1
|
Gupta Y, de la Cruz F, Rieger K, di Giuliano M, Gaser C, Cole J, Breithaupt L, Holsen LM, Eddy KT, Thomas JJ, Cetin-Karayumak S, Kubicki M, Lawson EA, Miller KK, Misra M, Schumann A, Bär KJ. Does restrictive anorexia nervosa impact brain aging? A machine learning approach to estimate age based on brain structure. Comput Biol Med 2025; 194:110484. [PMID: 40516452 DOI: 10.1016/j.compbiomed.2025.110484] [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: 01/08/2025] [Revised: 05/01/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025]
Abstract
Anorexia nervosa (AN), a severe eating disorder marked by extreme weight loss and malnutrition, leads to significant alterations in brain structure. This study used machine learning (ML) to estimate brain age from structural MRI scans and investigated brain-predicted age difference (brain-PAD) as a potential biomarker in AN. Structural MRI scans were collected from female participants aged 10-40 years across two institutions (Boston, USA, and Jena, Germany), including acute AN (acAN; n=113), weight-restored AN (wrAN; n=35), and age-matched healthy controls (HC; n=90). The ML model was trained on 3487 healthy female participants (ages 5-45 years) from ten datasets, using 377 neuroanatomical features extracted from T1-weighted MRI scans. The model achieved strong performance with a mean absolute error (MAE) of 1.93 years and a correlation of r = 0.88 in HCs. In acAN patients, brain age was overestimated by an average of +2.25 years, suggesting advanced brain aging. In contrast, wrAN participants showed significantly lower brain-PAD than acAN (+0.26 years, p=0.0026) and did not differ from HC (p=0.98), suggesting normalization of brain age estimates following weight restoration. A significant group-by-age interaction effect on predicted brain age (p<0.001) indicated that brain age deviations were most pronounced in younger acAN participants. Brain-PAD in acAN was significantly negatively associated with BMI (r = -0.291, pfdr = 0.005), but not in wrAN or HC groups. Importantly, no significant associations were found between brain-PAD and clinical symptom severity. These findings suggest that acute AN is linked to advanced brain aging during the acute stage, and that may partially normalize following weight recovery.
Collapse
Affiliation(s)
- Yubraj Gupta
- Department of Psychosomatic Medicine and Psychotherapy, Jena University Hospital, Jena, Germany.
| | - Feliberto de la Cruz
- Department of Psychosomatic Medicine and Psychotherapy, Jena University Hospital, Jena, Germany; Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Katrin Rieger
- Department of Psychosomatic Medicine and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Monica di Giuliano
- Department of Psychosomatic Medicine and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Christian Gaser
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Department of Neurology, Jena University Hospital, Jena, Germany; German Center for Mental Health (DZPG), Germany
| | - James Cole
- Centre for Medical Imaging Computer, University College London, London, UK; Dementia Research Centre, University College London, London, UK
| | - Lauren Breithaupt
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, USA; Department of Psychiatry, Harvard Medical School, Boston, USA; Mass General Brigham Multidisciplinary Eating Disorder Research Collaborative, Massachusetts General Hospital, Boston, USA
| | - Laura M Holsen
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Mass General Brigham Multidisciplinary Eating Disorder Research Collaborative, Massachusetts General Hospital, Boston, USA; Harvard Medical School, Boston, USA
| | - Kamryn T Eddy
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, USA; Department of Psychiatry, Harvard Medical School, Boston, USA; Mass General Brigham Multidisciplinary Eating Disorder Research Collaborative, Massachusetts General Hospital, Boston, USA
| | - Jennifer J Thomas
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, USA; Department of Psychiatry, Harvard Medical School, Boston, USA; Mass General Brigham Multidisciplinary Eating Disorder Research Collaborative, Massachusetts General Hospital, Boston, USA
| | - Suheyla Cetin-Karayumak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Department of Psychiatry, Harvard Medical School, Boston, USA
| | - Marek Kubicki
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Department of Psychiatry, Harvard Medical School, Boston, USA; Athinoula A. Martinos Center, Massachusetts General Hospital, Boston, USA
| | - Elizabeth A Lawson
- Mass General Brigham Multidisciplinary Eating Disorder Research Collaborative, Massachusetts General Hospital, Boston, USA; Harvard Medical School, Boston, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, USA
| | - Karen K Miller
- Harvard Medical School, Boston, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, USA
| | - Madhusmita Misra
- Mass General Brigham Multidisciplinary Eating Disorder Research Collaborative, Massachusetts General Hospital, Boston, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, USA; Division of Pediatric Endocrinology, University of Virginia, Charlottesville, USA; Department of Pediatrics, University of Virginia, Charlottesville, USA
| | - Andy Schumann
- Department of Psychosomatic Medicine and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Karl-Jürgen Bär
- Department of Psychosomatic Medicine and Psychotherapy, Jena University Hospital, Jena, Germany
| |
Collapse
|
2
|
Pai V, Singh BN, Singh AK. Transformative advances in modeling brain aging and longevity: Success, challenges and future directions. Ageing Res Rev 2025; 108:102753. [PMID: 40222396 DOI: 10.1016/j.arr.2025.102753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/30/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Research on brain aging is crucial for understanding age-related neurodegenerative disorders and developing several therapeutic interventions. Numerous models ranging from two-dimensional (2D) cell-based, invertebrate, vertebrate, and sophisticated three-dimensional (3D) models have been used to understand the process of brain aging. Invertebrate models are ideal for researching conserved aging processes because of their simplicity, short lifespans, and genetic tractability. Moreover, vertebrate models, including zebrafish and rodents, exhibit more complex nervous systems and behaviors, enabling the exploration of age-related neurodegeneration and cognitive decline. 2D cell culture models derived from primary cells or immortalized cell lines are widely used for mechanistic studies at the cellular level but lack the physiological complexity of brain tissue. Recent advancements have shifted focus to 3D models, which better recapitulate the brain's microenvironment. Organoids derived from induced pluripotent stem cells mimic human brain architecture and enable the study of cell-cell interactions and aging in a human-specific context. Brain-on-a-chip systems integrate microfluidics and 3D cultures to model blood-brain barrier dynamics and neuronal networks. Additionally, scaffold-based 3D cultures and spheroids provide intermediate complexity, allowing researchers to study extracellular matrix interactions and age-related changes in neuronal function. These 3D models bridge the gap between traditional 2D cultures and animal-based in vivo studies, offering unprecedented insights into brain aging mechanisms. By combining these diverse models, researchers can unravel the multifaceted processes of brain aging and accelerate the development of targeted therapies for age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Varsha Pai
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Bhisham Narayan Singh
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
3
|
Zhu Z, He C, Yao H, Liao G, Gan Y, Deng L. Assessment of the combined utility of S100B, GFAP, and IL-6 in predicting long-term cognitive impairment in survivors of sepsis. Neurol Res 2025:1-10. [PMID: 40424558 DOI: 10.1080/01616412.2025.2511084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 05/15/2025] [Indexed: 05/29/2025]
Abstract
OBJECTIVE The main purpose of this study was to comprehensively investigate the combined utility of S100B, GFAP, and IL-6 as predictors of long-term cognitive impairment in sepsis survivors. Specifically, we aimed to determine whether these biomarkers, either individually or in combination, could effectively predict the occurrence of long-term cognitive impairment in this patient population, and to explore their potential as valuable clinical tools for early detection and intervention. METHODS This retrospective study enrolled 114 sepsis patients. Patients were divided into non-cognitive impairment and cognitive impairment groups. Serum biomarker levels were compared, and correlations between biomarkers and cognitive impairment were explored. ROC analysis evaluated the predictive value of S100B, GFAP, and IL-6 levels, and the combined diagnosis of the three biomarkers was studied. RESULTS The non-cognitive impairment group had a younger age, higher education level, employment rate, married rate, and presence of family members (p < 0.05). Correlation analysis showed positive correlations between cognitive impairment and CRP, IL-10, S100B, GFAP, and IL-6 (p < 0.05). AUC values of S100B, GFAP, and IL-6 were 0.792, 0.752, and 0.732, respectively, indicating significant predictive value for cognitive impairment. Combined prediction using the three biomarkers had an AUC value of 0.887, with a specificity of 88% and sensitivity of 89%. CONCLUSION Long-term cognitive impairment in sepsis survivors is influenced by various cytokines. Significant differences were found in biomarker levels between non-cognitive impairment and cognitive impairment groups. The combined utility of S100B, GFAP, and IL-6 showed significant predictive value for cognitive impairment in survivors of sepsis.
Collapse
Affiliation(s)
- Zigui Zhu
- The Nanhua Affiliated Hospital, Department of Intensive Care, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chengjian He
- The Nanhua Affiliated Hospital, Department of Intensive Care, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hongyi Yao
- The Nanhua Affiliated Hospital, Department of Intensive Care, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guqing Liao
- The Nanhua Affiliated Hospital, Department of Intensive Care, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yanqing Gan
- The Nanhua Affiliated Hospital, Department of Intensive Care, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lipu Deng
- The Nanhua Affiliated Hospital, Emergency Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
4
|
Reheman M, Buch S, He N, Huang P, Yu Q, Wang X, Liu Y, Zhang Y, Jin Z, Li Y, Liu P, Chen S, Haacke EM, Yan F. White matter hyperintensity tissue property spatial variations as a function of cognitive status in Parkinson's disease. Neuroimage 2025; 312:121236. [PMID: 40280215 DOI: 10.1016/j.neuroimage.2025.121236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 04/13/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND AND PURPOSE The pathological relationship between white matter hyperintensities (WMH) and cognitive impairment in Parkinson's disease (PD) remains unclear due to their variable locations, heterogeneity, and limited assessment of underlying tissue properties. This study integrates T2-FLAIR and quantitative MRI (qMRI) to investigate burden, spatial distribution, and extent of tissue alterations in WMH, aiming to elucidate their role in cognitive decline among PD patients. METHODS A total of 122 age- and sex-matched PD patients and 65 healthy controls (HC) were recruited, with PD patients grouped by Montreal Cognitive Assessment (MoCA) score including normal, mild cognitive impairment (MCI) or PD with dementia (PDD). WMH burden was compared across groups and cognitive status. Water content, T1, and T2* measures were derived from qMRI data and tissue property heatmaps and periventricular distance profiles were constructed for all groups to visualize location-dependent tissue alterations of WMH relative to the lateral ventricles. In addition, voxel-wise analysis was performed to examine the correlation between WMH lesion tissue properties and MoCA scores. RESULTS WMH volume was significantly higher in PDD compared to other groups (p < 0.05) and negatively correlated with MoCA scores (r = -0.352, p < 0.001). WMH appeared predominantly around the lateral ventricles, with anterior horn involvement common to all groups and posterior horn involvement specific to PDD. qMRI measures were significantly elevated in WMH compared to normal appearing white matter (NAWM) (p < 0.001), with heatmaps showing a negative gradient of tissue property changes from the lateral ventricles to the NAWM. Voxel-wise analysis revealed a significant negative correlation between the qMRI tissue properties of periventricular WMH and MoCA scores, with the strongest association observed in the periventricular WM situated just beyond the boundary of the lateral ventricles. CONCLUSION Over and above volume differences, the spatial distribution and tissue property variations of WMH were closely linked to cognitive impairment in PD patients, with distinct patterns across different cognitive stages.
Collapse
Affiliation(s)
- Mariyemuguli Reheman
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Sagar Buch
- Department of Neurology, Wayne State University, 3990 John R, Detroit, MI, USA
| | - Naying He
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Pei Huang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Qiurong Yu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China; Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinhui Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Yu Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Youmin Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Zhijia Jin
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Yan Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Peng Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China.
| | - E Mark Haacke
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China; Department of Neurology, Wayne State University, 3990 John R, Detroit, MI, USA; Department of Biomedical Engineering, Wayne State University, 3990 John R, Detroit, MI, USA; Department of Radiology, Wayne State University, 3990 John R, Detroit, MI, USA.
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai 200025, China; Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Jouvencel A, Dilharreguy B, Baillet M, Pérès K, Dartigues JF, Amieva H, Mayo W, Catheline G. Age-related disturbances in rest-activity rhythms and integrity of the hippocampal network: An exploratory study. Neurobiol Sleep Circadian Rhythms 2025; 18:100111. [PMID: 39834591 PMCID: PMC11743803 DOI: 10.1016/j.nbscr.2024.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
To better understand the relationship between the rest-activity rhythms and cognitive impairments during aging, we assessed the longitudinal changes in the rest-activity rhythms in an elderly population and their possible detrimental effect on the hippocampal network. This was done longitudinally in a rural cohort with two actigraphic assessments and brain imaging examinations, seven years apart. A segmentation of the hippocampus and its related structures was used to assess volumes and functional connectivity in this network based on anatomical and resting state functional data. Regression models were carried out to investigate the potential association of the evolution of sleep and rest-activity rhythms parameters with the structural and functional integrity of the hippocampal network. Our sample was composed of 33 subjects aged 75.2 ± 2.4 years old at the first time point with 40% of women. After seven years, the sleep of our participants did not change but their rest-activity rhythms did (p < 0.05), with a decrease in relative amplitude (∂RA = -0.021) and stability (∂IS = -0.044) as well as an increase in fragmentation (∂IV = +0.072). The deterioration of rest-activity rhythms was correlated with a lower anterior hippocampal volume (p corrected <0.05) while no correlation with functional connectivity was observed. These findings suggest that a degradation of rest-activity rhythms in people over 70 years old could constitute a factor of hippocampal vulnerability. Preventive interventions should consider rest-activity rhythms in the oldest-old population.
Collapse
Affiliation(s)
- Aurore Jouvencel
- INCIA, EPHE, Université PSL, Univ Bordeaux, CNRS, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Bixente Dilharreguy
- INCIA, EPHE, Université PSL, Univ Bordeaux, CNRS, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Marion Baillet
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Karine Pérès
- INSERM, Bordeaux Population Health Research Center, University of Bordeaux, UMR U1219, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Jean-François Dartigues
- INSERM, Bordeaux Population Health Research Center, University of Bordeaux, UMR U1219, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Hélène Amieva
- INSERM, Bordeaux Population Health Research Center, University of Bordeaux, UMR U1219, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Willy Mayo
- INCIA, EPHE, Université PSL, Univ Bordeaux, CNRS, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Gwenaëlle Catheline
- INCIA, EPHE, Université PSL, Univ Bordeaux, CNRS, 146, Rue Léo Saignat, 33076, Bordeaux, France
| |
Collapse
|
6
|
Kierońska-Siwak S, Filipiak P, Jabłońska M, Sokal P. A comparison of diffusion tensor imaging tractography approaches to identify the Frontal Aslant Tract in neurosurgical patients. Front Neurosci 2025; 19:1543032. [PMID: 40356699 PMCID: PMC12066519 DOI: 10.3389/fnins.2025.1543032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/20/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction This study aims to present various tractography methods for delineating the Frontal Aslant Tract (FAT) and to quantify morphological features of FAT based on diffusion tensor imaging. Methods The study includes 68 patients, for which FAT was reconstructed using the Region Of Interest (ROI)-based approach. The ROIs were defined in either SFG - Superior Frontal Gyrus (ROI 1), or SMA-Supplementary Motor Area (ROI 2). The respective endpoints were located in the Inferior Frontal Gyrus (IFG)-either in pars opercularis or in pars triangularis. For each patient, FAT was delineated using four combinations of the above ROI-endpoint pairs. Results The highest streamline counts and fiber volumes of FAT were obtained using ROI 1 (i.e., SFG) with the endpoint in IFG pars opercularis. All subjects expressed left dominance of the pathway quantified by the higher streamline counts and fiber volumes regardless of gender. Additionally, higher Mean Diffusivity (MD) and lower Fractional Anisotropy (FA) values were observed in patients above 55 years of age than in younger patients. Discussion FAT is a neural pathway that can be tracked based on various anatomical landmarks. Clinically, it appears that delineating FAT between SFG and the pars opercularis region of IFG is optimal, as it is directly associated with the highest number of fibers and the greatest volume of the tract contained between these points.
Collapse
Affiliation(s)
- Sara Kierońska-Siwak
- Department of Neurosurgery, Functional and Stereotactic Neurosurgery, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
- Department of Neurosurgery and Neurology, Jan Biziel University Hospital No 2, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Patryk Filipiak
- Center for Advanced Imaging Innovation and Research (CAIR), NYU Langone Health, New York, NY, United States
| | - Magdalena Jabłońska
- Department of Neurosurgery, Functional and Stereotactic Neurosurgery, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
- Doctoral School of Medical and Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Paweł Sokal
- Department of Neurosurgery, Functional and Stereotactic Neurosurgery, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
- Department of Neurosurgery and Neurology, Jan Biziel University Hospital No 2, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
7
|
Vandersmissen J, Dewachter I, Cuypers K, Hansen D. The Impact of Exercise Training on the Brain and Cognition in Type 2 Diabetes, and its Physiological Mediators: A Systematic Review. SPORTS MEDICINE - OPEN 2025; 11:42. [PMID: 40274715 PMCID: PMC12022206 DOI: 10.1186/s40798-025-00836-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/16/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Type 2 diabetes (T2DM) affects brain structure and function, and is associated with an increased risk of dementia and mild cognitive impairment. It is known that exercise training has a beneficial effect on cognition and brain structure and function, at least in healthy people, but the impact of exercise training on these aspects remains to be fully elucidated in patients with T2DM. OBJECTIVE To determine the impact of exercise training on cognition and brain structure and function in T2DM, and identify the involved physiological mediators. METHODS This paper systematically reviews studies that evaluate the effect of exercise training on cognition in T2DM, and aims to indicate the most beneficial exercise modality for improving or preserving cognition in this patient group. In addition, the possible physiological mediators and targets involved in these improvements are narratively described in the second part of this review. Papers published up until the 14th of January 2025 were searched by means of the electronic databases PubMed, Embase, and Web of Science. Studies directly investigating the effect of any kind of exercise training on the brain or cognition in patients with T2DM, or animal models thereof, were included, with the exception of human studies assessing cognition only at one time point, and studies combining exercise training with other interventions (e.g. dietary changes, cognitive training, etc.). Study quality was assessed by means of the TESTEX tool for human studies, and the CAMARADES tool for animal studies. RESULTS For the systematic part of the review, 22 papers were found to be eligible. 18 out of 22 papers (81.8%) showed a significant positive effect of exercise training on cognition in T2DM, of which two studies only showed significant improvements in the minority of the cognitive tests. Four papers (18.2%) could not find a significant effect of exercise on cognition in T2DM. Resistance and endurance exercise were found to be equally effective for achieving cognitive improvement. Machine-based power training is seemingly more effective than resistance training with body weight and elastic bands to reach cognitive improvement. In addition, BDNF, lactate, leptin, adiponectin, GSK3β, GLP-1, the AMPK/SIRT1 pathway, and the PI3K/Akt pathway were identified as plausible mediators directly from studies investigating the effect of exercise training on brain structure and function in T2DM. Via these mediators, exercise training induces multiple beneficial brain changes, such as increased neuroplasticity, increased insulin sensitivity, and decreased inflammation. CONCLUSION Overall, exercise training beneficially affects cognition and brain structure and function in T2DM, with resistance and endurance exercise having similar effects. However, there is a need for additional studies, and more methodological consistency between different studies in order to define an exercise program optimal for improving cognition in T2DM. Furthermore, we were able to define several mediators involved in the effect of exercise training on cognition in T2DM, but further research is necessary to unravel the entire process.
Collapse
Affiliation(s)
- Jitske Vandersmissen
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium.
| | - Ilse Dewachter
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | - Koen Cuypers
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium
- Movement Control and Neuroplasticity Research Group, Department of Movement Sciences, Group Biomedical Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Dominique Hansen
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium
- Heart Centre Hasselt, Jessa Hospital, 3500, Hasselt, Belgium
| |
Collapse
|
8
|
Taskaya F, Amini A, Swiatek VM, Al-Hamid S, Reiser J, Rashidi A, Stein KP, Sandalcioglu IE, Neyazi B. Age-Dependent Changes of the Sylvian Fissure Configuration. World Neurosurg 2025; 196:123825. [PMID: 40015675 DOI: 10.1016/j.wneu.2025.123825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE To investigate age-related morphological changes of the Sylvian fissure (SF) and their implications for neurosurgical procedures. METHODS A cohort of 150 individuals across the age groups 10-20, 40-50, and 80-90 years was analyzed using Brainlab software for 3-dimensional visualization and volumetric analysis of the SF and various brain regions. We compared SF volumes between age groups and investigated dynamic changes in SF configuration over time. Correlation analyses were performed to identify how atrophy in specific brain regions affects the SF volume and configuration. RESULTS Atrophy was evident in all measured regions of the brain. The frontoparietal lobe underwent the strongest atrophy, while the occipital lobe showed the least. Each age group exhibited a consistent distribution of lobe volumes, although a marginal decrease in frontoparietal lobe proportion was observed in the groups of higher age. The annual atrophy rate in the frontoparietal and temporal lobes was steady. Additionally, ventricular expansion, which may influence white matter atrophy patterns, correlated with age. A consistent increase in SF volume in relation to the intracranial volume was observed across all age groups, with a notable increase in SF volume in older patients. This expansion, especially at the anterior-superior point, might be influenced by gravity, cerebral elasticity, and lobe torque. CONCLUSIONS Our investigation highlights the significance of age-dependent changes in SF volume and configuration due to brain atrophy throughout life. These changes, influenced by physical factors, underscore the need for tailored surgical approaches. Additionally, brain pathologies affecting volume could significantly alter SF configuration.
Collapse
Affiliation(s)
- Firat Taskaya
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany
| | - Amir Amini
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany
| | - Vanessa M Swiatek
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany
| | - Sifian Al-Hamid
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany
| | - Julius Reiser
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany
| | - Ali Rashidi
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany
| | - Klaus-Peter Stein
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany
| | - I Erol Sandalcioglu
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany
| | - Belal Neyazi
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Saxony-Anhalt, Germany.
| |
Collapse
|
9
|
Rocchi A, Wollebo HS, Khalili K. Neurotropic Viruses as Acute and Insidious Drivers of Aging. Biomolecules 2025; 15:514. [PMID: 40305226 PMCID: PMC12025245 DOI: 10.3390/biom15040514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Aging is the result of various compounding stresses that gradually overcome the homeostatic regulation of the cell, resulting in irreversible damage. This manifests as many acute and chronic conditions, the most common of which are neurodegeneration and dementia. Epidemiological studies have shown significant, strong correlations between viral infection and neurodegenerative diseases. This review overlays the characteristics of viral pathogenesis with the hallmarks of aging to discuss how active and latent viruses contribute to aging. Through our contextualization of myriad basic science papers, we offer explanations for premature aging via viral induction of common stress response pathways. Viruses induce many stresses: dysregulated homeostasis by exogenous viral proteins and overwhelmed protein quality control mechanisms, DNA damage through direct integration and epigenetic manipulation, immune-mediated oxidative stress and immune exhaustion, and general energy theft that is amplified in an aging system. Overall, this highlights the long-term importance of vaccines and antivirals in addition to their acute benefits.
Collapse
Affiliation(s)
| | - Hassen S. Wollebo
- Center for Neurovirology and Gene Editing, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Kamel Khalili
- Center for Neurovirology and Gene Editing, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| |
Collapse
|
10
|
Stephen J, Kharkongor R, Khan U, Kathirvel M, Radhakrishnan R. Cognitive Training and Enrichment Modulates Neural Plasticity and Enhances Cognitive Reserve in Aging Rats. Exp Aging Res 2025:1-24. [PMID: 40116649 DOI: 10.1080/0361073x.2025.2476331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/03/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVES Cognitive decline in non-pathological aging is widely prevalent among the aging population. The current study assessed the impact of cognitive training (Ct) with multiple modules targeting various facets of learning and memory and the additional influence of an enriched environment (Ct+ee) on hippocampal subfields of aging male rats. METHODS Male Wistar rats aged 18 months were sorted into Control, Ct, and Ct+ee groups and were exposed to the respective modules for 30 days. Spontaneous behavioral tasks to assess working memory and recognition memory were performed. The hippocampal proper (CA1, CA3) and dentate gyrus (DG) neurons were analyzed for dendrite length, arborization, and spine density. The Synaptophysin, PSD 95 and BDNF, p53 and p-tau levels in the hippocampus were quantified. RESULTS The Ct group and Ct+ee group performed significantly better than the control group in behavioural tasks and had improved dendrite profiles of DG and basal tree of CA1 region of hippocampus. The Ct+ee group had increased dendrite length, arborization, and spine density in CA1, CA3 and DG neurons. Ct and Ct+ee groups showed increased expression of synaptophysin, PSD95 and BDNF and decreased p53 and p-tau levels in the hippocampus. CONCLUSION Cognitive training modules targeting specific mnemonic functions and enriched environment with diverse cognitive stimulators had a comprehensive effect on the neuronal health augmenting the impoverished cognitive reserve in aging rats.
Collapse
Affiliation(s)
- JenishaChris Stephen
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Ronyson Kharkongor
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - UlfathTasneem Khan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Muniraj Kathirvel
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Rameshkumar Radhakrishnan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| |
Collapse
|
11
|
Komleva Y, Shpiliukova K, Bondar N, Salmina A, Khilazheva E, Illarioshkin S, Piradov M. Decoding brain aging trajectory: predictive discrepancies, genetic susceptibilities, and emerging therapeutic strategies. Front Aging Neurosci 2025; 17:1562453. [PMID: 40177249 PMCID: PMC11962000 DOI: 10.3389/fnagi.2025.1562453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The global extension of human lifespan has intensified the focus on aging, yet its underlying mechanisms remain inadequately understood. The article highlights aspects of genetic susceptibility to impaired brain bioenergetics, trends in age-related gene expression related to neuroinflammation and brain senescence, and the impact of stem cell exhaustion and quiescence on accelerated brain aging. We also review the accumulation of senescent cells, mitochondrial dysfunction, and metabolic disturbances as central pathological processes in aging, emphasizing how these factors contribute to inflammation and disrupt cellular competition defining the aging trajectory. Furthermore, we discuss emerging therapeutic strategies and the future potential of integrating advanced technologies to refine aging assessments. The combination of several methods including genetic analysis, neuroimaging techniques, cognitive tests and digital twins, offer a novel approach by simulating and monitoring individual health and aging trajectories, thereby providing more accurate and personalized insights. Conclusively, the accurate estimation of brain aging trajectories is crucial for understanding and managing aging processes, potentially transforming preventive and therapeutic strategies to improve health outcomes in aging populations.
Collapse
Affiliation(s)
| | | | - Nikolai Bondar
- Research Center of Neurology, Moscow, Russia
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Elena Khilazheva
- Department of Biological Chemistry with Courses in Medical, Research Institute of Molecular Medicine and Pathobiochemistry, Pharmaceutical and Toxicological Chemistry Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Healthcare of the Russian Federation, Krasnoyarsk, Russia
| | | | | |
Collapse
|
12
|
Tieze SM, Esqueda A, McAllister R, Lagator M, Yücel B, Sun E, Lam TT, Lockyer N, Gupta K, Chandra SS. Molecular elucidation of brain lipofuscin in aging and Neuronal Ceroid Lipofuscinosis. RESEARCH SQUARE 2025:rs.3.rs-6010379. [PMID: 40166029 PMCID: PMC11957193 DOI: 10.21203/rs.3.rs-6010379/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Lipofuscin is an autofluorescent material that accrues in brain tissues with age and in Neuronal Ceroid Lipofuscinosis (NCL), a neurodegenerative disease with pediatric onset. The distribution, composition, and organellar origin of lipofuscin have remained unclear despite its widespread presence in aged tissues and involvement in neurodegeneration. Here, we elucidate lipofuscin composition and report the spatiotemporal dynamics of lipofuscin accumulation in aging and NCL on a neuroanatomical atlas. Multimodal mass spectrometry, ultrastructural analyses, and assays of metabolic flux identify a primary role of the lysosomal-mitochondrial axis in lipofuscin formation. Dissection of implicated molecular pathways reveals protein S-acylation and lipid homeostasis as central processes involved in aging and NCL.
Collapse
Affiliation(s)
- Sofia Massaro Tieze
- Departments of Neurology & Neuroscience, Yale University, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| | - Alexander Esqueda
- Departments of Neurology & Neuroscience, Yale University, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| | | | - Matija Lagator
- Photon Science Institute, Department of Chemistry, University of Manchester, Manchester, UK
- Rosalind Franklin Institute, Rutherford Appleton Laboratory, Didcot, Oxfordshire, UK
| | - Betül Yücel
- Departments of Neurology & Neuroscience, Yale University, New Haven, CT, USA
| | - Eric Sun
- Yale College, Yale University, New Haven, CT, USA
| | - TuKiet T. Lam
- Departments of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Keck Mass Spectrometry & Proteomics Resource, W.M. Keck Biotechnology Resource Laboratory, New Haven, CT, USA
| | - Nicholas Lockyer
- Photon Science Institute, Department of Chemistry, University of Manchester, Manchester, UK
| | - Kallol Gupta
- Department of Cell Biology, Yale University, New Haven, CT, USA
| | - Sreeganga S. Chandra
- Departments of Neurology & Neuroscience, Yale University, New Haven, CT, USA
- Senior author
- Lead contact
| |
Collapse
|
13
|
Abstract
OBJECTIVES This study compares auditory processing performance across different decades of adulthood, including young adults and middle-aged individuals with normal hearing and no spontaneous auditory complaints. DESIGN We assessed 80 participants with normal hearing, at least 10 years of education, and normal global cognition. The participants completed various auditory tests, including speech-in-noise, dichotic digits, duration, pitch pattern sequence, gap in noise, and masking level difference. In addition, we conducted working memory assessments and administered a questionnaire on self-perceived hearing difficulties. RESULTS Our findings revealed significant differences in auditory test performance across different age groups, except for the masking level difference. The youngest group outperformed all other age groups in the speech-in-noise test, while differences in dichotic listening and temporal resolution emerged from the age of 40 and in temporal ordering from the age of 50. Moreover, higher education levels and better working memory test scores were associated with better auditory performance as individuals aged. However, the influence of these factors varied across different auditory tests. It is interesting that we observed increased self-reported hearing difficulties with age, even in participants without spontaneous auditory complaints. CONCLUSIONS Our study highlights significant variations in auditory test performance, with noticeable changes occurring from age 30 and becoming more pronounced from age 40 onward. As individuals grow older, they tend to perceive more hearing difficulties. Furthermore, the impact of age on auditory processing performance is influenced by factors such as education and working memory.
Collapse
Affiliation(s)
- Pamela P Lunardelo
- Department of Psychology, School of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Brazil
| | - Marisa T H Fukuda
- Department of Psychology, School of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Brazil
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sthella Zanchetta
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
14
|
Sidhu AS, Duarte KTN, Shahid TH, Sharkey RJ, Lauzon ML, Salluzzi M, McCreary CR, Protzner AB, Goodyear BG, Frayne R. Age- and Sex-Specific Patterns in Adult Brain Network Segregation. Hum Brain Mapp 2025; 46:e70169. [PMID: 40084534 PMCID: PMC11907239 DOI: 10.1002/hbm.70169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
The human brain is organized into several segregated associative and sensory functional networks, each responsible for various aspects of cognitive and sensory processing. These functional networks become less segregated over the adult lifespan, possibly contributing to cognitive decline that is observed during advanced age. To date, a comprehensive understanding of decreasing network segregation with age has been hampered by (1) small sample sizes, (2) lack of investigation at different spatial scales, (3) the limited age range of participants, and more importantly (4) an inadequate consideration of sex (biological females and males) differences. This study aimed to address these shortcomings. Resting-state functional magnetic resonance imaging data were collected from 357 cognitively intact participants (18.2-91.8 years; 49.9 ± 17.1 years; 27.70 ± 1.72 MoCA score, 203 [56.8%] females), and the segregation index (defined as one minus the ratio of between-network connectivity to within-network connectivity) was calculated at three spatial scales of brain networks: whole-brain network, intermediate sensory and associative networks, as well as core visual (VIS), sensorimotor (SMN), frontoparietal (FPN), ventral attention (VAN), dorsal attention (DAN), and default mode networks (DMN). Where applicable, secondary within-, between-, and pairwise connectivity analyses were also conducted to investigate the origin of any observed age and sex effects on network segregation. For any given functional metric, linear and quadratic age effects, sex effects, and respective age by sex interaction effects were assessed using backwards iterative linear regression modeling. Replicating previous work, brain networks were found to become less segregated across adulthood. Specifically, negative quadratic decreases in whole-brain network, intermediate associative network, VAN, and DMN segregation index were observed. Intermediate sensory networks, VIS, and SMN exhibited negative linear decreases in segregation index. Secondary analysis revealed that this process of age-related functional reorganization was preferential as functional connectivity was observed to increase either between anatomically adjacent associative networks (DMN-DAN, FPN-DAN) or between anterior associative and posterior sensory networks (VIS-DAN, VIS-DMN, VIS-FPN, SMN-DMN, and SMN-FPN). Inherent sex differences in network segregation index were also observed. Specifically, whole-brain, associative, DMN, VAN, and FPN segregation index was greater in females compared to males, irrespective of age. Secondary analysis found that females have reduced functional connectivity between associative networks (DAN-VAN, VAN-FPN) compared to males and independent of age. A notable linear age-related decrease in FPN SI was also only observed for females and not males. The observed findings support the notion that functional networks reorganize across the adult lifespan, becoming less segregated. This decline may reflect underlying neurocognitive aging mechanisms like neural dedifferentiation, inefficiency, and compensation. The aging trajectories and rates of decreasing network segregation, however, vary across associative and sensory networks. This study also provides preliminary evidence of inherent sex differences in network organization, where associative networks are more segregated in females than males. These inherent sex differences suggest that female functional networks may be more efficient and functionally specialized compared to males across adulthood. Given these findings, future studies should take a more focused approach to examining sex differences across the lifespan, incorporating multimodal methodologies.
Collapse
Affiliation(s)
- Abhijot Singh Sidhu
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Research Centre, Calgary, Alberta, Canada
| | - Kaue T N Duarte
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Research Centre, Calgary, Alberta, Canada
- Calgary Image Processing and Analysis Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Talal H Shahid
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Research Centre, Calgary, Alberta, Canada
| | - Rachel J Sharkey
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Research Centre, Calgary, Alberta, Canada
| | - M Louis Lauzon
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Research Centre, Calgary, Alberta, Canada
| | - Marina Salluzzi
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Calgary Image Processing and Analysis Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Cheryl R McCreary
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Research Centre, Calgary, Alberta, Canada
| | - Andrea B Protzner
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
- The Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | - Bradley G Goodyear
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Research Centre, Calgary, Alberta, Canada
| | - Richard Frayne
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Research Centre, Calgary, Alberta, Canada
- Calgary Image Processing and Analysis Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| |
Collapse
|
15
|
Perez FP, Walker B, Morisaki J, Kanakri H, Rizkalla M. Neurostimulation devices to treat Alzheimer's disease. EXPLORATION OF NEUROSCIENCE 2025; 4:100674. [PMID: 40084342 PMCID: PMC11904933 DOI: 10.37349/en.2025.100674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025]
Abstract
The use of neurostimulation devices for the treatment of Alzheimer's disease (AD) is a growing field. In this review, we examine the mechanism of action and therapeutic indications of these neurostimulation devices in the AD process. Rapid advancements in neurostimulation technologies are providing non-pharmacological relief to patients affected by AD pathology. Neurostimulation therapies include electrical stimulation that targets the circuitry-level connection in important brain areas such as the hippocampus to induce therapeutic neuromodulation of dysfunctional neural circuitry and electromagnetic field (EMF) stimulation that targets anti-amyloid molecular pathways to promote the degradation of beta-amyloid (Aβ). These devices target specific or diffuse cortical and subcortical brain areas to modulate neuronal activity at the electrophysiological or molecular pathway level, providing therapeutic effects for AD. This review attempts to determine the most effective and safe neurostimulation device for AD and provides an overview of potential and current clinical indications. Several EMF devices have shown a beneficial or harmful effect in cell cultures and animal models but not in AD human studies. These contradictory results may be related to the stimulation parameters of these devices, such as frequency, penetration depth, power deposition measured by specific absorption rate, time of exposure, type of cell, and tissue dielectric properties. Based on this, determining the optimal stimulation parameters for EMF devices in AD and understanding their mechanism of action is essential to promote their clinical application, our review suggests that repeated EMF stimulation (REMFS) is the most appropriate device for human AD treatments. Before its clinical application, it is necessary to consider the complicated and interconnected genetic and epigenetic effects of REMFS-biological system interaction. This will move forward the urgently needed therapy of EMF in human AD.
Collapse
Affiliation(s)
- Felipe P. Perez
- Department of Medicine, Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brett Walker
- Department of Medicine, Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jorge Morisaki
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Haitham Kanakri
- Department of Electrical and Computer Engineering, Purdue University, Indianapolis, IN 46202, USA
| | - Maher Rizkalla
- Department of Electrical and Computer Engineering, Purdue University, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Quan J, Liu Q, Li P, Yang Z, Zhang Y, Zhao F, Zhu G. Mesenchymal stem cell exosome therapy: current research status in the treatment of neurodegenerative diseases and the possibility of reversing normal brain aging. Stem Cell Res Ther 2025; 16:76. [PMID: 39985030 PMCID: PMC11846194 DOI: 10.1186/s13287-025-04160-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/21/2025] [Indexed: 02/23/2025] Open
Abstract
With the exacerbation of the aging population trend, a series of neurodegenerative diseases caused by brain aging have become increasingly common, significantly impacting the daily lives of the elderly and imposing heavier burdens on nations and societies. Brain aging is a complex process involving multiple mechanisms, including oxidative stress, apoptosis of damaged neuronal cells, chronic inflammation, and mitochondrial dysfunction, and research into new therapeutic strategies to delay brain aging has gradually become a research focus in recent years. Mesenchymal stem cells (MSCs) have been widely used in cell therapy due to their functions such as antioxidative stress, anti-inflammation, and tissue regeneration. However, accompanying safety issues such as immune rejection, tumor development, and pulmonary embolism cannot be avoided. Studies have shown that using exosome derived from mesenchymal stem cells (MSC-Exo) for the treatment of neurodegenerative diseases is a safe and effective method. It not only has the therapeutic effects of stem cells but also avoids the risks associated with cell therapy. Therefore, exploring new therapeutic strategies to delay normal brain aging from the mechanism of MSC-Exo in the treatment of neurodegenerative diseases is feasible. This review summarizes the characteristics of MSC-Exo and their clinical progress in the treatment of neurodegenerative diseases, aiming to explore the possibility and potential mechanisms of MSC-Exo in reversing brain aging.
Collapse
Affiliation(s)
- Jinglan Quan
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Qing Liu
- Department of Library, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Pinghui Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Zhiyu Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Yaohui Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Fuxing Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Gaohong Zhu
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China.
| |
Collapse
|
17
|
Bower AE, Chung JW, Burciu RG. Mapping the aging brain: Insights into microstructural changes from free water-corrected fractional anisotropy. Neurosci Lett 2025; 849:138120. [PMID: 39862921 PMCID: PMC11851011 DOI: 10.1016/j.neulet.2025.138120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Aging has a significant impact on brain structure, demonstrated by numerous MRI studies using diffusion tensor imaging (DTI). While these studies reveal changes in fractional anisotropy (FA) across different brain regions, they tend to focus on white matter tracts and cognitive regions, often overlooking gray matter and motor areas. Additionally, traditional DTI metrics can be affected by partial volume effects. To address these limitations and gain a better understanding of microstructural changes across the whole brain, we utilized free water-corrected fractional anisotropy (FAt) to examine aging-related microstructural changes in a group of 20 young adults (YA) and 24 older adults (OA). A voxel-wise analysis revealed that YA had higher FAt values predominantly in white matter tracts associated with both motor and non-motor functions. In contrast, OA showed higher levels of FAt primarily in gray matter regions, including both subcortical and cortical motor areas, and occipital and temporal cortices. Complementing these cross-sectional results, correlation analyses within the OA group showed that many of these changes are further exacerbated with increasing age, underscoring the progressive nature of these microstructural alterations. In summary, the distinct patterns of FAt changes in gray versus white matter with aging suggest different underlying mechanisms. While white matter FAt values decrease, likely due to axonal degeneration, the increase in gray matter FAt could reflect either compensatory processes or pathological changes. Including behavioral data in future studies will be crucial for understanding the functional implications of these microstructural gray matter changes and their effects on cognitive and motor functions.
Collapse
Affiliation(s)
- Abigail E Bower
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA
| | - Jae Woo Chung
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Roxana G Burciu
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA.
| |
Collapse
|
18
|
Chau MT, Agzarian M, Wilcox RA, Todd G. Does age, sex, and area of substantia nigra echogenicity predict the MRI appearance of nigrosome-1? J Neurol Sci 2025; 469:123383. [PMID: 39787958 DOI: 10.1016/j.jns.2024.123383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/02/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
The appearance of the substantia nigra (SN) can aid diagnosis of Parkinson's disease (PD). The effect of age and sex on the appearance of nigrosome-1 (SN subregion) on magnetic resonance imaging (MRI), and the relationship between nigrosome-1 (viewed with MRI) and SN echogenicity (viewed with transcranial ultrasound) is unknown. The study aimed to address these knowledge gaps. It was hypothesized that age, sex, and area of SN echogenicity predict area of MRI hyperintense signal in nigrosome-1. The cross-sectional study involved a group of healthy adults (n = 105; aged 21-79 years) and adults diagnosed with PD (n = 37; aged 51-80 years). Multilevel mixed-effects regression analysis was used to investigate if age, sex, side, and area of SN echogenicity predict area of nigrosome-1 hyperintense signal. Area of nigrosome-1 hyperintense signal increased by an average of 0.045 mm2/year in healthy adults (p = 0.016) but not in adults living with PD. Sex, side, and area of SN echogenicity did not predict area of nigrosome-1 hyperintense signal in either group. Disease duration predicted area of nigrosome-1 hyperintense signal in the PD group (coefficient = -0.41, p = 0.014). The expected between-group differences (p < 0.001) in median area of nigrosome-1 hyperintense signal (smallest area across sides: healthy = 7.6 mm2, PD = 0.0 mm2) and SN echogenicity (largest area across sides: healthy = 14.3 mm2, PD = 29.3 mm2) were observed. The results suggest that the MRI appearance of nigrosome-1 can vary with age but not sex or area of SN echogenicity. The results further understanding of the nigrosome-1 biomarker for PD and aid interpretation of nigrosome-1 MRI findings in clinical settings.
Collapse
Affiliation(s)
- Minh T Chau
- UniSA Allied Health & Human Performance and Alliance for Research in Exercise, Nutrition and Activity (ARENA), City East Campus, University of South Australia, GPO Box 2471, Adelaide, South Australia 5001, Australia; Faculty of Science and Health, 475 Bangala Way, Charles Sturt University, Boorooma, New South Wales 2678, Australia; South Australia Medical Imaging, Flinders Medical Centre, 1 Flinders Drive, Bedford Park, South Australia 5042, Australia.
| | - Marc Agzarian
- South Australia Medical Imaging, Flinders Medical Centre, 1 Flinders Drive, Bedford Park, South Australia 5042, Australia; College of Medicine and Public Health, Flinders University, 1 Flinders Drive, Bedford Park, South Australia 5042, Australia.
| | - Robert A Wilcox
- College of Medicine and Public Health, Flinders University, 1 Flinders Drive, Bedford Park, South Australia 5042, Australia; Neurology Department, Flinders Medical Centre, 1 Flinders Drive, Bedford Park, South Australia 5042, Australia; UniSA Clinical & Health Sciences and Alliance for Research in Exercise, Nutrition and Activity (ARENA), City East Campus, University of South Australia, GPO Box 2471, Adelaide, South Australia 5001, Australia.
| | - Gabrielle Todd
- UniSA Clinical & Health Sciences and Alliance for Research in Exercise, Nutrition and Activity (ARENA), City East Campus, University of South Australia, GPO Box 2471, Adelaide, South Australia 5001, Australia.
| |
Collapse
|
19
|
Drewelies J, Fiedler A, Brick TR, Kühn S. Investigating associations between the physical living environment and hippocampus in adulthood and older age. ENVIRONMENTAL RESEARCH 2025; 267:120728. [PMID: 39733985 DOI: 10.1016/j.envres.2024.120728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 12/22/2024] [Accepted: 12/27/2024] [Indexed: 12/31/2024]
Abstract
It is by now well known that the physical living environment has a major impact on people's life, but the neural structures involved in this relationship remain to be explored. Most studies investigating this relationship only focus on single environmental predictors. In order to understand how the multitude of factors constituting the living environment relate to brain structure we used data from the UK Biobank (n = 21,094; age Mean = 63.35 years; SD = 7.46; range = 45-82) to examine how individuals' immediate characteristics around the home address (e.g., green space; air pollution in the neighborhood) are associated with hippocampal volume, a brain region known to be highly plastic. We accounted for common demographic factors that have been shown to be associated with brain structure and known factors such as sex, income, education, and age. We made use of an analytical paradigm based on the feature importance estimation and recursive feature elimination with decision tree ensembles as well as linear regression analysis. Results identified a subset of environmental measures (e.g., pollution, green space, noise) most strongly associated with hippocampal volume across adulthood. Findings highlight the importance of the environment for individuals' brain structure.
Collapse
Affiliation(s)
- Johanna Drewelies
- Center for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany; Humboldt Universität zu, Berlin, Germany
| | - Angela Fiedler
- Center for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany
| | - Timothy R Brick
- Institute for Computational and Data Sciences, The Pennsylvania State University, State College, PA, USA
| | - Simone Kühn
- Center for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany; University Medical Center Hamburg-Eppendorf, Clinic and Policlinic for Psychiatry and Psychotherapy, Hamburg, Germany; Max Planck-UCL Center for Computational Psychiatry and Ageing Research, Germany.
| |
Collapse
|
20
|
Malkinson G, Henriques CM. Cerebrovascular ageing: how zebrafish can contribute to solving the puzzle. Front Physiol 2025; 16:1548242. [PMID: 39995479 PMCID: PMC11849178 DOI: 10.3389/fphys.2025.1548242] [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: 12/19/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
The mean life expectancy continues to increase world-wide. However, this extended lifespan trend is not accompanied by health span, or years of healthy life. Understanding the fundamental mechanisms responsible for the switch from health to morbidity with ageing are key to identifying potential therapeutic targets to decrease age-associated morbidity and increase years spent in good health. The leading cause of morbidity in Europe are diseases of the circulatory system and diseases of the nervous system and cognitive disorders are among the top-ten. Cerebrovascular ageing is therefore of particular importance as it links circulatory disease to brain functions, cognition, and behavior. Despite major progress in brain research and related technologies, little is known on how the cerebrovascular network changes its properties as ageing proceeds. Importantly, we do not understand why this is different in different individuals in what concerns rate of dysfunction and its downstream impact on brain function. Here we explore how the zebrafish has evolved as an attractive complementary ageing model and how it could provide key insights to understanding the mechanisms underlying cerebrovascular ageing and downstream consequences.
Collapse
Affiliation(s)
- Guy Malkinson
- Université de Lorraine, Inserm, DCAC, Nancy, France
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Catarina M. Henriques
- Department of Oncology and Metabolism, Healthy Lifespan Institute and MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
21
|
Abdi H, Sanchez-Molina D, Garcia-Vilana S, Rahimi-Movaghar V. Biomechanical perspectives on traumatic brain injury in the elderly: a comprehensive review. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2025; 7:022001. [PMID: 39761631 DOI: 10.1088/2516-1091/ada654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/06/2025] [Indexed: 02/05/2025]
Abstract
Traumatic brain injuries (TBIs) pose a significant health concern among the elderly population, influenced by age-related physiological changes and the prevalence of neurodegenerative diseases. Understanding the biomechanical dimensions of TBIs in this demographic is vital for developing effective preventive strategies and optimizing clinical management. This comprehensive review explores the intricate biomechanics of TBIs in the elderly, integrating medical and aging studies, experimental biomechanics of head tissues, and numerical simulations. Research reveals that global brain atrophy in normal aging occurs at annual rates of -0.2% to -0.5%. In contrast, neurodegenerative diseases such as Alzheimer's, Parkinson's, and multiple sclerosis are associated with significantly higher rates of brain atrophy. These variations in atrophy rates underscore the importance of considering differing brain atrophy patterns when evaluating TBIs among the elderly. Experimental studies further demonstrate that age-related changes in the mechanical properties of critical head tissues increase vulnerability to head injuries. Numerical simulations provide insights into the biomechanical response of the aging brain to traumatic events, aiding in injury prediction and preventive strategy development tailored to the elderly. Biomechanical analysis is essential for understanding injury mechanisms and forms the basis for developing effective preventive strategies. By incorporating local atrophy and age-specific impact characteristics into biomechanical models, researchers can create targeted interventions to reduce the risk of head injuries in vulnerable populations. Future research should focus on refining these models and integrating clinical data to better predict outcomes and enhance preventive care. Advancements in this field promise to improve health outcomes and reduce injury risks for the aging population.
Collapse
Affiliation(s)
- Hamed Abdi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Shen X, Zhang W, Li X, Zhang X, Li Q, Wu M, Fu L, Lu J, Zhu Z, Zhang B. Cerebral Small Vessel Disease Outperforms Brain Atrophy as an Imaging Biomarker in Diabetic Retinopathy. J Diabetes 2025; 17:e70058. [PMID: 39968694 PMCID: PMC11836613 DOI: 10.1111/1753-0407.70058] [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: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
AIM This study aimed to examine microvascular lesions and neurodegenerative changes in diabetic retinopathy (DR) compared to type 2 diabetes mellitus (T2DM) without DR (NDR) using structural MRI and to explore their associations with DR. METHODS 243 patients with NDR and 122 patients with DR were included. Participants underwent conventional brain MRI scans, clinical measurements, and fundus examinations. Cerebral small vessel disease (CSVD) imaging parameters were obtained using AI-based software, manually verified, and corrected for accuracy. Volumes of major cortical and subcortical regions representing neurodegeneration were assessed using automated brain segmentation and quantitative techniques. Statistical analysis included T-test, chi-square test, Mann-Whitney U test, multivariate analysis of variance (MANCOVA), multivariate logistic regression, area under the receiver operating characteristic curve (AUC), and Delong test. RESULTS DR group exhibited significant differences in 11 CSVD features. Meanwhile, DR showed an atrophy trend in the frontal cortex, occipital cortex, and subcortical gray matter (GM) compared to NDR. After adjustment, DR patients exhibited greater perivascular spaces (PVS) numbers in the parietal lobe (OR = 1.394) and deep brain regions (OR = 1.066), greater dilated perivascular spaces (DPVS) numbers in the left basal ganglia (OR = 2.006), greater small subcortical infarcts (SSI) numbers in the right hemisphere (OR = 3.104), and decreased left frontal PVS (OR = 0.824), total left DPVS (OR = 0.714), and frontal cortex volume (OR = 0.959) compared to NDR. Further, the CSVD model showed a larger AUC (0.823, 95% CI: 0.781-0.866) than the brain atrophy model (AUC = 0.757, 95% CI: 0.706-0.808). CONCLUSION Microvascular and neurodegeneration are significantly associated with DR. CSVD is a better imaging biomarker for DR than brain atrophy.
Collapse
Affiliation(s)
- Xinyi Shen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Wen Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjingChina
- Medical Imaging Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Xin Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjingChina
- Medical Imaging Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Xin Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjingChina
- Medical Imaging Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Qian Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Min Wu
- Department of RadiologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Linqing Fu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Jiaming Lu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Zhengyang Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjingChina
- Medical Imaging Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Institute of Brain ScienceNanjing UniversityNanjingChina
| |
Collapse
|
23
|
Peric R, Romčević I, Mastilović M, Starčević I, Boban J. Age-related volume decrease in subcortical gray matter is a part of healthy brain aging in men. Ir J Med Sci 2025; 194:339-345. [PMID: 39531119 DOI: 10.1007/s11845-024-03840-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND With the global population aging, the number of individuals over 60 is expected to double by 2050. Brain volume increases until age 13, stabilizes between 18 and 35, then declines by 0.2% annually. Magnetic resonance imaging (MRI) studies highlight significant gray matter atrophy, necessitating differentiation between normal aging and neurodegeneration. AIMS This study assessed the impact of aging on subcortical gray matter in healthy males to identify biomarkers of physiological aging. METHODS A retrospective study of 106 healthy males who underwent brain MRI from 2012 to 2016, divided into two age groups: younger and older than 35 years. MRI scans were performed using a 3 T machine, and volumetric analysis was conducted with VolBrain software. Subcortical gray matter volumes were compared between groups. The Shapiro-Wilk test evaluated normality. Student's t-test and Mann-Whitney U test were used for statistical analysis, with significance defined as p < 0.05. RESULTS Total intracranial volume was comparable between age groups (p = 0.527). Significant volume reductions (p < 0.05) were observed in subcortical gray matter structures, including the nucleus accumbens, caudate nucleus, globus pallidus, putamen, thalamus, and ventral diencephalon, particularly on the right side in the elderly group. CONCLUSIONS Subcortical gray matter volume in healthy males shows significant differences between older and younger individuals (p < 0.05), with asymmetrical reduction and certain structures on the right aging more rapidly. These findings are significant for distinguishing healthy aging from neurodegeneration.
Collapse
Affiliation(s)
- Radmila Peric
- Department of Radiology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000, Novi Sad, Serbia.
- Centre for Radiology, University Clinical Center of Vojvodina, Hajduk Veljkova 7-9, 21000, Novi Sad, Serbia.
| | - Igor Romčević
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000, Novi Sad, Serbia
| | - Milica Mastilović
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000, Novi Sad, Serbia
| | - Ivana Starčević
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000, Novi Sad, Serbia
- Division of Nuclear Medicine, Oncology Institute of Vojvodina, Put Dr Goldmana 4, 21204, Sremska Kamenica, Serbia
| | - Jasmina Boban
- Department of Radiology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000, Novi Sad, Serbia
- Centre for Diagnostic Imaging, Oncology Institute of Vojvodina, Put Dr Goldmana 4, 21204, Sremska Kamenica, Serbia
| |
Collapse
|
24
|
Binner P, Starshynov I, Tejeda G, McFall A, Molloy C, Ciccone G, Walker M, Vassalli M, Tobin AB, Faccio D. Optical, contact-free assessment of brain tissue stiffness and neurodegeneration. BIOMEDICAL OPTICS EXPRESS 2025; 16:447-459. [PMID: 39958854 PMCID: PMC11828460 DOI: 10.1364/boe.545580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 02/18/2025]
Abstract
Dementia affects a large proportion of the world's population. Approaches that allow for early disease detection and non-invasive monitoring of disease progression are desperately needed. Current approaches are centred on costly imaging technologies such as positron emission tomography and magnetic resonance imaging. We propose an alternative approach to assess neurodegeneration based on diffuse correlation spectroscopy (DCS), a remote and optical sensing technique. We employ this approach to assess neurodegeneration in mouse brains from healthy animals and those with prion disease. We find a statistically significant difference in the optical speckle decorrelation times between prion-diseased and healthy animals. We directly calibrated our DCS technique using hydrogel samples of varying Young's modulus, indicating that we can optically measure changes in the brain tissue stiffness in the order of 60 Pa (corresponding to a 1 s change in speckle decorrelation time). DCS holds promise for contact-free assessment of tissue stiffness alteration due to neurodegeneration, with a similar sensitivity to contact-based (e.g. nanoindentation) approaches.
Collapse
Affiliation(s)
- Philip Binner
- School of Physics and Astronomy, University of Glasgow, Glasgow, United Kingdom
| | - Ilya Starshynov
- School of Physics and Astronomy, University of Glasgow, Glasgow, United Kingdom
| | - Gonzalo Tejeda
- School of Molecular Biosciences, University of Glasgow, Glasgow, United Kingdom
| | - Aisling McFall
- School of Molecular Biosciences, University of Glasgow, Glasgow, United Kingdom
| | - Colin Molloy
- School of Molecular Biosciences, University of Glasgow, Glasgow, United Kingdom
| | - Giuseppe Ciccone
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute for Science and Technology (BIST) Barcelona, Spain
- James Watt School of Engineering, University of Glasgow, Glasgow, United Kingdom
| | - Matthew Walker
- James Watt School of Engineering, University of Glasgow, Glasgow, United Kingdom
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Glasgow, United Kingdom
| | - Andrew B. Tobin
- School of Molecular Biosciences, University of Glasgow, Glasgow, United Kingdom
| | - Daniele Faccio
- School of Physics and Astronomy, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
25
|
Zhi Y, Shi Y, Lu D, Xu D. Neurodevelopmental Implications Underpinning Hereditary Spastic Paraplegia. CNS Neurosci Ther 2025; 31:e70260. [PMID: 39932116 PMCID: PMC11811889 DOI: 10.1111/cns.70260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/25/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Hereditary spastic paraplegia (HSP) is a group of rare genetic neurodegenerative disorders characterized by corticospinal tract abnormalities. But frequently, abnormalities of proteins implicated in HSP have been identified in brain disorders of childhood, raising the possibility that early brain developmental mechanism underlying HSP. RESULTS AND CONCLUSIONS Here we summarized the clinical features of 89 HSP subtypes and found most have onset of symptoms earliest reported in infancy or early childhood. Importantly, HSP patients showed early brain developmental related phenotypes such as microcephaly, ventricular enlargement, and corpus callosum dysplasia. In addition, the expression trajectories analysis showed HSP genes were diffusely expressed across all human prenatal cortical regions and most genes enriched from post-conception weeks 8-24, periods characterized by neuro progenitor proliferation and neurogenesis. Furthermore, studies utilizing patient derived induced pluripotent stem cells (iPSCs)/organoids and mouse models have suggested that most HSP proteins play either direct or indirect roles in the development of the central nervous system. Therefore, HSP possesses a neurodevelopmental aspect and is not merely a degenerative disease, which may aid in better understanding the pathogenesis of this disease.
Collapse
Affiliation(s)
- Yiqiang Zhi
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, School of Basic Medical SciencesFujian Medical UniversityFuzhouChina
| | - Yan Shi
- Fujian Key Laboratory of Molecular Neurology, Institute of NeuroscienceFujian Medical UniversityFuzhouChina
| | - Danping Lu
- College of Life SciencesFujian Agriculture and Forestry UniversityFuzhouChina
| | - Dan Xu
- Fujian Key Laboratory of Molecular Neurology, Institute of NeuroscienceFujian Medical UniversityFuzhouChina
| |
Collapse
|
26
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of chemokines in aging and age-related diseases. Mech Ageing Dev 2025; 223:112009. [PMID: 39631472 DOI: 10.1016/j.mad.2024.112009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Chemokines (chemotactic cytokines) play essential roles in developmental process, immune cell trafficking, inflammation, immunity, angiogenesis, cellular homeostasis, aging, neurodegeneration, and tumorigenesis. Chemokines also modulate response to immunotherapy, and consequently influence the therapeutic outcome. The mechanisms underlying these processes are accomplished by interaction of chemokines with their cognate cell surface G protein-coupled receptors (GPCRs) and subsequent cellular signaling pathways. Chemokines play crucial role in influencing aging process and age-related diseases across various tissues and organs, primarily through inflammatory responses (inflammaging), recruitment of macrophages, and orchestrated trafficking of other immune cells. Chemokines are categorized in four distinct groups based on the position and number of the N-terminal cysteine residues; namely, the CC, CXC, CX3C, and (X)C. They mediate inflammatory responses, and thereby considerably impact aging process across multiple organ-systems. Therefore, understanding the underlying mechanisms mediated by chemokines may be of crucial importance in delaying and/or modulating the aging process and preventing age-related diseases. In this review, we highlight recent progress accomplished towards understanding the role of chemokines and their cellular signaling pathways involved in aging and age-relaed diseases of various organs. Moreover, we explore potential therapeutic strategies involving anti-chemokines and chemokine receptor antagonists aimed at reducing aging and mitigating age-related diseases. One of the modern methods in this direction involves use of chemokine receptor antagonists and anti-chemokines, which suppress the pro-inflammatory response, thereby helping in resolution of inflammation. Considering the wide-spectrum of functional involvements of chemokines in aging and associated diseases, several clinical trials are being conducted to develop therapeutic approaches using anti-chemokine and chemokine receptor antagonists to improve life span and promote healthy aging.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi 110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Akshay Bhardwaj
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad Road, Faridabad, Haryana 121001, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
27
|
Jerez-Salas F, Campos-Jara C, Araya Sierralta S, Jerez-Mayorga D, Ramirez-Campillo R, Contreras-Díaz G, Carrasco-Alarcón V, Martínez-Cortés H, Arellano-Roco C, Hernández-Cifuentes V, Contreras-Osorio F. Effects of Resistance Training on Executive Functions of Cognitively Healthy Older Adults: A Systematic Review and Meta-Analysis Protocol. Healthcare (Basel) 2025; 13:165. [PMID: 39857192 PMCID: PMC11764570 DOI: 10.3390/healthcare13020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Aging involves a series of changes in non-pathological age-related conditions, some of which impact the cognitive functioning of older adults. Executive functions are cognitive skills that are often affected in this process, although they have been shown to improve after physical exercise interventions. This protocol aims to describe the procedures that will be carried out in a systematic literature review, including a meta-analysis of the effects of resistance interventions on the main dimensions of executive function in cognitively healthy older adults compared to active or passive control groups. Methods: The PRISMA-P guidelines will be followed. Eligibility criteria will be organized based on the PICOS strategy (older adults with normal cognition ≥60 years; chronic resistance interventions ≥4 weeks; active or passive control group; direct measures of executive function). The PubMed, EBSCO, Scopus, and Web of Science databases will be used. The risk of bias and quality of evidence will be measured using RoB2 and GRADE, respectively. The DerSimonian-Laird random effects model will be used for the meta-analysis. The effect size will be calculated using Hedges' g with a 95% confidence interval and p < 0.05 to indicate statistical significance. Discussion: The results of the proposed review may be useful to justify the design and implementation of treatment plans based on resistance training for the prevention and management of cognitive changes typical of aging among older adults. PROSPERO registry: CRD42024571127.
Collapse
Affiliation(s)
- Felipe Jerez-Salas
- Facultad de Salud y Ciencias Sociales, Universidad de Las Américas, Sede Providencia, Manuel Montt 948, Santiago 7500973, Chile;
| | - Christian Campos-Jara
- Exercise and Rehabilitation Sciences Institute, Faculty of Rehabilitation Sciences, Universidad Andres Bello, Santiago 7591538, Chile; (C.C.-J.); (D.J.-M.)
| | - Sergio Araya Sierralta
- Departamento de Educación Física, Universidad de Atacama, Copiapó 1531772, Chile; (S.A.S.); (H.M.-C.)
| | - Daniel Jerez-Mayorga
- Exercise and Rehabilitation Sciences Institute, Faculty of Rehabilitation Sciences, Universidad Andres Bello, Santiago 7591538, Chile; (C.C.-J.); (D.J.-M.)
- Strength and Conditioning Laboratory, CTS-642 Research Group, Department Physical Education and Sports, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
| | - Rodrigo Ramirez-Campillo
- Exercise and Rehabilitation Sciences Institute, School of Physical Therapy, Faculty of Rehabilitation Sciences, Universidad Andres Bello, Santiago 7591538, Chile; (R.R.-C.); (V.H.-C.)
| | - Guido Contreras-Díaz
- Escuela de Kinesiología, Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Lago Panguipulli 1390, Puerto Montt 5501842, Chile;
| | - Vanessa Carrasco-Alarcón
- Departamento de Educación Física, Deportes y Recreación, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Hugo Martínez-Cortés
- Departamento de Educación Física, Universidad de Atacama, Copiapó 1531772, Chile; (S.A.S.); (H.M.-C.)
| | | | - Victoria Hernández-Cifuentes
- Exercise and Rehabilitation Sciences Institute, School of Physical Therapy, Faculty of Rehabilitation Sciences, Universidad Andres Bello, Santiago 7591538, Chile; (R.R.-C.); (V.H.-C.)
| | - Falonn Contreras-Osorio
- Exercise and Rehabilitation Sciences Institute, Faculty of Rehabilitation Sciences, Universidad Andres Bello, Santiago 7591538, Chile; (C.C.-J.); (D.J.-M.)
| |
Collapse
|
28
|
Labarta-Bajo L, Allen NJ. Astrocytes in aging. Neuron 2025; 113:109-126. [PMID: 39788083 PMCID: PMC11735045 DOI: 10.1016/j.neuron.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/05/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
The mammalian nervous system is impacted by aging. Aging alters brain architecture, is associated with molecular damage, and can manifest with cognitive and motor deficits that diminish the quality of life. Astrocytes are glial cells of the CNS that regulate the development, function, and repair of neural circuits during development and adulthood; however, their functions in aging are less understood. Astrocytes change their transcriptome during aging, with astrocytes in areas such as the cerebellum, the hypothalamus, and white matter-rich regions being the most affected. While numerous studies describe astrocyte transcriptional changes in aging, many questions still remain. For example, how is astrocyte function altered by transcriptional changes that occur during aging? What are the mechanisms promoting astrocyte aged states? How do aged astrocytes impact brain function? This review discusses features of aged astrocytes and their potential triggers and proposes ways in which they may impact brain function and health span.
Collapse
Affiliation(s)
- Lara Labarta-Bajo
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Nicola J Allen
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
29
|
Fehrmann MLA, Meijer FJA, Mylanus EAM, Pennings RJE, Lanting CP, Huinck WJ. Evaluating cochlear implant outcomes in DFNA9 subjects: a comprehensive study on cerebral white matter lesions and vestibular abnormalities. Eur Arch Otorhinolaryngol 2025; 282:183-191. [PMID: 39271588 PMCID: PMC11735485 DOI: 10.1007/s00405-024-08933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024]
Abstract
PURPOSE This study assessed whether the Fazekas score could account for the variability in cochlear implantation (CI) outcomes among individuals with DFNA9 and evaluated signal loss in the semicircular canals (SCCs) on magnetic resonance imaging (MRI) among individuals with DFNA9. METHOD This retrospective cross-sectional study included CI recipients with DFNA9. Pre-implantation MRI-scans were reviewed to determine the Fazekas score, localizing and grading cerebral white matter lesions (WML), and identify abnormalities in the SCCs. CI performance was assessed by evaluating phoneme scores one year post-implantation. The function of the SCCs was evaluated using rotatory chair testing with electronystagmography (ENG) and the video Head Impulse Test (vHIT). RESULTS Forty-five subjects (49 ears) were enrolled. The phoneme scores significantly improved from 35% (IQR 11-50) pre-implantation to 84% (IQR 76-90) one year post-implantation. No correlation was observed between the Fazekas score and the one-year post-implantation phoneme score (rsp=0.003, p = 0.986). Signal loss in at least one SCCs was detected in 97.7% of subjects and 77.8% of ears. There was no correlation between vestibular test results and fluid signal loss in the SCCs on MRI. CONCLUSION Most individuals with DFNA9 show improved speech recognition with CI. The observed variability in CI outcomes was not linked to the Fazekas score. Additionally, our study confirms a high prevalence of focal sclerosis in DFNA9. Recognizing the limitations of this study, further research is needed to explore the predictive role of the Fazekas score on CI outcomes and its relationship with vestibular function.
Collapse
Affiliation(s)
- M L A Fehrmann
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - F J A Meijer
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - E A M Mylanus
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - R J E Pennings
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - C P Lanting
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - W J Huinck
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| |
Collapse
|
30
|
Su S, Huang R, Liu Y. The effects of transcranial direct current stimulation on global cognition in patients with Alzheimer's disease: An update meta-analysis. J Alzheimers Dis 2025; 103:19-37. [PMID: 39784680 DOI: 10.1177/13872877241298303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease. At present, there are currently no drugs that can cure AD. OBJECTIVE A number of empirical studies have shown that transcranial direct current stimulation (tDCS) may be used to treat cognitive abnormalities in patients with AD. We will through meta-analysis reviews tDCS overall research on the effects of cognitive function in patients with AD. METHODS Systematic searches were performed in the PubMed, Embase, and Cochrane Library databases from their creation until 8 March 2024. Using a fixed effect model and random effect model to evaluate the average difference between the treatment group and control group (MD) and its 95% confidence interval (CI). RESULTS The study included 10 randomized controlled trials (Nactive = 165, Nsham = 167). The results of the overall analysis showed that tDCS did not significantly improve the overall cognitive function (SMD = 0.17; 95%CI = -0.05, 0.39; p = 0.14; I² = 51%). Quality of life of AD patients after treatment was also evaluated, but no improvement was seen. Subgroup analysis showed no significant improvement in global cognitive function after tDCS treatment. The sensitivity analysis to confirm the reliability of the data, risk assessment did not find any high-risk projects. CONCLUSIONS The tDCS treatment did not improve cognitive function in patients with AD. Further empirical research in the future will help to explore new schemes for tDCS to improve cognitive function of patients.
Collapse
Affiliation(s)
- Siyuan Su
- School of Clinical Medicine, Qilu Medical University, Zibo, Shandong, China
| | - Ruihan Huang
- School of Public Health, Qilu Medical University, Zibo, Shandong, China
| | - Yongsheng Liu
- School of Clinical Medicine, Qilu Medical University, Zibo, Shandong, China
| |
Collapse
|
31
|
Ibrahim MFG, Ali FF, Ali SFES, Shaker ES, Mahmoud HI, Abdellatif FEM, Mokhemer SA. Neuroprotective effect of red dragon fruit extract ameliorates oxidative stress and inflammation in D-galactose-induced aging rat model: biochemical, histological and immunohistochemical study. J Mol Histol 2024; 56:51. [PMID: 39707017 DOI: 10.1007/s10735-024-10316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/17/2024] [Indexed: 12/23/2024]
Abstract
Aging is a worldwide socioeconomic burden. Cerebellar aging is an enigma contributing to many behavioral aging disorders, hence is its hindering by prophylactic measurements is a crucial geriatric research target. Red dragon fruit (RDF) is a tropical fruit with antioxidant, anti-inflammatory and anti-apoptotic properties. This study aimed to determine the protective effect of RDF extract against cerebellar aging. Thirty-two male albino rats were randomly allocated into 4 groups: Control, RDF, aged and RDF-aged groups. Aged group revealed structural distortion affecting cerebellar layers including a significant (P < 0.05) decrease in Purkinje cells number and decrease in granular cell layer thickness by comparison to the control and RDF groups. Additionally, distorted capillary endothelium, and defective myelination were noticed. Interestingly, cerebellar active caspase-3, iNOS, MDA and 3-NT and serum TNF-α levels significantly increased with aging by comparison to the control and RDF groups (all P < 0.05). Biochemical analysis revealed a significant (P < 0.05) decrease in cerebellar SOD and serum GSH levels in aged rats. RDF extract remarkably ameliorated most of the neuronal degenerative changes with a significant (P < 0.05) increase in Purkinje cells numbers, and granular cell layer thickness by comparison to the aged group. Furthermore, it resulted in a significant (P < 0.05) decrease in cerebellum expression of active caspase-3, iNOS, MDA, 3-NT, and serum TNF-α levels associated with a significant (P < 0.05) increase in cerebellar SOD and serum GSH levels by comparison to the aged group. To the best of our knowledge this is the first study showing a neuroprotective effect for RDF against cerebellar aging. RDF might be effective in attenuation of age-induced cerebellar degenerative changes through its anti-apoptotic, antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
| | - Fatma F Ali
- Medical Physiology Department, Faculty of Medicine, Minia University, Minia, Egypt
- Biochemistry, Molecular Biology and Physiology Department, Faculty of Medicine, Mutah University, Al-Karak, Jordan
| | | | - Emad S Shaker
- Agric. Chemistry Department, Faculty of Agriculture, Minia University, Minia, Egypt
| | - Hemdan I Mahmoud
- Agric. Chemistry Department, Faculty of Agriculture, Minia University, Minia, Egypt
| | | | - Sahar A Mokhemer
- Histology and Cell Biology Department, Faculty of Medicine, Minia University, Minia, Egypt.
| |
Collapse
|
32
|
Wang H, Shi C, Jiang L, Liu X, Tang R, Tang M. Neuroimaging techniques, gene therapy, and gut microbiota: frontier advances and integrated applications in Alzheimer's Disease research. Front Aging Neurosci 2024; 16:1485657. [PMID: 39691161 PMCID: PMC11649678 DOI: 10.3389/fnagi.2024.1485657] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder marked by cognitive decline, for which effective treatments remain elusive due to complex pathogenesis. Recent advances in neuroimaging, gene therapy, and gut microbiota research offer new insights and potential intervention strategies. Neuroimaging enables early detection and staging of AD through visualization of biomarkers, aiding diagnosis and tracking of disease progression. Gene therapy presents a promising approach for modifying AD-related genetic expressions, targeting amyloid and tau pathology, and potentially repairing neuronal damage. Furthermore, emerging evidence suggests that the gut microbiota influences AD pathology through the gut-brain axis, impacting inflammation, immune response, and amyloid metabolism. However, each of these technologies faces significant challenges, including concerns about safety, efficacy, and ethical considerations. This article reviews the applications, advantages, and limitations of neuroimaging, gene therapy, and gut microbiota research in AD, with a particular focus on their combined potential for early diagnosis, mechanistic insights, and therapeutic interventions. We propose an integrated approach that leverages these tools to provide a multi-dimensional framework for advancing AD diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Haitao Wang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Shi
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ling Jiang
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaozhu Liu
- Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Rui Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxi Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Yaan People’s Hospital (Yaan Hospital of West China Hospital of Sichuan University), Yaan, Sichuan, China
| |
Collapse
|
33
|
Gooijers J, Pauwels L, Hehl M, Seer C, Cuypers K, Swinnen SP. Aging, brain plasticity, and motor learning. Ageing Res Rev 2024; 102:102569. [PMID: 39486523 DOI: 10.1016/j.arr.2024.102569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
Motor skill learning, the process of acquiring new motor skills, is critically important across the lifespan, from early development through adulthood and into older age, as well as in pathological conditions (i.e., rehabilitation). Extensive research has demonstrated that motor skill acquisition in young adults is accompanied by significant neuroplastic changes, including alterations in brain structure (gray and white matter), function (i.e., activity and connectivity), and neurochemistry (i.e., levels of neurotransmitters). In the aging population, motor performance typically declines, characterized by slower and less accurate movements. However, despite these age-related changes, older adults maintain the capacity for skill improvement through training. In this review, we explore the extent to which the aging brain retains the ability to adapt in response to motor learning, specifically whether skill acquisition is accompanied by neural changes. Furthermore, we discuss the associations between inter-individual variability in brain structure and function and the potential for future learning in older adults. Finally, we consider the use of non-invasive brain stimulation techniques aimed at optimizing motor learning in this population. Our review provides insights into the neurobiological underpinnings of motor learning in older adults and emphasizes strategies to enhance their motor skill acquisition.
Collapse
Affiliation(s)
- Jolien Gooijers
- KU Leuven, Movement Control and Neuroplasticity Research Group, Group Biomedical Sciences, Leuven 3001, Belgium; Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium.
| | - Lisa Pauwels
- KU Leuven, Movement Control and Neuroplasticity Research Group, Group Biomedical Sciences, Leuven 3001, Belgium; Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Melina Hehl
- KU Leuven, Movement Control and Neuroplasticity Research Group, Group Biomedical Sciences, Leuven 3001, Belgium; Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium; Neuroplasticity and Movement Control Research Group, Rehabilitation Research Institute (REVAL), Hasselt University, Diepenbeek, Belgium
| | - Caroline Seer
- KU Leuven, Movement Control and Neuroplasticity Research Group, Group Biomedical Sciences, Leuven 3001, Belgium; Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Koen Cuypers
- KU Leuven, Movement Control and Neuroplasticity Research Group, Group Biomedical Sciences, Leuven 3001, Belgium; Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium; Neuroplasticity and Movement Control Research Group, Rehabilitation Research Institute (REVAL), Hasselt University, Diepenbeek, Belgium
| | - Stephan P Swinnen
- KU Leuven, Movement Control and Neuroplasticity Research Group, Group Biomedical Sciences, Leuven 3001, Belgium; Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Ostermann PN, Evering TH. The impact of aging on HIV-1-related neurocognitive impairment. Ageing Res Rev 2024; 102:102513. [PMID: 39307316 DOI: 10.1016/j.arr.2024.102513] [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: 04/02/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/25/2024]
Abstract
Depending on the population studied, HIV-1-related neurocognitive impairment is estimated to impact up to half the population of people living with HIV (PLWH) despite the availability of combination antiretroviral therapy (cART). Various factors contribute to this neurocognitive impairment, which complicates our understanding of the molecular mechanisms involved. Biological aging has been implicated as one factor possibly impacting the development and progression of HIV-1-related neurocognitive impairment. This is increasingly important as the life expectancy of PLWH with virologic suppression on cART is currently projected to be similar to that of individuals not living with HIV. Based on our increasing understanding of the biological aging process on a cellular level, we aim to dissect possible interactions of aging- and HIV-1 infection-induced effects and their role in neurocognitive decline. Thus, we begin by providing a brief overview of the clinical aspects of HIV-1-related neurocognitive impairment and review the accumulating evidence implicating aging in its development (Part I). We then discuss potential interactions between aging-associated pathways and HIV-1-induced effects at the molecular level (Part II).
Collapse
Affiliation(s)
- Philipp Niklas Ostermann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Teresa Hope Evering
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
35
|
Rodríguez-Fernández L, Zorzo C, Arias JL. Photobiomodulation in the aging brain: a systematic review from animal models to humans. GeroScience 2024; 46:6583-6623. [PMID: 38861125 PMCID: PMC11493890 DOI: 10.1007/s11357-024-01231-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024] Open
Abstract
Aging is a multifactorial biological process that may be associated with cognitive decline. Photobiomodulation (PBM) is a non-pharmacological therapy that shows promising results in the treatment or prevention of age-related cognitive impairments. The aim of this review is to compile the preclinical and clinical evidence of the effect of PBM during aging in healthy and pathological conditions, including behavioral analysis and neuropsychological assessment, as well as brain-related modifications. 37 studies were identified by searching in PubMed, Scopus, and PsycInfo databases. Most studies use wavelengths of 800, 810, or 1064 nm but intensity and days of application were highly variable. In animal studies, it has been shown improvements in spatial memory, episodic-like memory, social memory, while different results have been found in recognition memory. Locomotor activity improved in Parkinson disease models. In healthy aged humans, it has been outlined improvements in working memory, cognitive inhibition, and lexical/semantic access, while general cognition was mainly enhanced on Alzheimer disease or mild cognitive impairment. Anxiety assessment is scarce and shows mixed results. As for brain activity, results outline promising effects of PBM in reversing metabolic alterations and enhancing mitochondrial function, as evidenced by restored CCO activity and ATP levels. Additionally, PBM demonstrated neuroprotective, anti-inflammatory, immunomodulatory and hemodynamic effects. The findings suggest that PBM holds promise as a non-invasive intervention for enhancing cognitive function, and in the modulation of brain functional reorganization. It is necessary to develop standardized protocols for the correct, beneficial, and homogeneous use of PBM.
Collapse
Affiliation(s)
| | - Candela Zorzo
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain.
| | - Jorge L Arias
- ISPA, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
36
|
Haudry S, Turpin AL, Landeau B, Mézenge F, Delarue M, Hébert O, Marchant NL, Klimecki O, Collette F, Gonneaud J, de La Sayette V, Vivien D, Lutz A, Chételat G. Decoding meditation mechanisms underlying brain preservation and psycho-affective health in older expert meditators and older meditation-naive participants. Sci Rep 2024; 14:29521. [PMID: 39604423 PMCID: PMC11603193 DOI: 10.1038/s41598-024-79687-3] [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: 04/19/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Meditation is a mental training approach that can improve mental health and well-being in aging. Yet the underlying mechanisms remain unknown. The Medit-Ageing model stipulates that three mechanisms - attentional, constructive, and deconstructive - upregulate positive psycho-affective factors and downregulate negative ones. To test this hypothesis, we measured brain structural MRI and perfusion, negative and positive psycho-affective composite scores, and meditation mechanisms in 27 older expert meditators and 135 meditation-naive older controls. We identified brain and psycho-affective differences and performed mediation analyses to assess whether and which meditation mechanisms mediate their links.Meditators showed significantly higher volume in fronto-parietal areas and perfusion in temporo-occipito-parietal areas. They also had higher positive and lower negative psycho-affective scores. Attentional and constructive mechanisms both mediated the links between brain differences and the positive psycho-affective score whereas the deconstructive mechanism mediated the links between brain differences and the negative psycho-affective score.Our results corroborate the Medit-Ageing model, indicating that, in aging, meditation leads to brain changes that decrease negative psycho-affective factors and increase positive ones through relatively specific mechanisms. Shedding light on the neurobiological and psycho-affective mechanisms of meditation in aging, these findings provide insights to refine future interventions.
Collapse
Affiliation(s)
- Sacha Haudry
- U1237, PhIND, Neuropresage Team, Normandy University, UNICAEN, INSERM, GIP Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Anne-Laure Turpin
- U1237, PhIND, Neuropresage Team, Normandy University, UNICAEN, INSERM, GIP Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Brigitte Landeau
- U1237, PhIND, Neuropresage Team, Normandy University, UNICAEN, INSERM, GIP Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Florence Mézenge
- U1237, PhIND, Neuropresage Team, Normandy University, UNICAEN, INSERM, GIP Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Marion Delarue
- U1237, PhIND, Neuropresage Team, Normandy University, UNICAEN, INSERM, GIP Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Oriane Hébert
- U1237, PhIND, Neuropresage Team, Normandy University, UNICAEN, INSERM, GIP Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Natalie L Marchant
- Division of Psychiatry, Faculty of Brain Sciences, University College London, London, UK
| | - Olga Klimecki
- Developmental Psychology, Friedrich-Schiller-Universität Jena, Jena, Germany
- Biological Psychology, Faculty of Psychology, Technische Universität Dresden, Dresden, Germany
| | - Fabienne Collette
- GIGA-CRC In Vivo Imaging, University of Liège, Liège, Belgium
- Psychology and Neuroscience of Cognition Research Unit, University of Liège, Liège, Belgium
| | - Julie Gonneaud
- U1237, PhIND, Neuropresage Team, Normandy University, UNICAEN, INSERM, GIP Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | | | - Denis Vivien
- PhIND "Physiopathology and Imaging of Neurological Disorders", Normandy University, UNICAEN, INSERM, U1237, Institut Blood & Brain @ Caen, Cyceron, Caen, 14000, France
- Département de Recherche Clinique, CHU Caen-Normandie, Caen, France
| | - Antoine Lutz
- Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR5292, Lyon 1 University, Lyon, France
| | - Gaël Chételat
- U1237, PhIND, Neuropresage Team, Normandy University, UNICAEN, INSERM, GIP Cyceron, Boulevard Henri Becquerel, Caen, 14000, France.
| |
Collapse
|
37
|
Bej E, Cesare P, d’Angelo M, Volpe AR, Castelli V. Neuronal Cell Rearrangement During Aging: Antioxidant Compounds as a Potential Therapeutic Approach. Cells 2024; 13:1945. [PMID: 39682694 PMCID: PMC11639796 DOI: 10.3390/cells13231945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/02/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Aging is a natural process that leads to time-related changes and a decrease in cognitive abilities, executive functions, and attention. In neuronal aging, brain cells struggle to respond to oxidative stress. The structure, function, and survival of neurons can be mediated by different pathways that are sensitive to oxidative stress and age-related low-energy states. Mitochondrial impairment is one of the most noticeable signs of brain aging. Damaged mitochondria are thought to be one of the main causes that feed the inflammation related to aging. Also, protein turnover is involved in age-related impairments. The brain, due to its high oxygen usage, is particularly susceptible to oxidative damage. This review explores the mechanisms underlying neuronal cell rearrangement during aging, focusing on morphological changes that contribute to cognitive decline and increased susceptibility to neurodegenerative diseases. Potential therapeutic approaches are discussed, including the use of antioxidants (e.g., Vitamin C, Vitamin E, glutathione, carotenoids, quercetin, resveratrol, and curcumin) to mitigate oxidative damage, enhance mitochondrial function, and maintain protein homeostasis. This comprehensive overview aims to provide insights into the cellular and molecular processes of neuronal aging and highlight promising therapeutic avenues to counteract age-related neuronal deterioration.
Collapse
Affiliation(s)
- Erjola Bej
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
- Department of the Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, 1001 Tirana, Albania
| | - Patrizia Cesare
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
| | - Anna Rita Volpe
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
- Department of the Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, 1001 Tirana, Albania
| |
Collapse
|
38
|
Lazar M, Moroti R, Barbu EC, Chitu-Tisu CE, Tiliscan C, Erculescu TM, Rosca RR, Frasila S, Schmilevschi ET, Simion V, Duca GT, Padiu IF, Andreescu DI, Anton AN, Pacurar CG, Perdun PM, Petre AM, Oprea CA, Popescu AM, Maria E, Ion DA, Olariu MC. The Impact of HIV on Early Brain Aging-A Pathophysiological (Re)View. J Clin Med 2024; 13:7031. [PMID: 39685490 DOI: 10.3390/jcm13237031] [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: 10/21/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: This review aims to provide a comprehensive understanding of how HIV alters normal aging trajectories in the brain, presenting the HIV-related molecular mechanisms and pathophysiological pathways involved in brain aging. The review explores the roles of inflammation, oxidative stress, and viral persistence in the brain, highlighting how these factors contribute to neuronal damage and cognitive impairment and accelerate normal brain aging. Additionally, it also addresses the impact of antiretroviral therapy on brain aging and the biological markers associated with its occurrence. Methods: We extensively searched PubMed for English-language articles published from 2000 to 2024. The following keywords were used in the search: "HIV", "brain", "brain aging", "neuroinflammation", "HAART", and "HAND". This strategy yielded 250 articles for inclusion in our review. Results: A combination of blood-brain barrier dysfunction, with the direct effects of HIV on the central nervous system, chronic neuroinflammation, telomere shortening, neurogenesis impairments, and neurotoxicity associated with antiretroviral treatment (ART), alters and amplifies the mechanisms of normal brain aging. Conclusions: Current evidence suggests that HIV infection accelerates neurodegenerative processes of normal brain aging, leading to cognitive decline and structural brain changes at an earlier age than typically observed in the general population.
Collapse
Affiliation(s)
- Mihai Lazar
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
- National Institute for Infectious Diseases Prof. Dr. Matei Bals, No. 1, Calistrat Grozovici Street, Sector 2, 021105 Bucharest, Romania
| | - Ruxandra Moroti
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
- National Institute for Infectious Diseases Prof. Dr. Matei Bals, No. 1, Calistrat Grozovici Street, Sector 2, 021105 Bucharest, Romania
| | - Ecaterina Constanta Barbu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Cristina Emilia Chitu-Tisu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Catalin Tiliscan
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
- Faculty of Dental Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Teodora Maria Erculescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Ruxandra Raluca Rosca
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Stefan Frasila
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Emma Teodora Schmilevschi
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Vladimir Simion
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - George Theodor Duca
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Isabela Felicia Padiu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Darie Ioan Andreescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Andreea Nicoleta Anton
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Cosmina Georgiana Pacurar
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Patricia Maria Perdun
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Alexandru Mihai Petre
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Constantin Adrian Oprea
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Adelina Maria Popescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Enachiuc Maria
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Daniela Adriana Ion
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Mihaela Cristina Olariu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
- National Institute for Infectious Diseases Prof. Dr. Matei Bals, No. 1, Calistrat Grozovici Street, Sector 2, 021105 Bucharest, Romania
| |
Collapse
|
39
|
Roque-Carvalho B, Pereira-Macedo J, Arantes M, Sousa J, Romana-Dias L, Ribeiro H, Myrcha P, Rocha-Neves J. Basal Ganglia's influence on awake test in carotid endarterectomy. Eur J Radiol 2024; 181:111812. [PMID: 39531780 DOI: 10.1016/j.ejrad.2024.111812] [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: 07/22/2024] [Revised: 10/05/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND White matter changes (WMC) have been associated with the underlying presence of chronic cerebral ischemia, such as in carotid stenosis and age-related white matter changes (ARWMC). A low attenuation on computed tomography (CT) characterizes these alterations. Patients undergoing carotid endarterectomy (CEA) with severe WMC may be at increased risk of intraoperative neurologic deficits (IND) during carotid clamping. This study aims to determine the potential role of ARWMC as a predictor of IND during CEA with regional anesthesia (RA). METHODS Patients undergoing CEA under RA at a tertiary referral center, who presented with IND during CEA were prospectively and consecutively recruited between January 2011 and December 2023. The control group comprised the immediately consecutive patient who underwent the same procedure without IND. From this sample, patients with preoperative CT were selected and compared based on ARWMC score (≤ 1 and 2). Differences in demographics and comorbidities were assessed between the groups. A multivariable logistic regression was performed. RESULTS One hundred and twenty-one patients were enrolled. Patients with IND had a significantly higher ARWMC score in basal ganglia (ARWMC-BG ≥ 2) and posterior circulation disease was more frequent (27.8 %). No significant differences were observed in anatomical variations of the circle of Willis. For patients with ARWMC-BG ≥ 2, a significant burden of other comorbidities was associated, such as chronic kidney disease, coronary disease, and atrial fibrillation. After multivariable logistic regression analysis, ARWMC-BG score ≥ 2 was an independent risk factor for IND (aOR 3.472). CONCLUSION An ARWMC-BG score above 2 predicts positive intraoperative "awake tests" in CEA with RA, constituting a reliable tool to stratify patients according to their risk of adverse events. However, larger prospective cohorts are needed to validate these findings and offer a better selection and management of this subset of patients.
Collapse
Affiliation(s)
| | - Juliana Pereira-Macedo
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; Department of Surgery - Centro Hospitalar do Médio-Ave, 4780 371 Vila Nova de Famalicão, Portugal; RISE@Health, 4200‑450 Porto, Portugal.
| | - Mavilde Arantes
- RISE@Health, 4200‑450 Porto, Portugal; Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; Department of Neuroradiology - Instituto Português de Oncologia, 4200-072 Porto, Portugal.
| | - Jose Sousa
- Department of Neuroradiology - Unidade Local de Saúde de São João, EPE, 4200-319 Porto, Portugal.
| | - Lara Romana-Dias
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; Department of Vascular Surgery - Unidade Local de Saúde de São João, EPE, 4200-319 Porto, Portugal.
| | - Hugo Ribeiro
- Community Palliative Care Support Team Gaia, 4400-043 Vila Nova de Gaia, Portugal; Department of Medicina da Comunidade, Informação e Decisão em Saúde - Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; Centre for Innovative Biomedicine and Biotechnology, 3000-548 Coimbra, Portugal.
| | - Piotr Myrcha
- 1st Chair and Department of General and Vascular Surgery, Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; Department of General, Vascular and Oncological Surgery, Masovian Brodnowski Hospital, 03-242 Warsaw, Poland.
| | - João Rocha-Neves
- RISE@Health, 4200‑450 Porto, Portugal; Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.
| |
Collapse
|
40
|
Guo K, Chaudhari N, Jafar T, Chowdhury N, Bogdan P, Irimia A. Anatomic Interpretability in Neuroimage Deep Learning: Saliency Approaches for Typical Aging and Traumatic Brain Injury. RESEARCH SQUARE 2024:rs.3.rs-4960427. [PMID: 39483910 PMCID: PMC11527355 DOI: 10.21203/rs.3.rs-4960427/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The black box nature of deep neural networks (DNNs) makes researchers and clinicians hesitant to rely on their findings. Saliency maps can enhance DNN explainability by suggesting the anatomic localization of relevant brain features. This study compares seven popular attribution-based saliency approaches to assign neuroanatomic interpretability to DNNs that estimate biological brain age (BA) from magnetic resonance imaging (MRI). Cognitively normal (CN) adults ( N = 13,394 , 5,900 males; mean age: 65.82 ± 8.89 years) are included for DNN training, testing, validation, and saliency map generation to estimate BA. To study saliency robustness to the presence of anatomic deviations from normality, saliency maps are also generated for adults with mild traumatic brain injury (mTBI, N = 214 , 135 males; mean age: 55.3 ± 9.9 years). We assess saliency methods' capacities to capture known anatomic features of brain aging and compare them to a surrogate ground truth whose anatomic saliency is known a priori. Anatomic aging features are identified most reliably by the integrated gradients method, which outperforms all others through its ability to localize relevant anatomic features. Gradient Shapley additive explanations, input × gradient, and masked gradient perform less consistently but still highlight ubiquitous neuroanatomic features of aging (ventricle dilation, hippocampal atrophy, sulcal widening). Saliency methods involving gradient saliency, guided backpropagation, and guided gradient-weight class attribution mapping localize saliency outside the brain, which is undesirable. Our research suggests the relative tradeoffs of saliency methods to interpret DNN findings during BA estimation in typical aging and after mTBI.
Collapse
Affiliation(s)
- Kevin Guo
- Thomas Lord Department of Computer Science, Viterbi School of Engineering, University of Southern California
| | - Nikhil Chaudhari
- Corwin D. Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California
| | - Tamara Jafar
- Neuroscience Graduate Program, University of Southern California
| | - Nahian Chowdhury
- Neuroscience Graduate Program, University of Southern California
| | - Paul Bogdan
- Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California
| |
Collapse
|
41
|
Lepore G, Succu S, Cappai MG, Frau A, Senes A, Zedda M, Farina V, Gadau SD. Morphological and Metabolic Features of Brain Aging in Rodents, Ruminants, Carnivores, and Non-Human Primates. Animals (Basel) 2024; 14:2900. [PMID: 39409849 PMCID: PMC11482532 DOI: 10.3390/ani14192900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Brain aging in mammals is characterized by morphological and functional changes in neural cells. Macroscopically, this process, leading to progressive cerebral volume loss and functional decline, includes memory and motor neuron deficits, as well as behavioral disorders. Morphologically, brain aging is associated with aged neurons and astrocytes, appearing enlarged and flattened, and expressing enhanced pH-dependent β-galactosidase activity. Multiple mechanisms are considered hallmarks of cellular senescence in vitro, including cell cycle arrest, increased lysosomal activity, telomere shortening, oxidative stress, and DNA damage. The most common markers for senescence identification were identified in (i) proteins implicated in cell cycle arrest, such as p16, p21, and p53, (ii) increased lysosomal mass, and (iii) increased reactive oxygen species (ROS) and senescence-associated secretory phenotype (SASP) expression. Finally, dysfunctional autophagy, a process occurring during aging, contributes to altering brain homeostasis. The brains of mammals can be studied at cellular and subcellular levels to elucidate the mechanisms on the basis of age-related and degenerative disorders. The aim of this review is to summarize and update the most recent knowledge about brain aging through a comparative approach, where similarities and differences in some mammalian species are considered.
Collapse
Affiliation(s)
- Gianluca Lepore
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (S.S.); (M.G.C.); (A.F.); (A.S.); (M.Z.); (V.F.); (S.D.G.)
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Liu C, Jing J, Jiang J, Wen W, Zhu W, Li Z, Pan Y, Cai X, Liu H, Zhou Y, Meng X, Zhang J, Wang Y, Li H, Jiang Y, Zheng H, Wang S, Niu H, Kochan N, Brodaty H, Wei T, Sachdev P, Liu T, Wang Y. Relationships between brain structure-function coupling in normal aging and cognition: A cross-ethnicity population-based study. Neuroimage 2024; 299:120847. [PMID: 39265959 DOI: 10.1016/j.neuroimage.2024.120847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/19/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024] Open
Abstract
Increased efforts in neuroscience seek to understand how macro-anatomical and physiological connectomes cooperatively work to generate cognitive behaviors. However, the structure-function coupling characteristics in normal aging individuals remain unclear. Here, we developed an index, the Coupling in Brain Structural connectome and Functional connectome (C-BSF) index, to quantify regional structure-function coupling in a large community-based cohort. C-BSF used diffusion tensor imaging (DTI) and resting-state functional magnetic resonance imaging (fMRI) data from the Polyvascular Evaluation for Cognitive Impairment and Vascular Events study (PRECISE) cohort (2007 individuals, age: 61.15 ± 6.49 years) and the Sydney Memory and Ageing Study (MAS) cohort (254 individuals, age: 83.45 ± 4.33 years). We observed that structure-function coupling was the strongest in the visual network and the weakest in the ventral attention network. We also observed that the weaker structure-function coupling was associated with increased age and worse cognitive level of the participant. Meanwhile, the structure-function coupling in the visual network was associated with the visuospatial performance and partially mediated the connections between age and the visuospatial function. This work contributes to our understanding of the underlying brain mechanisms by which aging affects cognition and also help establish early diagnosis and treatment approaches for neurological diseases in the elderly.
Collapse
Affiliation(s)
- Chang Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jing Jing
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Jiyang Jiang
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Wei Wen
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia; Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, UNSW Medicine, Sydney NSW 2052, Australia
| | - Wanlin Zhu
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zixiao Li
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuesong Pan
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xueli Cai
- Department of Neurology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, China
| | - Hao Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yijun Zhou
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xia Meng
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jicong Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yilong Wang
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yong Jiang
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huaguang Zheng
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Suying Wang
- Cerebrovascular Research Lab, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, China
| | - Haijun Niu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Nicole Kochan
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia; Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, UNSW Medicine, Sydney NSW 2052, Australia
| | - Henry Brodaty
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia; Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, UNSW Medicine, Sydney NSW 2052, Australia
| | - Tiemin Wei
- Department of Cardiology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, China
| | - Perminder Sachdev
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia; Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, UNSW Medicine, Sydney NSW 2052, Australia
| | - Tao Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Yongjun Wang
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
43
|
Guo KH, Chaudhari NN, Jafar T, Chowdhury NF, Bogdan P, Irimia A. Anatomic Interpretability in Neuroimage Deep Learning: Saliency Approaches for Typical Aging and Traumatic Brain Injury. Neuroinformatics 2024; 22:591-606. [PMID: 39503843 PMCID: PMC11579113 DOI: 10.1007/s12021-024-09694-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 11/13/2024]
Abstract
The black box nature of deep neural networks (DNNs) makes researchers and clinicians hesitant to rely on their findings. Saliency maps can enhance DNN explainability by suggesting the anatomic localization of relevant brain features. This study compares seven popular attribution-based saliency approaches to assign neuroanatomic interpretability to DNNs that estimate biological brain age (BA) from magnetic resonance imaging (MRI). Cognitively normal (CN) adults (N = 13,394, 5,900 males; mean age: 65.82 ± 8.89 years) are included for DNN training, testing, validation, and saliency map generation to estimate BA. To study saliency robustness to the presence of anatomic deviations from normality, saliency maps are also generated for adults with mild traumatic brain injury (mTBI, N = 214, 135 males; mean age: 55.3 ± 9.9 years). We assess saliency methods' capacities to capture known anatomic features of brain aging and compare them to a surrogate ground truth whose anatomic saliency is known a priori. Anatomic aging features are identified most reliably by the integrated gradients method, which outperforms all others through its ability to localize relevant anatomic features. Gradient Shapley additive explanations, input × gradient, and masked gradient perform less consistently but still highlight ubiquitous neuroanatomic features of aging (ventricle dilation, hippocampal atrophy, sulcal widening). Saliency methods involving gradient saliency, guided backpropagation, and guided gradient-weight class attribution mapping localize saliency outside the brain, which is undesirable. Our research suggests the relative tradeoffs of saliency methods to interpret DNN findings during BA estimation in typical aging and after mTBI.
Collapse
Affiliation(s)
- Kevin H Guo
- Thomas Lord Department of Computer Science, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Nikhil N Chaudhari
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Corwin D. Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Tamara Jafar
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Nahian F Chowdhury
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Paul Bogdan
- Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
- Corwin D. Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Quantitative and Computational Biology, Dornsife College of Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA.
- Centre for Healthy Brain Aging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 de Crespigny Park, London, SE5 8AF, UK.
| |
Collapse
|
44
|
Jurcau MC, Jurcau A, Cristian A, Hogea VO, Diaconu RG, Nunkoo VS. Inflammaging and Brain Aging. Int J Mol Sci 2024; 25:10535. [PMID: 39408862 PMCID: PMC11476611 DOI: 10.3390/ijms251910535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Progress made by the medical community in increasing lifespans comes with the costs of increasing the incidence and prevalence of age-related diseases, neurodegenerative ones included. Aging is associated with a series of morphological changes at the tissue and cellular levels in the brain, as well as impairments in signaling pathways and gene transcription, which lead to synaptic dysfunction and cognitive decline. Although we are not able to pinpoint the exact differences between healthy aging and neurodegeneration, research increasingly highlights the involvement of neuroinflammation and chronic systemic inflammation (inflammaging) in the development of age-associated impairments via a series of pathogenic cascades, triggered by dysfunctions of the circadian clock, gut dysbiosis, immunosenescence, or impaired cholinergic signaling. In addition, gender differences in the susceptibility and course of neurodegeneration that appear to be mediated by glial cells emphasize the need for future research in this area and an individualized therapeutic approach. Although rejuvenation research is still in its very early infancy, accumulated knowledge on the various signaling pathways involved in promoting cellular senescence opens the perspective of interfering with these pathways and preventing or delaying senescence.
Collapse
Affiliation(s)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Vlad Octavian Hogea
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
45
|
Sanchez-Martinez J, Solis-Urra P, Olivares-Arancibia J, Plaza-Diaz J. Physical Exercise and Mechanism Related to Alzheimer's Disease: Is Gut-Brain Axis Involved? Brain Sci 2024; 14:974. [PMID: 39451988 PMCID: PMC11506766 DOI: 10.3390/brainsci14100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Alzheimer's disease is a progressive neurodegenerative disease characterized by structural changes in the brain, including hippocampal atrophy, cortical thinning, amyloid plaques, and tau tangles. Due to the aging of the global population, the burden of Alzheimer's disease is expected to increase, making the exploration of non-pharmacological interventions, such as physical exercise, an urgent priority. RESULTS There is emerging evidence that regular physical exercise may mitigate the structural and functional declines associated with Alzheimer's disease. The underlying mechanisms, however, remain poorly understood. Gut-brain axis research is a promising area for further investigation. This system involves bidirectional communication between the gut microbiome and the brain. According to recent studies, the gut microbiome may influence brain health through modulating neuroinflammation, producing neuroactive compounds, and altering metabolic processes. Exercise has been shown to alter the composition of the gut microbiome, potentially impacting brain structure and function. In this review, we aim to synthesize current research on the relationship between physical exercise, structural brain changes in Alzheimer's disease, and the gut-brain axis. CONCLUSIONS In this study, we will investigate whether changes in the gut microbiome induced by physical exercise can mediate its neuroprotective effects, offering new insights into the prevention and treatment of Alzheimer's disease. By integrating findings from neuroimaging studies, clinical trials, and microbiome research, this review will highlight potential mechanisms. It will also identify key gaps in the literature. This will pave the way for future research directions.
Collapse
Affiliation(s)
- Javier Sanchez-Martinez
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, 18071 Granada, Spain;
| | - Patricio Solis-Urra
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, 18071 Granada, Spain;
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 8370134, Chile
| | - Jorge Olivares-Arancibia
- AFySE Group, Research in Physical Activity and School Health, School of Physical Education, Faculty of Education, Universidad de Las Américas, Santiago 7500975, Chile;
| | - Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
46
|
Peng YJ, Kuo CY, Chang SW, Lin CP, Tsai YH. Acceleration of brain aging after small-volume infarcts. Front Aging Neurosci 2024; 16:1409166. [PMID: 39391585 PMCID: PMC11464776 DOI: 10.3389/fnagi.2024.1409166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Previous studies have shown that stroke patients exhibit greater neuroimaging-derived biological "brain age" than control subjects. This difference, known as the brain age gap (BAG), is calculated by comparing the chronological age with predicted brain age and is used as an indicator of brain health and aging. However, whether stroke accelerates the process of brain aging in patients with small-volume infarcts has not been established. By utilizing longitudinal data, we aimed to investigate whether small-volume infarctions can significantly increase the BAG, indicating accelerated brain aging. Methods A total of 123 stroke patients presenting with small-volume infarcts were included in this retrospective study. The brain age model was trained via established protocols within the field of machine learning and the structural features of the brain from our previous study. We used t-tests and regression analyses to assess longitudinal brain age changes after stroke and the associations between brain age, acute stroke severity, and poststroke outcome factors. Results Significant brain aging occurred between the initial and 6-month follow-ups, with a mean increase in brain age of 1.04 years (t = 3.066, p < 0.05). Patients under 50 years of age experienced less aging after stroke than those over 50 years of age (p = 0.245). Additionally, patients with a National Institute of Health Stroke Scale score >3 at admission presented more pronounced adverse effects on brain aging, even after adjusting for confounders such as chronological age, sex, and total intracranial volume (F 1,117 = 7.339, p = 0.008, η 2 = 0.059). There were significant differences in the proportional brain age difference at 6 months among the different functional outcome groups defined by the Barthel Index (F 2,118 = 4.637, p = 0.012, η 2 = 0.073). Conclusion Stroke accelerates the brain aging process, even in patients with relatively small-volume infarcts. This phenomenon is particularly accentuated in elderly patients, and both stroke severity and poststroke functional outcomes are closely associated with accelerated brain aging. Further studies are needed to explore the mechanisms underlying the accelerated brain aging observed in stroke patients, with a particular focus on the structural alterations and plasticity of the brain following minor strokes.
Collapse
Affiliation(s)
- Ying-Ju Peng
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
- Department of Diagnostic Radiology, Chang Gung University, Taoyuan, Taiwan
| | - Chen-Yuan Kuo
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Sheng-Wei Chang
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
- Department of Diagnostic Radiology, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Yuan-Hsiung Tsai
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
- Department of Diagnostic Radiology, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
47
|
Sun X, Yuan X, Chen H, Li W. PM2.5 is linked to Alzheimer's syndrome and delirium: a mendelian randomization analysis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:308-315. [PMID: 39399654 PMCID: PMC11470430 DOI: 10.62347/fmuc9744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/27/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Increasing air pollution has drawn our attention to particulate matter (PM2.5), which has been shown to correlate significantly with respiratory and cardiovascular systems. However, whether PM2.5 is causally associated with Alzheimer's syndrome or delirium is unclear. METHODS We retrieved the genetic summary data of PM2.5 from genome-wide association studies (GWAS). The genetic information for Alzheimer's disease was obtained from the IEU OpenGWAS project, and that for delirium was obtained from FinnGen. We used two-sample Mendelian randomization analysis (MR) to associate PM2.5 with Alzheimer's disease or delirium. RESULTS The odds ratio (OR) for Alzheimer's disease was 0.996 with a p-value of 0.443 using the inverse variance weighted algorithm, and the OR associated with the outcome variable of delirium was 0.393 with a p-value of 0.343. CONCLUSION With the exclusion of confounding factors, our findings do not support a genetic association between PM2.5 and Alzheimer's disease or delirium. Further population-based and experimental studies are needed to dissect the complex correlation between PM2.5 and Alzheimer's disease or delirium.
Collapse
Affiliation(s)
- Xiaojin Sun
- Department of Cardiology, Sihong HospitalSuqian 223900, Jiangsu, China
| | - Xiaofan Yuan
- Department of Radiology of The Second Affiliated Hospital of Nanjing Medical UniversityNanjing 210011, Jiangsu, China
| | - Haoyan Chen
- Department of Geriatrics, Jiangsu Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, Jiangsu, China
| | - Wenjie Li
- Department of Geriatrics, The First Affiliated Hospital of Ningbo UniversityNingbo 315000, Zhejiang, China
| |
Collapse
|
48
|
Ma Y, Wang X, Li X, Chen X, Teng Z, Wang X, Yang J, Liu G. COP-22 Alleviates D-Galactose-Induced Brain Aging by Attenuating Oxidative Stress, Inflammation, and Apoptosis in Mice. Mol Neurobiol 2024; 61:6708-6720. [PMID: 38347285 PMCID: PMC11339142 DOI: 10.1007/s12035-024-03976-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/18/2024] [Indexed: 08/22/2024]
Abstract
Aging is a natural and inevitable process of organisms. With the intensification of population aging, research on aging has become a hot topic of global attention. The most obvious manifestation of human aging is the aging of brain function, which has been linked to the development of neurodegenerative diseases. In this study, COP-22, a mono-carbonyl curcumin derivative, was evaluated for its anti-aging ability, especially its ability to resist brain aging induced by D-galactose (D-gal) in mice. For brain protection, COP-22 could resist D-gal-induced oxidative stress by increasing the activity of antioxidative defense enzymes and enhancing antioxidant capacity in the brain tissue; COP-22 could improve the dysfunction of the cholinergic system by decreasing the increased activity of acetylcholinesterase and increasing the reduced content of acetylcholine induced by D-gal; and COP-22 could protect nerve cells of the brain. Further, western blot was used to determine related proteins of the brain. We found that COP-22 could effectively protect against brain injury (SIRT1, p53, p21, and p16) by inhibiting oxidative stress (Nrf2 and HO-1), inflammation (IL-6 and TNF-α), and apoptosis (Bax and caspase-3) in D-gal-induced aging mice. Additionally, COP-22 demonstrated the ability to reduce oxidative stress in serum and liver caused by D-gal, as well as relieve the damages in the liver and kidney induced by D-gal. These results indicated that COP-22 had potential anti-aging activity and could be used in the therapy of aging and aging-associated diseases like Alzheimer disease.
Collapse
Affiliation(s)
- Yazhong Ma
- School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China
| | - Xiaotong Wang
- School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China
| | - Xin Li
- School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China
| | - Xi Chen
- School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China
| | - Zhifeng Teng
- School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China
| | - Xuekun Wang
- School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China.
| | - Jie Yang
- School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China.
- Liaocheng Key Laboratory of Quality Control and Pharmacodynamic Evaluation of Ganoderma Lucidum, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China.
| | - Guoyun Liu
- School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China.
- Liaocheng Key Laboratory of Quality Control and Pharmacodynamic Evaluation of Ganoderma Lucidum, Liaocheng University, 1 Hunan Street, Liaocheng, 252059, Shandong, China.
| |
Collapse
|
49
|
Steffens DC. Applying the construct of social determinants of health to imaging-based brain changes. Int Psychogeriatr 2024; 36:710-713. [PMID: 37039439 DOI: 10.1017/s1041610223000339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 04/12/2023]
Affiliation(s)
- David C Steffens
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
50
|
Vásquez E, Oresti GM, Paez MD, Callegari EA, Masone D, Muñoz EM. Impact of aging on the GABA B receptor-mediated connectome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606013. [PMID: 39131332 PMCID: PMC11312617 DOI: 10.1101/2024.07.31.606013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
GABA B receptors (GABABRs) are heterodimeric seven-transmembrane receptors that interact with a range of proteins and form large protein complexes on cholesterol-rich membrane microdomains. As the brain ages, membrane cholesterol levels exhibit alterations, although it remains unclear how these changes impact protein-protein interactions and downstream signaling. Herein, we studied the structural bases for the interaction between GABABR and the KCC2 transporter, including their protein expression and distribution, and we compared data between young and aged rat cerebella. Also, we analyzed lipid profiles for both groups, and we used molecular dynamics simulations on three plasma membrane systems with different cholesterol concentrations, to further explore the GABABR-transporter interaction. Based on our results, we report that a significant decrease in GABAB2 subunit expression occurs in the aged rat cerebella. After performing a comparative co-immunoprecipitation analysis, we confirm that GABABR and KCC2 form a protein complex in adult and aged rat cerebella, although their interaction levels are reduced substantially as the cerebellum ages. On the other hand, our lipid analyses reveal a significant increase in cholesterol and sphingomyelin levels of the aged cerebella. Finally, we used the Martini coarse-grained model to conduct molecular dynamics simulations, from which we observed that membrane cholesterol concentrations can dictate whether the GABABR tail domains physically establish G protein-independent contacts with a transporter, and the timing when those associations eventually occur. Taken together, our findings illustrate how age-related alterations in membrane cholesterol levels affect protein-protein interactions, and how they could play a crucial role in regulating GABABR's interactome-mediated signaling. Significance Statement This study elucidates age-related changes in cerebellar GABAB receptors (GABABRs), KCC2, and plasma membrane lipids, shedding light on mechanisms underlying neurological decline. Molecular dynamics simulations reveal how membrane lipids influence protein-protein interactions, offering insights into age-related neurodegeneration. The findings underscore the broader impact of cerebellar aging on motor functions, cognition, and emotional processing in the elderly. By elucidating plasma membrane regulation and GABAergic dynamics, this research lays the groundwork for understanding aging-related neurological disorders and inspires further investigation into therapeutic interventions.
Collapse
Affiliation(s)
- Elena Vásquez
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Gerardo M. Oresti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - María D. Paez
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD, USA
| | - Eduardo A. Callegari
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD, USA
| | - Diego Masone
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Estela M. Muñoz
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| |
Collapse
|