kirstin N Sterner
- Department of Anthropology, University of Oregon, Eugene, OR 97403, USA
| | - Angelina V Ruiz-Lambides
- Caribbean Primate Research Center, Unit of Comparative Medicine, University of Puerto Rico, San Juan, PR 00936, USA
| | - Melween I Martínez
- Caribbean Primate Research Center, Unit of Comparative Medicine, University of Puerto Rico, San Juan, PR 00936, USA
| | - James P Higham
- Department of Anthropology, New York University, New York, NY 10003, USA.,New York Consortium in Evolutionary Primatology, New York, NY, USA
| | - Lauren J N Brent
- Centre for Research in Animal Behaviour, University of Exeter, Exeter EX4 4QG, UK
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Marketing, Wharton School of Business, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Noah Snyder-Mackler
- Department of Psychology, University of Washington, Seattle, WA 98195, USA.,Department of Pathology, Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Washington, Seattle, WA 98195, USA.,Department of Biology, University of Washington, Seattle, WA 98195, USA.,Center for Studies in Demography and Ecology, University of Washington, Seattle, WA 98195, USA.,Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85281, USA.,School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
30
|
Wang X, Chen K, Pan M, Ge W, He Z. Comparison of proteome alterations during aging in the temporal lobe of humans and rhesus macaques. Exp Brain Res 2020; 238:1963-1976. [PMID: 32572507 DOI: 10.1007/s00221-020-05855-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/11/2020] [Indexed: 01/06/2023]
Abstract
Rhesus macaques are widely used as animal models for studies of the nervous system; however, it is unknown whether the alterations in the protein profile of the brain during aging are conserved between humans and rhesus macaques. In this study, temporal cortex samples from old and young humans (84 vs. 34 years, respectively) or rhesus macaques (20 vs. 6 years, respectively) were subjected to tandem mass tag-labeled proteomic analysis followed by bioinformatic analysis. A total of 3861 homologous pairs of proteins were identified during the aging process. The conservatively upregulated proteins (n = 190) were involved mainly in extracellular matrix (ECM), focal adhesion and coagulation; while, the conservatively downregulated proteins (n = 56) were enriched in ribosome. Network analysis showed that these conservatively regulated proteins interacted with each other with respect to protein synthesis and cytoskeleton-ECM connection. Many proteins in the focal adhesion, blood clotting, complement and coagulation, and cytoplasmic ribosomal protein pathways were regulated in the same direction in human and macaque; while, proteins involved in oligodendrocyte specification and differentiation pathways were downregulated during human aging, and many proteins in the electron transport chain pathway showed differences in the altered expression profiles. Data are available via ProteomeXchange with identifier PXD013597. Our findings suggest similarities in some changes in brain protein profiles during aging both in humans and macaques, although other changes are unique to only one of these species.
Collapse
Affiliation(s)
- Xia Wang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Kang Chen
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Meng Pan
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China. .,Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China.
| | - Zhanlong He
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
| |
Collapse
|
31
|
Damisah EC, Hill RA, Rai A, Chen F, Rothlin CV, Ghosh S, Grutzendler J. Astrocytes and microglia play orchestrated roles and respect phagocytic territories during neuronal corpse removal in vivo. SCIENCE ADVANCES 2020; 6:eaba3239. [PMID: 32637606 PMCID: PMC7319765 DOI: 10.1126/sciadv.aba3239] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/12/2020] [Indexed: 05/09/2023]
Abstract
Cell death is prevalent throughout life; however, the coordinated interactions and roles of phagocytes during corpse removal in the live brain are poorly understood. We developed photochemical and viral methodologies to induce death in single cells and combined this with intravital optical imaging. This approach allowed us to track multicellular phagocytic interactions with precise spatiotemporal resolution. Astrocytes and microglia engaged with dying neurons in an orchestrated and synchronized fashion. Each glial cell played specialized roles: Astrocyte processes rapidly polarized and engulfed numerous small dendritic apoptotic bodies, while microglia migrated and engulfed the soma and apical dendrites. The relative involvement and phagocytic specialization of each glial cell was plastic and controlled by the receptor tyrosine kinase Mertk. In aging, there was a marked delay in apoptotic cell removal. Thus, a precisely orchestrated response and cross-talk between glial cells during corpse removal may be critical for maintaining brain homeostasis.
Collapse
Affiliation(s)
- Eyiyemisi C. Damisah
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Robert A. Hill
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Anupama Rai
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Fuyi Chen
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Carla V. Rothlin
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Sourav Ghosh
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
32
|
Brasó-Vives M, Povolotskaya IS, Hartasánchez DA, Farré X, Fernandez-Callejo M, Raveendran M, Harris RA, Rosene DL, Lorente-Galdos B, Navarro A, Marques-Bonet T, Rogers J, Juan D. Copy number variants and fixed duplications among 198 rhesus macaques (Macaca mulatta). PLoS Genet 2020; 16:e1008742. [PMID: 32392208 PMCID: PMC7241854 DOI: 10.1371/journal.pgen.1008742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 05/21/2020] [Accepted: 03/27/2020] [Indexed: 01/01/2023] Open
Abstract
The rhesus macaque is an abundant species of Old World monkeys and a valuable model organism for biomedical research due to its close phylogenetic relationship to humans. Copy number variation is one of the main sources of genomic diversity within and between species and a widely recognized cause of inter-individual differences in disease risk. However, copy number differences among rhesus macaques and between the human and macaque genomes, as well as the relevance of this diversity to research involving this nonhuman primate, remain understudied. Here we present a high-resolution map of sequence copy number for the rhesus macaque genome constructed from a dataset of 198 individuals. Our results show that about one-eighth of the rhesus macaque reference genome is composed of recently duplicated regions, either copy number variable regions or fixed duplications. Comparison with human genomic copy number maps based on previously published data shows that, despite overall similarities in the genome-wide distribution of these regions, there are specific differences at the chromosome level. Some of these create differences in the copy number profile between human disease genes and their rhesus macaque orthologs. Our results highlight the importance of addressing the number of copies of target genes in the design of experiments and cautions against human-centered assumptions in research conducted with model organisms. Overall, we present a genome-wide copy number map from a large sample of rhesus macaque individuals representing an important novel contribution concerning the evolution of copy number in primate genomes.
Collapse
Affiliation(s)
- Marina Brasó-Vives
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Parc de Recerca Biomèdica de Barcelona, Barcelona, Catalonia, Spain
- Laboratoire de Biométrie et Biologie Évolutive UMR 5558, Université de Lyon, Université Lyon 1, CNRS, Villeurbanne, France
| | - Inna S. Povolotskaya
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov Russian National Research Medical University, Moscow, Russia
| | - Diego A. Hartasánchez
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Parc de Recerca Biomèdica de Barcelona, Barcelona, Catalonia, Spain
| | - Xavier Farré
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Parc de Recerca Biomèdica de Barcelona, Barcelona, Catalonia, Spain
| | - Marcos Fernandez-Callejo
- National Centre for Genomic Analysis-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Muthuswamy Raveendran
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - R. Alan Harris
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Douglas L. Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Belen Lorente-Galdos
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Arcadi Navarro
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Parc de Recerca Biomèdica de Barcelona, Barcelona, Catalonia, Spain
- National Institute for Bioinformatics (INB), Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain
| | - Tomas Marques-Bonet
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Parc de Recerca Biomèdica de Barcelona, Barcelona, Catalonia, Spain
- National Centre for Genomic Analysis-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain
- Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Catalonia, Spain
| | - Jeffrey Rogers
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - David Juan
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Parc de Recerca Biomèdica de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
33
|
Neurons, Glia, Extracellular Matrix and Neurovascular Unit: A Systems Biology Approach to the Complexity of Synaptic Plasticity in Health and Disease. Int J Mol Sci 2020; 21:ijms21041539. [PMID: 32102370 PMCID: PMC7073232 DOI: 10.3390/ijms21041539] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023] Open
Abstract
The synaptic cleft has been vastly investigated in the last decades, leading to a novel and fascinating model of the functional and structural modifications linked to synaptic transmission and brain processing. The classic neurocentric model encompassing the neuronal pre- and post-synaptic terminals partly explains the fine-tuned plastic modifications under both pathological and physiological circumstances. Recent experimental evidence has incontrovertibly added oligodendrocytes, astrocytes, and microglia as pivotal elements for synapse formation and remodeling (tripartite synapse) in both the developing and adult brain. Moreover, synaptic plasticity and its pathological counterpart (maladaptive plasticity) have shown a deep connection with other molecular elements of the extracellular matrix (ECM), once considered as a mere extracellular structural scaffold altogether with the cellular glue (i.e., glia). The ECM adds another level of complexity to the modern model of the synapse, particularly, for the long-term plasticity and circuit maintenance. This model, called tetrapartite synapse, can be further implemented by including the neurovascular unit (NVU) and the immune system. Although they were considered so far as tightly separated from the central nervous system (CNS) plasticity, at least in physiological conditions, recent evidence endorsed these elements as structural and paramount actors in synaptic plasticity. This scenario is, as far as speculations and evidence have shown, a consistent model for both adaptive and maladaptive plasticity. However, a comprehensive understanding of brain processes and circuitry complexity is still lacking. Here we propose that a better interpretation of the CNS complexity can be granted by a systems biology approach through the construction of predictive molecular models that enable to enlighten the regulatory logic of the complex molecular networks underlying brain function in health and disease, thus opening the way to more effective treatments.
Collapse
|
34
|
Ibañez S, Luebke JI, Chang W, Draguljić D, Weaver CM. Network Models Predict That Pyramidal Neuron Hyperexcitability and Synapse Loss in the dlPFC Lead to Age-Related Spatial Working Memory Impairment in Rhesus Monkeys. Front Comput Neurosci 2020; 13:89. [PMID: 32009920 PMCID: PMC6979278 DOI: 10.3389/fncom.2019.00089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/18/2019] [Indexed: 01/04/2023] Open
Abstract
Behavioral studies have shown spatial working memory impairment with aging in several animal species, including humans. Persistent activity of layer 3 pyramidal dorsolateral prefrontal cortex (dlPFC) neurons during delay periods of working memory tasks is important for encoding memory of the stimulus. In vitro studies have shown that these neurons undergo significant age-related structural and functional changes, but the extent to which these changes affect neural mechanisms underlying spatial working memory is not understood fully. Here, we confirm previous studies showing impairment on the Delayed Recognition Span Task in the spatial condition (DRSTsp), and increased in vitro action potential firing rates (hyperexcitability), across the adult life span of the rhesus monkey. We use a bump attractor model to predict how empirically observed changes in the aging dlPFC affect performance on the Delayed Response Task (DRT), and introduce a model of memory retention in the DRSTsp. Persistent activity-and, in turn, cognitive performance-in both models was affected much more by hyperexcitability of pyramidal neurons than by a loss of synapses. Our DRT simulations predict that additional changes to the network, such as increased firing of inhibitory interneurons, are needed to account for lower firing rates during the DRT with aging reported in vivo. Synaptic facilitation was an essential feature of the DRSTsp model, but it did not compensate fully for the effects of the other age-related changes on DRT performance. Modeling pyramidal neuron hyperexcitability and synapse loss simultaneously led to a partial recovery of function in both tasks, with the simulated level of DRSTsp impairment similar to that observed in aging monkeys. This modeling work integrates empirical data across multiple scales, from synapse counts to cognitive testing, to further our understanding of aging in non-human primates.
Collapse
Affiliation(s)
- Sara Ibañez
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Jennifer I. Luebke
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Wayne Chang
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Danel Draguljić
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
| | - Christina M. Weaver
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
| |
Collapse
|
35
|
Cui D, Zhang L, Zheng F, Wang H, Meng Q, Lu W, Liu Z, Yin T, Qiu J. Volumetric reduction of cerebellar lobules associated with memory decline across the adult lifespan. Quant Imaging Med Surg 2020; 10:148-159. [PMID: 31956538 DOI: 10.21037/qims.2019.10.19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background The human cerebellum plays an essential role in motor control, is involved in cognitive function and helps to regulate emotional responses. However, little is known about the relationship between cerebellar lobules and age-related memory decline. We aimed to investigate volume alterations in cerebellar lobules at different ages and assess their correlations with reduced memory recall abilities. Methods A sample of 275 individuals were divided into the following four groups: 20-35 years (young), 36-50 years (early-middle age), 51-65 years (late-middle age), and 66-89 years (old). Volumes of the cerebellar lobules were obtained using volBrain software. Analysis of covariance and post hoc analysis were used to analyze group differences in cerebellar lobular volumes, and multiple comparisons were performed using the Bonferroni method. Spearman correlation was used to investigate the relationship between lobular volumes and memory recall scores. Results In this study, we found that older adults had smaller cerebellar volumes than the other subjects. Volumetric decreases in size were noted in bilateral lobule VI and lobule crus I. Moreover, the volumes of bilateral lobule crus I, lobule VI, and right lobule IV were significantly associated with memory recall scores. Conclusions In the present study, we found that some lobules of the cerebellum appear more predisposed to age-related changes than other lobules. These findings provide further evidence that specific regions of the cerebellum could be used to assess the risk of memory decline across the adult lifespan.
Collapse
Affiliation(s)
- Dong Cui
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.,College of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Li Zhang
- College of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.,Medical Engineering and Technology Research Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.,Imaging-X Joint Laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Fenglian Zheng
- College of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.,Medical Engineering and Technology Research Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.,Imaging-X Joint Laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Huiqin Wang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Qingjian Meng
- College of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Wen Lu
- College of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Zhipeng Liu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Tao Yin
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Jianfeng Qiu
- College of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.,Medical Engineering and Technology Research Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.,Imaging-X Joint Laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| |
Collapse
|
36
|
Chapman TW, Hill RA. Myelin plasticity in adulthood and aging. Neurosci Lett 2019; 715:134645. [PMID: 31765728 DOI: 10.1016/j.neulet.2019.134645] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/06/2019] [Accepted: 11/21/2019] [Indexed: 12/31/2022]
Abstract
The central nervous system maintains the potential for molecular and cellular plasticity throughout life. This flexibility underlies fundamental features of neural circuitry including the brain's ability to sense, store, and properly adapt to everchanging external stimuli on time scales from seconds to years. Evidence for most forms of plasticity are centered around changes in neuronal structure and synaptic strength, however recent data suggests that myelinating oligodendrocytes exhibit certain forms of plasticity in the adult. This plasticity ranges from the generation of entirely new myelinating cells to more subtle changes in myelin sheath length, thickness, and distribution along axons. The extent to which these changes dynamically modify axonal function and neural circuitry and whether they are directly related to mechanisms of learning and memory remains an open question. Here we describe different forms of myelin plasticity, highlight some recent evidence for changes in myelination throughout life, and discuss how defects in these forms of plasticity could be associated with cognitive decline in aging.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
37
|
3D Electron Microscopy Study of Synaptic Organization of the Normal Human Transentorhinal Cortex and Its Possible Alterations in Alzheimer's Disease. eNeuro 2019; 6:ENEURO.0140-19.2019. [PMID: 31217195 PMCID: PMC6620390 DOI: 10.1523/eneuro.0140-19.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/23/2019] [Accepted: 05/27/2019] [Indexed: 01/10/2023] Open
Abstract
The transentorhinal cortex (TEC) is an obliquely oriented cortex located in the medial temporal lobe and, together with the entorhinal cortex, is one of the first affected areas in Alzheimer’s disease (AD). One of the most widely accepted hypotheses is that synaptopathy (synaptic alterations and loss) represents the major structural correlate of the cognitive decline observed in AD. However, very few electron microscope (EM) studies are available; the most common method to estimate synaptic density indirectly is by counting, at the light microscopic level, immunoreactive puncta using synaptic markers. To investigate synaptic morphology and possible alterations related to AD, a detailed three-dimensional (3D) ultrastructural analysis using focused ion beam/scanning EM (FIB/SEM) was performed in the neuropil of Layer II of the TEC in human brain samples from non-demented subjects and AD patients. Evaluation of the proportion and shape of asymmetric synapses (AS) and symmetric synapses (SS) targeting spines or dendritic shafts was performed using 3D reconstructions of every synapse. The 3D analysis of 4722 synapses revealed that the preferable targets were spine heads for AS and dendritic shafts for SS, both in control and AD cases. However, in AD patients, we observed a reduction in the percentage of synapses targeting spine heads. Regarding the shape of synapses, in both control cases and AD samples, the vast majority of synapses had a macular shape, followed by perforated or horseshoe-shaped synapses, with fragmented synapses being the least frequent type. Moreover, comparisons showed an increased number of fragmented AS in AD patients.
Collapse
|
38
|
Hill RA, Grutzendler J. Uncovering the biology of myelin with optical imaging of the live brain. Glia 2019; 67:2008-2019. [PMID: 31033062 DOI: 10.1002/glia.23635] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/26/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Myelin has traditionally been considered a static structure that is produced and assembled during early developmental stages. While this characterization is accurate in some contexts, recent studies have revealed that oligodendrocyte generation and patterns of myelination are dynamic and potentially modifiable throughout life. Unique structural and biochemical properties of the myelin sheath provide opportunities for the development and implementation of multimodal label-free and fluorescence optical imaging approaches. When combined with genetically encoded fluorescent tags targeted to distinct cells and subcellular structures, these techniques offer a powerful methodological toolbox for uncovering mechanisms of myelin generation and plasticity in the live brain. Here, we discuss recent advances in these approaches that have allowed the discovery of several forms of myelin plasticity in developing and adult nervous systems. Using these techniques, long-standing questions related to myelin generation, remodeling, and degeneration can now be addressed.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire
| | - Jaime Grutzendler
- Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
39
|
Kounakis K, Tavernarakis N. The Cytoskeleton as a Modulator of Aging and Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:227-245. [PMID: 31493230 DOI: 10.1007/978-3-030-25650-0_12] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cytoskeleton consists of filamentous protein polymers that form organized structures, contributing to a multitude of cell life aspects. It includes three types of polymers: the actin microfilaments, the microtubules and the intermediate filaments. Decades of research have implicated the cytoskeleton in processes that regulate cellular and organismal aging, as well as neurodegeneration associated with injury or neurodegenerative disease, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, or Charcot Marie Tooth disease. Here, we provide a brief overview of cytoskeletal structure and function, and discuss experimental evidence linking cytoskeletal function and dynamics with aging and neurodegeneration.
Collapse
Affiliation(s)
- Konstantinos Kounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece.,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece. .,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece.
| |
Collapse
|
40
|
Seixas AI, Azevedo MM, Paes de Faria J, Fernandes D, Mendes Pinto I, Relvas JB. Evolvability of the actin cytoskeleton in oligodendrocytes during central nervous system development and aging. Cell Mol Life Sci 2019; 76:1-11. [PMID: 30302529 PMCID: PMC11105620 DOI: 10.1007/s00018-018-2915-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/24/2018] [Accepted: 09/04/2018] [Indexed: 01/23/2023]
Abstract
The organization of actin filaments into a wide range of subcellular structures is a defining feature of cell shape and dynamics, important for tissue development and homeostasis. Nervous system function requires morphological and functional plasticity of neurons and glial cells, which is largely determined by the dynamic reorganization of the actin cytoskeleton in response to intrinsic and extracellular signals. Oligodendrocytes are specialized glia that extend multiple actin-based protrusions to form the multilayered myelin membrane that spirally wraps around axons, increasing conduction speed and promoting long-term axonal integrity. Myelination is a remarkable biological paradigm in development, and maintenance of myelin is essential for a healthy adult nervous system. In this review, we discuss how structure and dynamics of the actin cytoskeleton is a defining feature of myelinating oligodendrocytes' biology and function. We also review "old and new" concepts to reflect on the potential role of the cytoskeleton in balancing life and death of myelin membranes and oligodendrocytes in the aging central nervous system.
Collapse
Affiliation(s)
- Ana Isabel Seixas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal.
| | - Maria Manuela Azevedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Joana Paes de Faria
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Diogo Fernandes
- Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- International Iberian Nanotechnology Laboratory - INL, Braga, Portugal
| | - Inês Mendes Pinto
- International Iberian Nanotechnology Laboratory - INL, Braga, Portugal
| | - João Bettencourt Relvas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
- The Discoveries Centre for Regeneration and Precision Medicine, Porto Campus, Porto, Portugal
| |
Collapse
|
41
|
Guerreri M, Palombo M, Caporale A, Fasano F, Macaluso E, Bozzali M, Capuani S. Age-related microstructural and physiological changes in normal brain measured by MRI γ-metrics derived from anomalous diffusion signal representation. Neuroimage 2018; 188:654-667. [PMID: 30583064 DOI: 10.1016/j.neuroimage.2018.12.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 12/29/2022] Open
Abstract
Nowadays, increasing longevity associated with declining cerebral nervous system functions, suggests the need for continued development of new imaging contrast mechanisms to support the differential diagnosis of age-related decline. In our previous papers, we developed a new imaging contrast metrics derived from anomalous diffusion signal representation and obtained from diffusion-weighted (DW) data collected by varying diffusion gradient strengths. Recently, we highlighted that the new metrics, named γ-metrics, depended on the local inhomogeneity due to differences in magnetic susceptibility between tissues and diffusion compartments in young healthy subjects, thus providing information about myelin orientation and iron content within cerebral regions. The major structural modifications occurring in brain aging are myelinated fibers damage in nerve fibers and iron accumulation in gray matter nuclei. Therefore, we investigated the potential of γ-metrics in relation to other conventional diffusion metrics such as DTI, DKI and NODDI in detecting age-related structural changes in white matter (WM) and subcortical gray matter (scGM). DW-images were acquired in 32 healthy subjects, adults and elderly (age range 20-77 years) using 3.0T and 12 b-values up to 5000 s/mm2. Association between diffusion metrics and subjects' age was assessed using linear regression. A decline in mean γ (Mγ) in the scGM and a complementary increase in radial γ (γ⊥) in frontal WM, genu of corpus callosum and anterior corona radiata with advancing age were found. We suggested that the increase in γ⊥ might reflect declined myelin density, and Mγ decrease might mirror iron accumulation. An increase in D// and a decrease in the orientation dispersion index (ODI) were associated with axonal loss in the pyramidal tracts, while their inverted trends within the thalamus were thought to be linked to reduced architectural complexity of nerve fibers. γ-metrics together with conventional diffusion-metrics can more comprehensively characterize the complex mechanisms underlining age-related changes than conventional diffusion techniques alone.
Collapse
Affiliation(s)
- Michele Guerreri
- SAIMLAL Department, Sapienza, Piazzale Aldo Moro, 5, 00185, Roma, RM, Italy; Institute for Complex Systems, CNR, Rome, Italy.
| | - Marco Palombo
- Institute for Complex Systems, CNR, Rome, Italy; Department of Computer Science & Centre for Medical Image Computing, University College London, London, United Kingdom
| | - Alessandra Caporale
- Institute for Complex Systems, CNR, Rome, Italy; Laboratory for Structural, Physiologic and Functional Imaging, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Marco Bozzali
- Neuroimaging Laboratory, Santa Lucia Foundation, Rome, Italy
| | - Silvia Capuani
- Institute for Complex Systems, CNR, Rome, Italy; Neuroimaging Laboratory, Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
42
|
Benitez A, Jensen JH, Falangola MF, Nietert PJ, Helpern JA. Modeling white matter tract integrity in aging with diffusional kurtosis imaging. Neurobiol Aging 2018; 70:265-275. [PMID: 30055412 PMCID: PMC6195210 DOI: 10.1016/j.neurobiolaging.2018.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/03/2018] [Accepted: 07/10/2018] [Indexed: 01/25/2023]
Abstract
Myelin breakdown and neural fiber loss occur in aging. This study used white matter tract integrity metrics derived from biophysical modeling using Diffusional Kurtosis Imaging to assess loss of myelin (i.e., extraaxonal diffusivity, radial direction, De,⊥) and axonal density (i.e., axonal water fraction) in cognitively unimpaired older adults. Tract-based spatial statistics and region of interest analyses sought to identify ontogenic differences and age-related changes in white matter tracts using cross-sectional and longitudinal data analyzed with general linear and mixed-effects models. In addition to pure diffusion parameters (i.e., fractional anisotropy, mean diffusivity, mean kurtosis), we found that white matter tract integrity metrics significantly differentiated early- from late-myelinating tracts, correlated with age in spatially distinct regions, and identified primarily extraaxonal changes over time. Percent metric changes were |0.3-0.9|% and |0.0-1.9|% per year using cross-sectional data and longitudinal data, respectively. There was accelerated decline in some late- versus early-myelinating tracts in older age. These results demonstrate that these metrics may inform further study of the transition from age-related changes to neurodegenerative decline.
Collapse
Affiliation(s)
- Andreana Benitez
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA; Department of Neurology, Medical University of South Carolina, Charleston, SC, USA; Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA.
| | - Jens H Jensen
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA; Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Maria Fatima Falangola
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA; Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Paul J Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Joseph A Helpern
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA; Department of Neurology, Medical University of South Carolina, Charleston, SC, USA; Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
43
|
Koo BB, Calderazzo S, Bowley BGE, Kolli A, Moss MB, Rosene DL, Moore TL. Long-term effects of curcumin in the non-human primate brain. Brain Res Bull 2018; 142:88-95. [PMID: 29981358 DOI: 10.1016/j.brainresbull.2018.06.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/11/2018] [Accepted: 06/22/2018] [Indexed: 01/08/2023]
Abstract
Curcumin has recently been shown to be a potential treatment for slowing or ameloriating cognitive decline during aging in our nonhuman primate model of normal aging. In these same monkeys, we studied for the first time the neurological impacts of long-term curcumin treatments using longitudinal magnetic resonance imaging (MRI). Sixteen rhesus monkeys received curcumin or a vehicle control for 14-18 months. We applied a combination of structural and diffusion MRI to determine whether the curcumin resulted in structural or functional changes in focal regions of the brain. The longitudinal imaging revealed decreased microscale diffusivity (mD) measurements mainly in the hippocampus and basal forebrain structures of curcumin treated animals. Changes in generalized fractional anisotropy (GFA) and grey matter density (GMd) measurements indicated an increased grey matter density in cortical ROIs with improved white matter integrity in limbic, cerebellar, and brain stem regions. These findings suggest that noticeable changes in the neuronal environment could be induced from long-term curcumin treatments. Results may provide a neurological basis on the recent findings demonstrating improved spatial working memory and motor function in nonhuman primates.
Collapse
Affiliation(s)
- Bang-Bon Koo
- Department of Anatomy and Neurobiology, School of Medicine, Boston University, Boston, MA, USA.
| | - Samantha Calderazzo
- Department of Anatomy and Neurobiology, School of Medicine, Boston University, Boston, MA, USA
| | - Bethany G E Bowley
- Department of Anatomy and Neurobiology, School of Medicine, Boston University, Boston, MA, USA
| | - Alekha Kolli
- BA/MD Program, Boston University, Boston, MA, USA
| | - Mark B Moss
- Department of Anatomy and Neurobiology, School of Medicine, Boston University, Boston, MA, USA; BA/MD Program, Boston University, Boston, MA, USA; Department of Neurology, School of Medicine, Boston University, Boston, MA, USA
| | - Douglas L Rosene
- Department of Anatomy and Neurobiology, School of Medicine, Boston University, Boston, MA, USA; Department of Neurology, School of Medicine, Boston University, Boston, MA, USA
| | - Tara L Moore
- Department of Anatomy and Neurobiology, School of Medicine, Boston University, Boston, MA, USA; BA/MD Program, Boston University, Boston, MA, USA
| |
Collapse
|
44
|
Boisgontier MP, Cheval B, van Ruitenbeek P, Cuypers K, Leunissen I, Sunaert S, Meesen R, Zivari Adab H, Renaud O, Swinnen SP. Cerebellar gray matter explains bimanual coordination performance in children and older adults. Neurobiol Aging 2018; 65:109-120. [DOI: 10.1016/j.neurobiolaging.2018.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 01/12/2018] [Accepted: 01/21/2018] [Indexed: 02/02/2023]
|
45
|
Lifelong cortical myelin plasticity and age-related degeneration in the live mammalian brain. Nat Neurosci 2018; 21:683-695. [PMID: 29556031 PMCID: PMC5920745 DOI: 10.1038/s41593-018-0120-6] [Citation(s) in RCA: 285] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/05/2018] [Indexed: 01/30/2023]
Abstract
Axonal myelin increases neural processing speed and efficiency. It is unknown whether patterns of myelin distribution are fixed or whether myelinating oligodendrocytes are continually generated in adulthood and maintain the capacity for structural remodeling. Using high-resolution, intravital label-free and fluorescence optical imaging in mouse cortex, we demonstrate lifelong oligodendrocyte generation occurring in parallel with structural plasticity of individual myelin internodes. Continuous internode formation occurred on both partially myelinated and unmyelinated axons, and the total myelin coverage along individual axons progressed up to two years of age. After peak myelination, gradual oligodendrocyte death and myelin degeneration in aging were associated with pronounced internode loss and myelin debris accumulation within microglia. Thus, cortical myelin remodeling is protracted throughout life, potentially playing critical roles in neuronal network homeostasis. The gradual loss of internodes and myelin degeneration in aging could contribute significantly to brain pathogenesis.
Collapse
|
46
|
Hughes EG, Orthmann-Murphy JL, Langseth AJ, Bergles DE. Myelin remodeling through experience-dependent oligodendrogenesis in the adult somatosensory cortex. Nat Neurosci 2018; 21:696-706. [PMID: 29556025 PMCID: PMC5920726 DOI: 10.1038/s41593-018-0121-5] [Citation(s) in RCA: 329] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/09/2018] [Indexed: 12/30/2022]
Abstract
The generation of oligodendrocytes in the adult CNS provides a means to adapt the properties of circuits to changes in life experience. However, little is known about the dynamics of oligodendrocytes and the extent of myelin remodeling in the mature brain. Using longitudinal in vivo two photon imaging of oligodendrocytes and their progenitors in the mouse cerebral cortex, we show that myelination is an inefficient and extended process, with half of the final complement of oligodendrocytes generated after four months of age. Oligodendrocytes that successfully integrated formed novel sheaths on unmyelinated and sparsely myelinated axons, and were extremely stable, gradually changing the pattern of myelination. Sensory enrichment robustly increased oligodendrocyte integration, but did not change the length of existing sheaths. This experience-dependent enhancement of myelination in the mature cortex may accelerate information transfer in these circuits and strengthen the ability of axons to sustain activity by providing additional metabolic support.
Collapse
Affiliation(s)
- Ethan G Hughes
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Jennifer L Orthmann-Murphy
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abraham J Langseth
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
47
|
Domínguez-Álvaro M, Montero-Crespo M, Blazquez-Llorca L, Insausti R, DeFelipe J, Alonso-Nanclares L. Three-dimensional analysis of synapses in the transentorhinal cortex of Alzheimer's disease patients. Acta Neuropathol Commun 2018; 6:20. [PMID: 29499755 PMCID: PMC5834884 DOI: 10.1186/s40478-018-0520-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/17/2018] [Indexed: 12/11/2022] Open
Abstract
Synaptic dysfunction or loss in early stages of Alzheimer’s disease (AD) is thought to be a major structural correlate of cognitive dysfunction. Early loss of episodic memory, which occurs at the early stage of AD, is closely associated with the progressive degeneration of medial temporal lobe (MTL) structures of which the transentorhinal cortex (TEC) is the first affected area. However, no ultrastructural studies have been performed in this region in human brain samples from AD patients. In the present study, we have performed a detailed three-dimensional (3D) ultrastructural analysis using focused ion beam/scanning electron microscopy (FIB/SEM) to investigate possible synaptic alterations in the TEC of patients with AD. Surprisingly, the analysis of the density, morphological features and spatial distribution of synapses in the neuropil showed no significant differences between AD and control samples. However, light microscopy studies showed that cortical thickness of the TEC was severely reduced in AD samples, but there were no changes in the volume occupied by neuronal and glial cell bodies, blood vessels, and neuropil. Thus, the present results indicate that there is a dramatic loss of absolute number of synapses, while the morphology of synaptic junctions and synaptic spatial distribution are maintained. How these changes affect cognitive impairment in AD remains to be elucidated.
Collapse
|
48
|
Robinson AA, Abraham CR, Rosene DL. Candidate molecular pathways of white matter vulnerability in the brain of normal aging rhesus monkeys. GeroScience 2018; 40:31-47. [PMID: 29357021 PMCID: PMC5832663 DOI: 10.1007/s11357-018-0006-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022] Open
Abstract
Mammalian aging is associated with decline in cognitive functions. Studies searching for a cause of cognitive aging initially focused on neuronal loss but quantitative investigations of rat, monkey, and human brain using stereology demonstrated that in normal aging, unlike in neurodegenerative disease, neurons are not lost. Instead, electron microscopic and MRI studies in normal aging monkeys revealed age-related damage to myelin sheaths, loss of axons, and reduction in white matter volume which correlates with cognitive impairments. However, little is known about the cause of myelin defects or associated axon loss. The present study investigates the effect of age on signaling pathways between oligodendroglia and neurons using a custom PCR array to assess the expression of 87 genes of interest in cortical gray matter and white matter from the inferior parietal lobe (IPL) of normal rhesus monkeys ranging in age from 4.2 to 30.4 years old. From this array data, five target genes of interest were selected for further analysis to confirm gene expression and measure protein expression. The most interesting target gene identified is brain-derived neurotrophic factor (BDNF), which was the only gene that was altered at both mRNA and protein levels. In gray matter, BDNF mRNA was decreased. While the level of the mature form of the protein was unchanged, there was a specific decrease in the precursor form of BDNF. These alterations in the BDNF in gray matter could contribute to the vulnerability and loss of the axons with age.
Collapse
Affiliation(s)
- Amy A. Robinson
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118 USA
| | - Carmela R. Abraham
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118 USA
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118 USA
| | - Douglas L. Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118 USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322 USA
| |
Collapse
|
49
|
A computational study of the impact of inhomogeneous internodal lengths on conduction velocity in myelinated neurons. PLoS One 2018; 13:e0191106. [PMID: 29329312 PMCID: PMC5766232 DOI: 10.1371/journal.pone.0191106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 12/28/2017] [Indexed: 11/30/2022] Open
Abstract
Age-related decreases in the conduction velocity (CV) of action potentials along myelinated axons have been linked to morphological changes in the myelin sheath. In particular, evidence suggests the presence of segmental demyelination and remyelination of axons. In remyelinated segments, the distance between adjacent nodes of Ranvier is typically shorter, and myelin sheaths are thinner. Both experimental and computational evidence indicates that shortened internodes slows CV. In this computational study, we determine the impact of progressive segmental demyelination and remyelination, modeled by shorter internodes with thinner myelin sheaths interspersed with normal ones, upon the CV. We find that CV progressively decreases as the number of remyelinated segments increases, but this decrease is greater than one would expect from an estimate of the CV based merely upon the number of short and long internodes. We trace the additional suppression of the CV to transitions between long and short internodes. Our study presents an important consideration for the precise modeling of neural circuits with remyelinated neurons.
Collapse
|
50
|
Toufexis D, King SB, Michopoulos V. Socially Housed Female Macaques: a Translational Model for the Interaction of Chronic Stress and Estrogen in Aging. Curr Psychiatry Rep 2017; 19:78. [PMID: 28905316 DOI: 10.1007/s11920-017-0833-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE OF REVIEW Estrogen's role in cognitive aging remains unclear. Despite evidence implicating stress in pathological aging, the interaction of stress with estrogen on cognition in older women has received little attention, and few animal models exist with which to examine this interaction. RECENT FINDINGS We present evidence that aging socially subordinate female macaques that experience chronic psychosocial stress constitute a suitable model to investigate this. First, we review studies showing that estrogen modulates cognition in animal models, as well as studies demonstrating that estrogen's action on certain types of cognition is impaired by stress. Next, we discuss data showing that middle-aged socially subordinate female macaques exhibit distinct stress-induced phenotypes, and review our investigations indicating that estrogen modulates behavior and physiology differently in subordinate female monkeys. We conclude that socially housed female macaques represent a translational animal model for investigating the interplay of chronic stress and estrogen on cognitive aging in women.
Collapse
Affiliation(s)
- Donna Toufexis
- Department of Psychological Science, The University of Vermont, Burlington, VT, USA.,Division of Development and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - S Bradley King
- Department of Psychological Science, The University of Vermont, Burlington, VT, USA
| | - Vasiliki Michopoulos
- Division of Development and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA. .,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|