1
|
Teschendorff AE, Horvath S. Epigenetic ageing clocks: statistical methods and emerging computational challenges. Nat Rev Genet 2025; 26:350-368. [PMID: 39806006 DOI: 10.1038/s41576-024-00807-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
Over the past decade, epigenetic clocks have emerged as powerful machine learning tools, not only to estimate chronological and biological age but also to assess the efficacy of anti-ageing, cellular rejuvenation and disease-preventive interventions. However, many computational and statistical challenges remain that limit our understanding, interpretation and application of epigenetic clocks. Here, we review these computational challenges, focusing on interpretation, cell-type heterogeneity and emerging single-cell methods, aiming to provide guidelines for the rigorous construction of interpretable epigenetic clocks at cell-type and single-cell resolution.
Collapse
Affiliation(s)
- Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | | |
Collapse
|
2
|
Liu R, Yang S, Zhong X, Zhu Z, Huang W, Wang W. Metabolomic signature of retinal ageing, polygenetic susceptibility, and major health outcomes. Br J Ophthalmol 2025; 109:619-627. [PMID: 39581638 DOI: 10.1136/bjo-2024-325846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND/AIMS To identify the metabolic underpinnings of retinal aging and examine how it is related to mortality and morbidity of common diseases. METHODS The retinal age gap has been established as essential aging indicator for mortality and systemic health. We applied neural network to train the retinal age gap among the participants in UK Biobank and used nuclear magnetic resonance (NMR) to profile plasma metabolites. The metabolomic signature of retinal ageing (MSRA) was identified using an elastic network model. Multivariable Cox regressions were used to assess associations between the signature with 12 serious health conditions. The participants in Guangzhou Diabetic Eye Study (GDES) cohort were analyzed for validation. RESULTS This study included 110 722 participants (mean age 56.5±8.1 years at baseline, 53.8% female), and 28 plasma metabolites associated with retinal ageing were identified. The MSRA revealed significant correlations with each 12 serious health conditions beyond traditional risk factors and genetic predispositions. Each SD increase in MSRA was linked to a 24%-76% higher risk of mortality, cardiovascular diseases, dementia and diabetes mellitus. MSRA showed dose-response relationships with risks of these diseases, with seven showing non-linear and five showing linear increases. Validation in the GDES further established the relation between retinal ageing-related metabolites and increased risks of cardiovascular and chronic kidney diseases (all p<0.05). CONCLUSIONS The metabolic connections between ocular and systemic health offer a novel tool for identifying individuals at high risk of premature ageing, promoting a more holistic view of human health.
Collapse
Affiliation(s)
- Riqian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shaopeng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaoying Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ziyu Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wenyong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, China
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, China
| |
Collapse
|
3
|
Nussinov R, Yavuz BR, Jang H. Tumors and their microenvironments: Learning from pediatric brain pathologies. Biochim Biophys Acta Rev Cancer 2025; 1880:189328. [PMID: 40254040 DOI: 10.1016/j.bbcan.2025.189328] [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: 12/12/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Early clues to tumors and their microenvironments come from embryonic development. Here we review the literature and consider whether the embryonic brain and its pathologies can serve as a better model. Among embryonic organs, the brain is the most heterogenous and complex, with multiple lineages leading to wide spectrum of cell states and types. Its dysregulation promotes neurodevelopmental brain pathologies and pediatric tumors. Embryonic brain pathologies point to the crucial importance of spatial heterogeneity over time, akin to the tumor microenvironment. Tumors dedifferentiate through genetic mutations and epigenetic modulations; embryonic brains differentiate through epigenetic modulations. Our innovative review proposes learning developmental brain pathologies to target tumor evolution-and vice versa. We describe ways through which tumor pharmacology can learn from embryonic brains and their pathologies, and how learning tumor, and its microenvironment, can benefit targeting neurodevelopmental pathologies. Examples include pediatric low-grade versus high-grade brain tumors as in rhabdomyosarcomas and gliomas.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
4
|
Kroemer G, Maier AB, Cuervo AM, Gladyshev VN, Ferrucci L, Gorbunova V, Kennedy BK, Rando TA, Seluanov A, Sierra F, Verdin E, López-Otín C. From geroscience to precision geromedicine: Understanding and managing aging. Cell 2025; 188:2043-2062. [PMID: 40250404 DOI: 10.1016/j.cell.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 04/20/2025]
Abstract
Major progress has been made in elucidating the molecular, cellular, and supracellular mechanisms underlying aging. This has spurred the birth of geroscience, which aims to identify actionable hallmarks of aging. Aging can be viewed as a process that is promoted by overactivation of gerogenes, i.e., genes and molecular pathways that favor biological aging, and alternatively slowed down by gerosuppressors, much as cancers are caused by the activation of oncogenes and prevented by tumor suppressors. Such gerogenes and gerosuppressors are often associated with age-related diseases in human population studies but also offer targets for modeling age-related diseases in animal models and treating or preventing such diseases in humans. Gerogenes and gerosuppressors interact with environmental, behavioral, and psychological risk factors to determine the heterogeneous trajectory of biological aging and disease manifestation. New molecular profiling technologies enable the characterization of gerogenic and gerosuppressive pathways, which serve as biomarkers of aging, hence inaugurating the era of precision geromedicine. It is anticipated that, pending results from randomized clinical trials and regulatory approval, gerotherapeutics will be tailored to each person based on their genetic profile, high-dimensional omics-based biomarkers of aging, clinical and digital biomarkers of aging, psychosocial profile, and past or present exposures.
Collapse
Affiliation(s)
- Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Andrea B Maier
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit, Amsterdam Movement Sciences, Amsterdam, the Netherlands; NUS Academy for Healthy Longevity, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Vadim N Gladyshev
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, USA; Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian K Kennedy
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, Centre for Healthy Longevity, National University Health System, National University of Singapore, Singapore, Singapore; Life Sciences Institute Neurobiology Programme, Centre for Life Sciences, National University of Singapore, Singapore, Singapore; Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Department of Neurology and Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, USA; Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Carlos López-Otín
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, Paris, France; Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain.
| |
Collapse
|
5
|
Liu X, Zhao J, Liu J, Huang Y, Deng W, Yan L, Cui M, Pan X, Xiao H, Liu X. Association of α-Klotho with anti-aging effects of Ganoderma lucidum in animal models. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119597. [PMID: 40057146 DOI: 10.1016/j.jep.2025.119597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aging is a complex, universal process characterized by structural and functional decline across multiple organs. Ganoderma lucidum (G. lucidum), a renowned traditional Chinese medicinal fungus, has long been recognized for its anti-aging properties. However, the underlying mechanisms remain incompletely understood. AIM OF THE STUDY This study aimed to investigate the anti-aging effects of G. lucidum and its underlying mechanisms. MATERIALS AND METHODS We investigated the anti-aging effects of G. lucidum sporoderm-broken spore powder (Gl-SBSP) on Caenorhabditis elegans (C. elegans) lifespan and aging across multiple organs using natural aging, D-galactose (D-gal)-induced aging, and radiation-induced premature senescence mouse models. In C. elegans, we assessed lifespan, reproductive capacity, body length, pharyngeal pumping, body bends, fat and lipofuscin levels, as well as reactive oxygen species (ROS) accumulation. In mice, histopathological staining, complete blood counts, and enzyme-linked immunosorbent assay (ELISA) were used to evaluate tissue damage, while quantitative real-time PCR (RT-qPCR) was employed to access small intestine barrier integrity. Western blot (WB) and immunohistochemistry (IHC) were utilized to analyze the distribution of alpha Klotho (α-Klotho) in the kidney, blood, and urine. RESULTS Gl-SBSP significantly extended C. elegans lifespan, improved reproductive capacity and mobility, and reduced lipofuscin and ROS levels. In naturally aged mice, Gl-SBSP enhanced physical appearance and performance. Additionally, Gl-SBSP alleviated aging-related structural and functional decline in multiple organs, including the colon, spleen, kidneys, liver, and small intestine, across all aging models. Biochemical analyses revealed that Gl-SBSP increased transmembrane α-Klotho (mα-Klotho) and soluble α-Klotho (sα-Klotho) levels in kidney tissue and elevated sα-Klotho levels in serum and urine. CONCLUSION This study is the first to demonstrate that G. lucidum exerts α-Klotho-associated anti-aging effects in animal models, highlighting its potential as an anti-aging intervention.
Collapse
Affiliation(s)
- Xiaojing Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jiamin Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jia Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yan Huang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Wei Deng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Luwen Yan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Xinhua Pan
- Jiangxi Xiankelai Biotechnology Co., Ltd., 10 Shacheng Road, Jiujiang, 332000, China
| | - Huiwen Xiao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Xingzhong Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
6
|
Kou M, Ma H, Wang X, Heianza Y, Qi L. Plasma proteomics-based brain aging signature and incident dementia risk. GeroScience 2025; 47:2335-2349. [PMID: 39532828 PMCID: PMC11978599 DOI: 10.1007/s11357-024-01407-6] [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: 08/07/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Investigating brain-enriched proteins with machine learning methods may enable a brain-specific understanding of brain aging and provide insights into the molecular mechanisms and pathological pathways of dementia. The study aims to analyze associations of brain-specific plasma proteomic aging signature with risks of incident dementia. In 45,429 dementia-free UK Biobank participants at baseline, we generated a brain-specific biological age using 63 brain-enriched plasma proteins with machine learning methods. The brain age gap was estimated, and Cox proportional hazards models were used to study the association with incident all-cause dementia, Alzheimer's disease (AD), and vascular dementia. Per-unit increment in the brain age gap z-score was associated with significantly higher risks of all-cause dementia (hazard ratio [95% confidence interval], 1.67 [1.56-1.79], P < 0.001), AD (1.85 [1.66-2.08], P < 0.001), and vascular dementia (1.86 [1.55-2.24], P < 0.001), respectively. Notably, 2.1% of the study population exhibited extreme old brain aging defined as brain age gap z-score > 2, correlating with over threefold increased risks of all-cause dementia and vascular dementia (3.42 [2.25-5.20], P < 0.001, and 3.41 [1.05-11.13], P = 0.042, respectively), and fourfold increased risk of AD (4.45 [2.32-8.54], P < 0.001). The associations were stronger among participants with healthier lifestyle factors (all P-interaction < 0.05). These findings were corroborated by magnetic resonance imaging assessments showing that a higher brain age gap aligns global pathophysiology of dementia, including global and regional atrophy in gray matter, and white matter lesions (P < 0.001). The brain-specific proteomic age gap is a powerful biomarker of brain aging, indicative of dementia risk and neurodegeneration.
Collapse
Affiliation(s)
- Minghao Kou
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Hao Ma
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Xuan Wang
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Yoriko Heianza
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lu Qi
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
7
|
Vujosevic S, Limoli C, Kozak I. Hallmarks of aging in age-related macular degeneration and age-related neurological disorders: novel insights into common mechanisms and clinical relevance. Eye (Lond) 2025; 39:845-859. [PMID: 39289517 PMCID: PMC11933422 DOI: 10.1038/s41433-024-03341-5] [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: 04/14/2024] [Revised: 08/13/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
Age-related macular degeneration (AMD) and age-related neurological diseases (ANDs), such as Alzheimer's and Parkinson's Diseases, are increasingly prevalent conditions that significantly contribute to global morbidity, disability, and mortality. The retina, as an accessible part of the central nervous system (CNS), provides a unique window to study brain aging and neurodegeneration. By examining the associations between AMD and ANDs, this review aims to highlight novel insights into fundamental mechanisms of aging and their role in neurodegenerative disease progression. This review integrates knowledge from the emerging field of aging research, which identifies common denominators of biological aging, specifically loss of proteostasis, impaired macroautophagy, mitochondrial dysfunction, and inflammation. Finally, we emphasize the clinical relevance of these pathways and the potential for cross-disease therapies that target common aging hallmarks. Identifying these shared pathways could open avenues to develop therapeutic strategies targeting mechanisms common to multiple degenerative diseases, potentially attenuating disease progression and promoting the healthspan.
Collapse
Affiliation(s)
- Stela Vujosevic
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
- Eye Clinic, IRCCS MultiMedica, Milan, Italy.
| | - Celeste Limoli
- Eye Clinic, IRCCS MultiMedica, Milan, Italy
- University of Milan, Milan, Italy
| | - Igor Kozak
- Moorfields Eye Hospital Centre, Abu Dhabi, UAE
- Ophthalmology and Vision Science, University of Arizona, Tucson, USA
| |
Collapse
|
8
|
Zeiss CJ, Huttner A, Nairn AC, Arnsten A, Datta D, Strittmatter SM, Wyk BV, Duque A. The neuropathologic basis for translational biomarker development in the macaque model of late-onset Alzheimer's disease. J Alzheimers Dis 2025; 104:1243-1258. [PMID: 40095666 DOI: 10.1177/13872877251323787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
BackgroundAccurate placement of the macaque within the Alzheimer's disease (AD) research framework is essential to discover early-stage predictive biomarkers.ObjectiveTo assess utility of the aging macaque in advancing translational biomarker development for preclinical AD, we evaluated relative signal strength of comparable neuropathologic phenomena in macaques and patients.MethodsWe compared pathology in patient and macaque formalin-fixed paraffin embedded (FFPE) tissues using identical criteria. We quantified expression of amyloid-β (Aβ), pTau, and inflammatory and senescence markers across species. Distribution of AD-relevant markers were compared in FFPE and perfused frozen macaque brain to assess expression of labile proteins that could inform in-life fluid biomarkers.ResultsAβ pathology in macaques closely approximated patient pathology. Complex plaque composition in macaques implied significant disruption of synaptic connectivity. In FFPE tissue, pretangle pTau immunoreactivity placed the macaque in Braak Stage 1b. In perfused frozen tissue, soluble pTau distribution approximated Braak Stage III-IV. In macaque, Aβ, pTau, and acetylcholinesterase labeling co-localized to AD-vulnerable circuits. Significant association of glial fibrillary acidic protein with Aβ occurred in humans only. The senescence marker p16 correlated positively with pTau expression and negatively with Aβ in patients only. Macaques lacked neuropathologic co-morbidities.ConclusionsAD-relevant neuropathologic signals in the macaque support biomarker discovery in the areas of Aβ plaque evolution and associated synaptic disruption as well as early-stage tau phosphorylation. Relative protection from accumulation of senescence markers, fibrillar tau and neuropathologic co-morbidities in macaque implicate species difference in rates of biological brain aging. We provide over 4000 digital slides for further study.
Collapse
Affiliation(s)
- Caroline J Zeiss
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Amy Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Dibyadeep Datta
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen M Strittmatter
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
9
|
Zhao R, Lu H, Yuan H, Chen S, Xu K, Zhang T, Liu Z, Jiang Y, Suo C, Chen X. Plasma proteomics-based organ-specific aging for all-cause mortality and cause-specific mortality: a prospective cohort study. GeroScience 2025; 47:1411-1423. [PMID: 39477866 PMCID: PMC11978558 DOI: 10.1007/s11357-024-01411-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] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/23/2024] [Indexed: 04/09/2025] Open
Abstract
Individual's aging rates vary across organs. However, there are few methods for assessing aging at organ levels and whether they contribute differently to mortalities remains unknown. We analyzed data from 45,821 adults in the UK Biobank, using plasma proteomics and machine learning to estimate biological ages for 12 major organs. The differences between biological age and chronological age, referred to as "age gaps," were calculated for each organ. Partial correlation analyses were used to assess the association between age gaps and modifiable factors. Adjusted multivariable Cox regression models were applied to examine the association of age gaps with all-cause mortality, cause-specific mortalities, and cancer-specific mortalities. We reveal a complex network of varied associations between multi-organ aging and modifiable factors. All age gaps increase the risk of all-cause mortality by 6-60%. The risk of death varied from 5.54 to 29.18 times depending on the number of aging organs. Cause-specific mortalities are associated with certain organs' aging. For mental diseases mortality, and nervous system mortality, only brain aging exhibited a significant increased risk of HR 2.38 (per SD, 95% CI: 2.06-2.74) and 1.99 (per SD, 95% CI: 1.84-2.16), respectively. Age gaps of stomach were also a specific indicator for gastric cancer. Eventually, we find that an organ's biological age selectively influences the aging of other organ systems. Our study demonstrates that accelerated aging in specific organs increases the risk of mortality from various causes. This provides a potential tool for early identification of at-risk populations, offering a relatively objective method for precision medicine.
Collapse
Affiliation(s)
- Renjia Zhao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China
| | - Heyang Lu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China
| | - Shuaizhou Chen
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Kelin Xu
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Tiejun Zhang
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Chen Suo
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China.
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China.
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Wen J, Skampardoni I, Tian YE, Yang Z, Cui Y, Erus G, Hwang G, Varol E, Boquet-Pujadas A, Chand GB, Nasrallah I, Satterthwaite TD, Shou H, Shen L, Toga AW, Zalesky A, Davatzikos C. Neuroimaging-AI endophenotypes reveal underlying mechanisms and genetic factors contributing to progression and development of four brain disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2023.08.16.23294179. [PMID: 37662256 PMCID: PMC10473785 DOI: 10.1101/2023.08.16.23294179] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Recent work leveraging artificial intelligence has offered promise to dissect disease heterogeneity by identifying complex intermediate brain phenotypes, called dimensional neuroimaging endophenotypes (DNEs). We advance the argument that these DNEs capture the degree of expression of respective neuroanatomical patterns measured, offering a dimensional neuroanatomical representation for studying disease heterogeneity and similarities of neurologic and neuropsychiatric diseases. We investigate the presence of nine DNEs derived from independent yet harmonized studies on Alzheimer's disease, autism spectrum disorder, late-life depression, and schizophrenia in the UK Biobank study. Phenome-wide associations align with genome-wide associations, revealing 31 genomic loci (P-value<5×10-8/9) associated with the nine DNEs.The nine DNEs, along with their polygenic risk scores, significantly enhanced the predictive accuracy for 14 systemic disease categories, particularly for conditions related to mental health and the central nervous system, as well as mortality outcomes. These findings underscore the potential of the nine DNEs to capture the expression of disease-related brain phenotypes in individuals of the general population and to relate such measures with genetics, lifestyle factors, and chronic diseases.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
- Department of Radiology, Columbia University, New York, NY, USA
- New York Genome Center (NYGC), New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Data Science Institute (DSI), Columbia University, New York, NY, USA
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Department of Radiology, Columbia University, New York, NY, USA
- Zuckerman Institute, Columbia University, New York, NY, 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, USA
| | - Ye Ella Tian
- Systems Lab, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - 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, 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, 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, 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, USA
| | - Erdem Varol
- Department of Computer Science and Engineering, New York University, New York, USA
| | - Aleix Boquet-Pujadas
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
| | - Ganesh B. Chand
- Department of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Ilya Nasrallah
- 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, USA
| | - Theodore D. Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 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, USA
- Department of Biostatistics, Epidemiology and Informatics University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics University of Pennsylvania Perelman School of Medicine, Philadelphia, 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 Zalesky
- Systems Lab, 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 (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
11
|
Liu KS, Wang B, Mak IL, Choi EP, Lam CL, Wan EY. Early onset of hypertension and increased relative risks of chronic kidney disease and mortality: two population-based cohort studies in United Kingdom and Hong Kong. Hypertens Res 2025:10.1038/s41440-025-02188-x. [PMID: 40140711 DOI: 10.1038/s41440-025-02188-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025]
Abstract
This study aimed to evaluate the association between hypertension (HT) onset age and later risks of chronic kidney diseases (CKD) and mortality. Adult patients without CKD from 2008 to 2013 were identified using electronic medical records from United Kingdom (UK) and Hong Kong (HK). Patients newly diagnosed with HT and those without were included in the HT and control groups, respectively. All subjects were stratified into six age groups (18-39, 40-49, 50-59, 60-69, 70-79, ≥80). Multivariable Cox proportional hazard regression, adjusted with baseline characteristics and fine stratification weights, was conducted to investigate the association between HT onset and risks of CKD, renal decline, end-stage renal disease (ESRD), and all-cause mortality. Subjects were followed up from baseline until an outcome event, death, or administrative end of the cohort, whichever occurred first. A total of 4,413,551 and 3,132,951 subjects were included in the UK and HK cohorts, respectively. HT was significantly associated with increased risks of outcome, but the hazard ratios (HRs) decreased with increasing onset age. In the UK cohort, the HRs (95% confidence intervals) for subjects aged 18-39 and ≥80 were 3.69 (3.53, 3.86) and 2.01 (1.96, 2.06) for CKD, 3.83 (3.60, 4.07) and 3.17 (2.97, 3.38) for renal decline, 17.26 (14.34, 20.77) and 2.55 (2.12, 3.07) for ESRD, 2.88 (2.66, 3.11) and 1.09 (1.07, 1.12) for mortality. The HK cohort exhibited a similar pattern. Our study concluded that early onset of HT significantly affects renal health later in life, while the contribution decreases with the onset age of HT.
Collapse
Affiliation(s)
- Kiki Sn Liu
- Department of Family Medicine and Primary Care, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
| | - Boyuan Wang
- Department of Family Medicine and Primary Care, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
| | - Ivy L Mak
- Department of Family Medicine and Primary Care, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
| | - Edmond Ph Choi
- School of Nursing, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
| | - Cindy Lk Lam
- Department of Family Medicine and Primary Care, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China
- Department of Family Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Eric Yf Wan
- Department of Family Medicine and Primary Care, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China.
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China.
- The Institute of Cardiovascular Science and Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China.
- Advanced Data Analytics for Medical Science (ADAMS.) Limited, Hong Kong S.A.R., China.
| |
Collapse
|
12
|
Hansen CW, Strulik H. How do we age? A decomposition of Gompertz law. JOURNAL OF HEALTH ECONOMICS 2025; 101:102988. [PMID: 40127516 DOI: 10.1016/j.jhealeco.2025.102988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025]
Abstract
A strong regularity of human life is Gompertz's law, which predicts a near-perfect exponential increase in mortality with age. In this paper, we take into account that chronological age is not a cause of death and decompose Gompertz's law into two equally strong laws: (i) an exponential increase in health deficits as measured by the frailty index, and (ii) a power law association between the frailty index and the mortality rate. We show how the increase in the frailty index can be derived from the feature of self-productivity of health deficits. We explore the robustness of the Gompertz decomposition across countries, sex, and over time and show how information about mortality rates can be used to infer the state of health of an age-structured population. Finally, we use this method to infer the biological ages of past populations, such as Australians in 1940 and Swedes in 1770.
Collapse
Affiliation(s)
| | - Holger Strulik
- University of Goettingen, Department of Economics, Platz der Goettinger Sieben 3, 37073 Goettingen, Germany.
| |
Collapse
|
13
|
McDonagh J, Ferguson C, Hilmer SN, Hubbard RE, Lindley RI, Driscoll A, Maiorana A, Wu L, Atherton JJ, Bajorek BV, Carr B, Delbaere K, Dent E, Duong MH, Hickman LD, Hopper I, Huynh Q, Jha SR, Keech A, Sim M, Singh GK, Villani A, Shang C, Hsu M, Vandenberg J, Davidson PM, Macdonald PS. An Expert Opinion on the Management of Frailty in Heart Failure from the Australian Cardiovascular Alliance National Taskforce. Heart Lung Circ 2025:S1443-9506(25)00169-6. [PMID: 40107957 DOI: 10.1016/j.hlc.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 03/22/2025]
Abstract
Approximately 50% of all adults with heart failure (HF) are classified as frail. Frailty is a clinical state of 'accelerated ageing' that complicates management and results in adverse health outcomes. Despite recommendations for frailty assessment in HF guidelines, its implementation into routine clinical practice has been slow. Further, evidence to inform models of care and pharmacological treatment for individuals with HF who are classified as frail is lacking. The complexity of management underscores the importance of tailoring models of care that can improve the focus on frailty through multidisciplinary care teams. Frailty can be reduced in some cases through the comprehensive geriatric assessment model of care, integrating treatment pillars such as exercise, nutrition, social engagement and support networks, and optimised medication use. A national agenda for action on frailty in the context of HF is needed to advance policy, practice, education, and research improve health outcomes for individuals affected. In November 2023 the Australian Cardiovascular Alliance (ACvA) facilitated a national workshop on frailty and HF with key experts. This has led to the development of a frailty and HF national taskforce with the aim to address major priorities and unmet needs. This statement is first step for the taskforce in implementing a national agenda for the management of frailty in HF. Here we outline key considerations for policy, practice, education, and research in Australia.
Collapse
Affiliation(s)
- Julee McDonagh
- School of Nursing, Faculty of Science, Medicine & Health, The University of Wollongong, Wollongong, NSW, Australia; Centre for Chronic and Complex Care Research, Blacktown Hospital, Western Sydney Local Health District, Blacktown, NSW, Australia.
| | - Caleb Ferguson
- School of Nursing, Faculty of Science, Medicine & Health, The University of Wollongong, Wollongong, NSW, Australia; Centre for Chronic and Complex Care Research, Blacktown Hospital, Western Sydney Local Health District, Blacktown, NSW, Australia
| | - Sarah N Hilmer
- Kolling Institute, Northern Sydney Local Health District and The University of Sydney, St Leonards, NSW, Australia
| | - Ruth E Hubbard
- The Australian Frailty Network, Centre for Health Services Research, The University of Queensland, Brisbane, Qld, Australia; Princess Alexandra Hospital, Woolloongabba, Brisbane, Qld, Australia
| | - Richard I Lindley
- Westmead Applied Research Centre, Faculty of Medicine and Health, The University of Sydney, NSW, Australia; George Institute for Global Health, Sydney, NSW, Australia
| | - Andrea Driscoll
- Centre for Quality and Patient Safety, Monash Health & Deakin University, Melbourne, Vic, Australia; Department of Cardiology, Austin Health, Heidelberg, Vic, Australia
| | - Andrew Maiorana
- Curtin School of Allied Health, Faculty of Health Sciences, Perth, WA, Australia; Exercise Physiology Department, Fiona Stanley Hospital, Perth, WA, Australia
| | - Lindsay Wu
- Laboratory for Ageing Research, University of New South Wales, Sydney, NSW, Australia
| | - John J Atherton
- Department of Cardiology, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia
| | - Beata V Bajorek
- College of Health, Medicine, and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia; Pharmacy Department, John Hunter Hospital, Hunter New England Local Health District, New Lambton, NSW, Australia
| | - Bridie Carr
- Agency for Clinical Innovation, Cardiac Network, NSW Government, NSW, Australia
| | - Kim Delbaere
- Falls, Balance and Injury Research Centre, Neuroscience Research Australia, Randwick, NSW, Australia; School of Population Health, University of New South Wales, Sydney, NSW, Australia
| | - Elsa Dent
- Institute for Evidence-Based Health Care, Bond University, Robina, Qld, Australia
| | - Mai H Duong
- Kolling Institute, Northern Sydney Local Health District and The University of Sydney, St Leonards, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Louise D Hickman
- School of Nursing, Faculty of Science, Medicine & Health, The University of Wollongong, Wollongong, NSW, Australia
| | - Ingrid Hopper
- Department of Cardiology and General Medicine Unit, Alfred Health, Melbourne, Vic, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Vic, Australia
| | - Quan Huynh
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia
| | | | - Anthony Keech
- Faculty Medicine and Health, Cardiovascular Research, NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Marc Sim
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Perth, WA, Australia; Medical School, The University of Western Australia, Perth, WA, Australia
| | - Gursharan K Singh
- Centre for Healthcare Transformation, Faculty of Health, Queensland University of Technology, Brisbane, Qld, Australia; Cancer and Palliative Care Outcomes Centre, School of Nursing, Queensland University of Technology, Brisbane, Qld, Australia
| | - Anthony Villani
- School of Health, University of the Sunshine Coast, Birtinya, Qld, Australia
| | | | - Meng Hsu
- Australian Cardiovascular Alliance, Sydney, NSW, Australia
| | - Jamie Vandenberg
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW Australia
| | - Patricia M Davidson
- School of Nursing, Faculty of Science, Medicine & Health, The University of Wollongong, Wollongong, NSW, Australia
| | - Peter S Macdonald
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW Australia; Heart Lung Clinic, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia; School of Clinical Medicine, University of New South Wales, NSW, Sydney, Australia
| |
Collapse
|
14
|
Xie H. A capsule network-based public health prediction system for chronic diseases: clinical and community implications. Front Public Health 2025; 13:1526360. [PMID: 40161025 PMCID: PMC11949884 DOI: 10.3389/fpubh.2025.1526360] [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: 11/11/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Objective To observe the role of a public health chronic disease prediction method based on capsule network and information system in clinical treatment and public health management. Methods Patients with hypertension, diabetes, and asthma admitted from May 2022 to October 2023 were incorporated into the research. They were grouped into hypertension group (n = 341), diabetes group (n = 341), and asthma group (n = 341). The established chronic disease prediction method was used to diagnose these types of public health chronic diseases. The key influencing factors obtained by the prediction method were compared with the regression analysis results. In addition, its diagnostic accuracy and specificity were analyzed, and the clinical diagnostic value of this method was explored. This method was applied to public health management and the management approach was improved based on the distribution and prevalence of chronic diseases. The effectiveness and residents' acceptance of public health management before and after improvement were compared, and the application value of this method in public health management was explored. Results The key factors affecting the three diseases obtained by the application of prediction methods were found to be significantly correlated with disease occurrence after regression analysis (p < 0.05). Compared with before application, the diagnostic accuracy, specificity and sensitivity values of the method were 88.6, 89 and 92%, respectively, which were higher than the empirical diagnostic methods of doctors (p < 0.05). Compared with other existing AI-based chronic disease prediction methods, the AUC value of the proposed method was significantly higher than theirs (p < 0.05). This indicates that the diagnostic method proposed in this study has higher accuracy. After applying this method to public health management, the wellbeing of individuals with chronic conditions in the community was notably improved, and the incidence rate was notably reduced (p < 0.05). The acceptance level of residents toward the management work of public health management departments was also notably raised (p < 0.05). Conclusion The public health chronic disease prediction method based on information systems and capsule network has high clinical value in diagnosis and can help physicians accurately diagnose patients' conditions. In addition, this method has high application value in public health management. Management departments can adjust management strategies in a timely manner through predictive analysis results and propose targeted management measures based on the characteristics of residents in the management community.
Collapse
Affiliation(s)
- Haiyan Xie
- Medical College of Changsha Social Work College, Changsha, China
| |
Collapse
|
15
|
Sun X, Xia M. Schizophrenia and Neurodevelopment: Insights From Connectome Perspective. Schizophr Bull 2025; 51:309-324. [PMID: 39209793 PMCID: PMC11908871 DOI: 10.1093/schbul/sbae148] [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] [Indexed: 09/04/2024]
Abstract
BACKGROUND Schizophrenia is conceptualized as a brain connectome disorder that can emerge as early as late childhood and adolescence. However, the underlying neurodevelopmental basis remains unclear. Recent interest has grown in children and adolescent patients who experience symptom onset during critical brain development periods. Inspired by advanced methodological theories and large patient cohorts, Chinese researchers have made significant original contributions to understanding altered brain connectome development in early-onset schizophrenia (EOS). STUDY DESIGN We conducted a search of PubMed and Web of Science for studies on brain connectomes in schizophrenia and neurodevelopment. In this selective review, we first address the latest theories of brain structural and functional development. Subsequently, we synthesize Chinese findings regarding mechanisms of brain structural and functional abnormalities in EOS. Finally, we highlight several pivotal challenges and issues in this field. STUDY RESULTS Typical neurodevelopment follows a trajectory characterized by gray matter volume pruning, enhanced structural and functional connectivity, improved structural connectome efficiency, and differentiated modules in the functional connectome during late childhood and adolescence. Conversely, EOS deviates with excessive gray matter volume decline, cortical thinning, reduced information processing efficiency in the structural brain network, and dysregulated maturation of the functional brain network. Additionally, common functional connectome disruptions of default mode regions were found in early- and adult-onset patients. CONCLUSIONS Chinese research on brain connectomes of EOS provides crucial evidence for understanding pathological mechanisms. Further studies, utilizing standardized analyses based on large-sample multicenter datasets, have the potential to offer objective markers for early intervention and disease treatment.
Collapse
Affiliation(s)
- Xiaoyi Sun
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
16
|
Yi F, Yuan J, Somekh J, Peleg M, Zhu YC, Jia Z, Wu F, Huang Z. Genetically supported targets and drug repurposing for brain aging: A systematic study in the UK Biobank. SCIENCE ADVANCES 2025; 11:eadr3757. [PMID: 40073132 PMCID: PMC11900869 DOI: 10.1126/sciadv.adr3757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025]
Abstract
Brain age gap (BAG), the deviation between estimated brain age and chronological age, is a promising marker of brain health. However, the genetic architecture and reliable targets for brain aging remains poorly understood. In this study, we estimate magnetic resonance imaging (MRI)-based brain age using deep learning models trained on the UK Biobank and validated with three external datasets. A genome-wide association study for BAG identified two unreported loci and seven previously reported loci. By integrating Mendelian Randomization (MR) and colocalization analysis on eQTL and pQTL data, we prioritized seven genetically supported druggable genes, including MAPT, TNFSF12, GZMB, SIRPB1, GNLY, NMB, and C1RL, as promising targets for brain aging. We rediscovered 13 potential drugs with evidence from clinical trials of aging and prioritized several drugs with strong genetic support. Our study provides insights into the genetic basis of brain aging, potentially facilitating drug development for brain aging to extend the health span.
Collapse
Affiliation(s)
- Fan Yi
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China
| | - Jing Yuan
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Judith Somekh
- Department of Information Systems, University of Haifa, Haifa, Israel
| | - Mor Peleg
- Department of Information Systems, University of Haifa, Haifa, Israel
| | - Yi-Cheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhilong Jia
- Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Fei Wu
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China
| | - Zhengxing Huang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
García-Domínguez M. Pathological and Inflammatory Consequences of Aging. Biomolecules 2025; 15:404. [PMID: 40149940 PMCID: PMC11939965 DOI: 10.3390/biom15030404] [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: 02/25/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Aging is a complex, progressive, and irreversible biological process that entails numerous structural and functional changes in the organism. These changes affect all bodily systems, reducing their ability to respond and adapt to the environment. Chronic inflammation is one of the key factors driving the development of age-related diseases, ultimately causing a substantial decline in the functional abilities of older individuals. This persistent inflammatory state (commonly known as "inflammaging") is characterized by elevated levels of pro-inflammatory cytokines, an increase in oxidative stress, and a perturbation of immune homeostasis. Several factors, including cellular senescence, contribute to this inflammatory milieu, thereby amplifying conditions such as cardiovascular disease, neurodegeneration, and metabolic disorders. Exploring the mechanisms of chronic inflammation in aging is essential for developing targeted interventions aimed at promoting healthy aging. This review explains the strong connection between aging and chronic inflammation, highlighting potential therapeutic approaches like pharmacological treatments, dietary strategies, and lifestyle changes.
Collapse
Affiliation(s)
- Mario García-Domínguez
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain;
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
18
|
Zhang Y, Tang D, Zhang N, Xiang Y, Hu Y, Qian W, Baima Y, Ding X, Wang Z, Yin J, Xiao X, Zhao X. Lifestyles and their relative contribution to biological aging across multiple-organ systems: Change analysis from the China Multi-Ethnic Cohort study. eLife 2025; 13:RP99924. [PMID: 40052974 PMCID: PMC11888596 DOI: 10.7554/elife.99924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
Background Biological aging exhibits heterogeneity across multi-organ systems. However, it remains unclear how is lifestyle associated with overall and organ-specific aging and which factors contribute most in Southwest China. Methods This study involved 8396 participants who completed two surveys from the China Multi-Ethnic Cohort (CMEC) study. The healthy lifestyle index (HLI) was developed using five lifestyle factors: smoking, alcohol, diet, exercise, and sleep. The comprehensive and organ-specific biological ages (BAs) were calculated using the Klemera-Doubal method based on longitudinal clinical laboratory measurements, and validation were conducted to select BA reflecting related diseases. Fixed effects model was used to examine the associations between HLI or its components and the acceleration of validated BAs. We further evaluated the relative contribution of lifestyle components to comprehension and organ systems BAs using quantile G-computation. Results About two-thirds of participants changed HLI scores between surveys. After validation, three organ-specific BAs (the cardiopulmonary, metabolic, and liver BAs) were identified as reflective of specific diseases and included in further analyses with the comprehensive BA. The health alterations in HLI showed a protective association with the acceleration of all BAs, with a mean shift of -0.19 (95% CI -0.34, -0.03) in the comprehensive BA acceleration. Diet and smoking were the major contributors to overall negative associations of five lifestyle factors, with the comprehensive BA and metabolic BA accounting for 24% and 55% respectively. Conclusions Healthy lifestyle changes were inversely related to comprehensive and organ-specific biological aging in Southwest China, with diet and smoking contributing most to comprehensive and metabolic BA separately. Our findings highlight the potential of lifestyle interventions to decelerate aging and identify intervention targets to limit organ-specific aging in less-developed regions. Funding This work was primarily supported by the National Natural Science Foundation of China (Grant No. 82273740) and Sichuan Science and Technology Program (Natural Science Foundation of Sichuan Province, Grant No. 2024NSFSC0552). The CMEC study was funded by the National Key Research and Development Program of China (Grant No. 2017YFC0907305, 2017YFC0907300). The sponsors had no role in the design, analysis, interpretation, or writing of this article.
Collapse
Affiliation(s)
- Yuan Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
| | - Dan Tang
- West China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
- Xiamen Center for Disease Control and PreventionXiamenChina
| | - Ning Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
| | - Yi Xiang
- West China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
| | - Yifan Hu
- West China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
| | - Wen Qian
- Chengdu Center for Disease Control and PreventionChengduChina
| | | | - Xianbin Ding
- Chongqing Municipal Centre for Disease Control and PreventionChongqingChina
| | - Ziyun Wang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical UniversityGuiyangChina
| | - Jianzhong Yin
- School of Public Health, Kunming Medical UniversityKunmingChina
| | - Xiong Xiao
- West China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
| | - Xing Zhao
- West China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
19
|
Pfeffer LK, Fischbach F, Heesen C, Friese MA. Current state and perspectives of CAR T cell therapy in central nervous system diseases. Brain 2025; 148:723-736. [PMID: 39530593 DOI: 10.1093/brain/awae362] [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: 07/19/2024] [Revised: 10/03/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
B cell-directed CAR T cell therapy has fundamentally changed the treatment of haematological malignancies, and its scope of application is rapidly expanding to include other diseases such as solid tumours or autoimmune disorders. Therapy-refractoriness remains an important challenge in various inflammatory and non-inflammatory disorders of the CNS. The reasons for therapy failure are diverse and include the limited access current therapies have to the CNS, as well as enormous inter- and intra-individual disease heterogeneity. The tissue-penetrating properties of CAR T cells make them a promising option for overcoming this problem and tackling pathologies directly within the CNS. First application of B cell-directed CAR T cells in neuromyelitis optica spectrum disorder and multiple sclerosis patients has recently revealed promising outcomes, expanding the potential of CAR T cell therapy to encompass CNS diseases. Additionally, the optimization of CAR T cells for the therapy of gliomas is a growing field. As a further prospect, preclinical data reveal the potential benefits of CAR T cell therapy in the treatment of primary neurodegenerative diseases such as Alzheimer's disease. Considering the biotechnological optimizations in the field of T cell engineering, such as extension to target different antigens or variation of the modified T cell subtype, new and promising fields of CAR T cell application are rapidly opening up. These innovations offer the potential to address the complex pathophysiological properties of CNS diseases. To use CAR T cell therapy optimally to treat CNS diseases in the future while minimizing therapy risks, further mechanistic research and prospective controlled trials are needed to assess seriously the disease and patient-specific risk-benefit ratio.
Collapse
Affiliation(s)
- Lena Kristina Pfeffer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Felix Fischbach
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christoph Heesen
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
20
|
Dai Q, Sun H, Yang X, Chen S, Zhang X, Yin Z, Zhao X, Wu S, Cao Z, Wu Y, Ma X. Association of clinical biomarker-based biological age and aging trajectory with cardiovascular disease and all-cause mortality in Chinese adults: a population-based cohort study. BMC Public Health 2025; 25:868. [PMID: 40038610 DOI: 10.1186/s12889-025-22114-7] [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: 11/21/2024] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Evidence on the association of clinical biomarker-based biological age (BA) with cardiovascular disease (CVD) and mortality remains insufficient, particularly concerning aging trajectories' relationship with these two outcomes. METHODS Seventy-five thousand five hundred thirty-seven Chinese adults from the Kailuan study who participated in the first checkup (2006-2007) were included. BA was predicted by 32 clinical indicators using deep neural networks models. Aging status was divided into decelerated, accelerated, and normal aging based on BA in the first checkup. Six aging trajectories were developed in the initial three checkups. CVD and mortality were followed up till December 31, 2021. RESULTS After adjusting for chronological age, sex, education level, occupation, physical activity, smoking status, alcohol consumption, salt consumption habit, history of hypertension, diabetes, and dyslipidemia, as well as the use of antihypertensive, antidiabetic, and lipid-lowering drugs, Cox proportional hazard models showed that relative to normal aging, accelerated aging was a risk factor for CVD (adjusted hazard ratio [aHR], 1.17 [95% CI 1.11-1.23]) and mortality (aHR, 1.17 [1.12-1.22]), while participants with decelerated aging had a lower risk for CVD (aHR, 0.85 [0.80-0.90]) and mortality (aHR, 0.86 [0.82-0.90]). Relative to low-stable trajectory, other aging trajectories associated with higher risk of CVD and death, and high-stable trajectory associated with the highest risk of CVD (aHR, 1.62 [1.45-1.81]) and mortality (aHR, 1.55 [1.41-1.71]). Relative to high-stable trajectory, high-decreasing trajectory was associated with lower risk of CVD (aHR, 0.76 [0.67-0.86]) and death (aHR, 0.78 [0.70-0.87]), and decreasing-increasing trajectory was associated with lower risk of death (aHR, 0.86 [0.75-0.98]). CONCLUSIONS Accelerated BA aging is associated with a higher risk of CVD and mortality, whereas decelerated aging is associated with a lower risk compared to normal aging. Those persistently at high aging levels are at the highest risk for both CVD and death; conversely, it is the act of lowering and continually maintaining a reduced aging state that effectively mitigates these risks.
Collapse
Affiliation(s)
- Qiaoyun Dai
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Sharing Service Platform, Beijing, China
| | - Huayu Sun
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
- Graduate School, North China University of Science and Technology, Tangshan, China
| | - Xueying Yang
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Sharing Service Platform, Beijing, China
| | - Shuohua Chen
- Graduate School, North China University of Science and Technology, Tangshan, China
| | - Xinyuan Zhang
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Sharing Service Platform, Beijing, China
| | - Zhe Yin
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Sharing Service Platform, Beijing, China
| | - Xiujuan Zhao
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
- Graduate School, North China University of Science and Technology, Tangshan, China
| | - Shouling Wu
- Graduate School, North China University of Science and Technology, Tangshan, China
| | - Zongfu Cao
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China.
- National Human Genetic Resources Sharing Service Platform, Beijing, China.
| | - Yuntao Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, China.
- Graduate School, North China University of Science and Technology, Tangshan, China.
| | - Xu Ma
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China.
- National Human Genetic Resources Sharing Service Platform, Beijing, China.
| |
Collapse
|
21
|
Ribeiro AH, Ribeiro ALP. AI-ECG and prediction of new atrial fibrillation: when the heart tells the age. Eur Heart J 2025; 46:853-855. [PMID: 39657904 DOI: 10.1093/eurheartj/ehae809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Affiliation(s)
- Antonio H Ribeiro
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Antonio Luiz P Ribeiro
- Telehealth Center and Cardiology Service, Hospital das Clínicas, Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena 110, 1° Andar, Ala Sul, Sala 107, Belo Horizonte MG 30130-100, Brazil
- Department of Internal Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
22
|
Wang J, Xu W, Dove A, Salami A, Yang W, Ma X, Bennett DA, Xu W. Influence of lung function on macro- and micro-structural brain changes in mid- and late-life. Int J Surg 2025; 111:2467-2477. [PMID: 39869397 DOI: 10.1097/js9.0000000000002228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/29/2024] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Lung function has been associated with cognitive decline and dementia, but the extent to which lung function impacts brain structural changes remains unclear. We aimed to investigate the association of lung function with structural macro- and micro-brain changes across mid- and late-life. METHODS The study included a total of 37 164 neurologic disorder-free participants aged 40-70 years from the UK Biobank, who underwent brain MRI scans 9 years after baseline. After 2.5 years, a subsample (n = 3895) underwent a second MRI scan. Lung function was assessed using a composite score based on forced expiratory volume in 1 second, forced vital capacity, and peak expiratory flow, and divided into tertiles (i.e., low, moderate, and high). Structural brain volumes (including total brain, gray matter, white matter, hippocampus, and white matter hyperintensities) and diffusion markers (fractional anisotropy [FA] and mean diffusivity [MD]) were assessed. Data were analyzed using linear regression and mixed-effects models. RESULTS Compared to high lung function, low lung function was associated with smaller total brain, gray matter, white matter, and hippocampal volume, as well as lower white matter integrity. Over the 2.5-year follow-up, low lung function was associated with reduced white matter and hippocampal volume, reduced FA, and increased white matter hyperintensity volume and MD. After stratification by age, the associations remained significant among adults aged 40-60 years and 60+ years. CONCLUSION Low lung function is associated with macro- and micro-structural brain changes involving both neurodegenerative and vascular pathologies. This association is significant in both mid- and late-life.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Epidemiology, College of Preventive Medicine,Third Military Medical University, Chongqing, China
| | - Weige Xu
- Department of Radiology, Tianjin Gongan Hospital, Tianjin, China
| | - Abigail Dove
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Alireza Salami
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Wenzhe Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiangyu Ma
- Department of Epidemiology, College of Preventive Medicine,Third Military Medical University, Chongqing, China
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Weili Xu
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Kivimäki M, Frank P, Pentti J, Jokela M, Nyberg ST, Blake A, Lindbohm JV, Oh HSH, Singh-Manoux A, Wyss-Coray T, Partridge L. Proteomic organ-specific ageing signatures and 20-year risk of age-related diseases: the Whitehall II observational cohort study. Lancet Digit Health 2025; 7:e195-e204. [PMID: 40015764 DOI: 10.1016/j.landig.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/02/2024] [Accepted: 01/06/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Biological ageing is known to vary among different organs within an individual, but the extent to which advanced ageing of specific organs increases the risk of age-related diseases in the same and other organs remains poorly understood. METHODS In this observational cohort study, to assess the biological age of an individual's organs relative to those of same-aged peers, ie, organ age gaps, we collected plasma samples from 6235 middle-aged (age 45-69 years) participants of the Whitehall II prospective cohort study in London, UK, in 1997-99. Age gaps of nine organs were determined from plasma proteins via SomaScan (SomaLogic; Boulder, CO, USA) using the Python package organage. Following this assessment, we tracked participants for 20 years through linkage to national health records. Study outcomes were 45 individual age-related diseases and multimorbidity. FINDINGS Over 123 712 person-years of observation (mean follow-up 19·8 years [SD 3·6]), after excluding baseline disease cases and adjusting for age, sex, ethnicity, and age gaps in organs other than the one under investigation, individuals with large organ age gaps showed an increased risk of 30 diseases. Six diseases were exclusively associated with accelerated ageing of their respective organ: liver failure (hazard ratio [HR] per SD increment in the organ age gap 2·13 [95% CI 1·41-3·22]), dilated cardiomyopathy (HR 1·65 [1·28-2·12]), chronic heart failure (HR 1·52 [1·40-1·65]), lung cancer (HR 1·29 [1·04-1·59]), agranulocytosis (HR 1·27 [1·07-1·51]), and lymphatic node metastasis (HR 1·23 [1·06-1·43]). 24 diseases were associated with more than one organ age gap or with organ age gaps not directly related to the disease location. Larger age gaps were also associated with elevated HRs of developing two or more diseases affecting different organs within the same individual (ie, multiorgan multimorbidity): 2·03 (1·51-2·74) for the arterial age gap, 1·78 (1·48-2·14) for the kidney age gap, 1·52 (1·38-1·68) for the heart age gap, 1·52 (1·12-2·06) for the brain age gap, 1·43 (1·16-1·78) for the pancreas age gap, 1·37 (1·17-1·61) for the lung age gap, 1·36 (1·26-1·46) for the immune system age gap, and 1·30 (1·18-1·42) for the liver age gap. INTERPRETATION Advanced proteomic organ ageing is associated with the long-term risk of age-related diseases. In most cases, faster ageing of a specific organ increases susceptibility to morbidity affecting multiple organs. FUNDING Wellcome Trust, UK Medical Research Council, National Institute for Aging, Academy of Finland.
Collapse
Affiliation(s)
- Mika Kivimäki
- UCL Brain Sciences, University College London, London, UK; Clinicum, University of Helsinki, Helsinki, Finland.
| | - Philipp Frank
- UCL Brain Sciences, University College London, London, UK
| | - Jaana Pentti
- Clinicum, University of Helsinki, Helsinki, Finland; Department of Public Health and Centre for Population Health Research, University of Turku, Turku, Finland
| | - Markus Jokela
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | | | - Acer Blake
- Institute of Healthy Ageing, University College London, London, UK; Research Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Joni V Lindbohm
- UCL Brain Sciences, University College London, London, UK; Clinicum, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, The Klarman Cell Observatory, Cambridge, MA, USA
| | - 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
| | - Archana Singh-Manoux
- UCL Brain Sciences, University College London, London, UK; Epidemiology of Ageing and Neurodegenerative Diseases, Université Paris Cité, Inserm U1153, Paris, France
| | - 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, Stanford, CA, USA
| | - Linda Partridge
- Institute of Healthy Ageing, University College London, London, UK; Research Department of Genetics, Evolution and Environment, University College London, London, UK
| |
Collapse
|
24
|
Liberale L, Tual-Chalot S, Sedej S, Ministrini S, Georgiopoulos G, Grunewald M, Bäck M, Bochaton-Piallat ML, Boon RA, Ramos GC, de Winther MPJ, Drosatos K, Evans PC, Ferguson JF, Forslund-Startceva SK, Goettsch C, Giacca M, Haendeler J, Kallikourdis M, Ketelhuth DFJ, Koenen RR, Lacolley P, Lutgens E, Maffia P, Miwa S, Monaco C, Montecucco F, Norata GD, Osto E, Richardson GD, Riksen NP, Soehnlein O, Spyridopoulos I, Van Linthout S, Vilahur G, Wentzel JJ, Andrés V, Badimon L, Benetos A, Binder CJ, Brandes RP, Crea F, Furman D, Gorbunova V, Guzik TJ, Hill JA, Lüscher TF, Mittelbrunn M, Nencioni A, Netea MG, Passos JF, Stamatelopoulos KS, Tavernarakis N, Ungvari Z, Wu JC, Kirkland JL, Camici GG, Dimmeler S, Kroemer G, Abdellatif M, Stellos K. Roadmap for alleviating the manifestations of ageing in the cardiovascular system. Nat Rev Cardiol 2025:10.1038/s41569-025-01130-5. [PMID: 39972009 DOI: 10.1038/s41569-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Ageing of the cardiovascular system is associated with frailty and various life-threatening diseases. As global populations grow older, age-related conditions increasingly determine healthspan and lifespan. The circulatory system not only supplies nutrients and oxygen to all tissues of the human body and removes by-products but also builds the largest interorgan communication network, thereby serving as a gatekeeper for healthy ageing. Therefore, elucidating organ-specific and cell-specific ageing mechanisms that compromise circulatory system functions could have the potential to prevent or ameliorate age-related cardiovascular diseases. In support of this concept, emerging evidence suggests that targeting the circulatory system might restore organ function. In this Roadmap, we delve into the organ-specific and cell-specific mechanisms that underlie ageing-related changes in the cardiovascular system. We raise unanswered questions regarding the optimal design of clinical trials, in which markers of biological ageing in humans could be assessed. We provide guidance for the development of gerotherapeutics, which will rely on the technological progress of the diagnostic toolbox to measure residual risk in elderly individuals. A major challenge in the quest to discover interventions that delay age-related conditions in humans is to identify molecular switches that can delay the onset of ageing changes. To overcome this roadblock, future clinical trials need to provide evidence that gerotherapeutics directly affect one or several hallmarks of ageing in such a manner as to delay, prevent, alleviate or treat age-associated dysfunction and diseases.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Myriam Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Magnus Bäck
- Translational Cardiology, Centre for Molecular Medicine, Department of Medicine Solna, and Department of Cardiology, Heart and Vascular Centre, Karolinska Institutet, Stockholm, Sweden
- Inserm, DCAC, Université de Lorraine, Nancy, France
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - Gustavo Campos Ramos
- Department of Internal Medicine I/Comprehensive Heart Failure Centre, University Hospital Würzburg, Würzburg, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis and Ischaemic Syndromes; Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location AMC, Amsterdam, Netherlands
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul C Evans
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mauro Giacca
- British Heart foundation Centre of Reseach Excellence, King's College London, London, UK
| | - Judith Haendeler
- Cardiovascular Degeneration, Medical Faculty, University Hospital and Heinrich-Heine University, Düsseldorf, Germany
| | - Marinos Kallikourdis
- Adaptive Immunity Lab, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Daniel F J Ketelhuth
- Cardiovascular and Renal Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rory R Koenen
- CARIM-School for Cardiovascular Diseases, Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine & Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Satomi Miwa
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Münster, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu y Sant Pau l, IIB-Sant Pau, Barcelona, Spain
| | - Jolanda J Wentzel
- Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBERCV, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Health and Innovation Research Foundation (FICSI) and Cardiovascular Health and Network Medicine Department, University of Vic (UVIC-UCC), Barcelona, Spain
| | - Athanase Benetos
- Department of Geriatrics, University Hospital of Nancy and Inserm DCAC, Université de Lorraine, Nancy, France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Roma, Italy
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph A Hill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche-DIMI, Università degli Studi di Genova, Genova, Italy
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kimon S Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Tavernarakis
- Medical School, University of Crete, and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Zoltan Ungvari
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm, Institut Universitaire de France, Paris, France
| | | | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
25
|
Amirmoezzi Y, Cropley V, Mansour L S, Seguin C, Zalesky A, Tian YE. Characterizing Brain-Cardiovascular Aging Using Multiorgan Imaging and Machine Learning. J Neurosci 2025; 45:e1440242024. [PMID: 39971581 PMCID: PMC11841759 DOI: 10.1523/jneurosci.1440-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/19/2024] [Accepted: 12/04/2024] [Indexed: 02/21/2025] Open
Abstract
The structure and function of the brain and cardiovascular system change over the lifespan. In this study, we aim to establish the extent to which age-related changes in these two vital organs are linked. Utilizing normative models and data from the UK Biobank, we estimate biological ages for the brain and heart for 2,904 middle-aged and older healthy adults, including both males and females. Biological ages were based on multiple structural, morphological, and functional features derived from brain and cardiovascular imaging modalities. We find that cardiovascular aging, particularly aging of its functional capacity and physiology, is selectively associated with the aging of specific brain networks, including the salience, default mode, and somatomotor networks as well as the subcortex. Our work provides unique insight into brain-heart relationships and may facilitate an improved understanding of the increased co-occurrence of brain and heart diseases in aging.
Collapse
Affiliation(s)
- Yalda Amirmoezzi
- Systems Lab, Department of Psychiatry, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Vanessa Cropley
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria 3052, Australia
- Orygen, Parkville, Victoria 3052, Australia
| | - Sina Mansour L
- Systems Lab, Department of Psychiatry, The University of Melbourne, Melbourne, Victoria 3010, Australia
- Centre for Sleep & Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117549, Singapore
| | - Caio Seguin
- Systems Lab, Department of Psychiatry, The University of Melbourne, Melbourne, Victoria 3010, Australia
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405
| | - Andrew Zalesky
- Systems Lab, Department of Psychiatry, The University of Melbourne, Melbourne, Victoria 3010, Australia
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Ye Ella Tian
- Systems Lab, Department of Psychiatry, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
26
|
Zhao YP, Liu WH, Zhang QC. Determinants of generalized anxiety and construction of a predictive model in patients with chronic obstructive pulmonary disease. World J Psychiatry 2025; 15:98447. [PMID: 39974476 PMCID: PMC11758039 DOI: 10.5498/wjp.v15.i2.98447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/06/2024] [Accepted: 12/26/2024] [Indexed: 01/14/2025] Open
Abstract
BACKGROUND Patients with chronic obstructive pulmonary disease (COPD) frequently experience exacerbations requiring multiple hospitalizations over prolonged disease courses, which predispose them to generalized anxiety disorder (GAD). This comorbidity exacerbates breathing difficulties, activity limitations, and social isolation. While previous studies predominantly employed the GAD 7-item scale for screening, this approach is somewhat subjective. The current literature on predictive models for GAD risk in patients with COPD is limited. AIM To construct and validate a GAD risk prediction model to aid healthcare professionals in preventing the onset of GAD. METHODS This retrospective analysis encompassed patients with COPD treated at our institution from July 2021 to February 2024. The patients were categorized into a modeling (MO) group and a validation (VA) group in a 7:3 ratio on the basis of the occurrence of GAD. Univariate and multivariate logistic regression analyses were utilized to construct the risk prediction model, which was visualized using forest plots. The model's performance was evaluated using Hosmer-Lemeshow (H-L) goodness-of-fit test and receiver operating characteristic (ROC) curve analysis. RESULTS A total of 271 subjects were included, with 190 in the MO group and 81 in the VA group. GAD was identified in 67 patients with COPD, resulting in a prevalence rate of 24.72% (67/271), with 49 cases (18.08%) in the MO group and 18 cases (22.22%) in the VA group. Significant differences were observed between patients with and without GAD in terms of educational level, average household income, smoking history, smoking index, number of exacerbations in the past year, cardiovascular comorbidities, disease knowledge, and personality traits (P < 0.05). Multivariate logistic regression analysis revealed that lower education levels, household income < 3000 China yuan, smoking history, smoking index ≥ 400 cigarettes/year, ≥ two exacerbations in the past year, cardiovascular comorbidities, complete lack of disease information, and introverted personality were significant risk factors for GAD in the MO group (P < 0.05). ROC analysis indicated that the area under the curve for predicting GAD in the MO and VA groups was 0.978 and 0.960. The H-L test yielded χ 2 values of 6.511 and 5.179, with P = 0.275 and 0.274. Calibration curves demonstrated good agreement between predicted and actual GAD occurrence risks. CONCLUSION The developed predictive model includes eight independent risk factors: Educational level, household income, smoking history, smoking index, number of exacerbations in the past year, presence of cardiovascular comorbidities, level of disease knowledge, and personality traits. This model effectively predicts the onset of GAD in patients with COPD, enabling early identification of high-risk individuals and providing a basis for early preventive interventions by nursing staff.
Collapse
Affiliation(s)
- Yi-Pu Zhao
- Department of Respiratory and Critical Care Medicine, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Wei-Hua Liu
- Department of Nursing, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Qun-Cheng Zhang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| |
Collapse
|
27
|
Fancourt D, Bloomberg M, Steptoe A. Social connections are differentially related to perceived and physiological age acceleration amongst older adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.03.25320261. [PMID: 39973988 PMCID: PMC11838645 DOI: 10.1101/2025.02.03.25320261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Human social connections are complex ecosystems formed of structural, functional and quality components. Deficits in social connections are associated with adverse age-related health outcomes, but we know little about the ageing-related mechanistic processes underlying this. Using data from 7,047 adults aged 50+ in the English Longitudinal Study of Ageing, we explored associations between diverse aspects of social deficits and both perceived and physiological age acceleration, which provide complementary psycho-behavioural and biological mechanistic explanations. We created and validated a novel physiological ageing index using clinical indicators pertaining to the cardiovascular, respiratory, haematologic, metaboloic and cognitive systems using principal component analysis. Doubly-robust estimations using inverse-probability-weighted regression adjustment estimators showed that living alone, low social integration and high social isolation were risk factors for physiological age acceleration, with those who lived alone on average 1.9 years older than those who lived with others (95% CI 0.9-3.0 years older; 32% greater age acceleration than people who live with others). However, social deficits were not related to accelerations in perceived age. Analyses were robust to multiple sensitivity analyses and maintained four years later. These findings provide important mechanistic insight that helps to explain the relationship between social deficits and age-related morbitidy and mortality outcomes.
Collapse
|
28
|
Peng T, Xiang J, Tian Y, Tang X, Wang L, Gao L, Luo OJ, Huang L, Chen G. Lycium barbarum glycopeptide ameliorates aging phenotypes and enhances cardiac metabolism by activating the PINK1/Parkin-mediated mitophagy pathway in D-galactose-induced mice. Exp Gerontol 2025; 200:112686. [PMID: 39827719 DOI: 10.1016/j.exger.2025.112686] [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: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Aging is a complex biological process that disrupts tissue structure and impairs physiological function, which contributes to the development of age-related diseases such as cardiovascular disorders. However, effective treatment strategies are lacking. OBJECTIVE To investigate the geroprotective effects of Lycium barbarum glycopeptide (LbGp) and its potential mechanisms in a D-galactose-induced accelerated aging mouse model. METHODS Mice were subcutaneously injected with D-galactose (500 mg/kg/day) for 12 weeks to induce aging, while LbGp was orally administered (100 mg/kg/day) throughout the study. The geroprotective effects of LbGp were assessed by behavioral tests, cardiac echocardiography, pathohistological and transcriptomic analyses. Transmission electron microscopy was used to observe the ultrastructure of mitochondria. Mitochondrial stress assays and JC-1 fluorescent probe were conducted to evaluate mitochondrial function. Flow cytometer and western blot were performed to assess mitophagy flux. RESULTS LbGp treatment improved the aging phenotypes of D-galactose-induced mice, with a pronounced enhancement in cardiac function compared to neurocognitive and skeletal muscle functions. Transcriptome analysis indicated that LbGp ameliorated energy metabolism in the heart. Mitochondrial assays revealed LbGp improved mitochondrial function and preserved structural integrity of the mitochondrial inner membrane. LbGp attenuated mitochondrial fission and restored impaired PINK1/Parkin-mediated mitophagy pathway caused by D-galactose in cardiomyocytes. CONCLUSION LbGp can ameliorate aging phenotypes and enhance cardiac metabolism by activating the PINK1/Parkin-mediated mitophagy pathway in D-galactose-induced mice. These findings underscore its potential as a therapeutic agent for aging and aging-related cardiovascular diseases.
Collapse
Affiliation(s)
- Tianchan Peng
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jian Xiang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yun Tian
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiaogen Tang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lina Wang
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China
| | - Oscar Junhong Luo
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China
| | - Li'an Huang
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China.
| |
Collapse
|
29
|
Stylianou O, Meixner JM, Schlick T, Krüger CM. Whole-body networks: a holistic approach for studying aging. GeroScience 2025:10.1007/s11357-025-01540-w. [PMID: 39875752 DOI: 10.1007/s11357-025-01540-w] [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: 08/28/2024] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
Aging is a multi-organ disease, yet the traditional approach has been to study each organ in isolation. Such organ-specific studies have provided invaluable information regarding its pathomechanisms. However, an overall picture of the whole-body network (WBN) during aging is still incomplete. In this study, we analyzed the functional magnetic resonance imaging blood-oxygen level-dependent, respiratory rate and heart rate time series of a young and an elderly group during eyes-open resting-state. We constructed WBNs by exploring the time-lagged coupling between the different organs. First, we showed that our analytical pipeline could identify regional differences in the networks of both cohorts, allowing us to proceed with the remaining analyses. The comparison of the WBNs revealed a complex relationship where some connections were stronger and some weaker in the elderly. Finally, the interconnectivity and segregation of the WBNs were negatively correlated with the short-term memory and verbal learning of the young participants. This study: i) validated our methodology, ii) identified differences in the WBNs of the two groups and iii) showed correlations of WBNs with behavioral measures. In conclusion, the concept of WBN shows great potential for the understanding of aging and age-related diseases.
Collapse
Affiliation(s)
- Orestis Stylianou
- Department of Surgery, Immanuel Clinic Rüdersdorf, University Clinic of Brandenburg Medical School, Berlin, Germany.
| | - Johannes M Meixner
- Department of Psychology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Tilman Schlick
- Department of Surgery, Immanuel Clinic Rüdersdorf, University Clinic of Brandenburg Medical School, Berlin, Germany
| | - Colin M Krüger
- Department of Surgery, Immanuel Clinic Rüdersdorf, University Clinic of Brandenburg Medical School, Berlin, Germany.
- Department of Surgery, Clinic of General-, Visceral-, Vascular and Thoracic Surgery, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
30
|
Harinath G, Lee V, Nyquist A, Moel M, Wouters M, Hagemeier J, Verkennes B, Tacubao C, Nasher S, Kauppi K, Morgan SL, Isman A, Zalzala S. The bioavailability and blood levels of low-dose rapamycin for longevity in real-world cohorts of normative aging individuals. GeroScience 2025:10.1007/s11357-025-01532-w. [PMID: 39873920 DOI: 10.1007/s11357-025-01532-w] [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: 08/05/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
Rapamycin, also known as sirolimus, has demonstrated great potential for application in longevity medicine. However, the dynamics of low-dose rapamycin bioavailability, and any differences in bioavailability for different formulations (e.g., compounded or commercial), remain poorly understood. We thus explored rapamycin bioavailability in two real-world cohorts to begin providing a foundational understanding of differences in effects between formulations over time. The small trial study cohort was utilized to explore the blood rapamycin levels of commercial (n = 44, dosages 2, 3, 6, or 8 mg) or compounded (n = 23, dosages 5, 10, or 15 mg) rapamycin 24 h after dose self-administration. Results suggested dose-to-blood level relationships were linear for both formulations, though compounded had a lower bioavailability per milligram of rapamycin (estimated to be 31.03% of the same dose of commercial). While substantial inter-individual heterogeneity in blood rapamycin levels was observed for both formulations, repeat tests for individuals over time demonstrated relative consistency. Extending exploration to 316 real-world longevity rapamycin users from the AgelessRx Observational Research Database produced similar findings, and additionally suggested that blood rapamycin levels peak after 2 days with gradual decline thereafter. Taken together, our findings suggest that individualized dosing and routine monitoring of blood rapamycin levels should be utilized to ensure optimal dosing and efficacy for healthy longevity.
Collapse
Affiliation(s)
- Girish Harinath
- AgelessRx, Ann Arbor, MI, USA
- Division of Research and Applied Sciences, AgelessRx, Ann Arbor, MI, USA
| | - Virginia Lee
- AgelessRx, Ann Arbor, MI, USA
- Division of Research and Applied Sciences, AgelessRx, Ann Arbor, MI, USA
| | | | | | | | | | - Brandon Verkennes
- AgelessRx, Ann Arbor, MI, USA
- Data and Analytics Division, AgelessRx, Ann Arbor, MI, USA
| | - Colleen Tacubao
- AgelessRx, Ann Arbor, MI, USA
- Data and Analytics Division, AgelessRx, Ann Arbor, MI, USA
| | - Sayem Nasher
- AgelessRx, Ann Arbor, MI, USA
- Data and Analytics Division, AgelessRx, Ann Arbor, MI, USA
| | - Krister Kauppi
- Rapamycin Longevity Lab, Gothenburg, Västra Götaland County, Sweden
| | - Stefanie L Morgan
- AgelessRx, Ann Arbor, MI, USA.
- Division of Research and Applied Sciences, AgelessRx, Ann Arbor, MI, USA.
| | | | | |
Collapse
|
31
|
Kou M, Ma H, Wang X, Heianza Y, Qi L. Joint association of objective and subjective aging with premature mortality. NPJ AGING 2025; 11:3. [PMID: 39863635 PMCID: PMC11762265 DOI: 10.1038/s41514-024-00190-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/15/2024] [Indexed: 01/27/2025]
Abstract
Objective and subjective aging indicators reflect diverse biological and psychosocial processes, yet their combined association with premature mortality remains underexplored. This study aimed to investigate the association between a multidomain framework of aging indicators and premature mortality, addressing gaps in understanding cumulative effects. We included 369,741 UK Biobank participants initially free of cardiovascular disease (CVD) and cancer, followed until December 31, 2022. Four indicators, hearing loss, tooth loss, falls and subjective aging, were counted, and their joint associations with all-cause and cause-specific premature mortality were analyzed using the Cox proportional hazard models. During a median follow-up of 13.74 years, we documented 22,934 premature mortality. Participants with all indicators had an 81% (95%CI: 59-107%), 96% (47-160%), 55% (26-91%), and 114% (73-165%) higher risk of all-cause, CVD, cancer, and other-cause premature mortality, respectively, compared to those without indicators. The associations were particularly elevated among younger participants, those with unhealthy lifestyles, and those of lower socioeconomic status (P for interactions <0.05). Additive interaction with frailty contributed an additional 16.08% (7.91-24.25%) risk of premature mortality. Findings were replicated in the Health and Retirement Study, supporting the robustness of the multidomain aging framework. This study highlights the potential of integrating objective and subjective aging indicators to refine risk assessments and inform interventions targeting aging-related diseases.
Collapse
Affiliation(s)
- Minghao Kou
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Hao Ma
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Xuan Wang
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Yoriko Heianza
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lu Qi
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
32
|
Zhang X, Wang C, Pi X, Li B, Ding Y, Yu H, Sun J, Wang P, Chen Y, Wang Q, Zhang C, Meng X, Chen G, Wang D, Wang Z, Mu Z, Song H, Zhang J, Niu S, Han Z, Ren L. Bionic Recognition Technologies Inspired by Biological Mechanosensory Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2418108. [PMID: 39838736 DOI: 10.1002/adma.202418108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/23/2024] [Indexed: 01/23/2025]
Abstract
Mechanical information is a medium for perceptual interaction and health monitoring of organisms or intelligent mechanical equipment, including force, vibration, sound, and flow. Researchers are increasingly deploying mechanical information recognition technologies (MIRT) that integrate information acquisition, pre-processing, and processing functions and are expected to enable advanced applications. However, this also poses significant challenges to information acquisition performance and information processing efficiency. The novel and exciting mechanosensory systems of organisms in nature have inspired us to develop superior mechanical information bionic recognition technologies (MIBRT) based on novel bionic materials, structures, and devices to address these challenges. Herein, first bionic strategies for information pre-processing are presented and their importance for high-performance information acquisition is highlighted. Subsequently, design strategies and considerations for high-performance sensors inspired by mechanoreceptors of organisms are described. Then, the design concepts of the neuromorphic devices are summarized in order to replicate the information processing functions of a biological nervous system. Additionally, the ability of MIBRT is investigated to recognize basic mechanical information. Furthermore, further potential applications of MIBRT in intelligent robots, healthcare, and virtual reality are explored with a view to solve a range of complex tasks. Finally, potential future challenges and opportunities for MIBRT are identified from multiple perspectives.
Collapse
Affiliation(s)
- Xiangxiang Zhang
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Changguang Wang
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Xiang Pi
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Bo Li
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
- The National Key Laboratory of Automotive Chassis Integration and Bionics (ACIB), College of Biological and Agricultural Engineering, Jilin University, Changchun, 130022, China
| | - Yuechun Ding
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Hexuan Yu
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Jialue Sun
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Pinkun Wang
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - You Chen
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Qun Wang
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Changchao Zhang
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Xiancun Meng
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Guangjun Chen
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Dakai Wang
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Ze Wang
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Zhengzhi Mu
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Honglie Song
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
| | - Junqiu Zhang
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
- The National Key Laboratory of Automotive Chassis Integration and Bionics (ACIB), College of Biological and Agricultural Engineering, Jilin University, Changchun, 130022, China
- Institute of Structured and Architected Materials, Liaoning Academy of Materials, Shenyang, 110167, China
| | - Shichao Niu
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
- The National Key Laboratory of Automotive Chassis Integration and Bionics (ACIB), College of Biological and Agricultural Engineering, Jilin University, Changchun, 130022, China
- Institute of Structured and Architected Materials, Liaoning Academy of Materials, Shenyang, 110167, China
| | - Zhiwu Han
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
- The National Key Laboratory of Automotive Chassis Integration and Bionics (ACIB), College of Biological and Agricultural Engineering, Jilin University, Changchun, 130022, China
- Institute of Structured and Architected Materials, Liaoning Academy of Materials, Shenyang, 110167, China
| | - Luquan Ren
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, Jilin, 130022, China
- The National Key Laboratory of Automotive Chassis Integration and Bionics (ACIB), College of Biological and Agricultural Engineering, Jilin University, Changchun, 130022, China
- Institute of Structured and Architected Materials, Liaoning Academy of Materials, Shenyang, 110167, China
| |
Collapse
|
33
|
Wilczok D. Deep learning and generative artificial intelligence in aging research and healthy longevity medicine. Aging (Albany NY) 2025; 17:251-275. [PMID: 39836094 PMCID: PMC11810058 DOI: 10.18632/aging.206190] [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: 09/23/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
With the global population aging at an unprecedented rate, there is a need to extend healthy productive life span. This review examines how Deep Learning (DL) and Generative Artificial Intelligence (GenAI) are used in biomarker discovery, deep aging clock development, geroprotector identification and generation of dual-purpose therapeutics targeting aging and disease. The paper explores the emergence of multimodal, multitasking research systems highlighting promising future directions for GenAI in human and animal aging research, as well as clinical application in healthy longevity medicine.
Collapse
Affiliation(s)
- Dominika Wilczok
- Duke University, Durham, NC 27708, USA
- Duke Kunshan University, Kunshan, Jiangsu 215316, China
| |
Collapse
|
34
|
Angelini A, Garcia Marquez G, Malovannaya A, Fiorotto ML, Saltzman A, Jain A, Trial J, Taffet GE, Cieslik KA. Sex Differences in Response to Diet Enriched With Glutathione Precursors in the Aging Heart. J Gerontol A Biol Sci Med Sci 2025; 80:glae258. [PMID: 39492659 PMCID: PMC11788829 DOI: 10.1093/gerona/glae258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Indexed: 11/05/2024] Open
Abstract
Common features of the aging heart are dysregulated metabolism, inflammation, and fibrosis. Elevated oxidative stress is another hallmark of cardiac aging that can exacerbate each of these conditions. We hypothesize that by increasing natural antioxidant levels (glutathione), we will improve cardiac function. Twenty-one-month-old mice were fed glycine and N-acetyl cysteine (GlyNAC; glutathione precursors)-supplemented or control diets for 12 weeks. Heart function was monitored longitudinally, and the exercise performance was determined at the end of the study. We found that the GlyNAC diet was beneficial for old male but not old female mice, leading to an increase of Ndufb8 expression (a subunit of the mitochondrial respiratory chain complex), and higher enzymatic activity for CPT1b and CrAT, 2 carnitine acyltransferases that are critical to cardiomyocyte metabolism. Although no quantifiable change of collagen turnover was detected, hearts from GlyNAC-fed old males exhibited a slight but significant enrichment in Fmod, a protein that can inhibit collagen fibril formation, possibly reducing extracellular matrix stiffness and thus improving diastolic function. Cardiac diastolic function was modestly improved in males but not females, and surprisingly GlyNAC-fed female mice showed a decline in exercise performance. In summary, our work supports the concept that aged male and female hearts are phenotypically different. These basic differences may affect the response to pharmacological and diet interventions, including antioxidants.
Collapse
Affiliation(s)
- Aude Angelini
- Department of Medicine, Houston Methodist Hospital and Houston Methodist Research Institute, Houston, Texas, USA
- Section of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Grecia Garcia Marquez
- Department of Medicine, Houston Methodist Hospital and Houston Methodist Research Institute, Houston, Texas, USA
- Section of Geriatrics and Palliative Medicine, Department of Medicine, and Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Anna Malovannaya
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - Marta L Fiorotto
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Alexander Saltzman
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - Antrix Jain
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - JoAnn Trial
- Department of Medicine, Houston Methodist Hospital and Houston Methodist Research Institute, Houston, Texas, USA
- Section of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - George E Taffet
- Department of Medicine, Houston Methodist Hospital and Houston Methodist Research Institute, Houston, Texas, USA
- Section of Geriatrics and Palliative Medicine, Department of Medicine, and Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Katarzyna A Cieslik
- Department of Medicine, Houston Methodist Hospital and Houston Methodist Research Institute, Houston, Texas, USA
- Section of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
35
|
Ji L, Zhang J. Complex interactions and composite burden of risk factors in vascular cognitive impairment. J Neurol Sci 2025; 468:123367. [PMID: 39733713 DOI: 10.1016/j.jns.2024.123367] [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: 09/10/2024] [Revised: 11/23/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
Vascular cognitive impairment (VCI) stresses the vascular contributions to cognitive decline, ranging from mild to major forms. Except for symptomatic treatment for relevant vascular diseases, the other recommended strategy is to intervene in key vascular risk factors (VRFs) as early as possible. A considerable amount of previous research delineated the association of a specific factor with dementia, involving each risk factor discussed in the present review. However, due to the heterogeneity and complexity of VCI, managing a single factor is insufficient to reduce its incidence and prevalence. Ongoing studies suggest differences in the impact of various combinations of risk factors on dementia. Here in this review, we aimed to provide an updated overview of clinical evidence and implications of complex interactions among various risk factors of VCI, including common VRFs and modifiable dementia-related risk factors. Understating the effect of comorbid risk factors on VCI and underlying mechanisms of them during VCI progression is essential for identifying high-risk population and developing preventive strategies. Furthermore, we summarized common composite risk scores and models used for risk evaluation and prediction of VCI, involving conventional risk scores, subclinical vascular composites, and novel risk models driven by intelligent algorithms. Lastly, we discussed potential gaps and research directions on searching specific clinical risk profiles, constructing effective risk scores, and implementing targeted risk interventions.
Collapse
Affiliation(s)
- Linna Ji
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
36
|
Jamadar SD, Behler A, Deery H, Breakspear M. The metabolic costs of cognition. Trends Cogn Sci 2025:S1364-6613(24)00319-X. [PMID: 39809687 DOI: 10.1016/j.tics.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025]
Abstract
Cognition and behavior are emergent properties of brain systems that seek to maximize complex and adaptive behaviors while minimizing energy utilization. Different species reconcile this trade-off in different ways, but in humans the outcome is biased towards complex behaviors and hence relatively high energy use. However, even in energy-intensive brains, numerous parsimonious processes operate to optimize energy use. We review how this balance manifests in both homeostatic processes and task-associated cognition. We also consider the perturbations and disruptions of metabolism in neurocognitive diseases.
Collapse
Affiliation(s)
- Sharna D Jamadar
- School of Psychological Sciences, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, Australia; Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia.
| | - Anna Behler
- School of Psychological Sciences, College of Engineering, Science, and the Environment, University of Newcastle, Newcastle, New South Wales, Australia
| | - Hamish Deery
- School of Psychological Sciences, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, Australia; Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Michael Breakspear
- School of Psychological Sciences, College of Engineering, Science, and the Environment, University of Newcastle, Newcastle, New South Wales, Australia; School of Public Health and Medicine, College of Medicine, Health and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
37
|
Santisteban MM, Iadecola C. The pathobiology of neurovascular aging. Neuron 2025; 113:49-70. [PMID: 39788087 DOI: 10.1016/j.neuron.2024.12.014] [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: 11/07/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025]
Abstract
As global life expectancy increases, age-related brain diseases such as stroke and dementia have become leading causes of death and disability. The aging of the neurovasculature is a critical determinant of brain aging and disease risk. Neurovascular cells are particularly vulnerable to aging, which induces significant structural and functional changes in arterial, venous, and lymphatic vessels. Consequently, neurovascular aging impairs oxygen and glucose delivery to active brain regions, disrupts endothelial transport mechanisms essential for blood-brain exchange, compromises proteostasis by reducing the clearance of potentially toxic proteins, weakens immune surveillance and privilege, and deprives the brain of key growth factors required for repair and renewal. In this review, we examine the effects of neurovascular aging on brain function and its role in stroke, vascular cognitive impairment, and Alzheimer's disease. Finally, we discuss key unanswered questions that must be addressed to develop neurovascular strategies aimed at promoting healthy brain aging.
Collapse
Affiliation(s)
- Monica M Santisteban
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
38
|
Xu W, Li L, Cao Z, Ye J, Gu X. Circadian Rhythms and Lung Cancer in the Context of Aging: A Review of Current Evidence. Aging Dis 2025:AD.2024.1188. [PMID: 39812541 DOI: 10.14336/ad.2024.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
Circadian rhythm is the internal homeostatic physiological clock that regulates the 24-hour sleep/wake cycle. This biological clock helps to adapt to environmental changes such as light, dark, temperature, and behaviors. Aging, on the other hand, is a process of physiological changes that results in a progressive decline in cells, tissues, and other vital systems of the body. Both aging and the circadian clock are highly interlinked phenomena with a bidirectional relationship. The process of aging leads to circadian disruptions while dysfunctional circadian rhythms promote age-related complications. Both processes involve diverse physiological, molecular, and cellular changes such as modifications in the DNA repair mechanisms, mechanisms, ROS generation, apoptosis, and cell proliferation. This review aims to examine the role of aging and circadian rhythms in the context of lung cancer. This will also review the existing literature on the role of circadian disruptions in the process of aging and vice versa. Various molecular pathways and genes such as BMAL1, SIRT1, HLF, and PER1 and their implications in aging, circadian rhythms, and lung cancer will also be discussed.
Collapse
Affiliation(s)
- Wenhui Xu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Lei Li
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Zhendong Cao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Jinghong Ye
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Xuyu Gu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
39
|
Roberts AL, Ratanatharathorn A, Chibnik L, Zhu Y, Jha S, Kang JH, Wolf EJ, Kubzansky LD, Koenen KC. No association of posttraumatic stress disorder with epigenetic aging in women at mid-life: A longitudinal cohort study. Brain Behav Immun 2025; 123:672-680. [PMID: 39424013 DOI: 10.1016/j.bbi.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/16/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024] Open
Abstract
Posttraumatic stress disorder (PTSD) is associated with mortality and increased risk of diseases of aging, but underlying mechanisms remain unclear. We examine associations of PTSD with one potential pathway, accelerated epigenetic aging. In a longitudinal cohort of trauma-exposed middle-aged women (n = 831, n observations = 1,516), we examined cross-sectional and longitudinal associations between PTSD, with and without comorbid depression, and epigenetic aging measured by six clocks at two time points approximately 13.5 years apart: Hannum, Horvath, PhenoAge, GrimAge, DunedinPoAM, and DunedinPACE. We further examined associations of 3 well-established predictors of aging and mortality also linked with PTSD, namely, body mass index (BMI), diet quality, and physical activity, with epigenetic aging. Cross-sectionally, across all six clocks, epigenetic aging in women with PTSD alone, depression alone, and co-occurring depression and PTSD did not differ from the reference group of women without PTSD or depression in analyses adjusted for age, self-reported race, cell proportions, and ancestry principal components. In longitudinal analyses, we similarly did not find any difference in change in epigenetic age over time by PTSD and depression status at baseline. Among the health factors, in cross-sectional analyses, higher BMI was significantly and consistently associated with greater epigenetic aging measured by the PhenoAge, GrimAge, DunedinPoAM, and DunedinPACE clocks, but not measured by the Hannum or Horvath clocks. Physical activity was not consistently associated with epigenetic aging measured by Hannum, Horvath, PhenoAge, or GrimAge. In analyses with the DunedinPoAm and DunedinPACE clocks, women who reported exercise equivalent to 1 or more hours/week walking had slower epigenetic aging than women with less exercise. Diet quality was not consistently associated with epigenetic aging measured by any of the clocks. Our data do not provide evidence that biological aging, as measured by any of the six epigenetic clocks, is a pathway linking PTSD with mortality and diseases of aging.
Collapse
Affiliation(s)
- Andrea L Roberts
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | | | - Lori Chibnik
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Yiwen Zhu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Shaili Jha
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jae H Kang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Erika J Wolf
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Laura D Kubzansky
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Stanley Center for Psychiatric Research, Boston Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Boston MA, USA
| |
Collapse
|
40
|
Fan G, Liu Q, Bi J, Fang Q, Luo F, Huang X, Li H, Guo W, Liu B, Yan L, Wang Y, Song L. Reproductive factors and biological aging: the association with all-cause and cause-specific premature mortality. Hum Reprod 2025; 40:148-156. [PMID: 39516182 DOI: 10.1093/humrep/deae250] [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: 07/02/2024] [Revised: 09/22/2024] [Indexed: 11/16/2024] Open
Abstract
STUDY QUESTION Are reproductive factors associated with biological aging, and does biological aging mediate the associations of reproductive factors with premature mortality? SUMMARY ANSWER Multiple reproductive factors are related to phenotypic age acceleration (PhenoAge-Accel), while adherence to a healthy lifestyle mitigates these harmful effects; PhenoAge-Accel mediated the associations between reproductive factors and premature mortality. WHAT IS KNOWN ALREADY Accelerated aging is a key contributor to mortality, but knowledge about the effect of reproductive factors on aging is limited. STUDY DESIGN, SIZE, DURATION This prospective cohort study included 223 729 women aged 40-69 years from the UK biobank in 2006-2010 and followed up until 12 November 2021. PARTICIPANTS/MATERIALS, SETTING, METHODS Reproductive factors were collected through a touchscreen questionnaire. Biological aging was assessed through PhenoAge-Accel. Multiple linear regression models were used to examine the relationships of reproductive factors with PhenoAge-Accel and estimate the modified effect of a healthy lifestyle. Furthermore, we applied mediation analysis to explore the mediating role of PhenoAge-Accel in the associations between reproductive factors and premature mortality. MAIN RESULTS AND THE ROLE OF CHANCE Early menarche (<12 years vs 13 years, β: 0.37, 95% CI: 0.30, 0.44), late menarche (≥15 years vs 13 years, β: 0.18, 95% CI: 0.11, 0.25), early menopause (<45 years vs 50-51 years, β: 0.62, 95% CI: 0.51, 0.72), short reproductive lifespan (<30 years vs 35-39 years, β: 0.81, 95% CI: 0.70, 0.92), nulliparity (vs two live births, β: 0.36, 95% CI: 0.30, 0.43), high parity (≥4 vs 2 live births, β: 0.49, 95% CI: 0.40, 0.59), early age at first live birth (<20 years vs 25-29 years, β: 0.66, 95% CI: 0.56, 0.75), and stillbirth (β: 0.51, 95% CI: 0.36, 0.65) were associated with increased PhenoAge-Accel. Furthermore, PhenoAge-Accel mediated 6.0%-29.7% of the associations between reproductive factors and premature mortality. Women with an unfavorable lifestyle and reproductive risk factors had the highest PhenoAge-Accel compared to those with a favorable lifestyle and without reproductive risk factors. LIMITATIONS, REASONS FOR CAUTION The participants in the UK Biobank were predominantly of White ethnicity; thus, caution is warranted when generalizing these findings to other ethnic groups. WIDER IMPLICATIONS OF THE FINDINGS Our findings reveal the harmful effects of multiple reproductive factors on biological aging and the mediating role of biological aging in the associations between reproductive factors and premature mortality. They highlight the significance of adhering to a healthy lifestyle to slow biological aging as a potential way to reduce premature mortality among women with reproductive risk factors. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the National Natural Science Foundation of China (82003479, 82073660, 72204215), Hubei Provincial Natural Science Foundation of China (2023AFB663), Zhejiang Province Public Welfare Technology Application Research Project (GF22H269155), and China Postdoctoral Science Foundation (2019M662646, 2020T130220). The authors have no competing interests to disclose. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Gaojie Fan
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Liu
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianing Bi
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Fang
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fei Luo
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaofeng Huang
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Heng Li
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenwen Guo
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Binghai Liu
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lianyan Yan
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Youjie Wang
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lulu Song
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
41
|
Sotomayor-Lugo F, Iglesias-Barrameda N, Casado-Hernandez I, Villegas-Valverde CA, Ventura-Carmenate Y, Rivero-Jimenez RA. Aging: Disease or "natural" process? A glimpse from regenerative medicine. Rev Esp Geriatr Gerontol 2025; 60:101543. [PMID: 39369641 DOI: 10.1016/j.regg.2024.101543] [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: 04/22/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 10/08/2024]
Abstract
We explore aging as a global phenomenon, questioning whether it constitutes a treatable condition or follows a natural course. Acknowledging its multifactorial nature, we delve into the challenges and opportunities inherent in this intricate biological process. The inclusion of old age in the 11th International Classification of Diseases sparks debate, categorizing it as a disease based on mechanistic explanations, blood-based biomarkers, and anti-aging products. Ethical dilemmas arise, emphasizing the difficulty of defining the transition from normal to pathological states during this process. We suggest that aging should be regarded as a treatable condition without necessarily labeling it a 'disease.' While anti-aging research unveils promising interventions like Metformin, Rapamycin, and cellular therapy, achieving biological immortality remains a formidable challenge. The future promises to prolong life and enhance quality by comprehensively understanding aging's implications for human health.
Collapse
Affiliation(s)
| | | | | | | | - Yendry Ventura-Carmenate
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates; Yas Clinic Khalifa City Hospital, Abu Dhabi, United Arab Emirates; United Arab Emirates University, Office of Research and Graduate Studies, College of Medicine and Health Science, Abu Dhabi, United Arab Emirates
| | - Rene Antonio Rivero-Jimenez
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates; United Arab Emirates University, Office of Research and Graduate Studies, College of Medicine and Health Science, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
42
|
Liu WS, You J, Chen SD, Zhang Y, Feng JF, Xu YM, Yu JT, Cheng W. Plasma proteomics identify biomarkers and undulating changes of brain aging. NATURE AGING 2025; 5:99-112. [PMID: 39653801 DOI: 10.1038/s43587-024-00753-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/17/2024] [Indexed: 12/15/2024]
Abstract
Proteomics enables the characterization of brain aging biomarkers and discernment of changes during brain aging. We leveraged multimodal brain imaging data from 10,949 healthy adults to estimate brain age gap (BAG), an indicator of brain aging. Proteome-wide association analysis across 4,696 participants of 2,922 proteins identified 13 significantly associated with BAG, implicating stress, regeneration and inflammation. Brevican (BCAN) (β = -0.838, P = 2.63 × 10-10) and growth differentiation factor 15 (β = 0.825, P = 3.48 × 10-11) showed the most significant, and multiple, associations with dementia, stroke and movement functions. Dysregulation of BCAN affected multiple cortical and subcortical structures. Mendelian randomization supported the causal association between BCAN and BAG. We revealed undulating changes in the plasma proteome across brain aging, and profiled brain age-related change peaks at 57, 70 and 78 years, implicating distinct biological pathways during brain aging. Our findings revealed the plasma proteomic landscape of brain aging and pinpointed biomarkers for brain disorders.
Collapse
Affiliation(s)
- Wei-Shi Liu
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| |
Collapse
|
43
|
An L, Zhang C, Wulan N, Zhang S, Chen P, Ji F, Ng KK, Chen C, Zhou JH, Yeo BTT. DeepResBat: Deep residual batch harmonization accounting for covariate distribution differences. Med Image Anal 2025; 99:103354. [PMID: 39368279 DOI: 10.1016/j.media.2024.103354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024]
Abstract
Pooling MRI data from multiple datasets requires harmonization to reduce undesired inter-site variabilities, while preserving effects of biological variables (or covariates). The popular harmonization approach ComBat uses a mixed effect regression framework that explicitly accounts for covariate distribution differences across datasets. There is also significant interest in developing harmonization approaches based on deep neural networks (DNNs), such as conditional variational autoencoder (cVAE). However, current DNN approaches do not explicitly account for covariate distribution differences across datasets. Here, we provide mathematical results, suggesting that not accounting for covariates can lead to suboptimal harmonization. We propose two DNN-based covariate-aware harmonization approaches: covariate VAE (coVAE) and DeepResBat. The coVAE approach is a natural extension of cVAE by concatenating covariates and site information with site- and covariate-invariant latent representations. DeepResBat adopts a residual framework inspired by ComBat. DeepResBat first removes the effects of covariates with nonlinear regression trees, followed by eliminating site differences with cVAE. Finally, covariate effects are added back to the harmonized residuals. Using three datasets from three continents with a total of 2787 participants and 10,085 anatomical T1 scans, we find that DeepResBat and coVAE outperformed ComBat, CovBat and cVAE in terms of removing dataset differences, while enhancing biological effects of interest. However, coVAE hallucinates spurious associations between anatomical MRI and covariates even when no association exists. Future studies proposing DNN-based harmonization approaches should be aware of this false positive pitfall. Overall, our results suggest that DeepResBat is an effective deep learning alternative to ComBat. Code for DeepResBat can be found here: https://github.com/ThomasYeoLab/CBIG/tree/master/stable_projects/harmonization/An2024_DeepResBat.
Collapse
Affiliation(s)
- Lijun An
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Chen Zhang
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Naren Wulan
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Shaoshi Zhang
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Pansheng Chen
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Fang Ji
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kwun Kei Ng
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
| | - B T Thomas Yeo
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore; Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore; Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
44
|
Zhu T, Mu D, Hu Y, Cao Y, Yuan M, Xu J, Ye HQ, Zhang W. Association of clinical phenotypes of depression with comorbid conditions, treatment patterns and outcomes: a 10-year region-based cohort study. Transl Psychiatry 2024; 14:504. [PMID: 39719438 DOI: 10.1038/s41398-024-03213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024] Open
Abstract
Depression is a heterogeneous and complex psychological syndrome with highly variable manifestations, which poses difficulties for treatment and prognosis. Depression patients are prone to developing various comorbidities, which stem from different pathophysiological mechanisms, remaining largely understudied. The current study focused on identifying comorbidity-specific phenotypes, and whether these clustered phenotypes are associated with different treatment patterns, clinical manifestations, physiological characteristics, and prognosis. We have conducted a 10-year retrospective observational cohort study using electronic medical records (EMR) for 11,818 patients diagnosed with depression and hospitalized at a large academic medical center in Chengdu, China. K-means clustering and visualization methods were performed to identify phenotypic categories. The association between phenotypic categories and clinical outcomes was evaluated using adjusted Cox proportional hazards model. We classified patients with depression into five stable phenotypic categories, including 15 statistically driven clusters in the discovery cohort (n = 9925) and the validation cohort (n = 1893), respectively. The categories include: (Category A) the lowest incidence of comorbidity, with prominent suicide, psychotic, and somatic symptoms (n = 3493/9925); (Category B) moderate comorbidity rate, with prominent anhedonia and anxious symptoms (n = 1795/9925); (Category C) the highest incidence of comorbidity of endocrine/metabolic and digestive system diseases (n = 1702/9925); (Category D) the highest incidence of comorbidity of neurological, mental and behavioral diseases (n = 881/9925); (Category E) other diseases comorbid with depression (n = 2054/9925). Patients in Category E had the lowest risk of psychiatric rehospitalization within 60-day follow-up, followed by Category C (HR, 1.57; 95% CI, 1.07-2.30), Category B (HR, 1.61; 95% CI, 1.10-2.40), Category A (HR, 1.82; 95% CI, 1.28-2.60), and Category D (HR, 2.38; 95% CI, 1.59-3.60) with P < 0.05, after adjustment for comorbidities, medications, and age. Regarding other longer observation windows (90-day, 180-day and 365-day), patients in Category D showed the highest rehospitalization risk all the time while there were notable shifts in rankings observed for Categories A, B and C over time. The results indicate that the higher the severity of mental illness in patients with five phenotypic categories, the greater the risk of rehospitalization. These phenotypes are associated with various pathways, including the cardiometabolic system, chronic inflammation, digestive system, neurological system, and mental and behavioral disorders. These pathways play a crucial role in connecting depression with other psychiatric and somatic diseases. The identified phenotypes exhibit notable distinctions in terms of comorbidity patterns, symptomology, biological characteristics, treatment approaches, and clinical outcomes.
Collapse
Affiliation(s)
- Ting Zhu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610041, China
| | - Di Mu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610041, China
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3053, Australia
| | - Yao Hu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610041, China
| | - Yang Cao
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Minlan Yuan
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jia Xu
- The First Psychiatric Hospital of Harbin, Harbin, 150056, China
| | - Heng-Qing Ye
- Faculty of Business, Hong Kong Polytechnic University, Hong Kong, 100872, China.
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Med-X Center for Informatics, Sichuan University, Chengdu, 610041, China.
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
45
|
Sarver DC, Saqib M, Chen F, Wong GW. Mitochondrial respiration atlas reveals differential changes in mitochondrial function across sex and age. eLife 2024; 13:RP96926. [PMID: 39704485 PMCID: PMC11661797 DOI: 10.7554/elife.96926] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Organ function declines with age, and large-scale transcriptomic analyses have highlighted differential aging trajectories across tissues. The mechanism underlying shared and organ-selective functional changes across the lifespan, however, still remains poorly understood. Given the central role of mitochondria in powering cellular processes needed to maintain tissue health, we therefore undertook a systematic assessment of respiratory activity across 33 different tissues in young (2.5 months) and old (20 months) mice of both sexes. Our high-resolution mitochondrial respiration atlas reveals: (1) within any group of mice, mitochondrial activity varies widely across tissues, with the highest values consistently seen in heart, brown fat, and kidney; (2) biological sex is a significant but minor contributor to mitochondrial respiration, and its contributions are tissue-specific, with major differences seen in the pancreas, stomach, and white adipose tissue; (3) age is a dominant factor affecting mitochondrial activity, especially across most brain regions, different fat depots, skeletal muscle groups, eyes, and different regions of the gastrointestinal tract; (4) age effects can be sex- and tissue-specific, with some of the largest effects seen in pancreas, heart, adipose tissue, and skeletal muscle; and (5) while aging alters the functional trajectories of mitochondria in a majority of tissues, some are remarkably resilient to age-induced changes. Altogether, our data provide the most comprehensive compendium of mitochondrial respiration and illuminate functional signatures of aging across diverse tissues and organ systems.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Fangluo Chen
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
46
|
Tokizane K, Imai SI. Inter-organ communication is a critical machinery to regulate metabolism and aging. Trends Endocrinol Metab 2024:S1043-2760(24)00320-5. [PMID: 39694728 DOI: 10.1016/j.tem.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
Inter-organ communication (IOC) is a complex mechanism involved in maintaining metabolic homeostasis and healthy aging. Dysregulation of distinct forms of IOC is linked to metabolic derangements and age-related pathologies, implicating these processes as a potential target for therapeutic intervention to promote healthy aging. In this review, we delve into IOC mediated by hormonal signaling, circulating factors, organelle signaling, and neuronal networks and examine their roles in regulating metabolism and aging. Given the role of the hypothalamus as a high-order control center for aging and longevity, we particularly emphasize the importance of its communication with peripheral organs and pave the way for a better understanding of this critical machinery in metabolism and aging.
Collapse
Affiliation(s)
- Kyohei Tokizane
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, 63110, MO, USA; Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA.
| |
Collapse
|
47
|
Santin Y, Chiesa M, Alfonso A, Doghri Y, Kang R, Haidar F, Oreja-Fuentes P, Fousset O, Zahreddine R, Guardia M, Lemmel L, Rigamonti M, Rosati G, Florian C, Gauzin S, Guyonnet S, Rolland Y, de Souto Barreto P, Vellas B, Guiard B, Parini A. Computational and digital analyses in the INSPIRE mouse cohort to define sex-specific functional determinants of biological aging. SCIENCE ADVANCES 2024; 10:eadt1670. [PMID: 39671481 PMCID: PMC11641001 DOI: 10.1126/sciadv.adt1670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/08/2024] [Indexed: 12/15/2024]
Abstract
Biological age, which reflects the physiological state of an individual, offers a better predictive value than chronological age for age-related diseases and mortality. Nonetheless, determining accurate functional features of biological age remains challenging due to the multifactorial nature of aging. Here, we established a unique mouse cohort comprising 1576 male and female outbred SWISS mice subjected or not to high-fat, high-sucrose diet to investigate multiorgan/system biological aging throughout adulthood. Comprehensive functional and biological phenotyping at ages of 6, 12, 18, and 24 months revealed notable sex-specific disparities in longitudinal locomotion patterns and multifunctional aging parameters. Topological data analysis enabled the identification of functionally similar mouse clusters irrespective of chronological age. Moreover, our study pinpointed critical functional markers of biological aging such as muscle function, anxiety characteristics, urinary patterns, reticulocyte maturation, cardiac remodeling and function, and metabolic alterations, underscoring muscle function as an early indicator of biological age in male mice.
Collapse
Affiliation(s)
- Yohan Santin
- Institut Hospitalo-Universitaire (IHU) HealthAge, Toulouse, France
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| | - Mattia Chiesa
- Bioinformatics and Artificial Intelligence Facility, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Amélie Alfonso
- Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS, University of Toulouse, UPS, Toulouse, France
| | - Yosra Doghri
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| | - Ryeonshi Kang
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| | - Fraha Haidar
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| | - Pilar Oreja-Fuentes
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| | - Occiane Fousset
- Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS, University of Toulouse, UPS, Toulouse, France
| | - Rana Zahreddine
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| | - Mégane Guardia
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| | - Lucas Lemmel
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| | | | | | - Cédrick Florian
- Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS, University of Toulouse, UPS, Toulouse, France
| | - Sébastien Gauzin
- Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS, University of Toulouse, UPS, Toulouse, France
| | - Sophie Guyonnet
- Institut Hospitalo-Universitaire (IHU) HealthAge, Toulouse, France
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France
- University of Toulouse III, Toulouse, France
- CERPOP Inserm UMR 1295, Toulouse, France
| | - Yves Rolland
- Institut Hospitalo-Universitaire (IHU) HealthAge, Toulouse, France
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France
- University of Toulouse III, Toulouse, France
- CERPOP Inserm UMR 1295, Toulouse, France
| | - Philipe de Souto Barreto
- Institut Hospitalo-Universitaire (IHU) HealthAge, Toulouse, France
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France
- University of Toulouse III, Toulouse, France
- CERPOP Inserm UMR 1295, Toulouse, France
| | - Bruno Vellas
- Institut Hospitalo-Universitaire (IHU) HealthAge, Toulouse, France
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France
- University of Toulouse III, Toulouse, France
- CERPOP Inserm UMR 1295, Toulouse, France
| | - Bruno Guiard
- Institut Hospitalo-Universitaire (IHU) HealthAge, Toulouse, France
- Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS, University of Toulouse, UPS, Toulouse, France
| | - Angelo Parini
- Institut Hospitalo-Universitaire (IHU) HealthAge, Toulouse, France
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, University of Toulouse, UPS, Toulouse, France
| |
Collapse
|
48
|
Haikal C, Weissert R. Editorial: Aging, peripheral inflammation, and neurodegeneration. Front Aging Neurosci 2024; 16:1529026. [PMID: 39720419 PMCID: PMC11666552 DOI: 10.3389/fnagi.2024.1529026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Affiliation(s)
- Caroline Haikal
- Neurological Surgery, Weill Cornell Medical Center, New York-Presbyterian, New York, NY, United States
| | - Robert Weissert
- Department of Neurology, University of Regensburg Hospital, Regensburg, Germany
| |
Collapse
|
49
|
Pan R, Yi X, Xu Y, Song J, Yi W, Liu J, Song R, Li X, Liu L, Yuan J, Wei N, Huang Y, Cui Z, Kuang L, Zhang Z, Li M, Cheng J, Zhang X, Su H. Association between indoor PM 2.5 components and accelerated biological aging in schizophrenia patients: Evidence from multi-omics mechanisms. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136162. [PMID: 39490163 DOI: 10.1016/j.jhazmat.2024.136162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/06/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024]
Abstract
Indoor fine particulate matter (PM2.5) poses a considerable hazard to the aging process, particularly in vulnerable populations such as schizophrenia patients who frequently spend extended periods in indoor environments. Currently, the evidence on which PM2.5 components contribute to accelerated aging remains unclear. To address these issues, we conducted a prospective, repeated-measurement study on 104 schizophrenia patients. Our findings indicated that exposure to PM2.5 components was significantly associated with accelerated biological aging in schizophrenia patients. Notably, the most prominent effects were observed for thallium (1.303, 95 % CI: 0.481-2.125), chromium (1.029, 95 % CI: 0.303-1.756), lead (1.021, 95 % CI: 0.296-1.746), antimony (0.915, 95 % CI: 0.233-1.597), selenium (0.854, 95 % CI: 0.209-1.499), and manganese (0.833, 95 % CI: 0.186-1.480). Multivariate analysis revealed that PM2.5 components predominantly induced alterations in serum glycerophospholipid metabolites, accelerating the aging process. This intricate connection was closely linked to the gut microbiota, particularly to species such as Dorea and Blautia. Mediation analysis showed that the Blautia-PC (16:0/0:0) pathway mediated the largest proportion (30.69 %) of the effect of manganese exposure on accelerating immune biological aging in schizophrenia patients, as measured using the Klemera-Doubal method. These results underscore the need to address pollution sources that harm health, and provide new evidence for improving regional air quality.
Collapse
Affiliation(s)
- Rubing Pan
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xingxu Yi
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yanlong Xu
- Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Jian Song
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Weizhuo Yi
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Jintao Liu
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Rong Song
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xuanxuan Li
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Li Liu
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Jiajun Yuan
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Ning Wei
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yuxing Huang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Zhiqian Cui
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Lingmei Kuang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Zichen Zhang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Ming Li
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Jian Cheng
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xulai Zhang
- Anhui Mental Health Center (Affiliated Psychological Hospital of Anhui Medical University), Hefei, Anhui, China.
| | - Hong Su
- School of Public Health, Anhui Medical University, Hefei, Anhui, China; Center for Big Data and Population Health of IHM, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China.
| |
Collapse
|
50
|
Moguilner S, Baez S, Hernandez H, Migeot J, Legaz A, Gonzalez-Gomez R, Farina FR, Prado P, Cuadros J, Tagliazucchi E, Altschuler F, Maito MA, Godoy ME, Cruzat J, Valdes-Sosa PA, Lopera F, Ochoa-Gómez JF, Hernandez AG, Bonilla-Santos J, Gonzalez-Montealegre RA, Anghinah R, d'Almeida Manfrinati LE, Fittipaldi S, Medel V, Olivares D, Yener GG, Escudero J, Babiloni C, Whelan R, Güntekin B, Yırıkoğulları H, Santamaria-Garcia H, Lucas AF, Huepe D, Di Caterina G, Soto-Añari M, Birba A, Sainz-Ballesteros A, Coronel-Oliveros C, Yigezu A, Herrera E, Abasolo D, Kilborn K, Rubido N, Clark RA, Herzog R, Yerlikaya D, Hu K, Parra MA, Reyes P, García AM, Matallana DL, Avila-Funes JA, Slachevsky A, Behrens MI, Custodio N, Cardona JF, Barttfeld P, Brusco IL, Bruno MA, Sosa Ortiz AL, Pina-Escudero SD, Takada LT, Resende E, Possin KL, de Oliveira MO, Lopez-Valdes A, Lawlor B, Robertson IH, Kosik KS, Duran-Aniotz C, Valcour V, Yokoyama JS, Miller B, Ibanez A. Brain clocks capture diversity and disparities in aging and dementia across geographically diverse populations. Nat Med 2024; 30:3646-3657. [PMID: 39187698 PMCID: PMC11645278 DOI: 10.1038/s41591-024-03209-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: 03/22/2024] [Accepted: 07/22/2024] [Indexed: 08/28/2024]
Abstract
Brain clocks, which quantify discrepancies between brain age and chronological age, hold promise for understanding brain health and disease. However, the impact of diversity (including geographical, socioeconomic, sociodemographic, sex and neurodegeneration) on the brain-age gap is unknown. We analyzed datasets from 5,306 participants across 15 countries (7 Latin American and Caribbean countries (LAC) and 8 non-LAC countries). Based on higher-order interactions, we developed a brain-age gap deep learning architecture for functional magnetic resonance imaging (2,953) and electroencephalography (2,353). The datasets comprised healthy controls and individuals with mild cognitive impairment, Alzheimer disease and behavioral variant frontotemporal dementia. LAC models evidenced older brain ages (functional magnetic resonance imaging: mean directional error = 5.60, root mean square error (r.m.s.e.) = 11.91; electroencephalography: mean directional error = 5.34, r.m.s.e. = 9.82) associated with frontoposterior networks compared with non-LAC models. Structural socioeconomic inequality, pollution and health disparities were influential predictors of increased brain-age gaps, especially in LAC (R² = 0.37, F² = 0.59, r.m.s.e. = 6.9). An ascending brain-age gap from healthy controls to mild cognitive impairment to Alzheimer disease was found. In LAC, we observed larger brain-age gaps in females in control and Alzheimer disease groups compared with the respective males. The results were not explained by variations in signal quality, demographics or acquisition methods. These findings provide a quantitative framework capturing the diversity of accelerated brain aging.
Collapse
Grants
- R01 AG075775 NIA NIH HHS
- R01AG083799 John E. Fogarty Foundation for Persons with Intellectual and Developmental Disabilities
- 75N95022C00031 NIDA NIH HHS
- P01 AG019724 NIA NIH HHS
- SG-20-725707 Alzheimer's Association
- R01 AG057234 NIA NIH HHS
- R01 AG083799 NIA NIH HHS
- U.S. Department of Health & Human Services | NIH | Fogarty International Center (FIC)
- Latin American Brain Health Institute (BrainLat) # BL-SRGP2020-02 ReDLat [National Institutes of Health and the Fogarty International Center (FIC), National Institutes of Aging (R01 AG057234, R01 AG075775, AG021051, R01AG083799, CARDS-NIH 75N95022C00031), Alzheimer's Association (SG-20-725707), Rainwater Charitable Foundation, The Bluefield project to cure FTD, and Global Brain Health Institute)], ANID/FONDECYT Regular (1210195, 1210176 and 1220995); and ANID/FONDAP/15150012
- National Institute on Aging of the National Institutes of Health (R01AG075775, R01AG083799, 2P01AG019724); ANID (FONDECYT Regular 1210176, 1210195); and DICYT-USACH (032351G_DAS)
Collapse
Affiliation(s)
- Sebastian Moguilner
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Cognitive Neuroscience Center, Universidad de San Andrés, Buenos Aires, Argentina
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sandra Baez
- Universidad de los Andes, Bogota, Colombia
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
| | - Hernan Hernandez
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
| | - Joaquín Migeot
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
| | - Agustina Legaz
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Cognitive Neuroscience Center, Universidad de San Andrés, Buenos Aires, Argentina
| | - Raul Gonzalez-Gomez
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
| | - Francesca R Farina
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- The University of California Santa Barbara (UCSB), Santa Barbara, CA, USA
| | - Pavel Prado
- Escuela de Fonoaudiología, Universidad San Sebastián, Santiago de Chile, Chile
| | - Jhosmary Cuadros
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Grupo de Bioingeniería, Decanato de Investigación, Universidad Nacional Experimental del Táchira, San Cristóbal, Venezuela
- Advanced Center for Electrical and Electronic Engineering, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | - Enzo Tagliazucchi
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- University of Buenos Aires, Buenos Aires, Argentina
| | - Florencia Altschuler
- Cognitive Neuroscience Center, Universidad de San Andrés, Buenos Aires, Argentina
| | - Marcelo Adrián Maito
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Cognitive Neuroscience Center, Universidad de San Andrés, Buenos Aires, Argentina
| | - María E Godoy
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Cognitive Neuroscience Center, Universidad de San Andrés, Buenos Aires, Argentina
| | - Josephine Cruzat
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
| | - Pedro A Valdes-Sosa
- The Clinical Hospital of Chengdu Brain Sciences Institute, University of Electronic Sciences and Technology of China, Chengdu, China
- Technology of China, Chengdu, China
- Cuban Neuroscience Center, La Habana, Cuba
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia (GNA), University of Antioquia, Medellín, Colombia
| | | | - Alfredis Gonzalez Hernandez
- Department of Psychology, Master Program of Clinical Neuropsychology, Universidad Surcolombiana Neiva, Neiva, Colombia
| | | | | | - Renato Anghinah
- Reference Center of Behavioural Disturbances and Dementia, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Traumatic Brain Injury Cognitive Rehabilitation Out-Patient Center, University of Sao Paulo, Sao Paulo, Brazil
| | - Luís E d'Almeida Manfrinati
- Reference Center of Behavioural Disturbances and Dementia, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Traumatic Brain Injury Cognitive Rehabilitation Out-Patient Center, University of Sao Paulo, Sao Paulo, Brazil
| | - Sol Fittipaldi
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
| | - Vicente Medel
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
| | - Daniela Olivares
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Center for Social and Cognitive Neuroscience, School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Program-Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, University of Chile, Santiago, Chile
- Centro de Neuropsicología Clínica (CNC), Santiago, Chile
| | - Görsev G Yener
- Faculty of Medicine, Izmir University of Economics, Izmir, Turkey
- Brain Dynamics Multidisciplinary Research Center, Dokuz Eylul University, Izmir, Turkey
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Javier Escudero
- School of Engineering, Institute for Imaging, Data and Communications, University of Edinburgh, Edinburgh, UK
| | - Claudio Babiloni
- Department of Physiology and Pharmacology 'V. Erspamer', Sapienza University of Rome, Rome, Italy
- Hospital San Raffaele Cassino, Cassino, Italy
| | - Robert Whelan
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Bahar Güntekin
- Department of Neurosciences, Health Sciences Institute, Istanbul Medipol University, İstanbul, Turkey
- Health Sciences and Technology Research Institute (SABITA), Istanbul Medipol University, Istanbul, Turkey
- Department of Biophysics, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Harun Yırıkoğulları
- Department of Neurosciences, Health Sciences Institute, Istanbul Medipol University, İstanbul, Turkey
- Health Sciences and Technology Research Institute (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Hernando Santamaria-Garcia
- Pontificia Universidad Javeriana (PhD Program in Neuroscience), Bogotá, Colombia
- Center of Memory and Cognition Intellectus, Hospital Universitario San Ignacio Bogotá, San Ignacio, Colombia
| | - Alberto Fernández Lucas
- Departamento de Medicina Legal, Psiquiatría y Patología, Universidad Complutense de Madrid, Madrid, Spain
| | - David Huepe
- Center for Social and Cognitive Neuroscience, School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Gaetano Di Caterina
- Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK
| | | | - Agustina Birba
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
| | | | - Carlos Coronel-Oliveros
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
| | - Amanuel Yigezu
- The University of California Santa Barbara (UCSB), Santa Barbara, CA, USA
| | - Eduar Herrera
- Departamento de Estudios Psicológicos, Universidad ICESI, Cali, Colombia
| | - Daniel Abasolo
- Centre for Biomedical Engineering, School of Mechanical Engineering Sciences, University of Surrey, Guildford, UK
| | - Kerry Kilborn
- School of Psychology, University of Glasgow, Glasgow, UK
| | - Nicolás Rubido
- Institute for Complex Systems and Mathematical Biology, University of Aberdeen, Aberdeen, UK
| | - Ruaridh A Clark
- Centre for Signal and Image Processing, Department of Electronic and Electrical Engineering, University of Strathclyde, Strathclyde, UK
| | - Ruben Herzog
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, InsermCNRS, Paris, France
| | - Deniz Yerlikaya
- Department of Neurosciences, Health Sciences Institute, Dokuz Eylül University, Izmir, Turkey
| | - Kun Hu
- Harvard Medical School, Boston, MA, USA
| | - Mario A Parra
- Department of Psychological Sciences and Health, University of Strathclyde, Glasgow, UK
- BrainLat, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Pablo Reyes
- Pontificia Universidad Javeriana (PhD Program in Neuroscience), Bogotá, Colombia
- Center of Memory and Cognition Intellectus, Hospital Universitario San Ignacio Bogotá, San Ignacio, Colombia
| | - Adolfo M García
- Cognitive Neuroscience Center, Universidad de San Andrés, Buenos Aires, Argentina
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Departamento de Lingüística y Literatura, Universidad de Santiago de Chile, Santiago, Chile
| | - Diana L Matallana
- Pontificia Universidad Javeriana (PhD Program in Neuroscience), Bogotá, Colombia
- Center of Memory and Cognition Intellectus, Hospital Universitario San Ignacio Bogotá, San Ignacio, Colombia
- Mental Health Department, Hospital Universitario Fundación Santa Fe, Bogota, Colombia
| | - José Alberto Avila-Funes
- Department of Geriatrics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Andrea Slachevsky
- Memory and Neuropsychiatric Center (CMYN), Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Program - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, University of Chile, Santiago, Chile
| | - María I Behrens
- Neurology and Psychiatry Department, Clínica Alemana-Universidad Desarrollo, Santiago, Chile
- Centro de Investigación Clínica Avanzada (CICA), Universidad de Chile, Santiago, Chile
- Departamento de Neurología y Neurocirugía, Hospital Clínico de la Universidad de Chile, Santiago, Chile
- Departamento de Neurociencia, Universidad de Chile, Santiago, Chile
| | - Nilton Custodio
- Servicio de Neurología, Instituto Peruano de Neurociencias, Lima, Perú
| | - Juan F Cardona
- Facultad de Psicología, Universidad del Valle, Cali, Colombia
| | - Pablo Barttfeld
- Cognitive Science Group, Instituto de Investigaciones Psicológicas (IIPsi), CONICET UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ignacio L Brusco
- Centro de Neuropsiquiatría y Neurología de la Conducta (CENECON), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Martín A Bruno
- Instituto de Ciencias Biomédicas (ICBM), Universidad Catoóica de Cuyo, San Juan, Argentina
| | - Ana L Sosa Ortiz
- Instituto Nacional de Neurologia y Neurocirugia MVS, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Stefanie D Pina-Escudero
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Leonel T Takada
- Cognitive and Behavioral Neurology Unit, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Elisa Resende
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Katherine L Possin
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Maira Okada de Oliveira
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Cognitive and Behavioral Neurology Unit, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Alejandro Lopez-Valdes
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- School of Engineering, Department of Electrical and Electronic Engineering, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Brian Lawlor
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
| | - Ian H Robertson
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Kenneth S Kosik
- Division of the Biological Sciences, The University of Chicago, Chicago, IL, USA
| | - Claudia Duran-Aniotz
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile
| | - Victor Valcour
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Jennifer S Yokoyama
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Bruce Miller
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Agustin Ibanez
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago de Chile, Chile.
- Cognitive Neuroscience Center, Universidad de San Andrés, Buenos Aires, Argentina.
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA.
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|