1
|
Sepúlveda M, Palomo I, Montecino-Garrido H, Wehinger S, Rodriguez-Mañas L, Trostchansky A, Fuentes E. Physiological changes associated with aging: Identification of novel biomarkers for frailty syndrome in women. Free Radic Biol Med 2024; 223:160-171. [PMID: 39059511 DOI: 10.1016/j.freeradbiomed.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
This study explores the physiological changes associated with aging that lead to frailty syndrome, characterized by reduced vitality and degeneration across multiple bodily systems, increasing susceptibility to various pathologies. While established scales like the Fried Phenotype and Frailty Trait Scale (FTS) are commonly used for assessing frailty, incorporating biomarkers is crucial for accurate diagnosis and prognosis. Our research examines plasma oxylipin levels in frail elderly individuals to identify novel biomarkers. Diagnostic criteria for frailty included assessments using the Fried Phenotype and FTS-5, with blood samples collected from 71 elderly participants (50 women and 21 men) with mean ages of 73.6 ± 5.9 and 76.2 ± 6.2 years, respectively. Women exhibited elevated platelet counts (p-value 0.0035). The significant differences in oxylipin concentrations associated with the Fried Phenotype were particularly noteworthy, predominantly observed in women. Specifically, in women, decreased grip strength (<15 kg) and slow gait speed (<0.8 m/s) correlated with increased levels of thromboxane B2 (TxB2) and 7-HDoHE (p-values 0.0404, 0.0300, 0.0033, and 0.0033, respectively). Additionally, elevated 7-HDoHE levels correlated with a BMI exceeding 28 kg/m2 (p-value 0.0123) and Physical Activity Scale for the Elderly (PASE) scores surpassing 5 points (p-value 0.0134) in women. In summary, our findings emphasize that frail older individuals, particularly women, exhibit higher levels of TxB2 and 7-HDoHE compared to their non-frail counterparts, aligning with established frailty classification and scale parameters, suggesting their potential as indicative biomarkers.
Collapse
Affiliation(s)
- Magdalena Sepúlveda
- Thrombosis and Healthy Aging Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca, 3480094, Chile
| | - Iván Palomo
- Thrombosis and Healthy Aging Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca, 3480094, Chile
| | - Héctor Montecino-Garrido
- Thrombosis and Healthy Aging Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca, 3480094, Chile
| | - Sergio Wehinger
- Thrombosis and Healthy Aging Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca, 3480094, Chile
| | - Leocadio Rodriguez-Mañas
- CIBER de Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Avda. Monforte de Lemos, 28029, Madrid, Madrid, Spain; Geriatrics Department, Hospital Universitario de Getafe, Ctra. Madrid-Toledo km. 12.5, 28905, Getafe, Madrid, Spain
| | - Andrés Trostchansky
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, 11800, Uruguay.
| | - Eduardo Fuentes
- Thrombosis and Healthy Aging Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca, 3480094, Chile.
| |
Collapse
|
2
|
Arauna D, Navarrete S, Albala C, Wehinger S, Pizarro-Mena R, Palomo I, Fuentes E. Understanding the Role of Oxidative Stress in Platelet Alterations and Thrombosis Risk among Frail Older Adults. Biomedicines 2024; 12:2004. [PMID: 39335518 PMCID: PMC11429027 DOI: 10.3390/biomedicines12092004] [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: 07/30/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Frailty and cardiovascular diseases are increasingly prevalent in aging populations, sharing common pathological mechanisms, such as oxidative stress. The evidence shows that these factors predispose frail individuals to cardiovascular diseases but also increase the risk of thrombosis. Considering this background, this review aims to explore advances regarding the relationship between oxidative stress, platelet alterations, and cardiovascular diseases in frailty, examining the role of reactive oxygen species overproduction in platelet activation and thrombosis. The current evidence shows a bidirectional relationship between frailty and cardiovascular diseases, emphasizing how frailty not only predisposes individuals to cardiovascular diseases but also accelerates disease progression through oxidative damage and increased platelet function. Thus, oxidative stress is the central axis in the increase in platelet activation and secretion and the inadequate response to acetylsalicylic acid observed in frail people by mitochondrial mechanisms. Also, key biomarkers of oxidative stress, such as isoprostanes and derivate reactive oxygen metabolites, can be optimal predictors of cardiovascular risk and potential targets for therapeutic intervention. The potential of antioxidant therapies in mitigating oxidative stress and improving cardiovascular clinical outcomes such as platelet function is promising in frailty, although further research is necessary to establish the efficacy of these therapies. Understanding these mechanisms could prove essential in improving the health and quality of life of an aging population faced with the dual burden of frailty and cardiovascular diseases.
Collapse
Affiliation(s)
- Diego Arauna
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| | - Simón Navarrete
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| | - Cecilia Albala
- Unidad de Nutrición Pública, Instituto de Nutrición y Tecnología de los Alimentos, Interuniversity Center for Healthy Aging, Universidad de Chile, Santiago 7810000, Chile
| | - Sergio Wehinger
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| | - Rafael Pizarro-Mena
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Sede Los Leones, Santiago 7500000, Chile
- Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Santiago 7810000, Chile
| | - Iván Palomo
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| | - Eduardo Fuentes
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
3
|
Arauna D, Araya-Maturana R, Urra FA, García Á, Palomo I, Fuentes E. Altered dynamics of calcium fluxes and mitochondrial metabolism in platelet activation-related disease and aging. Life Sci 2024; 351:122846. [PMID: 38880165 DOI: 10.1016/j.lfs.2024.122846] [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: 04/29/2024] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Understanding the mechanisms controlling platelet function is crucial for exploring potential therapeutic targets related to atherothrombotic pathologies and primary hemostasis disorders. Our research, which focuses on the role of platelet mitochondria and Ca2+ fluxes in platelet activation, the formation of the procoagulant phenotype, and thrombosis, has significant implications for the development of new therapeutic strategies. Traditionally, Ca2+-dependent cellular signaling has been recognized as a determinant process throughout the platelet activation, controlled primarily by store-operated Ca2+ entry and the PLC-PKC signaling pathway. However, despite the accumulated knowledge of these regulatory mechanisms, the effectiveness of therapy based on various commonly used antiplatelet drugs (such as acetylsalicylic acid and clopidogrel, among others) has faced challenges due to bleeding risks and reduced efficacy associated with the phenomenon of high platelet reactivity. Recent evidence suggests that platelet mitochondria could play a fundamental role in these aspects through Ca2+-dependent mechanisms linked to apoptosis and forming a procoagulant phenotype. In this context, the present review describes the latest advances regarding the role of platelet mitochondria and Ca2+ fluxes in platelet activation, the formation of the procoagulant phenotype, and thrombosis.
Collapse
Affiliation(s)
- Diego Arauna
- Thrombosis and Healthy Aging Research Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center of Healthy Aging (CIES), MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales, MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Universidad de Talca, Talca, Chile
| | - Félix A Urra
- Laboratory of Metabolic Plasticity and Bioenergetics, Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Interuniversity Center of Healthy Aging (CIES), MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Santiago, Chile
| | - Ángel García
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Iván Palomo
- Thrombosis and Healthy Aging Research Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center of Healthy Aging (CIES), MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Eduardo Fuentes
- Thrombosis and Healthy Aging Research Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center of Healthy Aging (CIES), MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Faculty of Health Sciences, Universidad de Talca, Talca, Chile.
| |
Collapse
|
4
|
León-González R, Ortolá R, Carballo-Casla A, Sotos-Prieto M, Buño-Soto A, Rodríguez-Sánchez I, Pastor-Barriuso R, Rodríguez-Artalejo F, García-Esquinas E. Growth Differentiation Factor 15 as a Biomarker of Cardiovascular Risk in Chronic Musculoskeletal Pain. J Gerontol A Biol Sci Med Sci 2024; 79:glae163. [PMID: 38975684 DOI: 10.1093/gerona/glae163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND It is unknown whether growth differentiation factor 15 (GDF-15) is associated with chronic musculoskeletal pain (CMP) and whether or not its association with incident cardiovascular disease (CVD) changes according to CMP status. METHODS In total, 1 957 randomly selected adults aged ≥65 years without prior CVD were followed up between 2015 and 2023. CMP was classified according to its intensity, frequency, and interference with daily activities. The association between GDF-15 levels and CMP was assessed using linear models with progressive inclusion of potential confounders, whereas the association between GDF-15 and CVD risk was evaluated with Cox proportional hazard models with similar adjustment and interaction terms between GDF-15 and CMP. The incremental predictive performance of GDF-15 over standard predictors was evaluated using discrimination and risk reclassification metrics. RESULTS GDF-15 concentrations were 6.90% (95% confidence interval [CI]: 2.56; 11.25) higher in individuals with CMP, and up to 8.89% (4.07; 15.71) and 15.79% (8.43; 23.16) higher in those with ≥3 CMP locations and interfering pain. These increased levels were influenced by a higher prevalence of cardiometabolic risk factors, functional impairments, depressive symptoms, and greater levels of inflammation in individuals with CMP. In fully adjusted models, a twofold increase in GDF-15 was associated with a 1.49 increased risk (95% CI: 1.08; 2.05) of a CVD event in individuals with CMP, but not among those without CMP (1.02 [0.77; 1.35]); p-interaction 0.041. Adding GDF-15 to models including the Framingham Risk Score improved predictive performance among individuals with CMP. CONCLUSIONS We provide evidence that GDF-15 could serve as a biomarker to assess CMP, as well as to predict CVD incidence in individuals with CMP.
Collapse
Affiliation(s)
- Rocío León-González
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rosario Ortolá
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Adrián Carballo-Casla
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Neurobiology, Aging Research Center, Care Sciences and Society Karolinska Institutet & Stockholm University, Stockholm, Sweden
| | - Mercedes Sotos-Prieto
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Environmental Health and Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Antonio Buño-Soto
- Department of Laboratory Medicine, La Paz University Hospital-IdiPaz, Madrid, Spain
| | | | - Roberto Pastor-Barriuso
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| | - Fernando Rodríguez-Artalejo
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| | - Esther García-Esquinas
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
5
|
Kamper RS, Nygaard H, Praeger‐Jahnsen L, Ekmann A, Ditlev SB, Schultz M, Hansen SK, Hansen P, Pressel E, Suetta C. GDF-15 is associated with sarcopenia and frailty in acutely admitted older medical patients. J Cachexia Sarcopenia Muscle 2024; 15:1549-1557. [PMID: 38890783 PMCID: PMC11294026 DOI: 10.1002/jcsm.13513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Growth differentiation factor-15 (GDF-15) has been associated with senescence, lower muscle strength, and physical performance in healthy older people. Still, it is not clear whether GDF-15 can be utilized as a biomarker of sarcopenia and frailty in the early stages of hospitalization. We investigated the association of plasma GDF-15 with sarcopenia and frailty in older, acutely admitted medical patients. METHODS The present study is based on secondary analyses of cross-sectional data from the Copenhagen PROTECT study, a prospective cohort study including 1071 patients ≥65 years of age admitted to the acute medical ward at Copenhagen University Hospital, Bispebjerg, Denmark. Muscle strength was assessed using handgrip strength, and lean mass was assessed using direct segmental multifrequency bioelectrical impedance analyses and used to clarify the potential presence of sarcopenia defined according to guidelines from the European Working Group on Sarcopenia in Older People. Frailty was evaluated using the Clinical Frailty Scale. Plasma GDF-15 was measured using electrochemiluminescence assays from Meso Scale Discovery (MSD, Rockville, MD, USA). RESULTS We included 1036 patients with completed blood samples (mean age 78.9 ± 7.8 years, 53% female). The median concentration of GDF-15 was 2669.3 pg/mL. Systemic GDF-15 was significantly higher in patients with either sarcopenia (P < 0.01) or frailty (P < 0.001) compared with patients without the conditions. Optimum cut-off points of GDF-15 relating to sarcopenia and frailty were 1541 and 2166 pg/mL, respectively. CONCLUSIONS Systemic GDF-15 was higher in acutely admitted older medical patients with sarcopenia and frailty compared with patients without. The present study defined the optimum cut-off for GDF-15, related to the presence of sarcopenia and frailty, respectively. When elevated above the derived cutoffs, GDF-15 was strongly associated with frailty and sarcopenia in both crude and fully adjusted models.
Collapse
Affiliation(s)
- Rikke S. Kamper
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Hanne Nygaard
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
- Department of Emergency MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
| | - Louis Praeger‐Jahnsen
- Copenhagen Center for Translational ResearchCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
| | - Anette Ekmann
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Sisse Bolm Ditlev
- Copenhagen Center for Translational ResearchCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
| | - Martin Schultz
- Department of GeriatricsCopenhagen University Hospital, Hvidovre and AmagerHvidovreDenmark
- Department of Clinical Medicine, Faculty of HealthUniversity of CopenhagenCopenhagenDenmark
| | - Sofie Krarup Hansen
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Pernille Hansen
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Eckart Pressel
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Clinical Medicine, Faculty of HealthUniversity of CopenhagenCopenhagenDenmark
| | - Charlotte Suetta
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical Medicine, Faculty of HealthUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
6
|
Guo X, Li Z, Long T, Cheng S, Yang C, Jiang C, Ma H, Gao R, Song C, Huang X, Wu Y. Physical Frailty and the Risk of Degenerative Valvular Heart Disease. Innov Aging 2024; 8:igae062. [PMID: 39131201 PMCID: PMC11310592 DOI: 10.1093/geroni/igae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Indexed: 08/13/2024] Open
Abstract
Background and Objectives The relationship between physical frailty, age-related conditions, and the incidence of degenerative valvular heart disease (VHD) remains unclear. This study aimed to investigate the potential association between physical frailty and the development of degenerative VHD. Research Design and Methods Participants from the UK Biobank who were initially free of VHD and heart failure were categorized into 3 groups based on the frailty phenotype: non-frailty, pre-frailty, and frailty. The frailty phenotype was determined by evaluating the following 5 components: weight loss, exhaustion, reduced physical activity, slow gait speed, and low grip strength. The incidence of degenerative VHD, including mitral valve regurgitation (MR), aortic valve regurgitation (AR), and aortic valve stenosis (AS), was assessed using hospital admission or death registries. Results Among the 331 642 participants, 11 885 (3.6%) exhibited frailty and 143 379 (43.2%) were categorized as pre-frailty. During a median follow-up of 13.8 years, there were 3 684 MR, 1 205 AR, and 3 166 AS events. Compared to non-frailty participants, those with pre-frailty and frailty showed significantly increased risks for MR (hazard ratio [HR], HRpre-frailty:1.19, 95% confidence interval [CI]: 1.11-1.28; HRfrailty: 1.50, 95% CI: 1.30-1.74), AR (HRpre-frailty:1.19, 95% CI: 1.05-1.34; HRfrailty: 1.58, 95% CI: 1.22-2.04), and AS (HRpre-frailty:1.19, 95% CI: 1.11-1.29; HRfrailty: 1.74, 95% CI: 1.51-2.00). Among the 5 components, slow gait speed showed the strongest association with the risk of various types of VHD (HRMR: 1.50, 95% CI: 1.34-1.65; HRAR: 1.50, 95% CI: 1.24-1.80; HRAS: 1.46, 95% CI: 1.32-1.62), followed by exhaustion, low grip strength, and weight loss. Discussion and Implications Pre-frailty and frailty were associated with a higher risk of all 3 types of degenerative VHD. Early detection and intervention for pre-frailty and frailty in middle-aged and older individuals may assist in preventing or delaying the onset of degenerative VHD.
Collapse
Affiliation(s)
- Xinli Guo
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ziang Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Tianxin Long
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Sijing Cheng
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Cheng Yang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Chenqing Jiang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haowen Ma
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ruixin Gao
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changpeng Song
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaohong Huang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongjian Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Ramonfaur D, Buckley LF, Arthur V, Yang Y, Claggett BL, Ndumele CE, Walker KA, Austin T, Odden MC, Floyd JS, Sanders-van Wijk S, Njoroge J, Kizer JR, Kitzman D, Konety SH, Schrack J, Liu F, Windham BG, Palta P, Coresh J, Yu B, Shah AM. High Throughput Plasma Proteomics and Risk of Heart Failure and Frailty in Late Life. JAMA Cardiol 2024; 9:649-658. [PMID: 38809565 PMCID: PMC11137660 DOI: 10.1001/jamacardio.2024.1178] [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: 10/16/2023] [Accepted: 04/05/2024] [Indexed: 05/30/2024]
Abstract
Importance Heart failure (HF) and frailty frequently coexist and may share a common pathobiology, although the underlying mechanisms remain unclear. Understanding these mechanisms may provide guidance for preventing and treating both conditions. Objective To identify shared pathways between incident HF and frailty in late life using large-scale proteomics. Design, Setting, and Participants In this cohort study, 4877 aptamers (Somascan v4) were measured among participants in the community-based longitudinal Atherosclerosis Risk In Communities (ARIC) cohort study at visit 3 (V3; 1993-1995; n = 10 638) and at visit 5 (V5; 2011-2013; n = 3908). Analyses were externally replicated among 3189 participants in the Cardiovascular Health Study (CHS). Data analysis was conducted from February 2022 to June 2023. Exposures Protein aptamers, measured at study V3 and V5. Main Outcomes and Measures Outcomes assessed included incident HF hospitalization after V3 and after V5, prevalent frailty at V5, and incident frailty between V5 and visit 6 (V6; 2016-2017; n = 4131). Frailty was assessed using the Fried criteria. Analyses were adjusted for age, gender, race, field center, hypertension, diabetes, smoking status, body mass index, estimated glomerular filtration rate, prevalent coronary heart disease, prevalent atrial fibrillation, and history of myocardial infarction. Mendelian randomization (MR) analysis was performed to assess potential causal effects of candidate proteins on HF and frailty. Results A total of 4877 protein aptamers were measured among 10 638 participants at V3 (mean [SD] age, 60 [6] years; 4886 [46%] men). Overall, 286 proteins were associated with incident HF after V3 (822 events; P < 1.0 × 10-5), 83 of which were also associated with incident after V5 (336 events; P < 1.7 × 10-4). Among HF-free participants at V5 (n = 3908; mean [SD] age, 75 [5] years; 1861 [42%] men), 48 of 83 HF-associated proteins were associated with prevalent frailty (223 cases; P < 6.0 × 10-4), 18 of which were also associated with incident frailty at V6 (152 cases; P < 1.0 × 10-3). These proteins enriched fibrosis and inflammation pathways and demonstrated stronger associations with incident HF with preserved ejection fraction (HFpEF) than HF with reduced ejection fraction. All 18 proteins were associated with both prevalent frailty and incident HF in CHS. MR identified potential causal effects of several proteins on frailty and HF. Conclusions and Relevance In this study, the proteins associated with risk of HF and frailty enrich for pathways related to inflammation and fibrosis as well as risk of HFpEF. Several of these proteins could potentially contribute to the shared pathophysiology of frailty and HF.
Collapse
Affiliation(s)
- Diego Ramonfaur
- University of Texas Southwestern Medical Center, Dallas
- Brigham and Women’s Hospital, Boston, Massachusetts
| | | | | | - Yimin Yang
- Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Chiadi E. Ndumele
- Johns Hopkins University School of Medicine, Baltimore, Maryland
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Thomas Austin
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle
| | - Michelle C. Odden
- Department of Epidemiology and Population Health, Stanford University, Stanford, California
| | - James S. Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle
| | - Sandra Sanders-van Wijk
- Division of Cardiology, Department of Medicine, Zuyderland Medical Center, Heerlen, the Netherlands
| | - Joyce Njoroge
- Division of Cardiology, Department of Medicine, Stanford University Medical Center, Palo Alto, California
| | - Jorge R. Kizer
- Division of Cardiology, San Francisco Veterans Affairs Health Care System, and Departments of Medicine, Epidemiology and Biostatistics, San Francisco, California
| | - Dalane Kitzman
- Wake Forest School of Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | | | - Jennifer Schrack
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Fangyu Liu
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Priya Palta
- University of North Carolina School of Medicine, Chapel Hill
| | - Josef Coresh
- Departments of Medicine and Population Health, NYU Langone Health, New York, New York
| | - Bing Yu
- The University of Texas Health Science Center at Houston School of Public Health, Houston
| | - Amil M. Shah
- University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
8
|
Harris ZC, Su HK, Xi AS, Somasundaram A, Sabouri AS. Anterior spinal cord syndrome from a spinal epidural hematoma following removal of an epidural catheter. Can J Anaesth 2024; 71:1037-1042. [PMID: 38750348 DOI: 10.1007/s12630-024-02768-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 07/24/2024] Open
Abstract
PURPOSE Spinal epidural hematoma (SEH) is a rare yet significant complication associated with neuraxial anesthesia. Here, we present the case of a 74-yr-old male who underwent open repair of an abdominal aortic aneurysm. Following the removal of an epidural catheter, the patient developed anterior spinal cord syndrome due to an SEH despite having a normal coagulation profile. CLINICAL FEATURES This patient's neurologic presentation was marked by a loss of motor function while maintaining fine touch sensation distal to the spinal cord injury. Initial truncal computed tomography (CT) angiography failed to detect vascular compromise or diagnose the SEH. Subsequently, delayed magnetic resonance imaging (MRI) revealed a multilevel thoracic epidural hematoma, spinal cord infarction, and ischemia. Immediate surgical decompression was performed, but unfortunately, the patient had a poor outcome. CONCLUSION Anterior spinal cord syndrome (ASCS) represents an uncommon neurologic manifestation of SEH, which is typically characterized by a triad of back pain and sensory and motor deficits. Although the initial CT scan was necessary to diagnose the postvascular surgery complication, it did not immediately detect the SEH. In cases of ASCS subsequent to thoracic epidural placement and removal, MRI is the preferred imaging modality for precise diagnosis and assessment of the need for surgical intervention. Despite adherence to anticoagulation guidelines, patients undergoing neuraxial anesthesia may face an elevated risk of developing SEH. Health care professionals should remain vigilant in monitoring for neurologic abnormalities following epidural catheter insertion or removal, particularly in the context of vascular surgery.
Collapse
Affiliation(s)
- Zechariah C Harris
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Henry K Su
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Amanda S Xi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alwin Somasundaram
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - A Sassan Sabouri
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
9
|
El Assar M, Rodríguez-Sánchez I, Álvarez-Bustos A, Rodríguez-Mañas L. Biomarkers of frailty. Mol Aspects Med 2024; 97:101271. [PMID: 38631189 DOI: 10.1016/j.mam.2024.101271] [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: 11/15/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/19/2024]
Abstract
Several biomarkers have been proposed to identify frailty, a multisystemic age-related syndrome. However, the complex pathophysiology and the absence of a consensus on a comprehensive and universal definition make it challenging to pinpoint a singular biomarker or set of biomarkers that conclusively characterize frailty. This review delves into the main laboratory biomarkers, placing special emphasis on those associated with various pathways closely tied to the frailty condition, such as inflammation, oxidative stress, mitochondrial dysfunction, metabolic and endocrine alterations and microRNA. Additionally, we provide a summary of different clinical biomarkers encompassing different tools that have been proposed to assess frailty. We further address various imaging biomarkers such as Dual Energy X-ray Absorptiometry, Bioelectrical Impedance analysis, Computed Tomography and Magnetic Resonance Imaging, Ultrasound and D3 Creatine dilution. Intervention to treat frailty, including non-pharmacological ones, especially those involving physical exercise and nutrition, and pharmacological interventions, that include those targeting specific mechanisms such as myostatin inhibitors, insulin sensitizer metformin and with special relevance for hormonal treatments are mentioned. We further address the levels of different biomarkers in monitoring the potential positive effects of some of these interventions. Despite the availability of numerous biomarkers, their performance and usefulness in the clinical arena are far from being satisfactory. Considering the multicausality of frailty, there is an increasing need to assess the role of sets of biomarkers and the combination between laboratory, clinical and image biomarkers, in terms of sensitivity, specificity and predictive values for the diagnosis and prognosis of the different outcomes of frailty to improve detection and monitoring of older people with frailty or at risk of developing it, being this a need in the everyday clinical practice.
Collapse
Affiliation(s)
- Mariam El Assar
- Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Alejandro Álvarez-Bustos
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Leocadio Rodríguez-Mañas
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Geriatría, Hospital Universitario de Getafe, Madrid, Spain.
| |
Collapse
|
10
|
Yousef A, Sosnowski DK, Fang L, Legaspi RJ, Korodimas J, Lee A, Magor KE, Seubert JM. Cardioprotective response and senescence in aged sEH null female mice exposed to LPS. Am J Physiol Heart Circ Physiol 2024; 326:H1366-H1385. [PMID: 38578240 DOI: 10.1152/ajpheart.00706.2023] [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: 11/08/2023] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Deterioration of physiological systems, like the cardiovascular system, occurs progressively with age impacting an individual's health and increasing susceptibility to injury and disease. Cellular senescence has an underlying role in age-related alterations and can be triggered by natural aging or prematurely by stressors such as the bacterial toxin lipopolysaccharide (LPS). The metabolism of polyunsaturated fatty acids by CYP450 enzymes produces numerous bioactive lipid mediators that can be further metabolized by soluble epoxide hydrolase (sEH) into diol metabolites, often with reduced biological effects. In our study, we observed age-related cardiac differences in female mice, where young mice demonstrated resistance to LPS injury, and genetic deletion or pharmacological inhibition of sEH using trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid attenuated LPS-induced cardiac dysfunction in aged female mice. Bulk RNA-sequencing analyses revealed transcriptomics differences in aged female hearts. The confirmatory analysis demonstrated changes to inflammatory and senescence gene markers such as Il-6, Mcp1, Il-1β, Nlrp3, p21, p16, SA-β-gal, and Gdf15 were attenuated in the hearts of aged female mice where sEH was deleted or inhibited. Collectively, these findings highlight the role of sEH in modulating the aging process of the heart, whereby targeting sEH is cardioprotective.NEW & NOTEWORTHY Soluble epoxide hydrolase (sEH) is an essential enzyme for converting epoxy fatty acids to their less bioactive diols. Our study suggests deletion or inhibition of sEH impacts the aging process in the hearts of female mice resulting in cardioprotection. Data indicate targeting sEH limits inflammation, preserves mitochondria, and alters cellular senescence in the aged female heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Liye Fang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Renald James Legaspi
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jacob Korodimas
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andy Lee
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Katharine E Magor
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Beydoun MA, Noren Hooten N, Fanelli-Kuczmaski MT, Maino Vieytes CA, Georgescu MF, Beydoun HA, Freeman DW, Evans MK, Zonderman AB. Growth Differentiation Factor 15 and Diet Quality Trajectory Interact to Determine Frailty Incidence among Middle-Aged Urban Adults. J Nutr 2024; 154:1652-1664. [PMID: 38479650 PMCID: PMC11130674 DOI: 10.1016/j.tjnut.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Elevated plasma growth differentiation factor 15 (GDF15) and poor diet quality may be associated with increased frailty incidence, although their interactive associations have not been assessed in urban middle-aged adults. OBJECTIVES We aimed to examine GDF15 and its interactive association with diet quality in relation to frailty incidence among a sample of middle-aged urban adults. METHODS The relationship between GDF15 and diet quality trajectories in relation to incident frailty was examined in a longitudinal study of participants in the Healthy Aging in Neighborhoods of Diversity across the Life Span (2004-2017). Serum GDF15 concentration and frailty incidence were primary exposure and outcome, respectively. Group-based trajectory models were used to assess diet quality trajectories (≤3 visits/participant, N = 945, N' = 2247 observations) using the Healthy Eating Index 2010 version (HEI-2010), Dietary Inflammatory Index, and mean adequacy ratio (MAR). Cox proportional hazards models were used, testing interactive associations of GDF15 and diet quality trajectories with frail/prefrail incidence (N = 400 frailty-free at first visit, N' = 604 observations, n = 168 incident frail/prefrail). RESULTS Both elevated GDF15 and lower diet quality trajectories were associated with a lower probability of remaining nonfrail (≤13 y follow-up). Among females, the "high diet quality" HEI-2010 trajectory had a hazard ratio (HR) of 0.15 [95% confidence interval (CI): 0.04, 0.54; P = 0.004; fully adjusted model] when compared with the "low diet quality" trajectory group. Among males only, there was an antagonistic interaction between lower HEI-2010 trajectory and elevated GDF15. Specifically, the HR for GDF15-frailty in the higher diet quality trajectory group (high/medium combined), and among males, was 2.69 (95% CI: 1.06, 6.62; P = 0.032), whereas among the lower diet quality trajectory group, the HR was 0.94 (95% CI: 0.49, 1.80; P = 0.86). Elevated GDF15 was independently associated with frailty among African American adults. CONCLUSIONS Pending replication, we found an antagonistic interaction between GDF15 and HEI-2010 trajectory in relation to frailty incidence among males.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, United States.
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, United States
| | | | | | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, United States
| | - Hind A Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC, United States; Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - David W Freeman
- Department of Oncological Sciences, School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, United States
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, United States
| |
Collapse
|
12
|
Palomo I, Wehinger S, Andrés V, García‐García FJ, Fuentes E. RhoA/rho kinase pathway activation in age-associated endothelial cell dysfunction and thrombosis. J Cell Mol Med 2024; 28:e18153. [PMID: 38568071 PMCID: PMC10989549 DOI: 10.1111/jcmm.18153] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 04/05/2024] Open
Abstract
The small GTPase RhoA and the downstream Rho kinase (ROCK) regulate several cell functions and pathological processes in the vascular system that contribute to the age-dependent risk of cardiovascular disease, including endothelial dysfunction, excessive permeability, inflammation, impaired angiogenesis, abnormal vasoconstriction, decreased nitric oxide production and apoptosis. Frailty is a loss of physiological reserve and adaptive capacity with advanced age and is accompanied by a pro-inflammatory and pro-oxidative state that promotes vascular dysfunction and thrombosis. This review summarises the role of the RhoA/Rho kinase signalling pathway in endothelial dysfunction, the acquisition of the pro-thrombotic state and vascular ageing. We also discuss the possible role of RhoA/Rho kinase signalling as a promising therapeutic target for the prevention and treatment of age-related cardiovascular disease.
Collapse
Affiliation(s)
- Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Medical Technology School, Thrombosis and Healthy Aging Research CenterUniversidad de TalcaTalcaChile
| | - Sergio Wehinger
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Medical Technology School, Thrombosis and Healthy Aging Research CenterUniversidad de TalcaTalcaChile
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Francisco J. García‐García
- Department of Geriatric MedicineHospital Universitario de Toledo, Instituto de Investigación de Castilla La Mancha (IDISCAM), CIBERFES (ISCIII)ToledoSpain
| | - Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Medical Technology School, Thrombosis and Healthy Aging Research CenterUniversidad de TalcaTalcaChile
| |
Collapse
|
13
|
Xu Q, Jia Y, Wang Y, Yang P, Sun L, Liu Y, Chang X, He Y, Guo D, Shi M, Zhang Y, Zhu Z. The bidirectional association between frailty index and cardiovascular disease: A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2024; 34:624-632. [PMID: 38176958 DOI: 10.1016/j.numecd.2023.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/22/2023] [Accepted: 10/19/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND AND AIM Observational studies have suggested a relationship between frailty and cardiovascular disease (CVD), but the causality is still uncertain. We used bidirectional Mendelian randomization (MR) design to investigate the potential causal associations between frailty and four main CVDs, including hypertension, myocardial infarction (MI), heart failure (HF), and atrial fibrillation (AF). METHODS AND RESULTS Independent single-nucleotide polymorphisms for frailty index (FI) and CVDs (hypertension, MI, HF, and AF) were selected as genetic instruments based on European-descent genome-wide association studies (GWASs). Summary-level data for outcomes on FI (n = 175,226), hypertension (n = 463,010), MI (n = 171,875), HF (n = 977323), and AF (n = 1,030,836) was derived from five large-scale GWASs of European ancestry. We used the inverse-variance weighted (IVW) method to examine the bidirectional associations between FI and CVDs in the main analyses. In the IVW MR analyses, genetically determined high FI was significantly associated with increased risks of hypertension (odds ratio [OR] per 1-SD increase: 1.07 [95 % confidence interval, 1.05-1.08]), MI (OR per 1-SD increase: 1.74 [1.21-2.51]), HF (OR per 1-SD increase: 1.28 [1.10-1.48]), and AF (OR per 1-SD increase: 1.20 [1.08-1.33]). In addition, genetically determined hypertension (beta: 1.406 [1.225-1.587]), MI (beta: 0.045 [0.023-0.067]), HF (beta: 0.105 [0.066-0.143]) and AF (beta: 0.021 [0.012-0.031]) were significantly associated with high FI. These findings were robustly supported by a series of sensitivity analyses with different MR models. CONCLUSIONS We found potential bidirectional causal associations between elevated FI and increased risks of CVD, suggesting mutual risk factors between frailty and CVD.
Collapse
Affiliation(s)
- Qingyun Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yiming Jia
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yinan Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lulu Sun
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yi Liu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xinyue Chang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yu He
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Daoxia Guo
- School of Nursing, Medical College of Soochow University, Suzhou, China
| | - Mengyao Shi
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
14
|
Qian Z, Huang Y, Zhang Y, Yang N, Fang Z, Zhang C, Zhang L. Metabolic clues to aging: exploring the role of circulating metabolites in frailty, sarcopenia and vascular aging related traits and diseases. Front Genet 2024; 15:1353908. [PMID: 38415056 PMCID: PMC10897029 DOI: 10.3389/fgene.2024.1353908] [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: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
Background: Physical weakness and cardiovascular risk increase significantly with age, but the underlying biological mechanisms remain largely unknown. This study aims to reveal the causal effect of circulating metabolites on frailty, sarcopenia and vascular aging related traits and diseases through a two-sample Mendelian Randomization (MR) analysis. Methods: Exposures were 486 metabolites analyzed in a genome-wide association study (GWAS), while outcomes included frailty, sarcopenia, arterial stiffness, atherosclerosis, peripheral vascular disease (PAD) and aortic aneurysm. Primary causal estimates were calculated using the inverse-variance weighted (IVW) method. Methods including MR Egger, weighted median, Q-test, and leave-one-out analysis were used for the sensitive analysis. Results: A total of 125 suggestive causative associations between metabolites and outcomes were identified. Seven strong causal links were ultimately identified between six metabolites (kynurenine, pentadecanoate (15:0), 1-arachidonoylglycerophosphocholine, androsterone sulfate, glycine and mannose) and three diseases (sarcopenia, PAD and atherosclerosis). Besides, metabolic pathway analysis identified 13 significant metabolic pathways in 6 age-related diseases. Furthermore, the metabolite-gene interaction networks were constructed. Conclusion: Our research suggested new evidence of the relationship between identified metabolites and 6 age-related diseases, which may hold promise as valuable biomarkers.
Collapse
Affiliation(s)
- Zonghao Qian
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhen Huang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yucong Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ni Yang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziwei Fang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Le Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Gu Y, Li Z, Dang A, Zhang W, Liu J, Han X, Li Y, Lv N. Obesity, birth weight, and lifestyle factors for frailty: a Mendelian randomization study. Aging (Albany NY) 2023; 15:14066-14085. [PMID: 38095641 PMCID: PMC10756094 DOI: 10.18632/aging.205290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/17/2023] [Indexed: 12/21/2023]
Abstract
Obesity, birth weight and lifestyle factors have been found associated with the risk of frailty in observational studies, but whether these associations are causal is uncertain. We conducted a two-sample Mendelian randomization study to investigate the associations. Genetic instruments associated with the exposures at the genome-wide significance level (p < 5 × 10-8) were selected from corresponding genome-wide association studies (n = 143,677 to 703,901 individuals). Summary-level data for the frailty index were obtained from the UK Biobank (n = 164,610) and Swedish TwinGene (n = 10,616). The β of the frailty index was 0.15 (p = 3.88 × 10-9) for 1 standard deviation increase in the prevalence of smoking initiation, 0.19 (p = 3.54 × 10-15) for leisure screen time, 0.13 (p = 5.26 × 10-7) for body mass index and 0.13 (p = 1.80 × 10-4) for waist circumference. There was a suggestive association between genetically predicted higher birth weight and moderate-to-vigorous intensity physical activity with the decreased risk of the frailty index. We observed no causal association between genetically predicted age of smoking initiation and alcoholic drinks per week with the frailty index. This study supports the causal roles of smoking initiation, leisure screen time, overall obesity, and abdominal obesity in frailty. The possible association between higher birth weight, proper physical activity and a decreased risk of frailty needs further confirmation.
Collapse
Affiliation(s)
- Yingzhen Gu
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zuozhi Li
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Aimin Dang
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei Zhang
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jinxing Liu
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaorong Han
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yifan Li
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Naqiang Lv
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Salvioli S, Basile MS, Bencivenga L, Carrino S, Conte M, Damanti S, De Lorenzo R, Fiorenzato E, Gialluisi A, Ingannato A, Antonini A, Baldini N, Capri M, Cenci S, Iacoviello L, Nacmias B, Olivieri F, Rengo G, Querini PR, Lattanzio F. Biomarkers of aging in frailty and age-associated disorders: State of the art and future perspective. Ageing Res Rev 2023; 91:102044. [PMID: 37647997 DOI: 10.1016/j.arr.2023.102044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
According to the Geroscience concept that organismal aging and age-associated diseases share the same basic molecular mechanisms, the identification of biomarkers of age that can efficiently classify people as biologically older (or younger) than their chronological (i.e. calendar) age is becoming of paramount importance. These people will be in fact at higher (or lower) risk for many different age-associated diseases, including cardiovascular diseases, neurodegeneration, cancer, etc. In turn, patients suffering from these diseases are biologically older than healthy age-matched individuals. Many biomarkers that correlate with age have been described so far. The aim of the present review is to discuss the usefulness of some of these biomarkers (especially soluble, circulating ones) in order to identify frail patients, possibly before the appearance of clinical symptoms, as well as patients at risk for age-associated diseases. An overview of selected biomarkers will be discussed in this regard, in particular we will focus on biomarkers related to metabolic stress response, inflammation, and cell death (in particular in neurodegeneration), all phenomena connected to inflammaging (chronic, low-grade, age-associated inflammation). In the second part of the review, next-generation markers such as extracellular vesicles and their cargos, epigenetic markers and gut microbiota composition, will be discussed. Since recent progresses in omics techniques have allowed an exponential increase in the production of laboratory data also in the field of biomarkers of age, making it difficult to extract biological meaning from the huge mass of available data, Artificial Intelligence (AI) approaches will be discussed as an increasingly important strategy for extracting knowledge from raw data and providing practitioners with actionable information to treat patients.
Collapse
Affiliation(s)
- Stefano Salvioli
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | | | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy
| | - Sara Carrino
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Eleonora Fiorenzato
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy
| | - Alessandro Gialluisi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Angelo Antonini
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), Department of Neurosciences, University of Padova, Padova, Italy
| | - Nicola Baldini
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Miriam Capri
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Scientific Institute of Telese Terme, Telese Terme, Italy
| | | | | |
Collapse
|
17
|
Main EN, Cruz TM, Bowlin GL. Mitochondria as a therapeutic: a potential new frontier in driving the shift from tissue repair to regeneration. Regen Biomater 2023; 10:rbad070. [PMID: 37663015 PMCID: PMC10468651 DOI: 10.1093/rb/rbad070] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/12/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
Fibrosis, or scar tissue development, is associated with numerous pathologies and is often considered a worst-case scenario in terms of wound healing or the implantation of a biomaterial. All that remains is a disorganized, densely packed and poorly vascularized bundle of connective tissue, which was once functional tissue. This creates a significant obstacle to the restoration of tissue function or integration with any biomaterial. Therefore, it is of paramount importance in tissue engineering and regenerative medicine to emphasize regeneration, the successful recovery of native tissue function, as opposed to repair, the replacement of the native tissue (often with scar tissue). A technique dubbed 'mitochondrial transplantation' is a burgeoning field of research that shows promise in in vitro, in vivo and various clinical applications in preventing cell death, reducing inflammation, restoring cell metabolism and proper oxidative balance, among other reported benefits. However, there is currently a lack of research regarding the potential for mitochondrial therapies within tissue engineering and regenerative biomaterials. Thus, this review explores these promising findings and outlines the potential for mitochondrial transplantation-based therapies as a new frontier of scientific research with respect to driving regeneration in wound healing and host-biomaterial interactions, the current successes of mitochondrial transplantation that warrant this potential and the critical questions and remaining obstacles that remain in the field.
Collapse
Affiliation(s)
- Evan N Main
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| | - Thaiz M Cruz
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| | - Gary L Bowlin
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| |
Collapse
|
18
|
Trostchansky A, Alarcon M. An Overview of Two Old Friends Associated with Platelet Redox Signaling, the Protein Disulfide Isomerase and NADPH Oxidase. Biomolecules 2023; 13:biom13050848. [PMID: 37238717 DOI: 10.3390/biom13050848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/30/2022] [Accepted: 01/18/2023] [Indexed: 05/28/2023] Open
Abstract
Oxidative stress participates at the baseline of different non-communicable pathologies such as cardiovascular diseases. Excessive formation of reactive oxygen species (ROS), above the signaling levels necessary for the correct function of organelles and cells, may contribute to the non-desired effects of oxidative stress. Platelets play a relevant role in arterial thrombosis, by aggregation triggered by different agonists, where excessive ROS formation induces mitochondrial dysfunction and stimulate platelet activation and aggregation. Platelet is both a source and a target of ROS, thus we aim to analyze both the platelet enzymes responsible for ROS generation and their involvement in intracellular signal transduction pathways. Among the proteins involved in these processes are Protein Disulphide Isomerase (PDI) and NADPH oxidase (NOX) isoforms. By using bioinformatic tools and information from available databases, a complete bioinformatic analysis of the role and interactions of PDI and NOX in platelets, as well as the signal transduction pathways involved in their effects was performed. We focused the study on analyzing whether these proteins collaborate to control platelet function. The data presented in the current manuscript support the role that PDI and NOX play on activation pathways necessary for platelet activation and aggregation, as well as on the platelet signaling imbalance produced by ROS production. Our data could be used to design specific enzyme inhibitors or a dual inhibition for these enzymes with an antiplatelet effect to design promising treatments for diseases involving platelet dysfunction.
Collapse
Affiliation(s)
- Andrés Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Marcelo Alarcon
- Thrombosis Research Center, Universidad de Talca, Talca 3460000, Chile
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
19
|
Growth Differentiation Factor 15 Is Associated with Platelet Reactivity in Patients with Acute Coronary Syndrome. J Clin Med 2023; 12:jcm12041627. [PMID: 36836162 PMCID: PMC9966081 DOI: 10.3390/jcm12041627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/01/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Bleeding events in patients with acute coronary syndrome (ACS) are a risk factor for adverse outcomes, including mortality. We investigated the association of growth differentiation factor (GDF)-15, an established predictor of bleeding complications, with on-treatment platelet reactivity in ACS patients undergoing coronary stenting receiving prasugrel or ticagrelor. Platelet aggregation was measured by multiple electrode aggregometry (MEA) in response to adenosine diphosphate (ADP), arachidonic acid (AA), thrombin receptor-activating peptide (TRAP, a protease-activated receptor-1 (PAR-1) agonist), AYPGKF (a PAR-4 agonist) and collagen (COL). GDF-15 levels were measured using a commercially available assay. GDF-15 correlated inversely with MEA ADP (r = -0.202, p = 0.004), MEA AA (r = -0.139, p = 0.048) and MEA TRAP (r = -0.190, p = 0.007). After adjustment, GDF-15 was significantly associated with MEA TRAP (β = -0.150, p = 0.044), whereas no significant associations were detectable for the other agonists. Patients with low platelet reactivity in response to ADP had significantly higher GDF-15 levels (p = 0.005). In conclusion, GDF-15 is inversely associated with TRAP-inducible platelet aggregation in ACS patients treated with state-of-the-art antiplatelet therapy and significantly elevated in patients with low platelet reactivity in response to ADP.
Collapse
|
20
|
Konstorum A, Mohanty S, Zhao Y, Melillo A, Vander Wyk B, Nelson A, Tsang S, Blevins TP, Belshe R, Chawla DG, Rondina MT, Gill TM, Montgomery RR, Allore HG, Kleinstein SH, Shaw AC. Platelet response to influenza vaccination reflects effects of aging. Aging Cell 2023; 22:e13749. [PMID: 36656789 PMCID: PMC9924941 DOI: 10.1111/acel.13749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/21/2022] [Accepted: 11/15/2022] [Indexed: 01/20/2023] Open
Abstract
Platelets are uniquely positioned as mediators of not only hemostasis but also innate immunity. However, how age and geriatric conditions such as frailty influence platelet function during an immune response remains unclear. We assessed the platelet transcriptome at baseline and following influenza vaccination in Younger (age 21-35) and Older (age ≥65) adults (including community-dwelling individuals who were largely non-frail and skilled nursing facility (SNF)-resident adults who nearly all met criteria for frailty). Prior to vaccination, we observed an age-associated increase in the expression of platelet activation and mitochondrial RNAs and decrease in RNAs encoding proteins mediating translation. Age-associated differences were also identified in post-vaccination response trajectories over 28 days. Using tensor decomposition analysis, we found increasing RNA expression of genes in platelet activation pathways in young participants, but decreasing levels in (SNF)-resident adults. Translation RNA trajectories were inversely correlated with these activation pathways. Enhanced platelet activation was found in community-dwelling older adults at the protein level, compared to young individuals both prior to and post-vaccination; whereas SNF residents showed decreased platelet activation compared to community-dwelling older adults that could reflect the influence of decreased translation RNA expression. Our results reveal alterations in the platelet transcriptome and activation responses that may contribute to age-associated chronic inflammation and the increased incidence of thrombotic and pro-inflammatory diseases in older adults.
Collapse
Affiliation(s)
- Anna Konstorum
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
| | - Subhasis Mohanty
- Department of Internal Medicine, Section of Infectious DiseasesYale School of MedicineNew HavenConnecticutUSA
| | - Yujiao Zhao
- Section of Rheumatology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Anthony Melillo
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
| | - Brent Vander Wyk
- Department of Internal Medicine, Section of Geriatrics and Program on AgingYale School of MedicineNew HavenConnecticutUSA
| | - Allison Nelson
- Department of Internal Medicine, Section of Infectious DiseasesYale School of MedicineNew HavenConnecticutUSA
| | - Sui Tsang
- Department of Internal Medicine, Section of Geriatrics and Program on AgingYale School of MedicineNew HavenConnecticutUSA
| | - Tamara P. Blevins
- Division of Infectious Diseases, Department of MedicineSaint Louis University School of MedicineSt. LouisMissouriUSA
| | - Robert B. Belshe
- Division of Infectious Diseases, Department of MedicineSaint Louis University School of MedicineSt. LouisMissouriUSA
| | - Daniel G. Chawla
- Program in Computational Biology and BioinformaticsYale UniversityNew HavenConnecticutUSA
| | - Matthew T. Rondina
- Departments of Internal Medicine and Pathology, and the Molecular Medicine ProgramUniversity of Utah HealthSalt Lake CityUtahUSA
- Department of Medicine and the GRECCGeorge E. Wahlen VAMCSalt Lake CityUtahUSA
| | - Thomas M. Gill
- Department of Internal Medicine, Section of Geriatrics and Program on AgingYale School of MedicineNew HavenConnecticutUSA
| | - Ruth R. Montgomery
- Section of Rheumatology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Heather G. Allore
- Department of Internal Medicine, Section of Geriatrics and Program on AgingYale School of MedicineNew HavenConnecticutUSA
| | - Steven H. Kleinstein
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
- Program in Computational Biology and BioinformaticsYale UniversityNew HavenConnecticutUSA
| | - Albert C. Shaw
- Department of Internal Medicine, Section of Infectious DiseasesYale School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
21
|
An Insight into Platelets at Older Age: Cellular and Clinical Perspectives. Subcell Biochem 2023; 102:343-363. [PMID: 36600139 DOI: 10.1007/978-3-031-21410-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Higher access to medical care, advanced diagnostic tools, and overall public health improvements have favored increased humans lifespan. With a growing proportion of older adults, the associated costs to care for ageing-associated conditions will continue to grow. This chapter highlights recent cellular and clinical evidence of platelets at an older age, from the hyperreactive phenotype associated with thrombosis to the well-known hallmarks of ageing identifiable in platelets and their potential functional implications on platelets at an older age. Therefore, it is imperative to understand platelets' molecular and cellular mechanisms during ageing in health and disease. New knowledge will favor the development of new ways to prevent some of the age-associated complications where platelets are key players.
Collapse
|
22
|
Gonçalves RSDSA, Maciel ÁCC, Rolland Y, Vellas B, de Souto Barreto P. Frailty biomarkers under the perspective of geroscience: A narrative review. Ageing Res Rev 2022; 81:101737. [PMID: 36162706 DOI: 10.1016/j.arr.2022.101737] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 01/31/2023]
Abstract
Cellular and molecular aging biomarkers might contribute to identify at-risk individuals for frailty before overt clinical manifestations appear. Although studies on the associations of aging biomarkers and frailty exist, no investigation has gathered this information using a structured framework for identifying aging biomarkers; as a result, the evidence on frailty and aging biomarkers is diffuse and incomplete. Therefore, this narrative review aimed to gather information on the associations of the hallmarks of aging and frailty under the perspective of geroscience. The literature on human studies on this topic is sparse and mainly composed of cross-sectional investigations performed in small study samples. The main putative aging biomarkers associated to frailty were: mitochondrial DNA copy number (genomic instability and mitochondrial dysfunction), telomere length (telomere attrition), global DNA methylation (epigenetic alterations), Hsp70 and Hsp72 (loss of proteostasis), IGF-1 and SIRT1 (deregulated nutrient-sensing), GDF-15 (mitochondrial dysfunction, cellular senescence and altered intercellular communication), CD4 + and CD8 + cell percentages (cellular senescence), circulating osteogenic progenitor (COP) cells (stem cell exhaustion), and IL-6, CRP and TNF-alpha (altered intercellular communication). IGF-1, SIRT1, GDF-15, IL-6, CRP and TNF-alpha presented more evidence among these biomarkers, highlighting the importance of inflammation and nutrient sensing on frailty. Further longitudinal studies investigating biomarkers across the hallmarks of aging would provide valuable information on this topic.
Collapse
Affiliation(s)
| | | | - Yves Rolland
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France.
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France.
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France.
| |
Collapse
|
23
|
De Paepe B. The Cytokine Growth Differentiation Factor-15 and Skeletal Muscle Health: Portrait of an Emerging Widely Applicable Disease Biomarker. Int J Mol Sci 2022; 23:ijms232113180. [PMID: 36361969 PMCID: PMC9654287 DOI: 10.3390/ijms232113180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 12/04/2022] Open
Abstract
Growth differentiation factor 15 (GDF-15) is a stress-induced transforming growth factor-β superfamily cytokine with versatile functions in human health. Elevated GDF-15 blood levels associate with multiple pathological conditions, and are currently extensively explored for diagnosis, and as a means to monitor disease progression and evaluate therapeutic responses. This review analyzes GDF-15 in human conditions specifically focusing on its association with muscle manifestations of sarcopenia, mitochondrial myopathy, and autoimmune and viral myositis. The use of GDF-15 as a widely applicable health biomarker to monitor muscle disease is discussed, and its potential as a therapeutic target is explored.
Collapse
Affiliation(s)
- Boel De Paepe
- Neuromuscular Reference Center, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| |
Collapse
|
24
|
Mensah E, Ali K, Banya W, Kirkham FA, Mengozzi M, Ghezzi P, Rajkumar C. FRailty and Arterial stiffness - the role of oXidative stress and Inflammation (FRAXI study). Biomark Insights 2022; 17:11772719221130719. [PMID: 36275839 PMCID: PMC9583202 DOI: 10.1177/11772719221130719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/16/2022] [Indexed: 11/08/2022] Open
Abstract
Objective There is an association between frailty and arterial stiffness. However, arterial stiffness does not uniformly correlate with the spectrum of frailty states. Both oxidative stress and inflammaging contribute to vascular ageing. There are no human studies exploring links between arterial stiffness, oxidative stress, inflammaging and frailty. Our objective is to investigate arterial stiffness and inflammaging as predictors of frailty states. Methods An observational longitudinal cohort study will be used to examine the association between arterial stiffness, oxidative stress and inflammation in 50 older adults (⩾70 years) with clinical frailty scores (CFS) ⩽6 over 6 months. All study measurements will be taken at baseline. Frailty assessment will include hand-grip strength, timed-up and go test, mini-mental state examination, geriatric depression scale and sarcopenia using body composition measurements with Tanita®. Arterial stiffness measurements will include carotid-femoral pulse wave velocity (cfPWV) and carotid-radial pulse wave velocity (crPWV) using Complior (Alam Medical, France). CAVI device will measure Cardio-ankle vascular index and ankle brachial index (ABI). Oxidative stress blood markers nitrotyrosine (NT) and 8-hydroxy-2'-deoxyguanosin (8-oxo-dG) and inflammation markers high-sensitive C-reactive protein (hs-CRP) and interlukin-6(IL-6) will be measured at baseline and 6 month along with lipid profile and glycated haemoglobin. Results data analysis plan Descriptive statistics for continuous data using means and standard deviations for normality distributed variables or medians and inter-quartile ranges for skewed variables will be used. Participants will be categorised into CFS 1-3, and CFS 4-6. Categorical data will use frequencies and comparison between groups. Change in frailty between the groups over 6 months will be compared using paired t-test. Simple linear regression will be done between frailty measures, arterial stiffness, inflammation and oxidative stress biomarkers. Significance will be at P < .05. Conclusion This study data will inform a larger, multi-centre study exploring further the interplay between frailty, biomarkers and arterial stiffness parameters.
Collapse
Affiliation(s)
- Ekow Mensah
- Brighton and Sussex Clinical Trials
Unit, University Hospitals Sussex NHS Trust, Brighton, UK,Ekow Mensah, Brighton and Sussex Clinical
Trials Unit, University Hospitals Sussex NHS Trust, Audrey Emerton Building,
Eastern Road, Brighton, BN2 5BE, UK.
| | - Khalid Ali
- Brighton and Sussex Clinical Trials
Unit, University Hospitals Sussex NHS Trust, Brighton, UK,Department of Medicine, Brighton and
Sussex Medical School, University of Sussex, Brighton, UK
| | - Winston Banya
- Research Office, Royal Brompton and
Harefield Clinical Group, Guy’s and St. Thomas’ NHS Foundation Trust, London,
UK
| | - Frances Ann Kirkham
- Brighton and Sussex Clinical Trials
Unit, University Hospitals Sussex NHS Trust, Brighton, UK
| | - Manuela Mengozzi
- Department of Medicine, Brighton and
Sussex Medical School, University of Sussex, Brighton, UK
| | | | - Chakravarthi Rajkumar
- Brighton and Sussex Clinical Trials
Unit, University Hospitals Sussex NHS Trust, Brighton, UK,Department of Medicine, Brighton and
Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
25
|
Batko-Szwaczka A, Francuz T, Kosowska A, Cogiel A, Dudzińska-Griszek J, Wilczyński K, Hornik B, Janusz-Jenczeń M, Włodarczyk I, Wnuk B, Szołtysek J, Durmała J, Dulawa J, Szewieczek J. Predictors of Adverse Outcomes in Healthy Aging Adults: Coronary Artery Disease, Lower Educational Status and Higher P-Selectin Levels. Clin Interv Aging 2022; 17:1173-1185. [PMID: 35957925 PMCID: PMC9362850 DOI: 10.2147/cia.s363881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/02/2022] [Indexed: 11/29/2022] Open
Abstract
Background Societal aging – as a global demographic phenomenon – shows no indication of abating. As a result, the problem of age-associated disability and related long-term care is emerging as a major public health challenge. It is important that methods for identifying older adults at risk of adverse outcomes are implemented early. Methods The study group consisted of 145 individuals, 44.1% women, who were randomized from community-dwelling 60–74-year-old adults. A comprehensive geriatric assessment was supplemented with Fried frailty phenotype evaluation and blood tests (including adhesion molecules, matrix metalloproteinases and neurotrophic factors). A follow-up by phone call was made for at least 3 years after the initial examination. Composite endpoint (CE) included falls, hospitalization, institutionalization and death. Results Mean study group age was 66.5 ± 4.1 years (\documentclass[12pt]{minimal}
\usepackage{wasysym}
\usepackage[substack]{amsmath}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage[mathscr]{eucal}
\usepackage{mathrsfs}
\DeclareFontFamily{T1}{linotext}{}
\DeclareFontShape{T1}{linotext}{m}{n} {linotext }{}
\DeclareSymbolFont{linotext}{T1}{linotext}{m}{n}
\DeclareSymbolFontAlphabet{\mathLINOTEXT}{linotext}
\begin{document}
$$\overline {\rm{X}} {\rm{ \pm SD}}$$
\end{document}) and mean number of diseases was 3.7 ± 2.2. Functional status of the subjects was good, as indicated by high Barthel Index scores of 99.1 ± 2.4, MMSE scores of 29.0 ±1.5 and no frailty case. During a three-year follow-up, 71 participants (49.0%) experienced any CE-events. The Wilcoxon-Gehan test indicates that a higher probability of three-year CE completion was associated with an age >65 years (P = 0.006), coronary artery disease (CAD) (P = 0.008), 6-Minute Walk Test <432 m (P = 0.034), serum glucose >120 mg/dL (P = 0.047), serum cortisol >10 μg/dL (P = 0.011), leptin ≥15 ng/mL (P = 0.018), P-selectin ≥23 ng/mL (P = 0.006) and GDNF ≥20 pg/mL (P = 0.004). CAD (OR = 3.64, 95% CI = 1.53−8.69, P = 0.004), educational status (OR = 0.87, 95% CI = 0.77−0.98, P = 0.022) and P-selectin levels (OR = 1.07, 95% CI = 1.02−1.13, P = 0.013) were independent measures predicting three-year CE occurrence in multivariate logistic regression analysis adjusted for clinical and functional measures, and blood tests. Conclusion Coronary artery disease, poorer lower educational status and higher P-selectin levels were predictive of adverse outcomes in the community-dwelling healthy-aging early-old adults during three-year follow-up.
Collapse
Affiliation(s)
- Agnieszka Batko-Szwaczka
- Department of Geriatrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
- Correspondence: Agnieszka Batko-Szwaczka, Department of Geriatrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, Poniatowskiego 15, Katowice, 40-055, Poland, Tel +48323598239, Fax +48322059483, Email
| | - Tomasz Francuz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agnieszka Kosowska
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Anna Cogiel
- Department of Geriatrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Joanna Dudzińska-Griszek
- Department of Geriatrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Krzysztof Wilczyński
- Department of Geriatrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Beata Hornik
- Department of Internal Nursing, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Magdalena Janusz-Jenczeń
- Department of Internal Nursing, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Iwona Włodarczyk
- Department of Internal Nursing, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Bartosz Wnuk
- Department of Rehabilitation, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Joanna Szołtysek
- Department of Rehabilitation, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Jacek Durmała
- Department of Rehabilitation, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Jan Dulawa
- Department of Internal Medicine and Metabolic Diseases, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Jan Szewieczek
- Department of Geriatrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
26
|
Sepúlveda M, Arauna D, García F, Albala C, Palomo I, Fuentes E. Frailty in Aging and the Search for the Optimal Biomarker: A Review. Biomedicines 2022; 10:1426. [PMID: 35740447 PMCID: PMC9219911 DOI: 10.3390/biomedicines10061426] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 01/09/2023] Open
Abstract
In the context of accelerated aging of the population worldwide, frailty has emerged as one of the main risk factors that can lead to loss of self-sufficiency in older people. This syndrome is defined as a reduced state of physiological reserve and functional capacity. The main diagnostic tools for frailty are based on scales that show deficits compared to their clinical application, such as the Fried frailty phenotype, among others. In this context, it is important to have one or more biomarkers with clinical applicability that can objectively and precisely determine the degree or risk of frailty in older people. The objective of this review was to analyze the biomarkers associated with frailty, classified according to the pathophysiological components of this syndrome (inflammation, coagulation, antioxidants, and liver function, among others). The evidence demonstrates that biomarkers associated with inflammation, oxidative stress, skeletal/cardiac muscle function, and platelet function represent the most promising markers of frailty due to their pathophysiological association with this syndrome. To a lesser extent but with the possibility of greater innovation, biomarkers associated with growth factors, vitamins, amino acids, and miRNAs represent alternatives as markers of this geriatric syndrome. Likewise, the incorporation of artificial intelligence represents an interesting approach to strengthening the diagnosis of frailty by biomarkers.
Collapse
Affiliation(s)
- Magdalena Sepúlveda
- Thrombosis Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca 3480094, Chile; (M.S.); (D.A.)
| | - Diego Arauna
- Thrombosis Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca 3480094, Chile; (M.S.); (D.A.)
| | - Francisco García
- Department of Geriatric Medicine, Complejo Hospitalario de Toledo, 45007 Toledo, Spain;
| | - Cecilia Albala
- Unidad de Nutrición Pública, Instituto de Nutrición y Tecnología de los Alimentos, Interuniversity Center for Healthy Aging, Universidad de Chile, Santiago 8320000, Chile;
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca 3480094, Chile; (M.S.); (D.A.)
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Faculty of Health Sciences, Interuniversity Center for Healthy Aging, Universidad de Talca, Talca 3480094, Chile; (M.S.); (D.A.)
| |
Collapse
|
27
|
Growth Differentiation Factor-15 Correlates Inversely with Protease-Activated Receptor-1-Mediated Platelet Reactivity in Patients with Left Ventricular Assist Devices. Pharmaceuticals (Basel) 2022; 15:ph15040484. [PMID: 35455481 PMCID: PMC9031879 DOI: 10.3390/ph15040484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 02/04/2023] Open
Abstract
Growth differentiation factor (GDF)-15 inhibits platelet activation, prevents thrombus formation, and has been linked to bleeding events. This was a prospective study including 51 left-ventricular assist device (LVAD) patients on aspirin and phenprocoumon. Platelet surface expression of activated glycoprotein (GP) IIb/IIIa was assessed by flow cytometry, and platelet aggregation was measured by multiple electrode aggregometry (MEA) in response to arachidonic acid (AA), adenosine diphosphate (ADP), and thrombin receptor-activating peptide (TRAP), a protease-activated-receptor-1 (PAR-1) agonist. GDF-15 was determined with a commercially-available assay. There was a trend towards an inverse correlation of GDF-15 with activated GPIIb/IIIa in response to TRAP (r = −0.275, p = 0.0532) but not in response to AA and ADP. Moreover, GDF-15 correlated with MEA TRAP (r = −0.326, p = 0.0194), whereas it did not correlate with MEA ADP and MEA AA. In a second step, GDF-15 levels in the fourth quartile were defined as high GDF-15. Patients with high GDF-15 showed significantly lower TRAP-inducible platelet aggregation by MEA compared to patients in the first quartile (63 AU vs. 113 AU, p = 0.0065). In conclusion, in LVAD patients receiving state-of-the-art antithrombotic therapy, GDF-15 correlates inversely with residual platelet reactivity via PAR-1.
Collapse
|
28
|
Cao L, Zhou Y, Liu H, Shi M, Wei Y, Xia Y. Bidirectional Longitudinal Study of Frailty and Depressive Symptoms Among Older Chinese Adults. Front Aging Neurosci 2022; 14:791971. [PMID: 35221990 PMCID: PMC8866966 DOI: 10.3389/fnagi.2022.791971] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveFrailty and depression, as two common conditions among older adults in China, have been shown to be closely related to each other. The aim of this study was to investigate the bidirectional effects between frailty and depressive symptoms in Chinese population.MethodsThe bidirectional effect of frailty with depressive symptoms was analyzed among 5,303 adults ≥ 60 years of age from the China Health and Retirement Longitudinal Study (CHARLS). Phenotype and a frailty index were used to measure frailty. Depressive symptoms were evaluated using the Epidemiological Studies Depression Scale (CES-D). Logistic regression and Cox proportional hazard regression models were used to determine the bidirectional effects of frailty and depressive symptoms in cross-sectional and cohort studies, respectively. Subgroup and sensitivity analyses were further used to further verify the associations.ResultsIn the cross-sectional study, the multivariate-adjusted ORs (95% CIs) for depressive symptoms among pre-frail and frail adults, as defined by the frailty index and phenotype, were 3.05 (2.68–3.49), and 9.78 (8.02–12.03), respectively. Depressed participants showed higher risks of pre-frailty and frailty [frailty index, 3.07 (2.69–3.50); and phenotypic frailty, 9.95 (8.15–12.24)]. During follow-up, the multivariate-adjusted HRs (95% CIs) for depressive symptoms among pre-frail and frail participants, as defined by the frailty index and phenotype, were 1.38 (1.22–1.57), and 1.30 (1.14–1.48), respectively. No significant relationship existed between baseline depressive symptoms and the incidence of frailty. Moreover, the results from subgroup and sensitivity analyses were consistent with the main results.ConclusionAlthough a cross-sectional bidirectional association between depressive symptom and frailty has been observed in older (≥60 years old) Chinese adults, frailty may be an independent predictor for subsequent depression. Moreover, no effect of depressive symptoms on subsequent frailty was detected. Additional bidirectional studies are warranted in China.
Collapse
Affiliation(s)
- Limin Cao
- The Third Central Hospital of Tianjin, Tianjin, China
| | - Yuhan Zhou
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huiyuan Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mengyuan Shi
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingliang Wei
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Yingliang Wei,
| | - Yang Xia
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Yang Xia, ,
| |
Collapse
|
29
|
Picca A, Coelho-Junior HJ, Calvani R, Marzetti E, Vetrano DL. Biomarkers shared by frailty and sarcopenia in older adults: A systematic review and meta-analysis. Ageing Res Rev 2022; 73:101530. [PMID: 34839041 DOI: 10.1016/j.arr.2021.101530] [Citation(s) in RCA: 160] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/04/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Physical frailty and sarcopenia show extensive clinical similarities. Whether biomarkers exist that are shared by the two conditions is presently unclear. METHODS We conducted a systematic review and meta-analysis of cross-sectional and longitudinal studies that investigated the association of frailty and/or sarcopenia with biomarkers as a primary or secondary outcome in adults aged 60 years and older. Only studies published in English that defined frailty using a validated scale and/or questionnaire and diagnosed sarcopenia according to the presence of muscle atrophy plus dynapenia or low physical function were included. Studies were identified from a systematic search of MEDLINE and SCOPUS databases from inception through August 2020. The quality of reporting of each study was assessed by using the Quality Assessment Tool for Observational Cohort, Cross-Sectional and Case-Control studies of the National Institute of Health. A meta-analysis was conducted when at least three studies investigated the same biomarker in both frailty and sarcopenia. Pooled effect size was calculated based on standard mean differences and random-effect models. Sensitivity analysis was performed based on age and the setting where the study was conducted. RESULTS Eighty studies (58 on frailty and 22 on sarcopenia) met the inclusion criteria and were included in the qualitative analysis. Studies on frailty included 33,160 community-dwellers, hospitalized, or institutionalized older adults (60-88 years) from 21 countries. Studies on sarcopenia involved 4904 community-living and institutionalized older adults (68-87.6 years) from 9 countries. Several metabolic, inflammatory, and hematologic markers were found to be shared between the two conditions. Albumin and hemoglobin were negatively associated with both frailty and sarcopenia. Interleukin 6 was associated with frailty and sarcopenia only in people aged < 75. Community-dwelling older adults with frailty and sarcopenia had higher levels of tumor necrosis factor alpha compared with their robust and non-sarcopenic counterparts. CONCLUSIONS A set of metabolic, hematologic, and inflammatory biomarkers was found to be shared by frailty and sarcopenia. These findings fill a knowledge gap in the quest of biomarkers for these conditions and provide a rationale for biomarker selection in studies on frailty and sarcopenia.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | | | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden.
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Department of Geriatrics and Orthopedics, Rome, Italy
| | - Davide Liborio Vetrano
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| |
Collapse
|
30
|
Alcazar J, Frandsen U, Prokhorova T, Kamper RS, Haddock B, Aagaard P, Suetta C. Changes in systemic GDF15 across the adult lifespan and their impact on maximal muscle power: the Copenhagen Sarcopenia Study. J Cachexia Sarcopenia Muscle 2021; 12:1418-1427. [PMID: 34617415 PMCID: PMC8718085 DOI: 10.1002/jcsm.12823] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/14/2021] [Accepted: 09/07/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Although growth differentiation factor 15 (GDF15) is known to increase with disease and is associated with low physical performance, the role of GDF15 in normal ageing is still not fully understood. Specifically, the influence of circulating GDF15 on impairments in maximal muscle power (a major contributor to functional limitations) and the underlying components has not been investigated. METHODS Data from 1305 healthy women and men aged 20 to 93 years from The Copenhagen Sarcopenia Study were analysed. Circulating levels of GDF15 and markers of inflammation (tumor necrosis factor-alpha, interleukin-6, and high-sensitivity C-reactive protein) were measured by ELISA (R&D Systems) and multiplex bead-based immunoassays (Bio-Rad). Relative (normalized to body mass), allometric (normalized to height squared), and specific (normalized to leg muscle mass) muscle power were assessed by the Nottingham power rig [leg extension power (LEP)] and the 30 s sit-to-stand (STS) muscle power test. Total body fat, visceral fat, and leg lean mass were assessed by dual energy X-ray absorptiometry. Leg skeletal muscle index was measured as leg lean mass normalized to body height squared. RESULTS Systemic levels of GDF15 increased progressively as a function of age in women (1.1 ± 0.4 pg·mL-1 ·year-1 ) and men (3.3 ± 0.6 pg·mL-1 ·year-1 ) (both P < 0.05). Notably, GDF15 increased at a faster rate from the age of 65 years in women (11.5 ± 1.2 pg·mL-1 ·year-1 , P < 0.05) and 70 years in men (19.3 ± 2.3 pg·mL-1 ·year-1 , P < 0.05), resulting in higher GDF15 levels in men compared with women above the age of 65 years (P < 0.05). Independently of age and circulatory markers of inflammation, GDF15 was negatively correlated to relative STS power (P < 0.05) but not LEP, in both women and men. These findings were mainly explained by negative associations of GDF15 with specific STS power in women and men (both P < 0.05). CONCLUSIONS A J-shaped relationship between age and systemic GDF15 was observed, with men at older age showing steeper increases and elevated GDF15 levels compared with women. Importantly, circulating GDF15 was independently and negatively associated with relative STS power, supporting the potential role of GDF15 as a sensitive biomarker of frailty in older people.
Collapse
Affiliation(s)
- Julian Alcazar
- GENUD Toledo Research Group, Universidad de Castilla-La Mancha, Toledo, Spain.,CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain.,CopenAge - Copenhagen Center for Clinical Age Research, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Frandsen
- Institute of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Tatyana Prokhorova
- Institute of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Rikke S Kamper
- CopenAge - Copenhagen Center for Clinical Age Research, University of Copenhagen, Copenhagen, Denmark.,Geriatric Research Unit, Department of Geriatric and Palliative Medicine, Bispebjerg-Frederiksberg University Hospital, Copenhagen, Denmark
| | - Bryan Haddock
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet University Hospital, Copenhagen, Denmark
| | - Per Aagaard
- Institute of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Charlotte Suetta
- CopenAge - Copenhagen Center for Clinical Age Research, University of Copenhagen, Copenhagen, Denmark.,Geriatric Research Unit, Department of Geriatric and Palliative Medicine, Bispebjerg-Frederiksberg University Hospital, Copenhagen, Denmark.,Geriatric Research Unit, Department of Internal Medicine, Herlev-Gentofte University Hospital, Copenhagen, Denmark
| |
Collapse
|
31
|
Gomez-Cabrero D, Walter S, Abugessaisa I, Miñambres-Herraiz R, Palomares LB, Butcher L, Erusalimsky JD, Garcia-Garcia FJ, Carnicero J, Hardman TC, Mischak H, Zürbig P, Hackl M, Grillari J, Fiorillo E, Cucca F, Cesari M, Carrie I, Colpo M, Bandinelli S, Feart C, Peres K, Dartigues JF, Helmer C, Viña J, Olaso G, García-Palmero I, Martínez JG, Jansen-Dürr P, Grune T, Weber D, Lippi G, Bonaguri C, Sinclair AJ, Tegner J, Rodriguez-Mañas L. A robust machine learning framework to identify signatures for frailty: a nested case-control study in four aging European cohorts. GeroScience 2021; 43:1317-1329. [PMID: 33599920 PMCID: PMC8190217 DOI: 10.1007/s11357-021-00334-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Phenotype-specific omic expression patterns in people with frailty could provide invaluable insight into the underlying multi-systemic pathological processes and targets for intervention. Classical approaches to frailty have not considered the potential for different frailty phenotypes. We characterized associations between frailty (with/without disability) and sets of omic factors (genomic, proteomic, and metabolomic) plus markers measured in routine geriatric care. This study was a prevalent case control using stored biospecimens (urine, whole blood, cells, plasma, and serum) from 1522 individuals (identified as robust (R), pre-frail (P), or frail (F)] from the Toledo Study of Healthy Aging (R=178/P=184/F=109), 3 City Bordeaux (111/269/100), Aging Multidisciplinary Investigation (157/79/54) and InCHIANTI (106/98/77) cohorts. The analysis included over 35,000 omic and routine laboratory variables from robust and frail or pre-frail (with/without disability) individuals using a machine learning framework. We identified three protective biomarkers, vitamin D3 (OR: 0.81 [95% CI: 0.68-0.98]), lutein zeaxanthin (OR: 0.82 [95% CI: 0.70-0.97]), and miRNA125b-5p (OR: 0.73, [95% CI: 0.56-0.97]) and one risk biomarker, cardiac troponin T (OR: 1.25 [95% CI: 1.23-1.27]). Excluding individuals with a disability, one protective biomarker was identified, miR125b-5p (OR: 0.85, [95% CI: 0.81-0.88]). Three risks of frailty biomarkers were detected: pro-BNP (OR: 1.47 [95% CI: 1.27-1.7]), cardiac troponin T (OR: 1.29 [95% CI: 1.21-1.38]), and sRAGE (OR: 1.26 [95% CI: 1.01-1.57]). Three key frailty biomarkers demonstrated a statistical association with frailty (oxidative stress, vitamin D, and cardiovascular system) with relationship patterns differing depending on the presence or absence of a disability.
Collapse
Affiliation(s)
- David Gomez-Cabrero
- Translational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Stefan Walter
- Dept. of Medicine and Public Health, Rey Juan Carlos University, Alcorcon, Spain
| | | | | | | | - Lee Butcher
- Department of Biomedical Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Jorge D Erusalimsky
- Department of Biomedical Sciences, Cardiff Metropolitan University, Cardiff, UK
| | | | - José Carnicero
- Dept. of Geriatric Medicine, Complejo Hospitalario Universitario de Toledo (CHUT), Toledo, Spain
| | | | - Harald Mischak
- Mosaiques Diagnostics GmbH, Rotenburger Str. 20, 30659, Hannover, Germany
| | - Petra Zürbig
- Mosaiques Diagnostics GmbH, Rotenburger Str. 20, 30659, Hannover, Germany
| | - Matthias Hackl
- Evercyte GmbH; BOKU-University of Natural Resources and Life Sciences Vienna, Department of Biotechnology, Ludwig Boltzmann Institute of Experimental and Clinical Traumatology, Vienna, Austria
| | - Johannes Grillari
- Evercyte GmbH; BOKU-University of Natural Resources and Life Sciences Vienna, Department of Biotechnology, Ludwig Boltzmann Institute of Experimental and Clinical Traumatology, Vienna, Austria
| | - Edoardo Fiorillo
- Instituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Francesco Cucca
- Instituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Matteo Cesari
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | | | | | - Catherine Feart
- Univ. Bordeaux, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - Karine Peres
- Univ. Bordeaux, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - Jean-François Dartigues
- Univ. Bordeaux, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - Catherine Helmer
- Univ. Bordeaux, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - José Viña
- Freshage, University of Valencia, Valencia, Spain
| | - Gloria Olaso
- Freshage, University of Valencia, Valencia, Spain
| | | | | | - Pidder Jansen-Dürr
- Research Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Tilman Grune
- German Institute for Human Nutrition, Potsdam, Germany
| | - Daniela Weber
- German Institute for Human Nutrition, Potsdam, Germany
| | - Giuseppe Lippi
- Clinical Biochemistry and Molecular Biology, Universita di Verona, Verona, Italy
| | - Chiara Bonaguri
- Laboratoy Medicine Technical Sciences, Parma University, Parma, Italy
| | | | - Jesper Tegner
- Dept. of Medicine, Karolinska Institute, Stockholm, Sweden
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Leocadio Rodriguez-Mañas
- CIBER of Frailty and Healthy Aging, Madrid, Spain.
- Dept. of Geriatric Medicine, Getafe University Hospital, Getafe, Spain.
| |
Collapse
|
32
|
Tavenier J, Rasmussen LJH, Andersen AL, Houlind MB, Langkilde A, Andersen O, Petersen J, Nehlin JO. Association of GDF15 With Inflammation and Physical Function During Aging and Recovery After Acute Hospitalization: A Longitudinal Study of Older Patients and Age-Matched Controls. J Gerontol A Biol Sci Med Sci 2021; 76:964-974. [DOI: 10.1093/gerona/glab011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Indexed: 12/20/2022] Open
Abstract
Abstract
Growth differentiation factor 15 (GDF15) is a stress-induced cytokine. Its plasma levels increase during aging and acute illness. In older Patients and age-matched Controls, we evaluated whether GDF15 levels (i) were associated with recovery after acute illness, and (ii) reflected different trajectories of aging and longitudinal changes in health measures. Fifty-two older Patients (≥65 years) were included upon admission to the emergency department (ED). At 30 days after discharge (time of matching), Patients were matched 1:1 on age and sex with Controls who had not been hospitalized within 2 years of inclusion. Both groups were followed up after 1 year. We assessed plasma levels of GDF15 and inflammatory biomarkers, frailty, nutritional status (mini nutritional assessment short-form), physical and cognitive function, and metabolic biomarkers. In Patients, elevated GDF15 levels at ED admission were associated with poorer resolution of inflammation (soluble urokinase plasminogen activator receptor [suPAR]), slowing of gait speed, and declining nutritional status between admission and 30-day follow-up. At time of matching, Patients were frailer and overall less healthy than age-matched Controls. GDF15 levels were significantly associated with participant group, on average Patients had almost 60% higher GDF15 than age-matched Controls, and this difference was partly mediated by reduced physical function. Increases in GDF15 levels between time of matching and 1-year follow-up were associated with increases in levels of interleukin-6 in Patients, and tumor necrosis factor-α and suPAR in age-matched Controls. In older adults, elevated GDF15 levels were associated with signs of accelerated aging and with poorer recovery after acute illness.
Collapse
Affiliation(s)
- Juliette Tavenier
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | | | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Anne Langkilde
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Denmark
- Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Janne Petersen
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Denmark
- Center for Clinical Research and Prevention, Copenhagen University Hospital Frederiksberg, Denmark
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Denmark
| | - Jan O Nehlin
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Denmark
| |
Collapse
|
33
|
Faria AVS, Andrade SS, Peppelenbosch MP, Ferreira-Halder CV, Fuhler GM. Platelets in aging and cancer-"double-edged sword". Cancer Metastasis Rev 2020; 39:1205-1221. [PMID: 32869161 PMCID: PMC7458881 DOI: 10.1007/s10555-020-09926-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
Platelets control hemostasis and play a key role in inflammation and immunity. However, platelet function may change during aging, and a role for these versatile cells in many age-related pathological processes is emerging. In addition to a well-known role in cardiovascular disease, platelet activity is now thought to contribute to cancer cell metastasis and tumor-associated venous thromboembolism (VTE) development. Worldwide, the great majority of all patients with cardiovascular disease and some with cancer receive anti-platelet therapy to reduce the risk of thrombosis. However, not only do thrombotic diseases remain a leading cause of morbidity and mortality, cancer, especially metastasis, is still the second cause of death worldwide. Understanding how platelets change during aging and how they may contribute to aging-related diseases such as cancer may contribute to steps taken along the road towards a "healthy aging" strategy. Here, we review the changes that occur in platelets during aging, and investigate how these versatile blood components contribute to cancer progression.
Collapse
Affiliation(s)
- Alessandra V S Faria
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA, Rotterdam, The Netherlands
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP, 13083-862, Brazil
| | | | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA, Rotterdam, The Netherlands
| | - Carmen V Ferreira-Halder
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP, 13083-862, Brazil
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|