101
|
Monnat RJ. James German and the Quest to Understand Human RECQ Helicase Deficiencies. Cells 2024; 13:1077. [PMID: 38994931 PMCID: PMC11240319 DOI: 10.3390/cells13131077] [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] [Revised: 05/10/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
James German's work to establish the natural history and cancer risk associated with Bloom syndrome (BS) has had a strong influence on the generation of scientists and clinicians working to understand other RECQ deficiencies and heritable cancer predisposition syndromes. I summarize work by us and others below, inspired by James German's precedents with BS, to understand and compare BS with the other heritable RECQ deficiency syndromes with a focus on Werner syndrome (WS). What we know, unanswered questions and new opportunities are discussed, as are potential ways to treat or modify WS-associated disease mechanisms and pathways.
Collapse
Affiliation(s)
- Raymond J Monnat
- Departments of Laboratory Medicine/Pathology and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
102
|
Yusri K, Kumar S, Fong S, Gruber J, Sorrentino V. Towards Healthy Longevity: Comprehensive Insights from Molecular Targets and Biomarkers to Biological Clocks. Int J Mol Sci 2024; 25:6793. [PMID: 38928497 PMCID: PMC11203944 DOI: 10.3390/ijms25126793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Aging is a complex and time-dependent decline in physiological function that affects most organisms, leading to increased risk of age-related diseases. Investigating the molecular underpinnings of aging is crucial to identify geroprotectors, precisely quantify biological age, and propose healthy longevity approaches. This review explores pathways that are currently being investigated as intervention targets and aging biomarkers spanning molecular, cellular, and systemic dimensions. Interventions that target these hallmarks may ameliorate the aging process, with some progressing to clinical trials. Biomarkers of these hallmarks are used to estimate biological aging and risk of aging-associated disease. Utilizing aging biomarkers, biological aging clocks can be constructed that predict a state of abnormal aging, age-related diseases, and increased mortality. Biological age estimation can therefore provide the basis for a fine-grained risk stratification by predicting all-cause mortality well ahead of the onset of specific diseases, thus offering a window for intervention. Yet, despite technological advancements, challenges persist due to individual variability and the dynamic nature of these biomarkers. Addressing this requires longitudinal studies for robust biomarker identification. Overall, utilizing the hallmarks of aging to discover new drug targets and develop new biomarkers opens new frontiers in medicine. Prospects involve multi-omics integration, machine learning, and personalized approaches for targeted interventions, promising a healthier aging population.
Collapse
Affiliation(s)
- Khalishah Yusri
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sanjay Kumar
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sheng Fong
- Department of Geriatric Medicine, Singapore General Hospital, Singapore 169608, Singapore
- Clinical and Translational Sciences PhD Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jan Gruber
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Science Division, Yale-NUS College, Singapore 138527, Singapore
| | - Vincenzo Sorrentino
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Neuroscience Cellular & Molecular Mechanisms, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| |
Collapse
|
103
|
Wen J, Tian YE, Skampardoni I, Yang Z, Cui Y, Anagnostakis F, Mamourian E, Zhao B, Toga AW, Zaleskey A, Davatzikos C. The Genetic Architecture of Biological Age in Nine Human Organ Systems. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.06.08.23291168. [PMID: 37398441 PMCID: PMC10312870 DOI: 10.1101/2023.06.08.23291168] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Understanding the genetic basis of biological aging in multi-organ systems is vital for elucidating age-related disease mechanisms and identifying therapeutic interventions. This study characterized the genetic architecture of the biological age gap (BAG) across nine human organ systems in 377,028 individuals of European ancestry from the UK Biobank. We discovered 393 genomic loci-BAG pairs (P-value<5×10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary, and renal systems. We observed BAG-organ specificity and inter-organ connections. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system while exerting pleiotropic effects on traits linked to multiple organ systems. A gene-drug-disease network confirmed the involvement of the metabolic BAG-associated genes in drugs targeting various metabolic disorders. Genetic correlation analyses supported Cheverud's Conjecture1 - the genetic correlation between BAGs mirrors their phenotypic correlation. A causal network revealed potential causal effects linking chronic diseases (e.g., Alzheimer's disease), body weight, and sleep duration to the BAG of multiple organ systems. Our findings shed light on promising therapeutic interventions to enhance human organ health within a complex multi-organ network, including lifestyle modifications and potential drug repositioning strategies for treating chronic diseases. All results are publicly available at https://labs-laboratory.com/medicine.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Filippos Anagnostakis
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Andrew Zaleskey
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
104
|
Oh HSH, Le Guen Y, Rappoport N, Urey DY, Rutledge J, Brunet A, Greicius MD, Wyss-Coray T. Plasma proteomics in the UK Biobank reveals youthful brains and immune systems promote healthspan and longevity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597771. [PMID: 38915561 PMCID: PMC11195058 DOI: 10.1101/2024.06.07.597771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Organ-derived plasma protein signatures derived from aptamer protein arrays track organ-specific aging, disease, and mortality in humans, but the robustness and clinical utility of these models and their biological underpinnings remain unknown. Here, we estimate biological age of 11 organs from 44,526 individuals in the UK Biobank using an antibody-based proteomics platform to model disease and mortality risk. Organ age estimates are associated with future onset of heart failure (heart age HR=1.83), chronic obstructive pulmonary disease (lung age HR=1.39), type II diabetes (kidney age HR=1.58), and Alzheimer's disease (brain age HR=1.81) and sensitive to lifestyle factors such as smoking and exercise, hormone replacement therapy, or supplements. Remarkably, the accrual of aged organs progressively increases mortality risk while a youthful brain and immune system are uniquely associated with disease-free longevity. These findings support the use of plasma proteins for monitoring organ health and the efficacy of drugs targeting organ aging disease.
Collapse
Affiliation(s)
- Hamilton Se-Hwee Oh
- Graduate Program in Stem Cell and Regenerative Medicine, Stanford University, Stanford, CA, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Deniz Yagmur Urey
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jarod Rutledge
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anne Brunet
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA, USA
| | - Michael D. Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
105
|
Pu F, Chen W, Li C, Fu J, Gao W, Ma C, Cao X, Zhang L, Hao M, Zhou J, Huang R, Ma Y, Hu K, Liu Z. Heterogeneous associations of multiplexed environmental factors and multidimensional aging metrics. Nat Commun 2024; 15:4921. [PMID: 38858361 PMCID: PMC11164970 DOI: 10.1038/s41467-024-49283-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/31/2024] [Indexed: 06/12/2024] Open
Abstract
Complicated associations between multiplexed environmental factors and aging are poorly understood. We manipulated aging using multidimensional metrics such as phenotypic age, brain age, and brain volumes in the UK Biobank. Weighted quantile sum regression was used to examine the relative individual contributions of multiplexed environmental factors to aging, and self-organizing maps (SOMs) were used to examine joint effects. Air pollution presented a relatively large contribution in most cases. We also found fair heterogeneities in which the same environmental factor contributed inconsistently to different aging metrics. Particulate matter contributed the most to variance in aging, while noise and green space showed considerable contribution to brain volumes. SOM identified five subpopulations with distinct environmental exposure patterns and the air pollution subpopulation had the worst aging status. This study reveals the heterogeneous associations of multiplexed environmental factors with multidimensional aging metrics and serves as a proof of concept when analyzing multifactors and multiple outcomes.
Collapse
Affiliation(s)
- Fan Pu
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Weiran Chen
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Chenxi Li
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Jingqiao Fu
- Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Weijing Gao
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Chao Ma
- School of Economics and Management, Southeast University, Nanjing, 211189, Jiangsu, China
| | - Xingqi Cao
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Lingzhi Zhang
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Meng Hao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Jin Zhou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University; Department of Biostatistics and Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Rong Huang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University; Department of Biostatistics and Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yanan Ma
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University; Department of Biostatistics and Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Kejia Hu
- Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| | - Zuyun Liu
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
106
|
Wang M, Wei M, Wang L, Song J, Rominger A, Shi K, Jiang J. Tau Protein Accumulation Trajectory-Based Brain Age Prediction in the Alzheimer's Disease Continuum. Brain Sci 2024; 14:575. [PMID: 38928575 PMCID: PMC11201453 DOI: 10.3390/brainsci14060575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
Clinical cognitive advancement within the Alzheimer's disease (AD) continuum is intimately connected with sustained accumulation of tau protein pathology. The biological brain age and its gap show great potential for pathological risk and disease severity. In the present study, we applied multivariable linear support vector regression to train a normative brain age prediction model using tau brain images. We further assessed the predicted biological brain age and its gap for patients within the AD continuum. In the AD continuum, evaluated pathologic tau binding was found in the inferior temporal, parietal-temporal junction, precuneus/posterior cingulate, dorsal frontal, occipital, and inferior-medial temporal cortices. The biological brain age gaps of patients within the AD continuum were notably higher than those of the normal controls (p < 0.0001). Significant positive correlations were observed between the brain age gap and global tau protein accumulation levels for mild cognitive impairment (r = 0.726, p < 0.001), AD (r = 0.845, p < 0.001), and AD continuum (r = 0.797, p < 0.001). The pathologic tau-based age gap was significantly linked to neuropsychological scores. The proposed pathologic tau-based biological brain age model could track the tau protein accumulation trajectory of cognitive impairment and further provide a comprehensive quantification index for the tau accumulation risk.
Collapse
Affiliation(s)
- Min Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Min Wei
- Department of Neurology, XuanWu Hospital of Capital Medical University, Beijing 100053, China
| | - Luyao Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jun Song
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Computer Aided Medical Procedures, School of Computation, Information and Technology, Technical University of Munich, 85748 Munich, Germany
| | - Jiehui Jiang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
107
|
Jasper AA, Shah KH, Karim H, Gujral S, Miljkovic I, Rosano C, Barchowsky A, Sahu A. Regenerative rehabilitation measures to restore tissue function after arsenic exposure. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2024; 30:100529. [PMID: 40191583 PMCID: PMC11970924 DOI: 10.1016/j.cobme.2024.100529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Environmental exposure of arsenic impairs the cardiometabolic profile, skeletal muscle health, and neurological function. Such declining tissue health is observed as early as in one's childhood, where the exposure is prevalent, thereby accelerating the effect of time's arrow. Despite the known deleterious effects of arsenic exposure, there is a paucity of specific treatment plans for restoring tissue function in exposed individuals. In this review, we propose to harness the untapped potential of existing regenerative rehabilitation programs, such as stem cell therapeutics with rehabilitation, acellular therapeutics, and artificial intelligence/robotics technologies, to address this critical gap in environmental toxicology. With regenerative rehabilitation techniques showing promise in other injury paradigms, fostering collaboration between these scientific realms offers an effective means of mitigating the detrimental effects of arsenic on tissue function.
Collapse
Affiliation(s)
- Adam A Jasper
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - Kush H Shah
- The Lake Erie College of Osteopathic Medicine (LECOM), Erie, PA, USA
| | - Helmet Karim
- Department of Psychiatry, University of Pittsburgh, USA
- Department of Bioengineering, University of Pittsburgh, USA
| | - Swathi Gujral
- Department of Psychiatry, University of Pittsburgh, USA
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh, USA
| | | | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| |
Collapse
|
108
|
Li J, Zhang C, Meng Y, Yang S, Xia J, Chen H, Liao W. Morphometric brain organization across the human lifespan reveals increased dispersion linked to cognitive performance. PLoS Biol 2024; 22:e3002647. [PMID: 38900742 PMCID: PMC11189252 DOI: 10.1371/journal.pbio.3002647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 04/26/2024] [Indexed: 06/22/2024] Open
Abstract
The human brain is organized as segregation and integration units and follows complex developmental trajectories throughout life. The cortical manifold provides a new means of studying the brain's organization in a multidimensional connectivity gradient space. However, how the brain's morphometric organization changes across the human lifespan remains unclear. Here, leveraging structural magnetic resonance imaging scans from 1,790 healthy individuals aged 8 to 89 years, we investigated age-related global, within- and between-network dispersions to reveal the segregation and integration of brain networks from 3D manifolds based on morphometric similarity network (MSN), combining multiple features conceptualized as a "fingerprint" of an individual's brain. Developmental trajectories of global dispersion unfolded along patterns of molecular brain organization, such as acetylcholine receptor. Communities were increasingly dispersed with age, reflecting more disassortative morphometric similarity profiles within a community. Increasing within-network dispersion of primary motor and association cortices mediated the influence of age on the cognitive flexibility of executive functions. We also found that the secondary sensory cortices were decreasingly dispersed with the rest of the cortices during aging, possibly indicating a shift of secondary sensory cortices across the human lifespan from an extreme to a more central position in 3D manifolds. Together, our results reveal the age-related segregation and integration of MSN from the perspective of a multidimensional gradient space, providing new insights into lifespan changes in multiple morphometric features of the brain, as well as the influence of such changes on cognitive performance.
Collapse
Affiliation(s)
- Jiao Li
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Chao Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Yao Meng
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Siqi Yang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Xia
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Huafu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Liao
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
109
|
Lozada‐Martinez ID, Lozada‐Martinez LM, Anaya J. Gut microbiota in centenarians: A potential metabolic and aging regulator in the study of extreme longevity. Aging Med (Milton) 2024; 7:406-413. [PMID: 38975304 PMCID: PMC11222757 DOI: 10.1002/agm2.12336] [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: 04/12/2024] [Revised: 04/30/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024] Open
Abstract
Centenarians, those aged 100 years or older, are considered the most successful biological aging model in humans. This population is commonly characterized by a low prevalence of chronic diseases, with favorable maintenance of functionality and independence, thus determining a health phenotype of successful aging. There are many factors usually associated with extreme longevity: genetics, lifestyles, diet, among others. However, it is most likely a multifactorial condition where protective factors contribute individually to some extent. The gut microbiota (GM) has emerged as a potential factor associated with the establishment of a favorable health phenotype that allows for extreme longevity, as seen in centenarians. To understand the possible impact generated by the GM, its changes, and the probable causes for successful aging, the aim of this review was to synthesize evidence on the role of the GM as a potential protective factor for achieving extreme longevity, using its relationship with centenarians.
Collapse
Affiliation(s)
- Ivan David Lozada‐Martinez
- Health Research and Innovation Center at Coosalud EPSCartagenaColombia
- Universidad de la CostaBarranquillaColombia
| | | | - Juan‐Manuel Anaya
- Health Research and Innovation Center at Coosalud EPSCartagenaColombia
- Universidad de la CostaBarranquillaColombia
| |
Collapse
|
110
|
Yoo J, Hur J, Yoo J, Jurivich D, Lee KJ. A novel approach to quantifying individual's biological aging using Korea's national health screening program toward precision public health. GeroScience 2024; 46:3387-3403. [PMID: 38302843 PMCID: PMC11009216 DOI: 10.1007/s11357-024-01079-2] [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] [Received: 07/29/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
Accurate prediction of biological age can inform public health measures to extend healthy lifespans and reduce chronic conditions. Multiple theoretical models and methods have been developed; however, their applicability and accuracy are still not extensive. Here, we report Differential Aging and Health Index (DAnHI), a novel measure of age deviation, developed using physical and serum biomarkers from four million individuals in Korea's National Health Screening Program. Participants were grouped into aging statuses (< 26 vs. ≥ 26, < 27 vs. ≥ 27, …, < 75 vs. ≥ 75 years) as response variables in a binary logistic regression model with thirteen biomarkers as independent variables. DAnHI for each individual was calculated as the weighted mean of their relative probabilities of being classified into each older age status, based on model ages ranging from 26 to 75. DAnHI in our large study population showed a steady increase with the increase in age and was positively associated with death after adjusting for chronological age. However, the effect size of DAnHI on the risk of death varied according to the age group and sex. The hazard ratio was highest in the 50-59-year age group and then decreased as the individuals aged. This study demonstrates that routine health check-up biomarkers can be integrated into a quantitative measure for predicting aging-related health status and death via appropriate statistical models and methodology. Our DAnHI-based results suggest that the same level of aging-related health status does not indicate the same degree of risk for death.
Collapse
Affiliation(s)
- Jinho Yoo
- YooJin BioSoft, 24, Jeongbalsan-Ro Ilsandong-Gu, Goyang-Si Gyeonggi-Do, 10402, Korea
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Jintae Yoo
- YooJin BioSoft, 24, Jeongbalsan-Ro Ilsandong-Gu, Goyang-Si Gyeonggi-Do, 10402, Korea
| | - Donald Jurivich
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Kyung Ju Lee
- Department of Women's Rehabilitation, National Rehabilitation Center, 58, Samgaksan-Ro, Gangbuk-Gu, Seoul, 01022, Korea.
- Institute for Occupational & Environmental Health, Korea University, Seoul, 02841, Korea.
| |
Collapse
|
111
|
Sandalova E, Maier AB. Targeting the epigenetically older individuals for geroprotective trials: the use of DNA methylation clocks. Biogerontology 2024; 25:423-431. [PMID: 37968337 DOI: 10.1007/s10522-023-10077-4] [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: 09/08/2023] [Accepted: 10/15/2023] [Indexed: 11/17/2023]
Abstract
Chronological age is the most important risk factor for the incidence of age-related diseases. The pace of ageing determines the magnitude of that risk and can be expressed as biological age. Targeting fundamental pathways of human aging with geroprotectors has the potential to lower the biological age and therewith prolong the healthspan, the period of life one spends in good health. Target populations for geroprotective interventions should be chosen based on the ageing mechanisms being addressed and the expected effect of the geroprotector on the primary outcome. Biomarkers of ageing, such as DNA methylation age, can be used to select populations for geroprotective interventions and as a surrogate outcome. Here, the use of DNA methylation clocks for selecting target populations for geroprotective intervention is explored.
Collapse
Affiliation(s)
- Elena Sandalova
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore.
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore.
- Department of Human Movement Sciences, @AgeAmsterdam, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
112
|
Dashtmian AR, Darvishi FB, Arnold WD. Chronological and Biological Aging in Amyotrophic Lateral Sclerosis and the Potential of Senolytic Therapies. Cells 2024; 13:928. [PMID: 38891059 PMCID: PMC11171952 DOI: 10.3390/cells13110928] [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: 04/01/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a group of sporadic and genetic neurodegenerative disorders that result in losses of upper and lower motor neurons. Treatment of ALS is limited, and survival is 2-5 years after disease onset. While ALS can occur in younger individuals, the risk significantly increases with advancing age. Notably, both sporadic and genetic forms of ALS share pathophysiological features overlapping hallmarks of aging including genome instability/DNA damage, mitochondrial dysfunction, inflammation, proteostasis, and cellular senescence. This review explores chronological and biological aging in the context of ALS onset and progression. Age-related muscle weakness and motor unit loss mirror aspects of ALS pathology and coincide with peak ALS incidence, suggesting a potential link between aging and disease development. Hallmarks of biological aging, including DNA damage, mitochondrial dysfunction, and cellular senescence, are implicated in both aging and ALS, offering insights into shared mechanisms underlying disease pathogenesis. Furthermore, senescence-associated secretory phenotype and senolytic treatments emerge as promising avenues for ALS intervention, with the potential to mitigate neuroinflammation and modify disease progression.
Collapse
Affiliation(s)
- Anna Roshani Dashtmian
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA; (A.R.D.); (F.B.D.)
- NextGen Precision Health, Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, MO 65211, USA
| | - Fereshteh B. Darvishi
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA; (A.R.D.); (F.B.D.)
- NextGen Precision Health, Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, MO 65211, USA
| | - William David Arnold
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA; (A.R.D.); (F.B.D.)
- NextGen Precision Health, Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
113
|
Huang Y, Peng H, Wu Y, Deng S, Ge F, Ma W, Zhou X, Songyang Z. Rosa roxburghii Fruit Extracts Upregulate Telomerase Activity and Ameliorate Cell Replicative Senescence. Foods 2024; 13:1673. [PMID: 38890904 PMCID: PMC11171777 DOI: 10.3390/foods13111673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
Anti-aging functional foods benefit the elderly. Telomeres are chromosomal ends that maintain genome stability extended by telomerase catalytic subunit TERT. Due to the end-replication problem, telomeres shorten after each cell cycle without telomerase in most human cells, and eventually the cell enters the senescence stage. Natural products can attenuate the aging process by increasing telomerase activity, such as TA-65. However, TA-65 is expensive. Other Chinese natural products may achieve comparable effects. Here, we found that Rosa roxburghii fruit extracts effectively increase TERT expression and telomerase activity in cultured human mesenchymal stem cells. Both R. roxburghii fruit extracts obtained by freeze-drying and spray-drying increased the activity of telomerase. R. roxburghii fruit extracts were able to reduce reactive oxygen species levels, enhance superoxide dismutase activity, and reduce DNA damage caused by oxidative stress or radiation. R. roxburghii fruit extracts promoted cell proliferation, improved senescent cell morphology, delayed replicative cellular senescence, attenuated cell cycle suppressors, and alleviated the senescence-associated secretory phenotype. Transcriptome and metabolic profiling revealed that R. roxburghii fruit extracts promote DNA replication and telomere maintenance pathways and decrease triglyceride levels. Overall, we provide a theoretical basis for the application of R. roxburghii fruit as an anti-aging product.
Collapse
Affiliation(s)
- Yan Huang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (H.P.); (Y.W.); (S.D.); (W.M.); (Z.S.)
| | - Haoyue Peng
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (H.P.); (Y.W.); (S.D.); (W.M.); (Z.S.)
| | - Yifan Wu
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (H.P.); (Y.W.); (S.D.); (W.M.); (Z.S.)
| | - Shengcheng Deng
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (H.P.); (Y.W.); (S.D.); (W.M.); (Z.S.)
| | - Fahuan Ge
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China;
| | - Wenbin Ma
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (H.P.); (Y.W.); (S.D.); (W.M.); (Z.S.)
| | - Xue Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China;
| | - Zhou Songyang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (H.P.); (Y.W.); (S.D.); (W.M.); (Z.S.)
| |
Collapse
|
114
|
Jiang R, Noble S, Rosenblatt M, Dai W, Ye J, Liu S, Qi S, Calhoun VD, Sui J, Scheinost D. The brain structure, inflammatory, and genetic mechanisms mediate the association between physical frailty and depression. Nat Commun 2024; 15:4411. [PMID: 38782943 PMCID: PMC11116547 DOI: 10.1038/s41467-024-48827-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Cross-sectional studies have demonstrated strong associations between physical frailty and depression. However, the evidence from prospective studies is limited. Here, we analyze data of 352,277 participants from UK Biobank with 12.25-year follow-up. Compared with non-frail individuals, pre-frail and frail individuals have increased risk for incident depression independent of many putative confounds. Altogether, pre-frail and frail individuals account for 20.58% and 13.16% of depression cases by population attributable fraction analyses. Higher risks are observed in males and individuals younger than 65 years than their counterparts. Mendelian randomization analyses support a potential causal effect of frailty on depression. Associations are also observed between inflammatory markers, brain volumes, and incident depression. Moreover, these regional brain volumes and three inflammatory markers-C-reactive protein, neutrophils, and leukocytes-significantly mediate associations between frailty and depression. Given the scarcity of curative treatment for depression and the high disease burden, identifying potential modifiable risk factors of depression, such as frailty, is needed.
Collapse
Affiliation(s)
- Rongtao Jiang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06510, USA.
| | - Stephanie Noble
- Department of Psychology, Northeastern University, Boston, MA, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Center for Cognitive and Brain Health, Northeastern University, Boston, MA, USA
| | - Matthew Rosenblatt
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Wei Dai
- Department of Biostatistics, Yale University, New Haven, CT, 06520, USA
| | - Jean Ye
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06520, USA
| | - Shu Liu
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Shile Qi
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, GA, 30303, USA
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, GA, 30303, USA
| | - Jing Sui
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China.
| | - Dustin Scheinost
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06520, USA
- Department of Statistics & Data Science, Yale University, New Haven, CT, 06520, USA
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA
| |
Collapse
|
115
|
Hishikawa A, Nishimura ES, Yoshimoto N, Nakamichi R, Hama EY, Ito W, Maruki T, Nagashima K, Shimizu-Hirota R, Takaishi H, Itoh H, Hayashi K. Predicting exacerbation of renal function by DNA methylation clock and DNA damage of urinary shedding cells: a pilot study. Sci Rep 2024; 14:11530. [PMID: 38773208 PMCID: PMC11109093 DOI: 10.1038/s41598-024-62405-4] [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: 10/31/2023] [Accepted: 05/16/2024] [Indexed: 05/23/2024] Open
Abstract
Recent reports have shown the feasibility of measuring biological age from DNA methylation levels in blood cells from specific regions identified by machine learning, collectively known as the epigenetic clock or DNA methylation clock. While extensive research has explored the association of the DNA methylation clock with cardiovascular diseases, cancer, and Alzheimer's disease, its relationship with kidney diseases remains largely unexplored. In particular, it is unclear whether the DNA methylation clock could serve as a predictor of worsening kidney function. In this pilot study involving 20 subjects, we investigated the association between the DNA methylation clock and subsequent deterioration of renal function. Additionally, we noninvasively evaluated DNA damage in urinary shedding cells using a previously reported method to examine the correlation with the DNA methylation clock and worsening kidney function. Our findings revealed that patients with an accelerated DNA methylation clock exhibited increased DNA damage in urinary shedding cells, along with a higher rate of eGFR decline. Moreover, in cases of advanced CKD (G4-5), the DNA damage in urinary shedding cells was significantly increased, highlighting the interplay between elevated DNA damage and eGFR decline. This study suggests the potential role of the DNA methylation clock and urinary DNA damage as predictive markers for the progression of chronic kidney disease.
Collapse
Affiliation(s)
- Akihito Hishikawa
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Erina Sugita Nishimura
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Norifumi Yoshimoto
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Ran Nakamichi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Eriko Yoshida Hama
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Wataru Ito
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Tomomi Maruki
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Kengo Nagashima
- Biostatistics Unit, Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan
| | - Ryoko Shimizu-Hirota
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiromasa Takaishi
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kaori Hayashi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
116
|
Al-Hassinah S, Al-Daihan S, Alahmadi M, Alghamdi S, Almulhim R, Obeid D, Arabi Y, Alswaji A, Aldriwesh M, Alghoribi M. Interplay of Demographic Influences, Clinical Manifestations, and Longitudinal Profile of Laboratory Parameters in the Progression of SARS-CoV-2 Infection: Insights from the Saudi Population. Microorganisms 2024; 12:1022. [PMID: 38792852 PMCID: PMC11124088 DOI: 10.3390/microorganisms12051022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Understanding the factors driving SARS-CoV-2 infection progression and severity is complex due to the dynamic nature of human physiology. Therefore, we aimed to explore the severity risk indicators of SARS-CoV-2 through demographic data, clinical manifestations, and the profile of laboratory parameters. The study included 175 patients either hospitalized at King Abdulaziz Medical City-Riyadh or placed in quarantine at designated hotels in Riyadh, Saudi Arabia, from June 2020 to April 2021. Hospitalized patients were followed up through the first week of admission. Demographic data, clinical presentations, and laboratory results were retrieved from electronic patient records. Our results revealed that older age (OR: 1.1, CI: [1.1-1.12]; p < 0.0001), male gender (OR: 2.26, CI: [1.0-5.1]; p = 0.047), and blood urea nitrogen level (OR: 2.56, CI: [1.07-6.12]; p = 0.034) were potential predictors of severity level. In conclusion, the study showed that apart from laboratory parameters, age and gender could potentially predict the severity of SARS-CoV-2 infection in the early stages. To our knowledge, this study is the first in Saudi Arabia to explore the longitudinal profile of laboratory parameters among risk factors, shedding light on SARS-CoV-2 infection progression parameters.
Collapse
Affiliation(s)
- Sarah Al-Hassinah
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Sooad Al-Daihan
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Mashael Alahmadi
- Research Office, Saudi National Institute of Health (SNIH), Riyadh 12382, Saudi Arabia;
| | - Sara Alghamdi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
| | - Rawabi Almulhim
- Infection Prevention and Control Department, King Abdulaziz Medical City, Riyadh 14611, Saudi Arabia;
| | - Dalia Obeid
- King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia;
| | - Yaseen Arabi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Intensive Care Department, King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Abdulrahman Alswaji
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
| | - Marwh Aldriwesh
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Majed Alghoribi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Department of Basic Science, College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| |
Collapse
|
117
|
Anazodo UC, Plessis SD. Imaging without barriers. Science 2024; 384:623-624. [PMID: 38723100 DOI: 10.1126/science.adp0670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Low-field magnetic resonance imaging can be engineered for widespread point-of-care diagnostics.
Collapse
Affiliation(s)
- Udunna C Anazodo
- McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Insitute, McGill University, Montreal, QC, Canada
- Medical Artificial Intelligence Laboratory, Crestview Radiology Ltd., Lagos, Nigeria
| | - Stefan du Plessis
- Department of Psychiatry, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
118
|
Liang W, Wei T, Hu L, Chen M, Tong L, Zhou W, Duan X, Zhao X, Zhou W, Jiang Q, Xiao G, Zou W, Chen D, Zou Z, Bai X. An integrated multi-omics analysis reveals osteokines involved in global regulation. Cell Metab 2024; 36:1144-1163.e7. [PMID: 38574738 DOI: 10.1016/j.cmet.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/22/2024] [Accepted: 03/10/2024] [Indexed: 04/06/2024]
Abstract
Bone secretory proteins, termed osteokines, regulate bone metabolism and whole-body homeostasis. However, fundamental questions as to what the bona fide osteokines and their cellular sources are and how they are regulated remain unclear. In this study, we analyzed bone and extraskeletal tissues, osteoblast (OB) conditioned media, bone marrow supernatant (BMS), and serum, for basal osteokines and those responsive to aging and mechanical loading/unloading. We identified 375 candidate osteokines and their changes in response to aging and mechanical dynamics by integrating data from RNA-seq, scRNA-seq, and proteomic approaches. Furthermore, we analyzed their cellular sources in the bone and inter-organ communication facilitated by them (bone-brain, liver, and aorta). Notably, we discovered that senescent OBs secrete fatty-acid-binding protein 3 to propagate senescence toward vascular smooth muscle cells (VSMCs). Taken together, we identified previously unknown candidate osteokines and established a dynamic regulatory network among them, thus providing valuable resources to further investigate their systemic roles.
Collapse
Affiliation(s)
- Wenquan Liang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tiantian Wei
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Le Hu
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Meijun Chen
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Liping Tong
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wu Zhou
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xingwei Duan
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyang Zhao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weijie Zhou
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Di Chen
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, China.
| | - Zhipeng Zou
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Academy of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510630, China.
| |
Collapse
|
119
|
Goyal P, Maurer MS, Roh J. Aging in Heart Failure: Embracing Biology Over Chronology: JACC Family Series. JACC. HEART FAILURE 2024; 12:795-809. [PMID: 38597865 PMCID: PMC11331491 DOI: 10.1016/j.jchf.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 04/11/2024]
Abstract
Age is among the most potent risk factors for developing heart failure and is strongly associated with adverse outcomes. As the global population continues to age and the prevalence of heart failure rises, understanding the role of aging in the development and progression of this chronic disease is essential. Although chronologic age is on a fixed course, biological aging is more variable and potentially modifiable in patients with heart failure. This review describes the current knowledge on mechanisms of biological aging that contribute to the pathogenesis of heart failure. The discussion focuses on 3 hallmarks of aging-impaired proteostasis, mitochondrial dysfunction, and deregulated nutrient sensing-that are currently being targeted in therapeutic development for older adults with heart failure. In assessing existing and emerging therapeutic strategies, the review also enumerates the importance of incorporating geriatric conditions into the management of older adults with heart failure and in ongoing clinical trials.
Collapse
Affiliation(s)
- Parag Goyal
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Mathew S Maurer
- Department of Medicine, Columbia University Medical Center, New York, New York, USA.
| | - Jason Roh
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
120
|
Hama EY, Nakamichi R, Hishikawa A, Kihara M, Abe T, Yoshimoto N, Nishimura ES, Itoh H, Hayashi K. Podocyte Ercc1 is indispensable for glomerular integrity. Biochem Biophys Res Commun 2024; 704:149713. [PMID: 38428304 DOI: 10.1016/j.bbrc.2024.149713] [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: 02/01/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
As life expectancy continues to increase, age-related kidney diseases are becoming more prevalent. Chronic kidney disease (CKD) is not only a consequence of aging but also a potential accelerator of aging process. Here we report the pivotal role of podocyte ERCC1, a DNA repair factor, in maintaining glomerular integrity and a potential effect on multiple organs. Podocyte-specific ERCC1-knockout mice developed severe proteinuria, glomerulosclerosis, and renal failure, accompanied by a significant increase in glomerular DNA single-strand breaks (SSBs) and double-strand breaks (DSBs). ERCC1 gene transfer experiment in the knockout mice attenuated proteinuria and glomerulosclerosis with reduced DNA damage. Notably, CD44+CD8+ memory T cells, indicative of T-cell senescence, were already elevated in the peripheral blood of knockout mice at 10 weeks old. Additionally, levels of senescence-associated secretory phenotype (SASP) factors were significantly increased in both the circulation and multiple organs of the knockout mice. In older mice and human patients, we observed an accumulation of DSBs and an even greater buildup of SSBs in glomeruli, despite no significant reduction in ERCC1 expression with age in mice. Collectively, our findings highlight the crucial role of ERCC1 in repairing podocyte DNA damage, with potential implications for inflammation in various organs.
Collapse
Affiliation(s)
- Eriko Yoshida Hama
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ran Nakamichi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihito Hishikawa
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Miho Kihara
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Hyogo, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Hyogo, Japan
| | - Norifumi Yoshimoto
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Erina Sugita Nishimura
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Center for Preventive Medicine, School of Medicine, Keio University, Japan
| | - Kaori Hayashi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
121
|
Beck D, de Lange AG, Gurholt TP, Voldsbekk I, Maximov II, Subramaniapillai S, Schindler L, Hindley G, Leonardsen EH, Rahman Z, van der Meer D, Korbmacher M, Linge J, Leinhard OD, Kalleberg KT, Engvig A, Sønderby I, Andreassen OA, Westlye LT. Dissecting unique and common variance across body and brain health indicators using age prediction. Hum Brain Mapp 2024; 45:e26685. [PMID: 38647042 PMCID: PMC11034003 DOI: 10.1002/hbm.26685] [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: 12/29/2023] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Ageing is a heterogeneous multisystem process involving different rates of decline in physiological integrity across biological systems. The current study dissects the unique and common variance across body and brain health indicators and parses inter-individual heterogeneity in the multisystem ageing process. Using machine-learning regression models on the UK Biobank data set (N = 32,593, age range 44.6-82.3, mean age 64.1 years), we first estimated tissue-specific brain age for white and gray matter based on diffusion and T1-weighted magnetic resonance imaging (MRI) data, respectively. Next, bodily health traits, including cardiometabolic, anthropometric, and body composition measures of adipose and muscle tissue from bioimpedance and body MRI, were combined to predict 'body age'. The results showed that the body age model demonstrated comparable age prediction accuracy to models trained solely on brain MRI data. The correlation between body age and brain age predictions was 0.62 for the T1 and 0.64 for the diffusion-based model, indicating a degree of unique variance in brain and bodily ageing processes. Bayesian multilevel modelling carried out to quantify the associations between health traits and predicted age discrepancies showed that higher systolic blood pressure and higher muscle-fat infiltration were related to older-appearing body age compared to brain age. Conversely, higher hand-grip strength and muscle volume were related to a younger-appearing body age. Our findings corroborate the common notion of a close connection between somatic and brain health. However, they also suggest that health traits may differentially influence age predictions beyond what is captured by the brain imaging data, potentially contributing to heterogeneous ageing rates across biological systems and individuals.
Collapse
Affiliation(s)
- Dani Beck
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of Mental Health and Substance AbuseDiakonhjemmet HospitalOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
| | - Ann‐Marie G. de Lange
- Department of PsychologyUniversity of OsloOsloNorway
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesCHUV and University of LausanneLausanneSwitzerland
- Department of PsychiatryUniversity of OxfordOxfordUK
| | - Tiril P. Gurholt
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Irene Voldsbekk
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
| | - Ivan I. Maximov
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of Health and FunctioningWestern Norway University of Applied SciencesBergenNorway
| | - Sivaniya Subramaniapillai
- Department of PsychologyUniversity of OsloOsloNorway
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesCHUV and University of LausanneLausanneSwitzerland
| | - Louise Schindler
- Department of PsychologyUniversity of OsloOsloNorway
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesCHUV and University of LausanneLausanneSwitzerland
| | - Guy Hindley
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Esten H. Leonardsen
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
| | - Zillur Rahman
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Dennis van der Meer
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Max Korbmacher
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of Health and FunctioningWestern Norway University of Applied SciencesBergenNorway
| | - Jennifer Linge
- AMRA Medical ABLinköpingSweden
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring SciencesLinköping UniversityLinköpingSweden
| | - Olof D. Leinhard
- AMRA Medical ABLinköpingSweden
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring SciencesLinköping UniversityLinköpingSweden
| | | | - Andreas Engvig
- Department of Endocrinology, Obesity and Preventive Medicine, Section of Preventive CardiologyOslo University HospitalOsloNorway
| | - Ida Sønderby
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of Medical GeneticsOslo University HospitalOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of Oslo
| | - Ole A. Andreassen
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of Oslo
| | - Lars T. Westlye
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of Oslo
| |
Collapse
|
122
|
Fruscione S, Malta G, Verso MG, Calascibetta A, Martorana D, Cannizzaro E. Correlation among job-induced stress, overall well-being, and cardiovascular risk in Italian workers of logistics and distribution. Front Public Health 2024; 12:1358212. [PMID: 38655515 PMCID: PMC11035898 DOI: 10.3389/fpubh.2024.1358212] [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: 12/19/2023] [Accepted: 03/04/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Work-related stress is an occupational risk that has been linked to the development of cardiovascular disease (CVD). While previous studies have explored this association in various work contexts, none have focused specifically on logistics and distribution personnel. These workers may be exposed to significant job stress, which potentially increases the risk of CVD. Methods In this study, we aimed to examine the relationship between work-related stress and cardiovascular risk in a sample of 413 healthy workers of a logistics and distribution company. To assess work-related stress and cardiovascular risk, we used the organisational well-being questionnaire proposed by the Italian National Anti-Corruption Authority, the Framingham Heart Study General Cardiovascular Disease (CVD) Risk Prediction Score and the WHO General Wellbeing Index (WHO-5). Results Our results revealed that individuals with low job support had a significantly higher CVD risk score and lower well-being index than those reporting high job support. Furthermore, workers with high-stress tasks showed higher well-being index scores than those with passive tasks. Approximately 58% of the subjects were classified as low CVD risk (CVD risk <10%), approximately 31% were classified as moderate risk (CVD risk between 10 and 20%) and 11% were considered high risk (CVD risk >20%). The overall median CVD risk for the population was moderate (6.9%), with individual scores ranging from 1 to 58%. Discussion Further analyses confirmed the protective effect of work support, also identifying physical inactivity, regular alcohol consumption and low educational level as factors contributing to an increased risk of CVD. Interestingly, factors such as job control and work support demonstrated a positive impact on psychological well-being. These results emphasise the importance of intervention strategies aimed at promoting health in the workplace. By addressing these combined factors, organisations can effectively reduce the risk of CVD and improve the general well-being of their workforce.
Collapse
Affiliation(s)
| | - Ginevra Malta
- PROMISE Department, University of Palermo, Palermo, Italy
| | | | | | - Daniela Martorana
- Department of Orthopedics, Hospital Company ‘Ospedali Riuniti Villa Sofia-Cervello’, Palermo, Italy
| | | |
Collapse
|
123
|
Willows JW, Alshahal Z, Story NM, Alves MJ, Vidal P, Harris H, Rodrigo R, Stanford KI, Peng J, Reifsnyder PC, Harrison DE, David Arnold W, Townsend KL. Contributions of mouse genetic strain background to age-related phenotypes in physically active HET3 mice. Neurobiol Aging 2024; 136:58-69. [PMID: 38325031 DOI: 10.1016/j.neurobiolaging.2024.01.010] [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: 10/01/2023] [Revised: 01/06/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024]
Abstract
We assessed aging hallmarks in skin, muscle, and adipose in the genetically diverse HET3 mouse, and generated a broad dataset comparing these to individual animal diagnostic SNPs from the 4 founding inbred strains of the HET3 line. For middle- and old-aged HET3 mice, we provided running wheel exercise to ensure our observations were not purely representative of sedentary animals, but age-related phenotypes were not improved with running wheel activity. Adipose tissue fibrosis, peripheral neuropathy, and loss of neuromuscular junction integrity were consistent phenotypes in older-aged HET3 mice regardless of physical activity, but aspects of these phenotypes were moderated by the SNP% contributions of the founding strains for the HET3 line. Taken together, the genetic contribution of founder strain SNPs moderated age-related phenotypes in skin and muscle innervation and were dependent on biological sex and chronological age. However, there was not a single founder strain (BALB/cJ, C57BL/6J, C3H/HeJ, DBA/2J) that appeared to drive more protection or disease-risk across aging in this mouse line, but genetic diversity in general was more protective.
Collapse
Affiliation(s)
- Jake W Willows
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Zahra Alshahal
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Naeemah M Story
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Michele J Alves
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Pablo Vidal
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Hallie Harris
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Rochelle Rodrigo
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Juan Peng
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | | | | | - W David Arnold
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
124
|
Whitman ET, Ryan CP, Abraham WC, Addae A, Corcoran DL, Elliott ML, Hogan S, Ireland D, Keenan R, Knodt AR, Melzer TR, Poulton R, Ramrakha S, Sugden K, Williams BS, Zhou J, Hariri AR, Belsky DW, Moffitt TE, Caspi A. A blood biomarker of the pace of aging is associated with brain structure: replication across three cohorts. Neurobiol Aging 2024; 136:23-33. [PMID: 38301452 PMCID: PMC11017787 DOI: 10.1016/j.neurobiolaging.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/03/2024]
Abstract
Biological aging is the correlated decline of multi-organ system integrity central to the etiology of many age-related diseases. A novel epigenetic measure of biological aging, DunedinPACE, is associated with cognitive dysfunction, incident dementia, and mortality. Here, we tested for associations between DunedinPACE and structural MRI phenotypes in three datasets spanning midlife to advanced age: the Dunedin Study (age=45 years), the Framingham Heart Study Offspring Cohort (mean age=63 years), and the Alzheimer's Disease Neuroimaging Initiative (mean age=75 years). We also tested four additional epigenetic measures of aging: the Horvath clock, the Hannum clock, PhenoAge, and GrimAge. Across all datasets (total N observations=3380; total N individuals=2322), faster DunedinPACE was associated with lower total brain volume, lower hippocampal volume, greater burden of white matter microlesions, and thinner cortex. Across all measures, DunedinPACE and GrimAge had the strongest and most consistent associations with brain phenotypes. Our findings suggest that single timepoint measures of multi-organ decline such as DunedinPACE could be useful for gauging nervous system health.
Collapse
Affiliation(s)
- Ethan T Whitman
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA.
| | - Calen P Ryan
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, USA
| | | | - Angela Addae
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - David L Corcoran
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maxwell L Elliott
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Sean Hogan
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - David Ireland
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Ross Keenan
- Brain Research New Zealand-Rangahau Roro Aotearoa, Centre of Research Excellence, Universities of Auckland and Otago, New Zealand; Christchurch Radiology Group, Christchurch, New Zealand
| | - Annchen R Knodt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Tracy R Melzer
- Brain Research New Zealand-Rangahau Roro Aotearoa, Centre of Research Excellence, Universities of Auckland and Otago, New Zealand; Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Richie Poulton
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Sandhya Ramrakha
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | | | - Jiayi Zhou
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, USA
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Daniel W Belsky
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, USA; Department of Epidemiology, Columbia University Mailman School of Public Health, New York, USA
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Center for Genomic and Computational Biology, Duke University, Durham, NC, USA; King's College London, Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, London, UK; PROMENTA, Department of Psychology, University of Oslo, Norway; Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Center for Genomic and Computational Biology, Duke University, Durham, NC, USA; King's College London, Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, London, UK; PROMENTA, Department of Psychology, University of Oslo, Norway; Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| |
Collapse
|
125
|
Prattichizzo F, Frigé C, Pellegrini V, Scisciola L, Santoro A, Monti D, Rippo MR, Ivanchenko M, Olivieri F, Franceschi C. Organ-specific biological clocks: Ageotyping for personalized anti-aging medicine. Ageing Res Rev 2024; 96:102253. [PMID: 38447609 DOI: 10.1016/j.arr.2024.102253] [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: 12/12/2023] [Revised: 02/11/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
Aging is a complex multidimensional, progressive remodeling process affecting multiple organ systems. While many studies have focused on studying aging across multiple organs, assessment of the contribution of individual organs to overall aging processes is a cutting-edge issue. An organ's biological age might influence the aging of other organs, revealing a multiorgan aging network. Recent data demonstrated a similar yet asynchronous inter-organs and inter-individuals progression of aging, thereby providing a foundation to track sources of declining health in old age. The integration of multiple omics with common clinical parameters through artificial intelligence has allowed the building of organ-specific aging clocks, which can predict the development of specific age-related diseases at high resolution. The peculiar individual aging-trajectory, referred to as ageotype, might provide a novel tool for a personalized anti-aging, preventive medicine. Here, we review data relative to biological aging clocks and omics-based data, suggesting different organ-specific aging rates. Additional research on longitudinal data, including young subjects and analyzing sex-related differences, should be encouraged to apply ageotyping analysis for preventive purposes in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Aurelia Santoro
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Daniela Monti
- Department of Experimental and Clinical, Biomedical Sciences "Mario Serio" University of Florence, Florence, Italy
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Mikhail Ivanchenko
- Institute of Information Technologies, Mathematics and Mechanics, and Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod 603022, Russia
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy.
| | - Claudio Franceschi
- Institute of Information Technologies, Mathematics and Mechanics, and Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod 603022, Russia
| |
Collapse
|
126
|
Seitz-Holland J, Haas SS, Penzel N, Reichenberg A, Pasternak O. BrainAGE, brain health, and mental disorders: A systematic review. Neurosci Biobehav Rev 2024; 159:105581. [PMID: 38354871 PMCID: PMC11119273 DOI: 10.1016/j.neubiorev.2024.105581] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
The imaging-based method of brainAGE aims to characterize an individual's vulnerability to age-related brain changes. The present study systematically reviewed brainAGE findings in neuropsychiatric conditions and discussed the potential of brainAGE as a marker for biological age. A systematic PubMed search (from inception to March 6th, 2023) identified 273 articles. The 30 included studies compared brainAGE between neuropsychiatric and healthy groups (n≥50). We presented results qualitatively and adapted a bias risk assessment questionnaire. The imaging modalities, design, and input features varied considerably between studies. While the studies found higher brainAGE in neuropsychiatric conditions (11 mild cognitive impairment/ dementia, 11 schizophrenia spectrum/ other psychotic and bipolar disorder, six depression/ anxiety, two multiple groups), the associations with clinical characteristics were mixed. While brainAGE is sensitive to group differences, limitations include the lack of diverse training samples, multi-modal studies, and external validation. Only a few studies obtained longitudinal data, and all have used algorithms built solely to predict chronological age. These limitations impede the validity of brainAGE as a biological age marker.
Collapse
Affiliation(s)
- Johanna Seitz-Holland
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Shalaila S Haas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nora Penzel
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abraham Reichenberg
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
127
|
Ibanez A, Kringelbach ML, Deco G. A synergetic turn in cognitive neuroscience of brain diseases. Trends Cogn Sci 2024; 28:319-338. [PMID: 38246816 PMCID: PMC11927795 DOI: 10.1016/j.tics.2023.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/15/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Despite significant improvements in our understanding of brain diseases, many barriers remain. Cognitive neuroscience faces four major challenges: complex structure-function associations; disease phenotype heterogeneity; the lack of transdiagnostic models; and oversimplified cognitive approaches restricted to the laboratory. Here, we propose a synergetics framework that can help to perform the necessary dimensionality reduction of complex interactions between the brain, body, and environment. The key solutions include low-dimensional spatiotemporal hierarchies for brain-structure associations, whole-brain modeling to handle phenotype diversity, model integration of shared transdiagnostic pathophysiological pathways, and naturalistic frameworks balancing experimental control and ecological validity. Creating whole-brain models with reduced manifolds combined with ecological measures can improve our understanding of brain disease and help identify novel interventions. Synergetics provides an integrated framework for future progress in clinical and cognitive neuroscience, pushing the boundaries of brain health and disease toward more mature, naturalistic approaches.
Collapse
Affiliation(s)
- Agustin Ibanez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile; Global Brain Health Institute (GBHI), University California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland; Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina; Department of Psychiatry, University of Oxford, Oxford, UK.
| | - Morten L Kringelbach
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, UK
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Institució Catalana de la Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
128
|
Dohm-Hansen S, English JA, Lavelle A, Fitzsimons CP, Lucassen PJ, Nolan YM. The 'middle-aging' brain. Trends Neurosci 2024; 47:259-272. [PMID: 38508906 DOI: 10.1016/j.tins.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/22/2024]
Abstract
Middle age has historically been an understudied period of life compared to older age, when cognitive and brain health decline are most pronounced, but the scope for intervention may be limited. However, recent research suggests that middle age could mark a shift in brain aging. We review emerging evidence on multiple levels of analysis indicating that midlife is a period defined by unique central and peripheral processes that shape future cognitive trajectories and brain health. Informed by recent developments in aging research and lifespan studies in humans and animal models, we highlight the utility of modeling non-linear changes in study samples with wide subject age ranges to distinguish life stage-specific processes from those acting linearly throughout the lifespan.
Collapse
Affiliation(s)
- Sebastian Dohm-Hansen
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; INFANT Research Centre, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jane A English
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; INFANT Research Centre, University College Cork, Cork, Ireland
| | - Aonghus Lavelle
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Carlos P Fitzsimons
- Swammerdam Institute for Life Sciences, Brain Plasticity Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Brain Plasticity Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
129
|
Wen J, Zhao B, Yang Z, Erus G, Skampardoni I, Mamourian E, Cui Y, Hwang G, Bao J, Boquet-Pujadas A, Zhou Z, Veturi Y, Ritchie MD, Shou H, Thompson PM, Shen L, Toga AW, Davatzikos C. The genetic architecture of multimodal human brain age. Nat Commun 2024; 15:2604. [PMID: 38521789 PMCID: PMC10960798 DOI: 10.1038/s41467-024-46796-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 03/06/2024] [Indexed: 03/25/2024] Open
Abstract
The complex biological mechanisms underlying human brain aging remain incompletely understood. This study investigated the genetic architecture of three brain age gaps (BAG) derived from gray matter volume (GM-BAG), white matter microstructure (WM-BAG), and functional connectivity (FC-BAG). We identified sixteen genomic loci that reached genome-wide significance (P-value < 5×10-8). A gene-drug-disease network highlighted genes linked to GM-BAG for treating neurodegenerative and neuropsychiatric disorders and WM-BAG genes for cancer therapy. GM-BAG displayed the most pronounced heritability enrichment in genetic variants within conserved regions. Oligodendrocytes and astrocytes, but not neurons, exhibited notable heritability enrichment in WM and FC-BAG, respectively. Mendelian randomization identified potential causal effects of several chronic diseases on brain aging, such as type 2 diabetes on GM-BAG and AD on WM-BAG. Our results provide insights into the genetics of human brain aging, with clinical implications for potential lifestyle and therapeutic interventions. All results are publicly available at https://labs.loni.usc.edu/medicine .
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Zhen Zhou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yogasudha Veturi
- Department of Biobehavioral Health and Statistics, Penn State University, University Park, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
130
|
Salinas-Rodríguez A, Fernández-Niño JA, Rivera-Almaraz A, Manrique-Espinoza B. Intrinsic capacity trajectories and socioeconomic inequalities in health: the contributions of wealth, education, gender, and ethnicity. Int J Equity Health 2024; 23:48. [PMID: 38462637 PMCID: PMC10926672 DOI: 10.1186/s12939-024-02136-0] [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: 08/02/2023] [Accepted: 02/25/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Life-long health inequalities exert enduring impacts and are governed by social determinants crucial for achieving healthy aging. A fundamental aspect of healthy aging, intrinsic capacity, is the primary focus of this study. Our objective is to evaluate the social inequalities connected with the trajectories of intrinsic capacity, shedding light on the impacts of socioeconomic position, gender, and ethnicity. METHODS Our dynamic cohort study was rooted in three waves (2009, 2014, 2017) of the World Health Organization's Study on Global AGEing and Adult Health in Mexico. We incorporated a nationally representative sample comprising 2722 older Mexican adults aged 50 years and over. Baseline measurements of socioeconomic position, gender, and ethnicity acted as the exposure variables. We evaluated intrinsic capacity across five domains: cognition, psychological, sensory, vitality, and locomotion. The Relative Index of Inequality and Slope Index of Inequality were used to quantify socioeconomic disparities. RESULTS We discerned three distinct intrinsic capacity trajectories: steep decline, moderate decline, and slight increase. Significant disparities based on wealth, educational level, gender, and ethnicity were observed. Older adults with higher wealth and education typically exhibited a trajectory of moderate decrease or slight increase in intrinsic capacity. In stark contrast, women and indigenous individuals were more likely to experience a steeply declining trajectory. CONCLUSIONS These findings underscore the pressing need to address social determinants, minimize gender and ethnic discrimination to ensure equal access to resources and opportunities across the lifespan. It is imperative for policies and interventions to prioritize these social determinants in order to promote healthy aging and alleviate health disparities. This approach will ensure that specific demographic groups receive customized support to sustain their intrinsic capacity during their elder years.
Collapse
Affiliation(s)
- Aaron Salinas-Rodríguez
- Center for Evaluation and Surveys Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Julián Alfredo Fernández-Niño
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Room E8532, Baltimore, MD, 21205, USA.
- Department of Public Health, Universidad del Norte, Barranquilla, Atlántico, Colombia.
| | - Ana Rivera-Almaraz
- Center for Evaluation and Surveys Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Betty Manrique-Espinoza
- Center for Evaluation and Surveys Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| |
Collapse
|
131
|
Chaudhari AS. AI in osteoarthritis: Illuminating the meandering path forward. Osteoarthritis Cartilage 2024; 32:227-228. [PMID: 38013138 DOI: 10.1016/j.joca.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/13/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023]
Affiliation(s)
- Akshay S Chaudhari
- Department of Radiology, Stanford University, 1201 Welch Road P269, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
132
|
Ribeiro BC, de Athayde Costa E Silva A, de Souza LBR, de Araújo Moraes JB, Carneiro SR, Neves LMT. Risk stratification for frailty, impairment and assessment of sleep disorders in community-dwelling older adults. Exp Gerontol 2024; 187:112370. [PMID: 38310982 DOI: 10.1016/j.exger.2024.112370] [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: 12/06/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Frailty is associated with an increased susceptibility to functional decline, impairment, hospitalization, and mortality among the older adults. However, the potential reversibility of frailty lies in identifying modifiable factors that could prevent, mitigate, or interrupt its progression. While there is a suggestion that sleep disorders may increase the risk of frailty and impairment, the risk stratification of this relationship remains inconclusive. OBJECTIVE Stratify the risk of frailty and impairment and investigate potential connections with sleep quality, excessive daytime sleepiness, and the risk of obstructive sleep apnea in older adults dwelling in the community. METHODS This was a quantitative cross-sectional investigation. Frailty risk and impairment were stratified using the Frail Non-disabled Questionnaire (for impairment) and the FRAIL Scale (for Frailty). The assessment of excessive daytime sleepiness, sleep quality, and the risk of obstructive sleep apnea involved the employment of the Epworth Sleepiness Scale, Pittsburgh Sleep Quality Index, and the STOP-BANG questionnaire, respectively. RESULTS A total of 109 older adults living in the urban area (86 %, p = 0.010), females (61 %; p = 0.030), median age 68 (64-75) years, with overweight (36 %, p < 0.010) and self-identified as belonging to other racial or ethnic categories (71 %, p < 0.010). According to the impairment assessment, 32 % of participants were classified as disable (p < 0.01). Conversely, as per the frailty evaluation, 33 % were pre-frail and 25 % were identified as frail. Additionally, a substantial proportion experienced poor sleep quality (80 %, p = 0.010), exhibited a moderate risk of obstructive sleep apnea (49 %, p < 0.010), and showed no signs of excessive daytime sleepiness (62 %, p < 0.010). There was a modest correlation between frailty and impairment with poor sleep quality (rho = 0.39; p < 0.001) and the risk of obstructive sleep apnea (rho = 0.26; p = 0.000). However, the was no significant relationship was observed between frailty and impairment and excessive daytime sleepiness (rho = 0.04; p = 0.660). Similarly, a modest correlation was observed between sleep quality (rho = 0.33; p < 0.001), the risk of obstructive sleep apnea (rho = 0.27; p = 0.001), and frailty. Conversely, no correlation was found with excessive daytime sleepiness (rho = 0.05; p = 0.590). Also, the poor sleep quality and the risk of obstructive sleep apnea explain 14 % of the risk of frailty in the population of community-dwelling older adults (r2 = 0.14; p = 0.04). CONCLUSION This study reveals a modest risk of frailty and impairment with sleep quality and the risk of obstructive sleep apnea, but not with excessive daytime sleepiness in community-dwelling older adults.
Collapse
Affiliation(s)
- Breno Caldas Ribeiro
- Graduate Program in Human Movement Sciences, Federal University of Pará, Belém, Pará, Brazil
| | | | | | | | - Saul Rassy Carneiro
- Graduate Program in Human Movement Sciences, Federal University of Pará, Belém, Pará, Brazil
| | | |
Collapse
|
133
|
Gao C, Wu X, Wang Y, Li G, Ma L, Wang C, Xie S, Chu C, Madsen KH, Hou Z, Fan L. Prior-guided individualized thalamic parcellation based on local diffusion characteristics. Hum Brain Mapp 2024; 45:e26646. [PMID: 38433705 PMCID: PMC10910286 DOI: 10.1002/hbm.26646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024] Open
Abstract
Comprising numerous subnuclei, the thalamus intricately interconnects the cortex and subcortex, orchestrating various facets of brain functions. Extracting personalized parcellation patterns for these subnuclei is crucial, as different thalamic nuclei play varying roles in cognition and serve as therapeutic targets for neuromodulation. However, accurately delineating the thalamic nuclei boundary at the individual level is challenging due to intersubject variability. In this study, we proposed a prior-guided parcellation (PG-par) method to achieve robust individualized thalamic parcellation based on a central-boundary prior. We first constructed probabilistic atlas of thalamic nuclei using high-quality diffusion MRI datasets based on the local diffusion characteristics. Subsequently, high-probability voxels in the probabilistic atlas were utilized as prior guidance to train unique multiple classification models for each subject based on a multilayer perceptron. Finally, we employed the trained model to predict the parcellation labels for thalamic voxels and construct individualized thalamic parcellation. Through a test-retest assessment, the proposed prior-guided individualized thalamic parcellation exhibited excellent reproducibility and the capacity to detect individual variability. Compared with group atlas registration and individual clustering parcellation, the proposed PG-par demonstrated superior parcellation performance under different scanning protocols and clinic settings. Furthermore, the prior-guided individualized parcellation exhibited better correspondence with the histological staining atlas. The proposed prior-guided individualized thalamic parcellation method contributes to the personalized modeling of brain parcellation.
Collapse
Affiliation(s)
- Chaohong Gao
- Sino‐Danish CollegeSino‐Danish Center for Education and ResearchUniversity of Chinese Academy of SciencesBeijingChina
- Brainnetome Center, Institute of AutomationChinese Academy of SciencesBeijingChina
- Department of Applied Mathematics and Computer ScienceTechnical University of DenmarkKongens LyngbyDenmark
| | - Xia Wu
- Brainnetome Center, Institute of AutomationChinese Academy of SciencesBeijingChina
| | - Yaping Wang
- Sino‐Danish CollegeSino‐Danish Center for Education and ResearchUniversity of Chinese Academy of SciencesBeijingChina
- Brainnetome Center, Institute of AutomationChinese Academy of SciencesBeijingChina
- Department of Applied Mathematics and Computer ScienceTechnical University of DenmarkKongens LyngbyDenmark
| | - Gang Li
- Brainnetome Center, Institute of AutomationChinese Academy of SciencesBeijingChina
| | - Liang Ma
- Brainnetome Center, Institute of AutomationChinese Academy of SciencesBeijingChina
| | - Changshuo Wang
- Sino‐Danish CollegeSino‐Danish Center for Education and ResearchUniversity of Chinese Academy of SciencesBeijingChina
- Brainnetome Center, Institute of AutomationChinese Academy of SciencesBeijingChina
| | - Sangma Xie
- Institute of Biomedical Engineering and Instrumentation, School of AutomationHangzhou Dianzi UniversityHangzhouChina
| | - Congying Chu
- Brainnetome Center, Institute of AutomationChinese Academy of SciencesBeijingChina
| | - Kristoffer Hougaard Madsen
- Sino‐Danish CollegeSino‐Danish Center for Education and ResearchUniversity of Chinese Academy of SciencesBeijingChina
- Department of Applied Mathematics and Computer ScienceTechnical University of DenmarkKongens LyngbyDenmark
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and ResearchCopenhagen University Hospital—Amager and HvidovreHvidovreDenmark
| | - Zhongyu Hou
- Department of Medical ImagingShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Lingzhong Fan
- Sino‐Danish CollegeSino‐Danish Center for Education and ResearchUniversity of Chinese Academy of SciencesBeijingChina
- Brainnetome Center, Institute of AutomationChinese Academy of SciencesBeijingChina
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of AutomationChinese Academy of SciencesBeijingChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoShandongChina
| |
Collapse
|
134
|
Thalén A, Ledberg A. Consequences of heterogeneity in aging: parental age at death predicts midlife all-cause mortality and hospitalization in a Swedish national birth cohort. BMC Geriatr 2024; 24:207. [PMID: 38424528 PMCID: PMC10903026 DOI: 10.1186/s12877-024-04786-9] [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: 08/07/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND The processes that underlie aging may advance at different rates in different individuals and an advanced biological age, relative to the chronological age, is associated with increased risk of disease and death. Here we set out to quantify the extent to which heterogeneous aging shapes health outcomes in midlife by following a Swedish birth-cohort and using parental age at death as a proxy for biological age in the offspring. METHODS We followed a nationwide Swedish birth cohort (N = 89,688) between the ages of 39 and 66 years with respect to hospitalizations and death. Cox regressions were used to quantify the association, in the offspring, between parental age at death and all-cause mortality, as well as hospitalization for conditions belonging to the 10 most common ICD-10 chapters. RESULTS Longer parental lifespan was consistently associated with reduced risks of hospitalization and all-cause mortality. Differences in risk were mostly evident from before the age of 50 and persisted throughout the follow-up. Each additional decade of parental survival decreased the risk of offspring all-cause mortality by 22% and risks of hospitalizations by 9 to 20% across the 10 diseases categories considered. The number of deaths and hospitalizations attributable to having parents not living until old age were 1500 (22%) and 11,000 (11%) respectively. CONCLUSIONS Our findings highlight that increased parental lifespan is consistently associated with health benefits in the offspring across multiple outcomes and suggests that heterogeneous aging processes have clinical implications already in midlife.
Collapse
Affiliation(s)
- Anna Thalén
- Department of Public Health Sciences, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Anders Ledberg
- Department of Public Health Sciences, Stockholm University, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
135
|
Pitrez PR, Monteiro LM, Borgogno O, Nissan X, Mertens J, Ferreira L. Cellular reprogramming as a tool to model human aging in a dish. Nat Commun 2024; 15:1816. [PMID: 38418829 PMCID: PMC10902382 DOI: 10.1038/s41467-024-46004-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
The design of human model systems is highly relevant to unveil the underlying mechanisms of aging and to provide insights on potential interventions to extend human health and life span. In this perspective, we explore the potential of 2D or 3D culture models comprising human induced pluripotent stem cells and transdifferentiated cells obtained from aged or age-related disorder-affected donors to enhance our understanding of human aging and to catalyze the discovery of anti-aging interventions.
Collapse
Affiliation(s)
- Patricia R Pitrez
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Luis M Monteiro
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
- IIIUC-institute of Interdisciplinary Research, University of Coimbra, Casa Costa Alemão, Coimbra, 3030-789, Portugal
| | - Oliver Borgogno
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Xavier Nissan
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic diseases, Evry cedex, France
| | - Jerome Mertens
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal.
| |
Collapse
|
136
|
Okada D. Plasma proteins as potential biomarkers of aging of single tissue and cell type. Biogerontology 2024; 25:177-181. [PMID: 37707684 DOI: 10.1007/s10522-023-10065-8] [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: 06/18/2023] [Accepted: 08/21/2023] [Indexed: 09/15/2023]
Abstract
Plasma proteins serve as biomarkers of aging and various age-related diseases. While a number of plasma proteins have been identified that increase or decrease with age, the interpretation of each protein is challenging. This is due to the nature of plasma, which is a mixture of factors secreted by many different tissues and cells. Therefore, the catalog of age-related proteins secreted by a single cell type in a single tissue would be useful for understanding tissue-specific aging patterns. In this study, the author addressed this challenge by integrative data mining of the Human Protein Atlas and the recently published result of large-scale aging proteomics research. Finally, we identified the 17 age-related proteins produced by a single tissue and a single cell type: MBL2 and HP in the liver (hepatocytes), SFTPC in the lung (type II alveolar cells), PRL and POMC in the pituitary (anterior cells), GCG, CUZD1 and CPA2 in the pancreas (pancreatic cells), MYBPC1 in skeletal muscle (myocytes), PTH in the parathyroid gland (glandular cells), LPO and AMY1A in the salivary gland (glandular cells), INSL3 in the male testis (Leydig cells), KLK3 and KLK4 in the male prostate (glandular cells), MPO and ACP5 in immune cells. This list of proteins would be potentially useful for understanding age-related changes in the plasma proteome and inter-tissue networks.
Collapse
Affiliation(s)
- Daigo Okada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, 53 Syogoin-Kawaramachi, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
137
|
Moqri M, Herzog C, Poganik JR, Ying K, Justice JN, Belsky DW, Higgins-Chen AT, Chen BH, Cohen AA, Fuellen G, Hägg S, Marioni RE, Widschwendter M, Fortney K, Fedichev PO, Zhavoronkov A, Barzilai N, Lasky-Su J, Kiel DP, Kennedy BK, Cummings S, Slagboom PE, Verdin E, Maier AB, Sebastiano V, Snyder MP, Gladyshev VN, Horvath S, Ferrucci L. Validation of biomarkers of aging. Nat Med 2024; 30:360-372. [PMID: 38355974 PMCID: PMC11090477 DOI: 10.1038/s41591-023-02784-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/19/2023] [Indexed: 02/16/2024]
Abstract
The search for biomarkers that quantify biological aging (particularly 'omic'-based biomarkers) has intensified in recent years. Such biomarkers could predict aging-related outcomes and could serve as surrogate endpoints for the evaluation of interventions promoting healthy aging and longevity. However, no consensus exists on how biomarkers of aging should be validated before their translation to the clinic. Here, we review current efforts to evaluate the predictive validity of omic biomarkers of aging in population studies, discuss challenges in comparability and generalizability and provide recommendations to facilitate future validation of biomarkers of aging. Finally, we discuss how systematic validation can accelerate clinical translation of biomarkers of aging and their use in gerotherapeutic clinical trials.
Collapse
Affiliation(s)
- Mahdi Moqri
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
| | - Jesse R Poganik
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kejun Ying
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Jamie N Justice
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Daniel W Belsky
- Department of Epidemiology, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | | | - Brian H Chen
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, San Diego, CA, USA
| | - Alan A Cohen
- Department of Environmental Health Sciences, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Martin Widschwendter
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, UK
- Department of Women's and Children's Health, Division of Obstetrics and Gynaecology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jessica Lasky-Su
- Department of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Douglas P Kiel
- Musculoskeletal Research Center, Hinda and Arthur Marcus Institute for Aging Research and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Brian K Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
| | - Steven Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - P Eline Slagboom
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
- Department of Human Movement Sciences, @AgeAmsterdam, Amsterdam Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Vittorio Sebastiano
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Michael P Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | |
Collapse
|
138
|
Jiang M, Zheng Z, Wang X, Chen Y, Qu J, Ding Q, Zhang W, Liu YS, Yang J, Tang W, Hou Y, He J, Wang L, Huang P, Li LC, He Z, Gao Q, Lu Q, Wei L, Wang YJ, Ju Z, Fan JG, Ruan XZ, Guan Y, Liu GH, Pei G, Li J, Wang Y. A biomarker framework for liver aging: the Aging Biomarker Consortium consensus statement. LIFE MEDICINE 2024; 3:lnae004. [PMID: 39872390 PMCID: PMC11749002 DOI: 10.1093/lifemedi/lnae004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/29/2024] [Indexed: 01/11/2025]
Abstract
In human aging, liver aging per se not only increases susceptibility to liver diseases but also increases vulnerability of other organs given its central role in regulating metabolism. Total liver function tends to be well maintained in the healthy elderly, so liver aging is generally difficult to identify early. In response to this critical challenge, the Aging Biomarker Consortium of China has formulated an expert consensus on biomarkers of liver aging by synthesizing the latest scientific literature, comprising insights from both scientists and clinicians. This consensus provides a comprehensive assessment of biomarkers associated with liver aging and presents a systematic framework to characterize these into three dimensions: functional, imaging, and humoral. For the functional domain, we highlight biomarkers associated with cholesterol metabolism and liver-related coagulation function. For the imaging domain, we note that hepatic steatosis and liver blood flow can serve as measurable biomarkers for liver aging. Finally, in the humoral domain, we pinpoint hepatokines and enzymatic alterations worthy of attention. The aim of this expert consensus is to establish a foundation for assessing the extent of liver aging and identify early signs of liver aging-related diseases, thereby improving liver health and the healthy life expectancy of the elderly population.
Collapse
Affiliation(s)
| | - Mengmeng Jiang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhuozhao Zheng
- Department of Radiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xuan Wang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - You-Shuo Liu
- Department of Geriatrics, the Second Xiangya Hospital, and the Institute of Aging and Geriatrics, Central South University, Changsha 410011, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yunlong Hou
- Yiling Pharmaceutical Academician Workstation, Shijiazhuang 050035, China
| | - Jinhan He
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Pengyu Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Engineering Research Center of Pulmonary and Critical Care Medicine Technology and Device (Ministry of Education), Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Lin-Chen Li
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Zhiying He
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200092, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qian Lu
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiong Zhong Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Pei
- Collaborative Innovation Center for Brain Science, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yunfang Wang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
| |
Collapse
|
139
|
Hermann DM, Peruzzotti-Jametti L, Giebel B, Pluchino S. Extracellular vesicles set the stage for brain plasticity and recovery by multimodal signalling. Brain 2024; 147:372-389. [PMID: 37768167 PMCID: PMC10834259 DOI: 10.1093/brain/awad332] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/07/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Extracellular vesicles (EVs) are extremely versatile naturally occurring membrane particles that convey complex signals between cells. EVs of different cellular sources are capable of inducing striking therapeutic responses in neurological disease models. Differently from pharmacological compounds that act by modulating defined signalling pathways, EV-based therapeutics possess multiple abilities via a variety of effectors, thus allowing the modulation of complex disease processes that may have very potent effects on brain tissue recovery. When applied in vivo in experimental models of neurological diseases, EV-based therapeutics have revealed remarkable effects on immune responses, cell metabolism and neuronal plasticity. This multimodal modulation of neuroimmune networks by EVs profoundly influences disease processes in a highly synergistic and context-dependent way. Ultimately, the EV-mediated restoration of cellular functions helps to set the stage for neurological recovery. With this review we first outline the current understanding of the mechanisms of action of EVs, describing how EVs released from various cellular sources identify their cellular targets and convey signals to recipient cells. Then, mechanisms of action applicable to key neurological conditions such as stroke, multiple sclerosis and neurodegenerative diseases are presented. Pathways that deserve attention in specific disease contexts are discussed. We subsequently showcase considerations about EV biodistribution and delineate genetic engineering strategies aiming at enhancing brain uptake and signalling. By sketching a broad view of EV-orchestrated brain plasticity and recovery, we finally define possible future clinical EV applications and propose necessary information to be provided ahead of clinical trials. Our goal is to provide a steppingstone that can be used to critically discuss EVs as next generation therapeutics for brain diseases.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Bernd Giebel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
140
|
Nielsen PY, Jensen MK, Mitarai N, Bhatt S. The Gompertz Law emerges naturally from the inter-dependencies between sub-components in complex organisms. Sci Rep 2024; 14:1196. [PMID: 38216698 PMCID: PMC10786855 DOI: 10.1038/s41598-024-51669-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/08/2024] [Indexed: 01/14/2024] Open
Abstract
Understanding and facilitating healthy aging has become a major goal in medical research and it is becoming increasingly acknowledged that there is a need for understanding the aging phenotype as a whole rather than focusing on individual factors. Here, we provide a universal explanation for the emergence of Gompertzian mortality patterns using a systems approach to describe aging in complex organisms that consist of many inter-dependent subsystems. Our model relates to the Sufficient-Component Cause Model, widely used within the field of epidemiology, and we show that including inter-dependencies between subsystems and modeling the temporal evolution of subsystem failure results in Gompertizan mortality on the population level. Our model also provides temporal trajectories of mortality-risk for the individual. These results may give insight into understanding how biological age evolves stochastically within the individual, and how this in turn leads to a natural heterogeneity of biological age in a population.
Collapse
Affiliation(s)
- Pernille Yde Nielsen
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, 2800, Kongens Lyngby, Denmark.
| | - Majken K Jensen
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Namiko Mitarai
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Samir Bhatt
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
141
|
Sol J, Ortega-Bravo M, Portero-Otín M, Piñol-Ripoll G, Ribas-Ripoll V, Artigues-Barberà E, Butí M, Pamplona R, Jové M. Human lifespan and sex-specific patterns of resilience to disease: a retrospective population-wide cohort study. BMC Med 2024; 22:17. [PMID: 38185624 PMCID: PMC10773063 DOI: 10.1186/s12916-023-03206-w] [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: 07/27/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Slower paces of aging are related to lower risk of developing diseases and premature death. Therefore, the greatest challenge of modern societies is to ensure that the increase in lifespan is accompanied by an increase in health span. To better understand the differences in human lifespan, new insight concerning the relationship between lifespan and the age of onset of diseases, and the ability to avoid them is needed. We aimed to comprehensively study, at a population-wide level, the sex-specific disease patterns associated with human lifespan. METHODS Observational data from the SIDIAP database of a cohort of 482,058 individuals that died in Catalonia (Spain) at ages over 50 years old between the 1st of January 2006 and the 30th of June 2022 were included. The time to the onset of the first disease in multiple organ systems, the prevalence of escapers, the percentage of life free of disease, and their relationship with lifespan were evaluated considering sex-specific traits. RESULTS In the study cohort, 50.4% of the participants were women and the mean lifespan was 83 years. The results show novel relationships between the age of onset of disease, health span, and lifespan. The key findings include: Firstly, the onset of both single and multisystem diseases is progressively delayed as lifespan increases. Secondly, the prevalence of escapers is lower in lifespans around life expectancy. Thirdly, the number of disease-free systems decreases until individuals reach lifespans around 87-88 years old, at which point it starts to increase. Furthermore, long-lived women are less susceptible to multisystem diseases. The associations between health span and lifespan are system-dependent, and disease onset and the percentage of life spent free of disease at the time of death contribute to explaining lifespan variability. Lastly, the study highlights significant system-specific disparities between women and men. CONCLUSIONS Health interventions focused on delaying aging and age-related diseases should be the most effective in increasing not only lifespan but also health span. The findings of this research highlight the relevance of Electronic Health Records in studying the aging process and open up new possibilities in age-related disease prevention that should assist primary care professionals in devising individualized care and treatment plans.
Collapse
Affiliation(s)
- Joaquim Sol
- Catalan Health Institute (ICS), Lleida Research Support Unit (USR), Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol), Lleida, Spain
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Marta Ortega-Bravo
- Catalan Health Institute (ICS), Lleida Research Support Unit (USR), Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol), Lleida, Spain.
| | - Manuel Portero-Otín
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Clinical Neuroscience Research, Santa Maria University Hospital, IRBLleida, Lleida, Spain
| | | | - Eva Artigues-Barberà
- Catalan Health Institute (ICS), Lleida Research Support Unit (USR), Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol), Lleida, Spain
| | - Miquel Butí
- Catalan Health Institute (ICS), Lleida Research Support Unit (USR), Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol), Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain.
| |
Collapse
|
142
|
Dartora C, Marseglia A, Mårtensson G, Rukh G, Dang J, Muehlboeck JS, Wahlund LO, Moreno R, Barroso J, Ferreira D, Schiöth HB, Westman E. A deep learning model for brain age prediction using minimally preprocessed T1w images as input. Front Aging Neurosci 2024; 15:1303036. [PMID: 38259636 PMCID: PMC10800627 DOI: 10.3389/fnagi.2023.1303036] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/04/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction In the last few years, several models trying to calculate the biological brain age have been proposed based on structural magnetic resonance imaging scans (T1-weighted MRIs, T1w) using multivariate methods and machine learning. We developed and validated a convolutional neural network (CNN)-based biological brain age prediction model that uses one T1w MRI preprocessing step when applying the model to external datasets to simplify implementation and increase accessibility in research settings. Our model only requires rigid image registration to the MNI space, which is an advantage compared to previous methods that require more preprocessing steps, such as feature extraction. Methods We used a multicohort dataset of cognitively healthy individuals (age range = 32.0-95.7 years) comprising 17,296 MRIs for training and evaluation. We compared our model using hold-out (CNN1) and cross-validation (CNN2-4) approaches. To verify generalisability, we used two external datasets with different populations and MRI scan characteristics to evaluate the model. To demonstrate its usability, we included the external dataset's images in the cross-validation training (CNN3). To ensure that our model used only the brain signal on the image, we also predicted brain age using skull-stripped images (CNN4). Results The trained models achieved a mean absolute error of 2.99, 2.67, 2.67, and 3.08 years for CNN1-4, respectively. The model's performance in the external dataset was in the typical range of mean absolute error (MAE) found in the literature for testing sets. Adding the external dataset to the training set (CNN3), overall, MAE is unaffected, but individual cohort MAE improves (5.63-2.25 years). Salience maps of predictions reveal that periventricular, temporal, and insular regions are the most important for age prediction. Discussion We provide indicators for using biological (predicted) brain age as a metric for age correction in neuroimaging studies as an alternative to the traditional chronological age. In conclusion, using different approaches, our CNN-based model showed good performance using one T1w brain MRI preprocessing step. The proposed CNN model is made publicly available for the research community to be easily implemented and used to study ageing and age-related disorders.
Collapse
Affiliation(s)
- Caroline Dartora
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Anna Marseglia
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Gustav Mårtensson
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Gull Rukh
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Junhua Dang
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - J-Sebastian Muehlboeck
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Olof Wahlund
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Moreno
- Department of Biomedical Engineering and Health Systems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - José Barroso
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, Las Palmas, España
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, Las Palmas, España
| | - Helgi B. Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
143
|
Wu Z, Qu J, Zhang W, Liu GH. Stress, epigenetics, and aging: Unraveling the intricate crosstalk. Mol Cell 2024; 84:34-54. [PMID: 37963471 DOI: 10.1016/j.molcel.2023.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023]
Abstract
Aging, as a complex process involving multiple cellular and molecular pathways, is known to be exacerbated by various stresses. Because responses to these stresses, such as oxidative stress and genotoxic stress, are known to interplay with the epigenome and thereby contribute to the development of age-related diseases, investigations into how such epigenetic mechanisms alter gene expression and maintenance of cellular homeostasis is an active research area. In this review, we highlight recent studies investigating the intricate relationship between stress and aging, including its underlying epigenetic basis; describe different types of stresses that originate from both internal and external stimuli; and discuss potential interventions aimed at alleviating stress and restoring epigenetic patterns to combat aging or age-related diseases. Additionally, we address the challenges currently limiting advancement in this burgeoning field.
Collapse
Affiliation(s)
- Zeming Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; China National Center for Bioinformation, Beijing 100101, China; CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; The Fifth People's Hospital of Chongqing, Chongqing 400062, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
144
|
Kitaeva KV, Solovyeva VV, Blatt NL, Rizvanov AA. Eternal Youth: A Comprehensive Exploration of Gene, Cellular, and Pharmacological Anti-Aging Strategies. Int J Mol Sci 2024; 25:643. [PMID: 38203812 PMCID: PMC10778954 DOI: 10.3390/ijms25010643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024] Open
Abstract
The improvement of human living conditions has led to an increase in average life expectancy, creating a new social and medical problem-aging, which diminishes the overall quality of human life. The aging process of the body begins with the activation of effector signaling pathways of aging in cells, resulting in the loss of their normal functions and deleterious effects on the microenvironment. This, in turn, leads to chronic inflammation and similar transformations in neighboring cells. The cumulative retention of these senescent cells over a prolonged period results in the deterioration of tissues and organs, ultimately leading to a reduced quality of life and an elevated risk of mortality. Among the most promising methods for addressing aging and age-related illnesses are pharmacological, genetic, and cellular therapies. Elevating the activity of aging-suppressing genes, employing specific groups of native and genetically modified cells, and utilizing senolytic medications may offer the potential to delay aging and age-related ailments over the long term. This review explores strategies and advancements in the field of anti-aging therapies currently under investigation, with a particular emphasis on gene therapy involving adeno-associated vectors and cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Kristina V. Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (K.V.K.); (V.V.S.); (N.L.B.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (K.V.K.); (V.V.S.); (N.L.B.)
| | - Nataliya L. Blatt
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (K.V.K.); (V.V.S.); (N.L.B.)
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (K.V.K.); (V.V.S.); (N.L.B.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| |
Collapse
|
145
|
Tenchov R, Sasso JM, Wang X, Zhou QA. Aging Hallmarks and Progression and Age-Related Diseases: A Landscape View of Research Advancement. ACS Chem Neurosci 2024; 15:1-30. [PMID: 38095562 PMCID: PMC10767750 DOI: 10.1021/acschemneuro.3c00531] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 01/04/2024] Open
Abstract
Aging is a dynamic, time-dependent process that is characterized by a gradual accumulation of cell damage. Continual functional decline in the intrinsic ability of living organisms to accurately regulate homeostasis leads to increased susceptibility and vulnerability to diseases. Many efforts have been put forth to understand and prevent the effects of aging. Thus, the major cellular and molecular hallmarks of aging have been identified, and their relationships to age-related diseases and malfunctions have been explored. Here, we use data from the CAS Content Collection to analyze the publication landscape of recent aging-related research. We review the advances in knowledge and delineate trends in research advancements on aging factors and attributes across time and geography. We also review the current concepts related to the major aging hallmarks on the molecular, cellular, and organismic level, age-associated diseases, with attention to brain aging and brain health, as well as the major biochemical processes associated with aging. Major age-related diseases have been outlined, and their correlations with the major aging features and attributes are explored. We hope this review will be helpful for apprehending the current knowledge in the field of aging mechanisms and progression, in an effort to further solve the remaining challenges and fulfill its potential.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M. Sasso
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Xinmei Wang
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
146
|
Nguyen H, Clément M, Mansencal B, Coupé P. Brain structure ages-A new biomarker for multi-disease classification. Hum Brain Mapp 2024; 45:e26558. [PMID: 38224546 PMCID: PMC10785199 DOI: 10.1002/hbm.26558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 01/17/2024] Open
Abstract
Age is an important variable to describe the expected brain's anatomy status across the normal aging trajectory. The deviation from that normative aging trajectory may provide some insights into neurological diseases. In neuroimaging, predicted brain age is widely used to analyze different diseases. However, using only the brain age gap information (i.e., the difference between the chronological age and the estimated age) can be not enough informative for disease classification problems. In this paper, we propose to extend the notion of global brain age by estimating brain structure ages using structural magnetic resonance imaging. To this end, an ensemble of deep learning models is first used to estimate a 3D aging map (i.e., voxel-wise age estimation). Then, a 3D segmentation mask is used to obtain the final brain structure ages. This biomarker can be used in several situations. First, it enables to accurately estimate the brain age for the purpose of anomaly detection at the population level. In this situation, our approach outperforms several state-of-the-art methods. Second, brain structure ages can be used to compute the deviation from the normal aging process of each brain structure. This feature can be used in a multi-disease classification task for an accurate differential diagnosis at the subject level. Finally, the brain structure age deviations of individuals can be visualized, providing some insights about brain abnormality and helping clinicians in real medical contexts.
Collapse
Affiliation(s)
- Huy‐Dung Nguyen
- Univ. Bordeaux, CNRS, Bordeaux INP, LaBRI, UMR 5800TalenceFrance
| | - Michaël Clément
- Univ. Bordeaux, CNRS, Bordeaux INP, LaBRI, UMR 5800TalenceFrance
| | - Boris Mansencal
- Univ. Bordeaux, CNRS, Bordeaux INP, LaBRI, UMR 5800TalenceFrance
| | - Pierrick Coupé
- Univ. Bordeaux, CNRS, Bordeaux INP, LaBRI, UMR 5800TalenceFrance
| |
Collapse
|
147
|
Yang Z, Wen J, Erus G, Govindarajan ST, Melhem R, Mamourian E, Cui Y, Srinivasan D, Abdulkadir A, Parmpi P, Wittfeld K, Grabe HJ, Bülow R, Frenzel S, Tosun D, Bilgel M, An Y, Yi D, Marcus DS, LaMontagne P, Benzinger TL, Heckbert SR, Austin TR, Waldstein SR, Evans MK, Zonderman AB, Launer LJ, Sotiras A, Espeland MA, Masters CL, Maruff P, Fripp J, Toga A, O’Bryant S, Chakravarty MM, Villeneuve S, Johnson SC, Morris JC, Albert MS, Yaffe K, Völzke H, Ferrucci L, Bryan NR, Shinohara RT, Fan Y, Habes M, Lalousis PA, Koutsouleris N, Wolk DA, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Five dominant dimensions of brain aging are identified via deep learning: associations with clinical, lifestyle, and genetic measures. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.29.23300642. [PMID: 38234857 PMCID: PMC10793523 DOI: 10.1101/2023.12.29.23300642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Brain aging is a complex process influenced by various lifestyle, environmental, and genetic factors, as well as by age-related and often co-existing pathologies. MRI and, more recently, AI methods have been instrumental in understanding the neuroanatomical changes that occur during aging in large and diverse populations. However, the multiplicity and mutual overlap of both pathologic processes and affected brain regions make it difficult to precisely characterize the underlying neurodegenerative profile of an individual from an MRI scan. Herein, we leverage a state-of-the art deep representation learning method, Surreal-GAN, and present both methodological advances and extensive experimental results that allow us to elucidate the heterogeneity of brain aging in a large and diverse cohort of 49,482 individuals from 11 studies. Five dominant patterns of neurodegeneration were identified and quantified for each individual by their respective (herein referred to as) R-indices. Significant associations between R-indices and distinct biomedical, lifestyle, and genetic factors provide insights into the etiology of observed variances. Furthermore, baseline R-indices showed predictive value for disease progression and mortality. These five R-indices contribute to MRI-based precision diagnostics, prognostication, and may inform stratification into clinical trials.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T. Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Paraskevi Parmpi
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University of Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Daniel S. Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R. Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Shari R. Waldstein
- Department of Psychology, University of Maryland, Baltimore County, Catonsville, MD, USA
| | - Michele K. Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Alan B. Zonderman
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Lenore J. Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute of Informatics, Washington University in St. Luis, St. Luis, MO63110, USA
| | - Mark A. Espeland
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Colin L. Masters
- Florey Institute, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Paul Maruff
- Florey Institute, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Arthur Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Sid O’Bryant
- Institute for Translational Research University of North Texas Health Science Center Fort Worth Texas USA
| | - Mallar M. Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Yaffe
- Departments of Neurology, Psychiatry and Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, USA
| | - Nick R. Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T. Shinohara
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Biggs Alzheimer’s Institute, University of Texas San Antonio Health Science Center, USA
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Nikolaos Koutsouleris
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Section for Precision Psychiatry, Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - David A. Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M. Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
148
|
Li D, Ge S, Liu Y, Pan M, Wang X, Han G, Zou S, Liu R, Niu K, Zhao C, Liu N, Qu L. Epitranscriptome analysis of NAD-capped RNA by spike-in-based normalization and prediction of chronological age. iScience 2023; 26:108558. [PMID: 38094247 PMCID: PMC10716591 DOI: 10.1016/j.isci.2023.108558] [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: 08/01/2023] [Revised: 10/14/2023] [Accepted: 11/20/2023] [Indexed: 01/20/2025] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) can be used as an initiating nucleotide in RNA transcription to produce NAD-capped RNA (NAD-RNA). RNA modification by NAD that links metabolite with expressed transcript is a poorly studied epitranscriptomic modification. Current NAD-RNA profiling methods involve multi-steps of chemo-enzymatic labeling and affinity-based enrichment, thus presenting a critical analytical challenge to remove unwanted variations, particularly batch effects. Here, we propose a computational framework, enONE, to remove unwanted variations. We demonstrate that designed spike-in RNA, together with modular normalization procedures and evaluation metrics, can mitigate technical noise, empowering quantitative and comparative assessment of NAD-RNA across different datasets. Using enONE and a human aging cohort, we reveal age-associated features of NAD-capping and further develop an accurate RNA-based aging clock that combines signatures from both transcriptome and NAD-modified epitranscriptome. enONE facilitates the discovery of NAD-RNA responsive to physiological changes, laying an important foundation for functional investigations into this modification.
Collapse
Affiliation(s)
- Dean Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Road, Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuwen Ge
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Road, Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yandong Liu
- Department of Vascular and Endovascular Surgery, Chang Zheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Miaomiao Pan
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 131 Dong An Road, Shanghai 200032, China
- Metalife Biotechnology, 1000 Zhen Chen Road, Baoshan, Shanghai 200444, China
| | - Xueting Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Road, Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guojing Han
- Department of Vascular and Endovascular Surgery, Chang Zheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Sili Zou
- Department of Vascular and Endovascular Surgery, Chang Zheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Rui Liu
- Singlera Genomics, 500 Fu Rong Hua Road, Pudong, Shanghai 201204, China
| | - Kongyan Niu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Road, Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Zhao
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Nan Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Road, Pudong, Shanghai 201210, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 131 Dong An Road, Shanghai 200032, China
- Shanghai Key Laboratory of Aging Studies, 100 Hai Ke Road, Pudong, Shanghai 201210, China
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Chang Zheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
149
|
Wang W, Peng H, Zeng M, Liu J, Liang G, He Z. Endothelial progenitor cells systemic administration alleviates multi-organ senescence by down-regulating USP7/p300 pathway in chronic obstructive pulmonary disease. J Transl Med 2023; 21:881. [PMID: 38057857 PMCID: PMC10699081 DOI: 10.1186/s12967-023-04735-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) has impacted approximately 390 million people worldwide and the morbidity is increasing every year. However, due to the poor treatment efficacy of COPD, exploring novel treatment has become the hotpot of study on COPD. Endothelial progenitor cells (EPCs) aging is a possible molecular way for COPD development. We aimed to explore the effector whether intravenous administration of EPCs has therapeutic effects in COPD mice. METHODS COPD mice model was induced by cigarette smoke exposure and EPCs were injected intravenously to investigate their effects on COPD mice. At day 127, heart, liver, spleen, lung and kidney tissues of mice were harvested. The histological effects of EPCs intervention on multiple organs of COPD mice were detected by morphology assay. Quantitative real-time PCR and Western blotting were used to detect the effect of EPCs intervention on the expression of multi-organ senescence-related indicators. And we explored the effect of EPCs systematically intervening on senescence-related USP7/p300 pathway. RESULTS Compared with COPD group, senescence-associated β-galactosidase activity was decreased, protein and mRNA expression of p16 was down-regulated, while protein and mRNA expression of cyclin D1 and TERT were up-regulated of multiple organs, including lung, heart, liver, spleen and kidney in COPD mice after EPCs system intervention. But the morphological alterations of the tissues described above in COPD mice failed to be reversed. Mechanistically, EPCs systemic administration inhibited the expression of mRNA and protein of USP7 and p300 in multiple organs of COPD mice, exerting therapeutic effects. CONCLUSIONS EPCs administration significantly inhibited the senescence of multiple organs in COPD mice via down-regulating USP7/p300 pathway, which presents a possibility of EPCs therapy for COPD.
Collapse
Affiliation(s)
- Wenhua Wang
- Department of Intensive Care Unit, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huaihuai Peng
- Department of Intensive Care Unit, Hunan Province Directly Affiliated Traditional Chinese Medicine Hospital, Zhuzhou, Hunan, China
| | - Menghao Zeng
- Department of Intensive Care Unit, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Liu
- Department of Intensive Care Unit, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guibin Liang
- Department of Intensive Care Unit, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhihui He
- Department of Intensive Care Unit, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
150
|
Henneicke S, Meuth SG, Schreiber S. [Cerebral Small Vessel Disease: Advances in Understanding its Pathophysiology]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2023; 91:494-502. [PMID: 38081163 DOI: 10.1055/a-2190-8957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Sporadic cerebral small vessel disease determines age- and vascular-risk-factor-related processes of the small brain vasculature. The underlying pathology develops in a stage-dependent manner - probably over decades - often already starting in midlife. Endothelial and pericyte activation precedes blood-brain barrier leaks, extracellular matrix remodeling and neuroinflammation, which ultimately result in bleeds, synaptic and neural dysfunction. Hemodynamic compromise of the small vessel walls promotes perivascular drainage failure and accumulation of neurotoxic waste products in the brain. Clinical diagnosis is mainly based on magnetic resonance imaging according to the Standards for Reporting Vascular Changes on Neuroimaging 2. Cerebral amyloid angiopathy is particularly stratified according to the Boston v2.0 criteria. Small vessel disease of the brain could be clinically silent, or manifested through a heterogeneous spectrum of diseases, where cognitive decline and stroke-related symptoms are the most common ones. Prevention and therapy are centered around vascular risk factor control, physically and cognitively enriched life style and, presumably, maintenance of a good sleep quality, which promotes sufficient perivascular drainage. Prevention of ischemic stroke through anticoagulation that carries at the same time an increased risk for large brain hemorrhages - particularly in the presence of disseminated cortical superficial siderosis - remains one of the main challenges. The cerebral small vessel disease field is rapidly evolving, focusing on the establishment of early disease stage imaging and biofluid biomarkers of neurovascular unit remodeling and the compromise of perivascular drainage. New prevention and therapy strategies will correspondingly center around the dedicated targeting of, e. g., cellular small vessel wall and perivascular tissue structures. Growing knowledge about brain microvasculature bridging neuroimmunological, neurovascular and neurodegenerative fields might lead to a rethink about apparently separate disease entities and the development of overarching concepts for a common line of prevention and treatment for several diseases.
Collapse
Affiliation(s)
- Solveig Henneicke
- Neurologie, Otto-von-Guericke-Universität Magdeburg Medizinische Fakultät, Magdeburg, Germany
| | | | - Stefanie Schreiber
- Neurologie, Otto-von-Guericke-Universität Magdeburg Medizinische Fakultät, Magdeburg, Germany
| |
Collapse
|