1
|
Curatolo M, Chiu AP, Chia C, Ward A, Khan S, Johnston SK, Klein RM, Henze DA, Zhu W, Raftery D. Multi-omics profiles of chronic low back pain and fibromyalgia-Study protocol. PLoS One 2025; 20:e0312061. [PMID: 40238742 PMCID: PMC12002477 DOI: 10.1371/journal.pone.0312061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/31/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Chronic low back pain (CLBP) and fibromyalgia (FM) are leading causes of suffering, disability, and social costs. Current pharmacological treatments do not target molecular mechanisms driving CLBP and FM, and no validated biomarkers are available, hampering the development of effective therapeutics. Omics research has the potential to substantially advance our ability to develop mechanism-specific therapeutics by identifying pathways involved in the pathophysiology of CLBP and FM, and facilitate the development of diagnostic, predictive, and prognostic biomarkers. We will conduct a blood and urine multi-omics study in comprehensively phenotyped and clinically characterized patients with CLBP and FM. Our aims are to identify molecular pathways potentially involved in the pathophysiology of CLBP and FM that would shift the focus of research to the development of target-specific therapeutics, and identify candidate diagnostic, predictive, and prognostic biomarkers. METHODS We are conducting a prospective cohort study of adults ≥18 years of age with CLBP (n=100) and FM (n=100), and pain-free controls (n=200). Phenotyping measures include demographics, medication use, pain-related clinical characteristics, physical function, neuropathic components (quantitative sensory tests and DN4 questionnaire), pain facilitation (temporal summation), and psychosocial function as moderator. Blood and urine samples are collected to analyze metabolomics, lipidomics and proteomics. We will integrate the overall omics data to identify common mechanisms and pathways, and associate multi-omics profiles to pain-related clinical characteristics, physical function, indicators of neuropathic pain, and pain facilitation, with psychosocial variables as moderators. DISCUSSION Our study addresses the need for a better understanding of the molecular mechanisms underlying chronic low back pain and fibromyalgia. Using a multi-omics approach, we hope to identify converging evidence for potential targets of future therapeutic developments, as well as promising candidate biomarkers for further investigation by biomarker validation studies. We believe that accurate patient phenotyping will be essential for the discovery process, as both conditions are characterized by high heterogeneity and complexity, likely rendering molecular mechanisms phenotype specific.
Collapse
Affiliation(s)
- Michele Curatolo
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, United States of America
- CLEAR Center for Musculoskeletal Research, University of Washington, Seattle, Washington, United States of America
| | - Abby P. Chiu
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, United States of America
- CLEAR Center for Musculoskeletal Research, University of Washington, Seattle, Washington, United States of America
| | - Catherine Chia
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, United States of America
- CLEAR Center for Musculoskeletal Research, University of Washington, Seattle, Washington, United States of America
| | - Ava Ward
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, United States of America
| | - Savera Khan
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, United States of America
| | - Sandra K. Johnston
- CLEAR Center for Musculoskeletal Research, University of Washington, Seattle, Washington, United States of America
- Department of Radiology, University of Washington, Seattle, Washington, United States of America
| | - Rebecca M. Klein
- Department of Neuroscience, Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Darrell A. Henze
- Department of Neuroscience, Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Wentao Zhu
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, United States of America
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
2
|
Marino C, Imarisio A, Gasparri C, Napolitano E, Di Maio A, Avenali M, Buongarzone G, Galandra C, Picascia M, Grimaldi M, Errico F, Rondanelli M, D'Ursi AM, Valente EM, Usiello A. 1H-NMR-based metabolomics identifies disrupted betaine metabolism as distinct serum signature of pre-frailty. NPJ AGING 2025; 11:26. [PMID: 40216769 PMCID: PMC11992162 DOI: 10.1038/s41514-025-00218-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Increasing evidence suggests that frailty results from a complex age-associated metabolic decline. Here, we investigated the serum metabolomic profile of a well-characterized cohort of elderly subjects encompassing the whole fit-to-frail continuum. Enrichment analyses revealed a complex dysregulation of amino acids and energy metabolism in both pre-frail and frail participants. Remarkably, upregulated betaine levels emerged as a specific biochemical signature of pre-frail females, holding promise for the development of novel targeted interventions.
Collapse
Affiliation(s)
- Carmen Marino
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Alberto Imarisio
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Neurogenetics Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona "Istituto Santa Margherita", University of Pavia, Pavia, Italy
| | - Enza Napolitano
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Anna Di Maio
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy
| | - Micol Avenali
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Parkinson's Disease and Movement Disorders Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Gabriele Buongarzone
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Parkinson's Disease and Movement Disorders Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Caterina Galandra
- Neurogenetics Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Marta Picascia
- Parkinson's Disease and Movement Disorders Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Agricultural Sciences, University of Naples "Federico II", Portici, Italy
| | - Mariangela Rondanelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Anna Maria D'Ursi
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Enza Maria Valente
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Neurogenetics Research Centre, IRCCS Mondino Foundation, Pavia, Italy.
| | - Alessandro Usiello
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy.
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
| |
Collapse
|
3
|
Jasinska W, Birenzweig Y, Sharav Y, Aframian DJ, Rettman A, Hanut A, Brotman Y, Haviv Y. Salivary Metabolomics as a Diagnostic Tool: Distinct Metabolic Profiles Across Orofacial Pain Subtypes. Int J Mol Sci 2025; 26:2260. [PMID: 40076882 PMCID: PMC11900362 DOI: 10.3390/ijms26052260] [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: 01/28/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Orofacial pain (OFP) includes chronic pain conditions categorized into musculoskeletal (MS), neurovascular (NV), and neuropathic (NP) pain types, encompassing temporomandibular disorders (TMD), migraines, trigeminal neuralgia (TN), post-traumatic neuropathies, and burning mouth syndrome (BMS). These conditions significantly affect quality of life; yet, their underlying metabolic disruptions remain inadequately explored. Salivary metabolomics provides a non-invasive method to investigate biochemical alterations associated with OFP subtypes. This study aimed to identify pain-specific salivary metabolites across chronic OFP types and examine their correlations with clinical characteristics. Saliva samples from 63 OFP patients (TMD, migraines, TN, post-traumatic neuropathies, BMS) and 37 pain-free controls were analyzed using liquid chromatography-mass spectrometry (LC-MS) targeting 28 metabolites linked to pain. Statistical analyses determined significant metabolite changes and associations with pain subtypes and patient characteristics. Among the 28 analyzed metabolites, 18 showed significant differences between OFP patients and controls. Key amino acids, including DL-glutamic acid, DL-aspartic acid, DL-citrulline, spermidine, and DL-ornithine, were significantly elevated in MS, NV, and NP pain types compared to controls. Additionally, DL-glutamine, DL-valine, and DL-phenylalanine were distinctively elevated in TMD and migraine patients. BMS displayed fewer alterations, with significantly lower levels of DL-proline, DL-tryptophan, DL-glutamic acid, DL-asparagine, and DL-aspartic acid compared to other pain types but elevated spermidine levels relative to controls. Salivary metabolomics revealed distinct metabolic alterations in OFP subtypes, providing insights into potential biomarkers for diagnosis and monitoring. These findings offer a foundation for personalized approaches in OFP management, although further research is required to validate and expand these results.
Collapse
Affiliation(s)
- Weronika Jasinska
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Yonatan Birenzweig
- Department of Oral Medicine, Sedation and Imaging, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem 91120, Israel; (Y.B.); (D.J.A.); (A.R.); (A.H.); (Y.H.)
| | - Yair Sharav
- Department of Oral Medicine, Sedation and Imaging, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem 91120, Israel; (Y.B.); (D.J.A.); (A.R.); (A.H.); (Y.H.)
| | - Doron J. Aframian
- Department of Oral Medicine, Sedation and Imaging, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem 91120, Israel; (Y.B.); (D.J.A.); (A.R.); (A.H.); (Y.H.)
| | - Andra Rettman
- Department of Oral Medicine, Sedation and Imaging, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem 91120, Israel; (Y.B.); (D.J.A.); (A.R.); (A.H.); (Y.H.)
| | - Aiham Hanut
- Department of Oral Medicine, Sedation and Imaging, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem 91120, Israel; (Y.B.); (D.J.A.); (A.R.); (A.H.); (Y.H.)
| | - Yariv Brotman
- School of Plant Sciences and Food Security, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Yaron Haviv
- Department of Oral Medicine, Sedation and Imaging, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem 91120, Israel; (Y.B.); (D.J.A.); (A.R.); (A.H.); (Y.H.)
| |
Collapse
|
4
|
Yi L, Liao T, Yuan M, Chen Q, Xiong W, Zhu H. Single-cell metabolomics profiling of somatosensory neurons in various stages of neuropathic pain. J Biol Chem 2025; 301:108309. [PMID: 39955065 PMCID: PMC11938157 DOI: 10.1016/j.jbc.2025.108309] [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: 06/05/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
Metabolic alterations in the somatosensory cortex (S1) play a crucial role in neuropathic pain development, as evidenced by magnetic resonance spectroscopy and mass spectrometry analyses of brain homogenates. However, investigating metabolic changes in specific neuronal subtypes during neuropathic pain development remains challenging. Here, utilizing a recently developed technique called single-cell mass spectrometry (SCMS), we investigated metabolomic alterations within excitatory glutamatergic neurons located in the primary S1 during various stages of neuropathic pain. Specifically, we induced neuropathic pain in mice using a spared nerve injury (SNI) model and observed activation of glutamatergic neurons in layer II/III of S1 through c-Fos staining and electrophysiology. We profiled metabolic changes and performed pathway enrichment analysis in these neurons by single-cell mass spectrometry during both acute and subchronic phases of SNI. Further analyses revealed metabolites whose alterations significantly correlated with changes in pain thresholds, as well as distinct temporal patterns of metabolite expression during pain progression. From these analyses, we identified several key metabolites (homogentisic acid, phosphatidylcholine, phosphorylcholine, and rhein) and validated their causal roles in pain modulation via pharmacological interventions. Thus, our study provides a valuable resource for elucidating the neurometabolic regulatory mechanisms underlying neuropathic pain from a single-cell perspective.
Collapse
Affiliation(s)
- Lin Yi
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tiepeng Liao
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Man Yuan
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qi Chen
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Xiong
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China.
| | - Hongying Zhu
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China.
| |
Collapse
|
5
|
Mackey S, Aghaeepour N, Gaudilliere B, Kao MC, Kaptan M, Lannon E, Pfyffer D, Weber K. Innovations in acute and chronic pain biomarkers: enhancing diagnosis and personalized therapy. Reg Anesth Pain Med 2025; 50:110-120. [PMID: 39909549 PMCID: PMC11877092 DOI: 10.1136/rapm-2024-106030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/17/2024] [Indexed: 02/07/2025]
Abstract
Pain affects millions worldwide, posing significant challenges in diagnosis and treatment. Despite advances in understanding pain mechanisms, there remains a critical need for validated biomarkers to enhance diagnosis, prognostication, and personalized therapy. This review synthesizes recent advancements in identifying and validating acute and chronic pain biomarkers, including imaging, molecular, sensory, and neurophysiological approaches. We emphasize the emergence of composite, multimodal strategies that integrate psychosocial factors to improve the precision and applicability of biomarkers in chronic pain management. Neuroimaging techniques like MRI and positron emission tomography provide insights into structural and functional abnormalities related to pain, while electrophysiological methods like electroencepholography and magnetoencepholography assess dysfunctional processing in the pain neuroaxis. Molecular biomarkers, including cytokines, proteomics, and metabolites, offer diagnostic and prognostic potential, though extensive validation is needed. Integrating these biomarkers with psychosocial factors into clinical practice can revolutionize pain management by enabling personalized treatment strategies, improving patient outcomes, and potentially reducing healthcare costs. Future directions include the development of composite biomarker signatures, advances in artificial intelligence, and biomarker signature integration into clinical decision support systems. Rigorous validation and standardization efforts are also necessary to ensure these biomarkers are clinically useful. Large-scale collaborative research will be vital to driving progress in this field and implementing these biomarkers in clinical practice. This comprehensive review highlights the potential of biomarkers to transform acute and chronic pain management, offering hope for improved diagnosis, treatment personalization, and patient outcomes.
Collapse
Affiliation(s)
- Sean Mackey
- Division of Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Nima Aghaeepour
- Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, California, USA
| | - Brice Gaudilliere
- Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, California, USA
| | - Ming-Chih Kao
- Division of Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Merve Kaptan
- Division of Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Edward Lannon
- Division of Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Dario Pfyffer
- Division of Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Kenneth Weber
- Division of Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
6
|
Vennu V, Alsaad SM, Alenazi AM, Bindawas SM. Association Between Musculoskeletal Pain and Frailty Over Time in Older Adults. J Geriatr Phys Ther 2025; 48:33-41. [PMID: 38427805 DOI: 10.1519/jpt.0000000000000411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
BACKGROUND AND PURPOSE There is a high frequency of frailty in patients with musculoskeletal pain. Pain from osteoarthritis and lower back pain may be associated with frailty. However, the future risk of frailty among older adults with pain remains unclear. Thus, the primary objective of this study was to examine the association between musculoskeletal pain and the risk of becoming prefrail and frail in older adults. PARTICIPANTS AND METHODS A secondary analysis was performed using data from baseline and 1-, 2-, 3-, 4-, 6-, and 8-year follow-ups of the Osteoarthritis Initiative (OAI). The OAI recruited participants from 4 clinical sites in the United States, between February 2004 and May 2006. A self-reported questionnaire was used to determine the baseline musculoskeletal pain status in older adults (n = 1780) 65 years and older, including pain in the lower back, hip, knee, and at 2 or more sites. Using the Fried phenotypic criteria, participants were classified as nonfrail, prefrail, and frail at each period over 8 years. RESULTS After adjusting for age, sex, race, education, marital status, annual income, smoking status, comorbidities, and body mass index, binary logistic regression modeling using generalized estimating equations revealed that in older adults musculoskeletal pain in the lower back and at multiple sites was associated with a slightly but significantly decreased risk of prefrailty over time (adjusted odds ratio [AOR] = 0.98, 95% CI = 0.95-0.99, P = .019; AOR = 0.96, CI = 0.92-0.99, P = .032). The association between musculoskeletal pain and frailty among older adults was not statistically significant (all P > .05). CONCLUSIONS Musculoskeletal pain did not independently significantly increase the risk of prefrailty or frailty over time. It remains possible that when musculoskeletal is combined with other factors, the risk of prefrailty and frailty may be heightened. Further research into the combination of characteristics that best predict prefrailty and frailty, including but not limited to musculoskeletal pain, is warranted.
Collapse
Affiliation(s)
- Vishal Vennu
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Saad M Alsaad
- Department of Family and Community Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Aqeel M Alenazi
- Department of Health and Rehabilitation Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Saad M Bindawas
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Zhong R, Chen Y, Zhong L, Huang G, Liang W, Zou Y. The vicious cycle of frailty and pain: a two-sided causal relationship revealed. Front Med (Lausanne) 2024; 11:1396328. [PMID: 39314224 PMCID: PMC11416971 DOI: 10.3389/fmed.2024.1396328] [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: 03/05/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Background The decline in physiological functions in the older people is frequently accompanied with pain and frailty, yet the causal connection between frailty and pain remains uncertain. In this study, we utilized a two-sample Mendelian randomization (MR) approach to investigate the potential causal association between frailty and pain. Methods Two-sample bidirectional MR was conducted using summary data from genome-wide association studies to examine the potential causal relationship between frailty (defined by the frailty index and frailty phenotype) and pain. Summary genome wide association statistics were extracted from populations of European ancestry. We also investigated the causal relationship between frailty and site-specific pain, including joint pain, limb pain, thoracic spine pain and low back pain. Causal effects were estimated using the inverse variance weighting method. Sensitivity analyses were performed to validate the robustness of the results. Results Genetic predisposition to frailty was associated with an increased risk of pain (frailty phenotype odds ratio [OR]: 1.73; P = 3.54 × 10-6, frailty index OR: 1.36; P = 2.43 × 10-4). Meanwhile, individuals with a genetic inclination toward pain had a higher risk of developing frailty. Regarding site-specific pain, genetic prediction of the frailty phenotype increased the occurrence risk of joint pain, limb pain and low back pain. Reverse MR analysis further showed that limb pain and low back pain were associated with an increased risk of frailty occurrence. Conclusion This study presented evidence supporting a bidirectional causal relationship between frailty and pain. We highlighted the significance of addressing pain to prevent frailty and recommend the inclusion of pain assessment in the evaluation system for frailty.
Collapse
Affiliation(s)
- Ruipeng Zhong
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou, China
| | - Yijian Chen
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou, China
| | - Lanhua Zhong
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou, China
| | - Guiming Huang
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou, China
| | - Weidong Liang
- Anesthesia Surgery Center, the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yun Zou
- Anesthesia Surgery Center, the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
8
|
Curatolo M, Chiu AP, Chia C, Ward A, Johnston SK, Klein RM, Henze DA, Zhu W, Raftery D. Multi-Omics Profiles of Chronic Low Back Pain and Fibromyalgia - Study Protocol. RESEARCH SQUARE 2024:rs.3.rs-4669838. [PMID: 39149502 PMCID: PMC11326421 DOI: 10.21203/rs.3.rs-4669838/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Chronic low back pain (CLBP) and fibromyalgia (FM) are leading causes of suffering, disability, and social costs. Current pharmacological treatments do not target molecular mechanisms driving CLBP and FM, and no validated biomarkers are available, hampering the development of effective therapeutics. Omics research has the potential to substantially advance our ability to develop mechanism-specific therapeutics by identifying pathways involved in the pathophysiology of CLBP and FM, and facilitate the development of diagnostic, predictive, and prognostic biomarkers. We will conduct a blood and urine multi-omics study in comprehensively phenotyped and clinically characterized patients with CLBP and FM. Our aims are to identify molecular pathways potentially involved in the pathophysiology of CLBP and FM that would shift the focus of research to the development of target-specific therapeutics, and identify candidate diagnostic, predictive, and prognostic biomarkers. Methods We are conducting a prospective cohort study of adults ≥18 years of age with CLBP (n=100) and FM (n=100), and pain-free controls (n=200). Phenotyping measures include demographics, medication use, pain-related clinical characteristics, physical function, neuropathiccomponents (quantitative sensory tests and DN4 questionnaire), pain facilitation (temporal summation), and psychosocial function as moderator. Blood and urine samples are collected to analyze metabolomics, lipidomics and proteomics. We will integrate the overall omics data to identify common mechanisms and pathways, and associate multi-omics profiles to pain-related clinical characteristics, physical function, indicators of neuropathic pain, and pain facilitation, with psychosocial variables as moderators. Discussion Our study addresses the need for a better understanding of the molecular mechanisms underlying chronic low back pain and fibromyalgia. Using a multi-omics approach, we hope to identify converging evidence for potential targets of future therapeutic developments, as well as promising candidate biomarkers for further investigation by biomarker validation studies. We believe that accurate patient phenotyping will be essential for the discovery process, as both conditions are characterized by high heterogeneity and complexity, likely rendering molecular mechanisms phenotype specific.
Collapse
|
9
|
Mak JKL, Skovgaard AC, Nygaard M, Kananen L, Reynolds CA, Wang Y, Kuja‐Halkola R, Karlsson IK, Pedersen NL, Hägg S, Soerensen M, Jylhävä J. Epigenome-wide analysis of frailty: Results from two European twin cohorts. Aging Cell 2024; 23:e14135. [PMID: 38414347 PMCID: PMC11166364 DOI: 10.1111/acel.14135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/11/2024] [Accepted: 02/17/2024] [Indexed: 02/29/2024] Open
Abstract
Epigenetics plays an important role in the aging process, but it is unclear whether epigenetic factors also influence frailty, an age-related state of physiological decline. In this study, we performed a meta-analysis of epigenome-wide association studies in four samples drawn from the Swedish Adoption/Twin Study of Aging (SATSA) and the Longitudinal Study of Aging Danish Twins (LSADT) to explore the association between DNA methylation and frailty. Frailty was defined using the frailty index (FI), and DNA methylation levels were measured in whole blood using Illumina's Infinium HumanMethylation450K and MethylationEPIC arrays. In the meta-analysis consisting of a total of 829 participants, we identified 589 CpG sites that were statistically significantly associated with either the continuous or categorical FI (false discovery rate <0.05). Many of these CpGs have previously been associated with age and age-related diseases. The identified sites were also largely directionally consistent in a longitudinal analysis using mixed-effects models in SATSA, where the participants were followed up to a maximum of 20 years. Moreover, we identified three differentially methylated regions within the MGRN1, MIR596, and TAPBP genes that have been linked to neuronal aging, tumor growth, and immune functions. Furthermore, our meta-analysis results replicated 34 of the 77 previously reported frailty-associated CpGs at p < 0.05. In conclusion, our findings demonstrate robust associations between frailty and DNA methylation levels in 589 novel CpGs, previously unidentified for frailty, and strengthen the role of neuronal/brain pathways in frailty.
Collapse
Affiliation(s)
- Jonathan K. L. Mak
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Asmus Cosmos Skovgaard
- The Danish Twin Registry and Epidemiology, Biostatistics and Biodemography, Department of Public HealthUniversity of Southern DenmarkOdense MDenmark
| | - Marianne Nygaard
- The Danish Twin Registry and Epidemiology, Biostatistics and Biodemography, Department of Public HealthUniversity of Southern DenmarkOdense MDenmark
| | - Laura Kananen
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Faculty of Social Sciences (Health Sciences) and Gerontology Research Center (GEREC)University of TampereTampereFinland
| | - Chandra A. Reynolds
- Institute for Behavioral Genetics, University of ColoradoBoulderColoradoUSA
- Department of PsychologyUniversity of CaliforniaRiversideCaliforniaUSA
| | - Yunzhang Wang
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Department of Clinical Sciences, Danderyd HospitalKarolinska InstitutetStockholmSweden
| | - Ralf Kuja‐Halkola
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Ida K. Karlsson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Nancy L. Pedersen
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Sara Hägg
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Mette Soerensen
- The Danish Twin Registry and Epidemiology, Biostatistics and Biodemography, Department of Public HealthUniversity of Southern DenmarkOdense MDenmark
- Department of Clinical GeneticsOdense University HospitalOdense CDenmark
| | - Juulia Jylhävä
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Faculty of Social Sciences (Health Sciences) and Gerontology Research Center (GEREC)University of TampereTampereFinland
| |
Collapse
|
10
|
Flint JP, Welstead M, Cox SR, Russ TC, Marshall A, Luciano M. Validation of a polygenic risk score for frailty in the Lothian Birth Cohort 1936 and English longitudinal study of ageing. Sci Rep 2024; 14:12586. [PMID: 38822050 PMCID: PMC11143351 DOI: 10.1038/s41598-024-63229-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/24/2024] [Indexed: 06/02/2024] Open
Abstract
Frailty is a complex trait. Twin studies and high-powered Genome Wide Association Studies conducted in the UK Biobank have demonstrated a strong genetic basis of frailty. The present study utilized summary statistics from a Genome Wide Association Study on the Frailty Index to create and test the predictive power of frailty polygenic risk scores (PRS) in two independent samples - the Lothian Birth Cohort 1936 (LBC1936) and the English Longitudinal Study of Ageing (ELSA) aged 67-84 years. Multiple regression models were built to test the predictive power of frailty PRS at five time points. Frailty PRS significantly predicted frailty, measured via the FI, at all-time points in LBC1936 and ELSA, explaining 2.1% (β = 0.15, 95%CI, 0.085-0.21) and 1.8% (β = 0.14, 95%CI, 0.10-0.17) of the variance, respectively, at age ~ 68/ ~ 70 years (p < 0.001). This work demonstrates that frailty PRS can predict frailty in two independent cohorts, particularly at early ages (~ 68/ ~ 70). PRS have the potential to be valuable instruments for identifying those at risk for frailty and could be important for controlling for genetic confounders in epidemiological studies.
Collapse
Affiliation(s)
- J P Flint
- Advanced Care Research Centre, School of Engineering, College of Science and Engineering, The University of Edinburgh, Edinburgh, UK.
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK.
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK.
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK.
| | - M Welstead
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - S R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| | - T C Russ
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - A Marshall
- Advanced Care Research Centre, School of Engineering, College of Science and Engineering, The University of Edinburgh, Edinburgh, UK
- School of Social and Political Science, University of Edinburgh, Edinburgh, UK
| | - M Luciano
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
11
|
Shekarchian A, Bandarian F, Hadizadeh A, Amirsardari Z, Sharifi Y, Ayati A, Varmaghani M, Shandiz AF, Sharifi F, Ghadery AH, Tayanloo A, Yavari T, Larijani B, Payab M, Ebrahimpur M. Exploring the metabolomics profile of frailty- a systematic review. J Diabetes Metab Disord 2024; 23:289-303. [PMID: 38932837 PMCID: PMC11196473 DOI: 10.1007/s40200-023-01379-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 12/19/2023] [Indexed: 06/28/2024]
Abstract
Background Frailty is a multifaceted geriatric syndrome characterized by an increased vulnerability to stressful events. metabolomics studies are valuable tool for better understanding the underlying mechanisms of pathologic conditions. This review aimed to elucidate the metabolomics profile of frailty. Method This systematic review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) 2020 statement. A comprehensive search was conducted across multiple databases. Initially, 5027 results were retrieved, and after removing duplicates, 1838 unique studies were subjected to screening. Subsequently, 248 studies underwent full-text screening, with 21 studies ultimately included in the analysis. Data extraction was performed meticulously by two authors, and the quality of the selected studies was assessed using the Critical Appraisal Skills Program (CASP) checklist. Results The findings revealed that certain Branched-chain amino acids (BCAAs) levels were lower in frail subjects compared to robust subjects, while levels of glutamate and glutamine were higher in frail individuals. Moreover, sphingomyelins and phosphatidylcholines (PC) displayed a decreasing trend as frailty advanced. Additionally, other metabolic derivatives, such as carnitine, exhibited significant associations with frailty. These metabolites were primarily interconnected through biochemical pathways related to the tricarboxylic acid and urea cycles. Notably, frailty was associated with a decrease in metabolic derivatives, including carnitine. Conclusion This study underscores the intricate relationship between essential metabolites, including amino acids and lipids, and their varying levels in frail individuals compared to their robust counterparts. It provides a comprehensive panel of metabolites, shedding light on their potential associations with frailty and expanding our understanding of this complex syndrome.
Collapse
Affiliation(s)
- Ahmadreza Shekarchian
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bandarian
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Hadizadeh
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Amirsardari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Sharifi
- Department of Radiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran university of medical sciences, Tehran, Iran
| | - Aryan Ayati
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Varmaghani
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Farshad Sharifi
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdolkarim Haji Ghadery
- Department of Radiology, Advanced Diagnostic, and Interventional Radiology Research Center (ADIR), Tehran, Iran
| | - Akram Tayanloo
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahereh Yavari
- Department of Internal Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran university of medical sciences, Tehran, Iran
| | - Moloud Payab
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- EMRI (Endocrinology and Metabolism Research Institute), First Floor, No 10, Jalal-Al-Ahmad Street, North Kargar Avenue, Tehran, 14117-13137 Iran
| | - Mahbube Ebrahimpur
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiology, Advanced Diagnostic, and Interventional Radiology Research Center (ADIR), Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- EMRI (Endocrinology and Metabolism Research Institute), First Floor, No 10, Jalal-Al-Ahmad Street, North Kargar Avenue, Tehran, 14117-13137 Iran
| |
Collapse
|
12
|
Mishra M, Wu J, Kane AE, Howlett SE. The intersection of frailty and metabolism. Cell Metab 2024; 36:893-911. [PMID: 38614092 PMCID: PMC11123589 DOI: 10.1016/j.cmet.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/15/2024]
Abstract
On average, aging is associated with unfavorable changes in cellular metabolism, which are the processes involved in the storage and expenditure of energy. However, metabolic dysregulation may not occur to the same extent in all older individuals as people age at different rates. Those who are aging rapidly are at increased risk of adverse health outcomes and are said to be "frail." Here, we explore the links between frailty and metabolism, including metabolic contributors and consequences of frailty. We examine how metabolic diseases may modify the degree of frailty in old age and suggest that frailty may predispose toward metabolic disease. Metabolic interventions that can mitigate the degree of frailty in people are reviewed. New treatment strategies developed in animal models that are poised for translation to humans are also considered. We suggest that maintaining a youthful metabolism into older age may be protective against frailty.
Collapse
Affiliation(s)
- Manish Mishra
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Judy Wu
- Institute for Systems Biology, Seattle, WA, USA
| | - Alice E Kane
- Institute for Systems Biology, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada; Department of Medicine (Geriatric Medicine), Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
13
|
Kondoh H, Kameda M. Metabolites in aging and aging-relevant diseases: Frailty, sarcopenia and cognitive decline. Geriatr Gerontol Int 2024; 24 Suppl 1:44-48. [PMID: 37837183 PMCID: PMC11503595 DOI: 10.1111/ggi.14684] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 10/15/2023]
Abstract
Aging shows biologically complex features with high individual variability, which reflects the exposure to several stimuli and the adaptation to them. Among them, metabolic changes are well observed as consequences or possible causes of aging. Calorie restriction extends organismal life span in experimental models. Several metabolites; for example, resveratrol or nicotinamide mononucleotide, are reported to mimic calorie restriction effects in vivo. Metabolomic research would be useful to evaluate metabolites as biomarkers in aging-relevant events and to identify metabolic regulation of aging. We recently developed the metabolomic approach for whole blood analysis, which functions as strong tool for this purpose. We review the update findings in aging-relevant metabolites detected by this method. Geriatr Gerontol Int 2024; 24: 44-48.
Collapse
Affiliation(s)
- Hiroshi Kondoh
- Geriatric Unit, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Masahiro Kameda
- Geriatric Unit, Graduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
14
|
Shen JX, Lu Y, Meng W, Yu L, Wang JK. Exploring causality between bone mineral density and frailty: A bidirectional Mendelian randomization study. PLoS One 2024; 19:e0296867. [PMID: 38271334 PMCID: PMC10810463 DOI: 10.1371/journal.pone.0296867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/23/2023] [Indexed: 01/27/2024] Open
Abstract
OBJECTIVE The bidirectional correlation between low bone mineral density (BMD) and frailty, despite its extensive documentation, still lacks a conclusive understanding. The objective of this Mendelian randomization (MR) study is to investigate the bidirectional causal relationship between BMD and frailty. METHODS We utilized summary statistics data for BMD at different skeletal sites-including heel BMD (e-BMD, N = 40,613), forearm BMD (FA-BMD, N = 8,143), femoral neck BMD (FN-BMD, N = 32,735), and lumbar spine BMD (LS-BMD, N = 28,489), alongside frailty index (FI, N = 175,226) data in participants of European ancestry. MR analysis in our study was conducted using well-established analytical methods, including inverse variance weighted (IVW), weighted median (WM), and MR-Egger approaches. RESULTS We observed negative causal estimates between genetically predicted e-BMD (IVW β = - 0.020, 95% confidence interval (CI) = - 0.038, - 0.002, P = 0.029) and FA-BMD (IVW β = -0.035, 95% CI = -0.066, -0.004, P = 0.028) with FI. However, the results did not reach statistical significance after applying the Bonferroni correction, with a significance threshold set at P < 0.0125 (0.05/4). There was no causal effect of FN-BMD (IVW β = - 0.024, 95% CI = -0.052, 0.004, P = 0.088) and LS-BMD (IVW β = - 0.005, 95% CI = -0.034, 0.024, P = 0.749) on FI. In the reverse Mendelian randomization (MR) analysis, we observed no causal effect of FI on BMD at various skeletal sites. CONCLUSION Our study provides support for the hypothesis that low BMD may be a potential causal risk factor for frailty, but further research is needed to confirm this relationship. However, our findings did not confirm reverse causality.
Collapse
Affiliation(s)
- Jue-xin Shen
- Department of Orthopedics, Chongming Branch, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Lu
- Department of Orthopedics, Chongming Branch, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Meng
- Department of Orthopedics, Chongming Branch, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lei Yu
- Department of Orthopedics, Chongming Branch, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun-kai Wang
- Department of Orthopedics, Chongming Branch, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Borim FSA, Assumpção DD, Yassuda MS, Costa HTDM, Batistoni SST, Neri AL, Voshaar RCO, Aprahamian I. Relationship between chronic pain, depressive symptoms, and functional disability in community-dwelling older adults: mediating role of frailty. EINSTEIN-SAO PAULO 2023; 21:eAO0284. [PMID: 38126546 DOI: 10.31744/einstein_journal/2023ao0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 06/12/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Borim et al. showed that older adults with chronic pain exhibited more depressive symptoms and frailty components. Depressive symptoms were associated with more frailty components, and those with more depressive symptoms and frailty faced greater limitations in IADL performance. Frailty appears to mediate the pathway from chronic pain to functional impairment Chronic pain is directly associated with depressive symptoms and frailty. Chronic pain is not directly associated with functional disability. Depression and frailty are both directly associated with functional disabilities. Frailty mediates the association between chronic pain and functional disability. Depression; Disability evaluation; Frailty; Frail elderly. OBJECTIVE To evaluate the direct and indirect effects of chronic pain, depressive symptoms, frailty components, and functional disability through a pathway analysis approach in a sample of community-dwelling older adults. METHODS Data of 419 participants were cross-sectionally evaluated for the presence of depressive symptoms (Geriatric Depression Scale [15 items]), physical frailty components (phenotype criteria), chronic pain, and limitations in performing instrumental activities of daily living (functional disability scale by Lawton and Brody). Structural equation modeling via path analysis was used to explore the direct and indirect effects among these four variables. Statistical significance was set at p<0.05. RESULTS Of the total participants, 69.8% were women and 59.3% had low education (1-4 years); the mean age was 80.3±4.6 years. Chronic pain and depressive symptoms were directly related and were associated to frailty. The number of frailty components and depressive symptoms were directly associated with functional disability. Frailty had an indirect effect on the association between chronic pain, depressive symptoms, and functional disabilities. CONCLUSION The pathway from chronic pain and depressive symptoms to functional disability is potentially mediated by the number of frailty components.
Collapse
Affiliation(s)
- Flávia Silva Arbex Borim
- Programa de pós-graduação em Gerontologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Daniela de Assumpção
- Programa de pós-graduação em Gerontologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Mônica Sanches Yassuda
- Programa de pós-graduação em Gerontologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Henrique Trajano de Moraes Costa
- Group of Investigation on Multimorbidity and Mental Health in Aging, Department of Internal Medicine, Faculdade de Medicina de Jundiaí, São Paulo, SP, Brazil
| | - Samila Sathler Tavares Batistoni
- Programa de pós-graduação em Gerontologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Anita Liberalesso Neri
- Programa de pós-graduação em Gerontologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Richard C Oude Voshaar
- Department of Psychiatry, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, Groningen, The Netherlands
| | - Ivan Aprahamian
- Group of Investigation on Multimorbidity and Mental Health in Aging, Department of Internal Medicine, Faculdade de Medicina de Jundiaí, São Paulo, SP, Brazil
- Department of Psychiatry, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
16
|
Flint JP, Welstead M, Cox SR, Russ TC, Marshall A, Luciano M. Validation of a polygenic risk score for Frailty in the Lothian Birth Cohort and English Longitudinal Study of Ageing. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.03.23288064. [PMID: 37066324 PMCID: PMC10104224 DOI: 10.1101/2023.04.03.23288064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Frailty is a complex trait. Twin studies and a high-powered Genome Wide Association Study (GWAS) conducted in the UK Biobank have demonstrated a strong genetic basis of frailty. The present study utilized summary statistics from this GWAS to create and test the predictive power of frailty polygenic risk scores (PRS) in two independent samples - the Lothian Birth Cohort 1936 (LBC1936) and the English Longitudinal Study of Ageing (ELSA) aged 67-84 years. Multiple regression models were built to test the predictive power of frailty PRS at five time points. Frailty PRS significantly predicted frailty at all-time points in LBC1936 and ELSA, explaining 2.1% (β = 0.15, 95%CI, 0.085-0.21) and 1.6% (β = 0.14, 95%CI, 0.10-0.17) of the variance, respectively, at age ~68/~70 years (p < 0.001). This work demonstrates that frailty PRS can predict frailty in two independent cohorts, particularly at early ages (~68/~70). PRS have the potential to be valuable instruments for identifying those at risk for frailty and could be important for controlling for genetic confounders in epidemiological studies.
Collapse
Affiliation(s)
- J P Flint
- Advanced Care Research Centre School of Engineering, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - M Welstead
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - S R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| | - T C Russ
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - A Marshall
- Advanced Care Research Centre School of Engineering, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- School of Social and Political Science, University of Edinburgh, Edinburgh, UK
| | - M Luciano
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
17
|
Nath T, Caffo B, Wager T, Lindquist MA. A machine learning based approach towards high-dimensional mediation analysis. Neuroimage 2023; 268:119843. [PMID: 36586543 PMCID: PMC10332048 DOI: 10.1016/j.neuroimage.2022.119843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/02/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022] Open
Abstract
Mediation analysis is used to investigate the role of intermediate variables (mediators) that lie in the path between an exposure and an outcome variable. While significant research has focused on developing methods for assessing the influence of mediators on the exposure-outcome relationship, current approaches do not easily extend to settings where the mediator is high-dimensional. These situations are becoming increasingly common with the rapid increase of new applications measuring massive numbers of variables, including brain imaging, genomics, and metabolomics. In this work, we introduce a novel machine learning based method for identifying high dimensional mediators. The proposed algorithm iterates between using a machine learning model to map the high-dimensional mediators onto a lower-dimensional space, and using the predicted values as input in a standard three-variable mediation model. Hence, the machine learning model is trained to maximize the likelihood of the mediation model. Importantly, the proposed algorithm is agnostic to the machine learning model that is used, providing significant flexibility in the types of situations where it can be used. We illustrate the proposed methodology using data from two functional Magnetic Resonance Imaging (fMRI) studies. First, using data from a task-based fMRI study of thermal pain, we combine the proposed algorithm with a deep learning model to detect distributed, network-level brain patterns mediating the relationship between stimulus intensity (temperature) and reported pain at the single trial level. Second, using resting-state fMRI data from the Human Connectome Project, we combine the proposed algorithm with a connectome-based predictive modeling approach to determine brain functional connectivity measures that mediate the relationship between fluid intelligence and working memory accuracy. In both cases, our multivariate mediation model links exposure variables (thermal pain or fluid intelligence), high dimensional brain measures (single-trial brain activation maps or resting-state brain connectivity) and behavioral outcomes (pain report or working memory accuracy) into a single unified model. Using the proposed approach, we are able to identify brain-based measures that simultaneously encode the exposure variable and correlate with the behavioral outcome.
Collapse
Affiliation(s)
- Tanmay Nath
- The Department of Biostatistics, Johns Hopkins University, Baltimore, MD, USA.
| | - Brian Caffo
- The Department of Biostatistics, Johns Hopkins University, Baltimore, MD, USA
| | - Tor Wager
- The Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Martin A Lindquist
- The Department of Biostatistics, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
18
|
Mak JKL, Kuja-Halkola R, Bai G, Hassing LB, Pedersen NL, Hägg S, Jylhävä J, Reynolds CA. Genetic and Environmental Influences on Longitudinal Frailty Trajectories From Adulthood into Old Age. J Gerontol A Biol Sci Med Sci 2023; 78:333-341. [PMID: 36124734 PMCID: PMC9951061 DOI: 10.1093/gerona/glac197] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Frailty is a complex, dynamic geriatric condition, but limited evidence has shown how genes and environment may contribute to its longitudinal changes. We sought to investigate sources of individual differences in the longitudinal trajectories of frailty, considering potential selection bias when including a sample of oldest-old twins. METHODS Data were from 2 Swedish twin cohort studies: a younger cohort comprising 1 842 adults aged 29-96 years followed up to 15 waves, and an older cohort comprising 654 adults aged ≥79 years followed up to 5 waves. Frailty was measured using the frailty index (FI). Age-based latent growth curve models were used to examine longitudinal trajectories, and extended to a biometric analysis to decompose variability into genetic and environmental etiologies. RESULTS A bilinear model with an inflection point at age 75 best described the data, indicating a fourfold to fivefold faster FI increase after 75 years. Twins from the older cohort had significantly higher mean FI at baseline but slower rate of increase afterward. FI level at age 75 was moderately heritable in both men (42%) and women (55%). Genetic influences were relatively stable across age for men and increasing for women, although the most salient amplification in FI variability after age 75 was due to individual-specific environmental influences for both men and women; conclusions were largely consistent when excluding the older cohort. CONCLUSION Increased heterogeneity of frailty in late life is mainly attributable to environmental influences, highlighting the importance of targeting environmental risk factors to mitigate frailty in older adults.
Collapse
Affiliation(s)
- Jonathan K L Mak
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ralf Kuja-Halkola
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ge Bai
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Linda B Hassing
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden
- Centre for Ageing and Health, University of Gothenburg, Gothenburg, Sweden
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Juulia Jylhävä
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Faculty of Social Sciences (Health Sciences) and Gerontology Research Center (GEREC), University of Tampere, Tampere, Finland
| | - Chandra A Reynolds
- Department of Psychology, University of California, Riverside, California, USA
| |
Collapse
|
19
|
Than KD, Mehta VA, Le V, Moss JR, Park P, Uribe JS, Eastlack RK, Chou D, Fu KM, Wang MY, Anand N, Passias PG, Shaffrey CI, Okonkwo DO, Kanter AS, Nunley P, Mundis GM, Fessler RG, Mummaneni PV. Role of obesity in less radiographic correction and worse health-related quality-of-life outcomes following minimally invasive deformity surgery. J Neurosurg Spine 2022; 37:222-231. [PMID: 35180705 DOI: 10.3171/2021.12.spine21703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/09/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Minimally invasive surgery (MIS) for adult spinal deformity (ASD) can offer deformity correction with less tissue manipulation and damage. However, the impact of obesity on clinical outcomes and radiographic correction following MIS for ASD is poorly understood. The goal of this study was to determine the role, if any, that obesity has on radiographic correction and health-related quality-of-life measures in MIS for ASD. METHODS Data were collected from a multicenter database of MIS for ASD. This was a retrospective review of a prospectively collected database. Patient inclusion criteria were age ≥ 18 years and coronal Cobb angle ≥ 20°, pelvic incidence-lumbar lordosis mismatch ≥ 10°, or sagittal vertical axis (SVA) > 5 cm. A group of patients with body mass index (BMI) < 30 kg/m2 was the control cohort; BMI ≥ 30 kg/m2 was used to define obesity. Obesity cohorts were categorized into BMI 30-34.99 and BMI ≥ 35. All patients had at least 1 year of follow-up. Preoperative and postoperative health-related quality-of-life measures and radiographic parameters, as well as complications, were compared via statistical analysis. RESULTS A total of 106 patients were available for analysis (69 control, 17 in the BMI 30-34.99 group, and 20 in the BMI ≥ 35 group). The average BMI was 25.24 kg/m2 for the control group versus 32.46 kg/m2 (p < 0.001) and 39.5 kg/m2 (p < 0.001) for the obese groups. Preoperatively, the BMI 30-34.99 group had significantly more prior spine surgery (70.6% vs 42%, p = 0.04) and worse preoperative numeric rating scale leg scores (7.71 vs 5.08, p = 0.001). Postoperatively, the BMI 30-34.99 cohort had worse Oswestry Disability Index scores (33.86 vs 23.55, p = 0.028), greater improvement in numeric rating scale leg scores (-4.88 vs -2.71, p = 0.012), and worse SVA (51.34 vs 26.98, p = 0.042) at 1 year postoperatively. Preoperatively, the BMI ≥ 35 cohort had significantly worse frailty (4.5 vs 3.27, p = 0.001), Oswestry Disability Index scores (52.9 vs 44.83, p = 0.017), and T1 pelvic angle (26.82 vs 20.71, p = 0.038). Postoperatively, after controlling for differences in frailty, the BMI ≥ 35 cohort had significantly less improvement in their Scoliosis Research Society-22 outcomes questionnaire scores (0.603 vs 1.05, p = 0.025), higher SVA (64.71 vs 25.33, p = 0.015) and T1 pelvic angle (22.76 vs 15.48, p = 0.029), and less change in maximum Cobb angle (-3.93 vs -10.71, p = 0.034) at 1 year. The BMI 30-34.99 cohort had significantly more infections (11.8% vs 0%, p = 0.004). The BMI ≥ 35 cohort had significantly more implant complications (30% vs 11.8%, p = 0.014) and revision surgery within 90 days (5% vs 1.4%, p = 0.034). CONCLUSIONS Obese patients who undergo MIS for ASD have less correction of their deformity, worse quality-of-life outcomes, more implant complications and infections, and an increased rate of revision surgery compared with their nonobese counterparts, although both groups benefit from surgery. Appropriate counseling should be provided to obese patients.
Collapse
Affiliation(s)
- Khoi D Than
- 1Departments of Neurosurgery and Orthopedics, Division of Spine, Duke University Medical Center, Durham, North Carolina
| | - Vikram A Mehta
- 1Departments of Neurosurgery and Orthopedics, Division of Spine, Duke University Medical Center, Durham, North Carolina
| | - Vivian Le
- 2Department of Neurosurgery, University of California, San Francisco, California
| | - Jonah R Moss
- 12Department of Neurological Surgery, Rush University Medical Center, Chicago, Illinois
| | - Paul Park
- 3Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Juan S Uribe
- 4Barrow Neurological Institute, Phoenix, Arizona
| | - Robert K Eastlack
- 5Department of Orthopedic Surgery, Scripps Clinic, La Jolla, California
| | - Dean Chou
- 2Department of Neurosurgery, University of California, San Francisco, California
| | - Kai-Ming Fu
- 6Department of Neurosurgery, Cornell Medical Center, New York, New York
| | - Michael Y Wang
- 7Department of Neurosurgery, University of Miami, Florida
| | - Neel Anand
- 8Department of Orthopaedics, Cedars-Sinai Medical Center, Los Angeles, California
| | - Peter G Passias
- 9Department of Orthopedics, New York University Langone Health, New York, New York
| | - Christopher I Shaffrey
- 1Departments of Neurosurgery and Orthopedics, Division of Spine, Duke University Medical Center, Durham, North Carolina
| | - David O Okonkwo
- 10Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Adam S Kanter
- 10Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Pierce Nunley
- 11Spine Institute of Louisiana, Shreveport, Louisiana; and
| | - Gregory M Mundis
- 5Department of Orthopedic Surgery, Scripps Clinic, La Jolla, California
| | - Richard G Fessler
- 12Department of Neurological Surgery, Rush University Medical Center, Chicago, Illinois
| | - Praveen V Mummaneni
- 2Department of Neurosurgery, University of California, San Francisco, California
| |
Collapse
|
20
|
Blodgett JM, Pérez-Zepeda MU, Godin J, Kehler DS, Andrew MK, Kirkland S, Rockwood K, Theou O. Frailty indices based on self-report, blood-based biomarkers and examination-based data in the Canadian Longitudinal Study on Aging. Age Ageing 2022; 51:6581611. [PMID: 35524747 PMCID: PMC9078045 DOI: 10.1093/ageing/afac075] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Frailty can be operationalised using the deficit accumulation approach, which considers health deficits across multiple domains. We aimed to develop, validate and compare three different frailty indices (FI) constructed from self-reported health measures (FI-Self Report), blood-based biomarkers (FI-Blood) and examination-based assessments (FI-Examination). METHODS Up to 30,027 participants aged 45-85 years from the baseline (2011-2015) comprehensive cohort of the Canadian Longitudinal Study on Aging were included in the analyses. Following standard criteria, three FIs were created: a 48-item FI-Self Report, a 23-item FI-Blood and a 47-item FI-Examination. In addition a 118-item FI-Combined was constructed. Mortality status was ascertained in July 2019. RESULTS FI-Blood and FI-Examination demonstrated broader distributions than FI-Self Report. FI-Self Report and FI-Blood scores were higher in females, whereas FI-Examination scores were higher in males. All FI scores increased nonlinearly with age and were highest at lower education levels. In sex and age-adjusted models, a 0.01 increase in FI score was associated with a 1.08 [95% confidence interval (CI): 1.07,1.10], 1.05 (1.04,1.06), 1.07 (1.05,1.08) and a 1.13 (1.11,1.16) increased odds of mortality for FI-Self Report, FI-Blood, FI-Examination and FI-Combined, respectively. Inclusion of the three distinct FI types in a single model yielded the best prognostic accuracy and model fit, even compared to the FI-Combined, with all FIs remaining independently associated with mortality. CONCLUSION Characteristics of all FIs were largely consistent with previously established FIs. To adequately capture frailty levels and to improve our understanding of the heterogeneity of ageing, FIs should consider multiple types of deficits including self-reported, blood and examination-based measures.
Collapse
Affiliation(s)
- Joanna M Blodgett
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mario U Pérez-Zepeda
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada,Instituto Nacional de Geriatria, Mexico City, Mexico,Centro de Investigacion en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac Mexico Campus Norte, Huixquilucan Mexico
| | - Judith Godin
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - D Scott Kehler
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada,School of Physiotherapy, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Melissa K Andrew
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Susan Kirkland
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada,Department of Community Health and Epidemiology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kenneth Rockwood
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Olga Theou
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada,School of Physiotherapy, Dalhousie University, Halifax, Nova Scotia, Canada,Address correspondence to: Olga Theou, School of Physiotherapy and Department of Medicine, Dalhousie University and Nova Scotia Health, Halifax, Nova Scotia, Canada. Tel: 902-473-4846; Fax: 902-473-1050.
| |
Collapse
|
21
|
Abstract
Frailty is a complex syndrome affecting a growing sector of the global population as medical developments have advanced human mortality rates across the world. Our current understanding of frailty is derived from studies conducted in the laboratory as well as the clinic, which have generated largely phenotypic information. Far fewer studies have uncovered biological underpinnings driving the onset and progression of frailty, but the stage is set to advance the field with preclinical and clinical assessment tools, multiomics approaches together with physiological and biochemical methodologies. In this article, we provide comprehensive coverage of topics regarding frailty assessment, preclinical models, interventions, and challenges as well as clinical frameworks and prevalence. We also identify central biological mechanisms that may be at play including mitochondrial dysfunction, epigenetic alterations, and oxidative stress that in turn, affect metabolism, stress responses, and endocrine and neuromuscular systems. We review the role of metabolic syndrome, insulin resistance and visceral obesity, focusing on glucose homeostasis, adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), and nicotinamide adenine dinucleotide (NAD+ ) as critical players influencing the age-related loss of health. We further focus on how immunometabolic dysfunction associates with oxidative stress in promoting sarcopenia, a key contributor to slowness, weakness, and fatigue. We explore the biological mechanisms involved in stem cell exhaustion that affect regeneration and may contribute to the frailty-associated decline in resilience and adaptation to stress. Together, an overview of the interplay of aging biology with genetic, lifestyle, and environmental factors that contribute to frailty, as well as potential therapeutic targets to lower risk and slow the progression of ongoing disease is covered. © 2022 American Physiological Society. Compr Physiol 12:1-46, 2022.
Collapse
Affiliation(s)
- Laís R. Perazza
- Department of Physical Therapy and Athletic Training, Boston University, Boston, Massachusetts, USA
| | - Holly M. Brown-Borg
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - LaDora V. Thompson
- Department of Physical Therapy and Athletic Training, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
22
|
Pan Y, Li Y, Liu P, Zhang Y, Li B, Liu Z, Shui G, Ma L. Metabolomics-Based Frailty Biomarkers in Older Chinese Adults. Front Med (Lausanne) 2022; 8:830723. [PMID: 35155487 PMCID: PMC8825494 DOI: 10.3389/fmed.2021.830723] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 11/14/2022] Open
Abstract
Background/Objectives Owing to accelerated population aging, health in older adults is becoming increasingly important. Frailty can reflect the health status and disease risks of older adults; however, appropriate biomarkers for early screening of frailty have not been identified. Here, we applied metabolomics to identify frailty biomarkers and potential pathogenic mechanisms of frailty. Methods Serum metabolic profiles from 25 frail and 49 non-frail (control) older adults were systematically investigated by liquid chromatography-mass spectrometry-based metabolomics. Results We identified 349 metabolites of 46 classes, with four increased and seven decreased metabolites in frail older adults. Pearson correlation analysis identified 11 and 21 metabolites that were positively and negatively correlated with grip strength, and 7 and 76 metabolites that were positively and negatively correlated with gait speed, respectively. Pathway analysis identified 10 metabolite sets and 13 pathways significantly associated with one or more frailty phenotype criteria. Conclusion These results revealed the metabolite characteristics of serum in frail older adults. Intermediates of carbohydrate metabolism (e.g., isocitrate, malate, fumarate, cis-aconitate, glucuronate, and pyruvate), saturated fatty acids (e.g., palmitic acid), unsaturated fatty acids (e.g., arachidonate and linoleic acid), and certain essential amino acids (e.g., tryptophan) may be candidate biomarkers for the early diagnosis of frailty. Mitochondrial function disorders, saturated fatty acid-mediated lipotoxicity, aberrant unsaturated fatty acid metabolism, and increased tryptophan degradation could be potential mechanisms of frailty.
Collapse
Affiliation(s)
- Yiming Pan
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yun Li
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Pan Liu
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yaxin Zhang
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Bowen Li
- LipidALL Technologies Company Limited, Changzhou, China
| | - Zuyun Liu
- Center for Clinical Big Data and Analytics, School of Public Health, Second Affiliated Hospital and Department of Big Data in Health Science, Zhejiang University School of Medicine, Hangzhou, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Meng L, Shi H, Wang DG, Shi J, Wu WB, Dang YM, Fan GQ, Shen J, Yu PL, Dong J, Yang RY, Xi H. Specific Metabolites Involved in Antioxidation and Mitochondrial Function Are Correlated With Frailty in Elderly Men. Front Med (Lausanne) 2022; 9:816045. [PMID: 35155500 PMCID: PMC8833032 DOI: 10.3389/fmed.2022.816045] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/04/2022] [Indexed: 01/14/2023] Open
Abstract
BackgroundAs an age-related syndrome, frailty may play a central role in poor health among older adults. Sarcopenia overlaps with the physical domain of frailty, and most existing studies have analyzed the associated factors of frailty and sarcopenia as an isolated state. Perturbations in metabolism may play an important role in the presence of frailty or sarcopenia; however, the metabolites associated with frailty, especially overlapping with sarcopenia remain unclear. In this study, we aimed to explore whether amino acids, carnitines, acylcarnitines and lysophosphatidylcholines, as specific panels, are significantly correlated with frailty, especially overlapping with sarcopenia, to gain insight into potential biomarkers and possible biological mechanisms and to facilitate their management.MethodsWe applied a targeted high-performance liquid chromatography-tandem mass spectrometry approach in serum samples from 246 Chinese older men (age 79.2 ± 7.8 years) with frailty (n = 150), non-frailty (n = 96), frailty and sarcopenia (n = 52), non-frail and non-sarcopenic control (n = 85). Frailty was evaluated using Freid phenotype criteria, sarcopenia was defined by diagnostic algorithm of Asian Working Group on Sarcopenia, and the participants were diagnosed as frailty and sarcopenia when they met the evaluation criteria of both frailty and sarcopenia. A panel of 29 metabolomic profiles was assayed and included different classes of amino acids, carnitines, acylcarnitines, and lysophosphatidylcholines (LPCs). Multivariate logistic regression was used to screen the metabolic factors contributing to frailty status, and orthogonal partial least squares discriminant analysis was used to explore important factors and distinguish different groups.ResultsIn older men demonstrating the frail phenotype, amino acid perturbations included lower tryptophan and higher glycine levels. With regard to lipid metabolism, the frailty phenotype was characterized by lower concentrations of isovalerylcarnitine (C5), LPC16:0 and LPC18:2, while higher levels of octanoyl-L-carnitine (C8), decanoyl-L-carnitine (C10), dodecanoyl-L-carnitine (C12) and tetradecanoyl-L-carnitine (C14). After adjusting for several clinical confounders, tryptophan, LPC18:2, LPC 16:0 and C5 were negatively correlated with frailty, and C8 and C12 were positively related to frailty. We preliminarily identified metabolic profiles (LPC16:0, LPC18:2, glycine and tryptophan) that may distinguish older men with frailty from those without frailty. Importantly, a set of serum amino acids and LPCs (LPC16:0, LPC18:2, and tryptophan) was characterized in the metabotype of older adults with an overlap of frailty and sarcopenia. The metabolites that were most discriminating of frailty status implied that the underlying mechanism might be involved in antioxidation and mitochondrial dysfunction.ConclusionsThese present metabolic analyses may provide valuable information on the potential biomarkers and possible biological mechanisms of frailty, and overlapping sarcopenia. The findings obtained may offer insight into their management in older adults.
Collapse
Affiliation(s)
- Li Meng
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Beijing Hospital/National Center of Gerontology of National Health Commission, Chinese Academy of Medical Sciences, Beijing, China
| | - Hong Shi
- Department of Geriatrics, National Clinical Research Center for Geriatrics, National Center of Gerontology, Institute of Geriatric Medicine, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Da-guang Wang
- Department of Laboratory Medicine, National Center of Gerontology, Institute of Geriatric Medicine, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Shi
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Beijing Hospital/National Center of Gerontology of National Health Commission, Chinese Academy of Medical Sciences, Beijing, China
| | - Wen-bin Wu
- Department of Geriatrics, National Clinical Research Center for Geriatrics, National Center of Gerontology, Institute of Geriatric Medicine, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ya-min Dang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Beijing Hospital/National Center of Gerontology of National Health Commission, Chinese Academy of Medical Sciences, Beijing, China
| | - Guo-qing Fan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Beijing Hospital/National Center of Gerontology of National Health Commission, Chinese Academy of Medical Sciences, Beijing, China
| | - Ji Shen
- Department of Geriatrics, National Clinical Research Center for Geriatrics, National Center of Gerontology, Institute of Geriatric Medicine, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Pu-lin Yu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Beijing Hospital/National Center of Gerontology of National Health Commission, Chinese Academy of Medical Sciences, Beijing, China
| | - Jun Dong
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Beijing Hospital/National Center of Gerontology of National Health Commission, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui-yue Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Beijing Hospital/National Center of Gerontology of National Health Commission, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Rui-yue Yang
| | - Huan Xi
- Department of Geriatrics, National Clinical Research Center for Geriatrics, National Center of Gerontology, Institute of Geriatric Medicine, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Huan Xi
| |
Collapse
|
24
|
D'Agnelli S, Amodeo G, Franchi S, Verduci B, Baciarello M, Panerai AE, Bignami EG, Sacerdote P. Frailty and pain, human studies and animal models. Ageing Res Rev 2022; 73:101515. [PMID: 34813977 DOI: 10.1016/j.arr.2021.101515] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 11/01/2022]
Abstract
The hypothesis that pain can predispose to frailty development has been recently investigated in several clinical studies suggesting that frailty and pain may share some mechanisms. Both pain and frailty represent important clinical and social problems and both lack a successful treatment. This circumstance is mainly due to the absence of in-depth knowledge of their pathological mechanisms. Evidence of shared pathways between frailty and pain are preliminary. Indeed, many clinical studies are observational and the impact of pain treatment, and relative pain-relief, on frailty onset and progression has never been investigated. Furthermore, preclinical research on this topic has yet to be performed. Specific researches on the pain-frailty relation are needed. In this narrative review, we will attempt to point out the most relevant findings present in both clinical and preclinical literature on the topic, with particular attention to genetics, epigenetics and inflammation, in order to underline the existing gaps and the potential future interventional strategies. The use of pain and frailty animal models discussed in this review might contribute to research in this area.
Collapse
|
25
|
Brunelli L, Davin A, Sestito G, Mimmi MC, De Simone G, Balducci C, Pansarasa O, Forloni G, Cereda C, Pastorelli R, Guaita A. Plasmatic Hippuric Acid as a Hallmark of Frailty in an Italian Cohort: The Mediation Effect of Fruit-Vegetable Intake. J Gerontol A Biol Sci Med Sci 2021; 76:2081-2089. [PMID: 34436596 PMCID: PMC8599087 DOI: 10.1093/gerona/glab244] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Frailty syndrome is an age-related condition involving a loss of resilience, susceptibility to adverse health outcomes, and poor quality of life. This study was conducted in the framework of InveCe.Ab, an ongoing longitudinal population-based study. Plasma from 130 older individuals (older adults aged 76–78 years) was analyzed and validated (on 303 participants) using mass spectrometry-based metabolomics approaches. Equivalence tests showed that metabolites from the central cellular metabolic pathways were equivalent in frail and fit participants. Hippuric acid was the only cometabolite that distinguished fit from frail older adults. Logistic regression analysis indicated that high hippuric acid levels are significantly associated with a reduction of the risk of frailty after 4 years. Mediation analysis using a Frailty Index, hippuric acid, and fruit–vegetable intake supported the role of fruit–vegetable consumption in the hippuric acid relationship with the Frailty Index. These data point to low plasma hippuric acid as a plausible hallmark of frailty status, associated with lower fruit–vegetable intakes.
Collapse
Affiliation(s)
- Laura Brunelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Giovanna Sestito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Giulia De Simone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Claudia Balducci
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | | | | | | |
Collapse
|
26
|
亀田 雅, 近藤 祥. [Metabolites for frailty biomarkers]. Nihon Ronen Igakkai Zasshi 2021; 58:333-340. [PMID: 34483155 DOI: 10.3143/geriatrics.58.333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
27
|
Atkins JL, Jylhävä J, Pedersen NL, Magnusson PK, Lu Y, Wang Y, Hägg S, Melzer D, Williams DM, Pilling LC. A genome-wide association study of the frailty index highlights brain pathways in ageing. Aging Cell 2021; 20:e13459. [PMID: 34431594 PMCID: PMC8441299 DOI: 10.1111/acel.13459] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 07/14/2021] [Accepted: 08/06/2021] [Indexed: 12/16/2022] Open
Abstract
Frailty is a common geriatric syndrome and strongly associated with disability, mortality and hospitalization. Frailty is commonly measured using the frailty index (FI), based on the accumulation of a number of health deficits during the life course. The mechanisms underlying FI are multifactorial and not well understood, but a genetic basis has been suggested with heritability estimates between 30 and 45%. Understanding the genetic determinants and biological mechanisms underpinning FI may help to delay or even prevent frailty. We performed a genome-wide association study (GWAS) meta-analysis of a frailty index in European descent UK Biobank participants (n = 164,610, 60-70 years) and Swedish TwinGene participants (n = 10,616, 41-87 years). FI calculation was based on 49 or 44 self-reported items on symptoms, disabilities and diagnosed diseases for UK Biobank and TwinGene, respectively. 14 loci were associated with the FI (p < 5*10-8 ). Many FI-associated loci have established associations with traits such as body mass index, cardiovascular disease, smoking, HLA proteins, depression and neuroticism; however, one appears to be novel. The estimated single nucleotide polymorphism (SNP) heritability of the FI was 11% (0.11, SE 0.005). In enrichment analysis, genes expressed in the frontal cortex and hippocampus were significantly downregulated (adjusted p < 0.05). We also used Mendelian randomization to identify modifiable traits and exposures that may affect frailty risk, with a higher educational attainment genetic risk score being associated with a lower degree of frailty. Risk of frailty is influenced by many genetic factors, including well-known disease risk factors and mental health, with particular emphasis on pathways in the brain.
Collapse
Affiliation(s)
- Janice L. Atkins
- Epidemiology and Public Health GroupUniversity of Exeter Medical SchoolExeterUK
| | - Juulia Jylhävä
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Nancy L. Pedersen
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Department of PsychologyUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Patrik K. Magnusson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Yi Lu
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Yunzhang Wang
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Sara Hägg
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - David Melzer
- Epidemiology and Public Health GroupUniversity of Exeter Medical SchoolExeterUK
- Center on AgingUniversity of ConnecticutFarmingtonCTUSA
| | - Dylan M. Williams
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | - Luke C. Pilling
- Epidemiology and Public Health GroupUniversity of Exeter Medical SchoolExeterUK
- Center on AgingUniversity of ConnecticutFarmingtonCTUSA
| |
Collapse
|
28
|
Minakata S, Manabe S, Inai Y, Ikezaki M, Nishitsuji K, Ito Y, Ihara Y. Protein C-Mannosylation and C-Mannosyl Tryptophan in Chemical Biology and Medicine. Molecules 2021; 26:molecules26175258. [PMID: 34500691 PMCID: PMC8433626 DOI: 10.3390/molecules26175258] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/25/2022] Open
Abstract
C-Mannosylation is a post-translational modification of proteins in the endoplasmic reticulum. Monomeric α-mannose is attached to specific Trp residues at the first Trp in the Trp-x-x-Trp/Cys (W-x-x-W/C) motif of substrate proteins, by the action of C-mannosyltransferases, DPY19-related gene products. The acceptor substrate proteins are included in the thrombospondin type I repeat (TSR) superfamily, cytokine receptor type I family, and others. Previous studies demonstrated that C-mannosylation plays critical roles in the folding, sorting, and/or secretion of substrate proteins. A C-mannosylation-defective gene mutation was identified in humans as the disease-associated variant affecting a C-mannosylation motif of W-x-x-W of ADAMTSL1, which suggests the involvement of defects in protein C-mannosylation in human diseases such as developmental glaucoma, myopia, and/or retinal defects. On the other hand, monomeric C-mannosyl Trp (C-Man-Trp), a deduced degradation product of C-mannosylated proteins, occurs in cells and extracellular fluids. Several studies showed that the level of C-Man-Trp is upregulated in blood of patients with renal dysfunction, suggesting that the metabolism of C-Man-Trp may be involved in human kidney diseases. Together, protein C-mannosylation is considered to play important roles in the biosynthesis and functions of substrate proteins, and the altered regulation of protein C-manosylation may be involved in the pathophysiology of human diseases. In this review, we consider the biochemical and biomedical knowledge of protein C-mannosylation and C-Man-Trp, and introduce recent studies concerning their significance in biology and medicine.
Collapse
Affiliation(s)
- Shiho Minakata
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
| | - Shino Manabe
- Pharmaceutical Department, The Institute of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa, Tokyo 142-8501, Japan;
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Science & Faculty of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Sendai, Miyagi 980-8578, Japan
| | - Yoko Inai
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
| | - Midori Ikezaki
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
| | - Kazuchika Nishitsuji
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
| | - Yukishige Ito
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan;
- RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshito Ihara
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
- Correspondence: ; Tel.: +81-73-441-0628
| |
Collapse
|
29
|
Freidin MB, Stalteri MA, Wells PM, Lachance G, Baleanu AF, Bowyer RCE, Kurilshikov A, Zhernakova A, Steves CJ, Williams FMK. An association between chronic widespread pain and the gut microbiome. Rheumatology (Oxford) 2021; 60:3727-3737. [PMID: 33331911 PMCID: PMC8328510 DOI: 10.1093/rheumatology/keaa847] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/12/2020] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVES Chronic widespread musculoskeletal pain (CWP) is a characteristic symptom of fibromyalgia, which has been shown to be associated with an altered gut microbiome. Microbiome studies to date have not examined the milder CWP phenotype specifically nor have they explored the role of raised BMI. The aim of this study was to investigate whether the microbiome is abnormal in CWP. METHODS CWP was assessed using a standardized screening questionnaire in female volunteers from the TwinsUK cohort including 113 CWP cases and 1623 controls. The stool microbiome was characterized using 16S rRNA amplicon sequencing and amplicon sequence variants, and associations with CWP examined using linear mixed-effects models adjusting for BMI, age, diet, family relatedness and technical factors. RESULTS Alpha diversity was significantly lower in CWP cases than controls (Mann-Whitney test, P-values 2.3e-04 and 1.2e-02, for Shannon and Simpson indices respectively). The species Coprococcus comes was significantly depleted in CWP cases (Padj = 3.04e-03). A genome-wide association study (GWAS) performed for C. comes in TwinsUK followed by meta-analysis with three Dutch cohorts (total n = 3521) resulted in nine suggestive regions, with the most convincing on chromosome 4 near the TRAM1L1 gene (rs76957229, P = 7.4e-8). A Mendelian randomization study based on the results of the GWAS did not support a causal role for C. comes on the development of CWP. CONCLUSIONS We have demonstrated reduced diversity in the microbiome in CWP, indicating an involvement of the gut microbiota in CWP; prospectively the microbiome may offer therapeutic opportunities for this condition.
Collapse
Affiliation(s)
- Maxim B Freidin
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Maria A Stalteri
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Philippa M Wells
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Genevieve Lachance
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Andrei-Florin Baleanu
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Ruth C E Bowyer
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| |
Collapse
|
30
|
Howlett SE, Rutenberg AD, Rockwood K. The degree of frailty as a translational measure of health in aging. NATURE AGING 2021; 1:651-665. [PMID: 37117769 DOI: 10.1038/s43587-021-00099-3] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 07/06/2021] [Indexed: 04/30/2023]
Abstract
Frailty is a multiply determined, age-related state of increased risk for adverse health outcomes. We review how the degree of frailty conditions the development of late-life diseases and modifies their expression. The risks for frailty range from subcellular damage to social determinants. These risks are often synergistic-circumstances that favor damage also make repair less likely. We explore how age-related damage and decline in repair result in cellular and molecular deficits that scale up to tissue, organ and system levels, where they are jointly expressed as frailty. The degree of frailty can help to explain the distinction between carrying damage and expressing its usual clinical manifestations. Studying people-and animals-who live with frailty, including them in clinical trials and measuring the impact of the degree of frailty are ways to better understand the diseases of old age and to establish best practices for the care of older adults.
Collapse
Affiliation(s)
- Susan E Howlett
- Geriatric Medicine Research Unit, Department of Medicine, Dalhousie University & Nova Scotia Health, Halifax, Nova Scotia, Canada
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew D Rutenberg
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kenneth Rockwood
- Geriatric Medicine Research Unit, Department of Medicine, Dalhousie University & Nova Scotia Health, Halifax, Nova Scotia, Canada.
| |
Collapse
|
31
|
Sphingomyelin is involved in multisite musculoskeletal pain: evidence from metabolomic analysis in 2 independent cohorts. Pain 2021; 162:1876-1881. [PMID: 33273416 DOI: 10.1097/j.pain.0000000000002163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 11/25/2020] [Indexed: 01/05/2023]
Abstract
ABSTRACT Metabolic dysfunction has been suggested to be involved in musculoskeletal pain; however, few studies have identified metabolic markers associated with multisite musculoskeletal pain (MSMP). This study sought to identify metabolic marker(s) for MSMP by metabolomic analysis. The Tasmanian Older Adult Cohort Study (TASOAC) provided the discovery cohort with the Newfoundland Osteoarthritis Study (NFOAS) providing the replication cohort. Multisite musculoskeletal pain was assessed by a self-reported pain questionnaire and defined as painful sites ≥4 in both the TASOAC and the NFOAS. Furthermore, MSMP was also defined as painful sites ≥7, whereas non-MSMP was defined as either painful sites <7 or ≤1 in the NFOAS. Serum samples of the TASOAC received metabolic profiling using The Metabolomics Innovation Centre Prime Metabolomics Profiling Assay. The data on the identified metabolites were retrieved from NFOAS metabolomic database for the purpose of replication. A total of 409 participants were included in the TASOAC, 38% of them had MSMP. Among the 143 metabolites assessed, 129 passed quality control and were included in the analysis. Sphingomyelin (SM) C18:1 was significantly associated with MSMP (odds ratio [OR] per log µM increase = 3.96, 95% confidence interval, 1.95-8.22; P = 0.0002). The significance remained in multivariable analysis (OR per log µM increase = 2.70, 95% confidence interval, 1.25-5.95). A total of 610 participants were included in the NFOAS, and the association with SM C18:1 was successfully replicated with 3 MSMP definitions (OR ranging from 1.89 to 2.82; all P < 0.03). Our findings suggest that sphingomyelin metabolism is involved in the pathogenesis of MSMP, and the circulating level of SM C18:1 could serve as a potential marker in the management of MSMP.
Collapse
|
32
|
Costenoble A, Knoop V, Vermeiren S, Vella RA, Debain A, Rossi G, Bautmans I, Verté D, Gorus E, De Vriendt P. A Comprehensive Overview of Activities of Daily Living in Existing Frailty Instruments: A Systematic Literature Search. THE GERONTOLOGIST 2021; 61:e12-e22. [PMID: 31872238 DOI: 10.1093/geront/gnz147] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The relationship between frailty and disability in activities of daily living (ADLs) can be seen in different ways, with disability being-to varying degrees-a characteristic, negative outcome, or predictor of frailty. This conflation of definitions is partly a result of the different frailty tools used in research. Aiming to provide a comprehensive overview, this systematic literature search analyzed (i) if, (ii) to what extent, and (iii) how ADLs are evaluated by frailty instruments. RESEARCH DESIGN AND METHODS A search was performed in PubMed, Web of Knowledge, and PsycINFO to identify all frailty instruments, followed by categorization of the ADL items into basic (b-), instrumental (i-), and advanced (a-) ADLs. RESULTS In total, 192 articles described 217 frailty instruments, from which 52.1% contained ADL items: 45.2% b-ADLs, 35.0% i-ADLs, and 10.1% a-ADLs. The most commonly included ADL items were bathing (b-ADLs); using transportation (i-ADLs); and semiprofessional work engagement in organized social life or leisure activities (a-ADLs). These instruments all had a multidomain origin (χ 2 = 122.4, p < .001). DISCUSSION AND IMPLICATIONS Because 52.1% of all instruments included ADL items, the concepts of frailty and disability appear to be highly entangled. This might lead to circular reasoning, serious concerns regarding contamination, and invalid research results.
Collapse
Affiliation(s)
- Axelle Costenoble
- Frailty in Ageing (FRIA) Research Department, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), Belgium
| | - Veerle Knoop
- Frailty in Ageing (FRIA) Research Department, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), Belgium
| | - Sofie Vermeiren
- Frailty in Ageing (FRIA) Research Department, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), Belgium
| | - Roberta Azzopardi Vella
- Frailty in Ageing (FRIA) Research Department, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), Belgium
| | - Aziz Debain
- Frailty in Ageing (FRIA) Research Department, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), Belgium
- Geriatrics Department, Universitair Ziekenhuis Brussel (UZ Brussel), Belgium
| | - Gina Rossi
- Personality and Psychopathology Research Group, Faculty of Psychology and Educational Sciences, Belgium
| | - Ivan Bautmans
- Frailty in Ageing (FRIA) Research Department, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), Belgium
- Geriatrics Department, Universitair Ziekenhuis Brussel (UZ Brussel), Belgium
| | - Dominique Verté
- Frailty in Ageing (FRIA) Research Department, Belgium
- Belgian Ageing Studies Research Group, Vrije Universiteit Brussel (VUB), Belgium
| | - Ellen Gorus
- Frailty in Ageing (FRIA) Research Department, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), Belgium
- Geriatrics Department, Universitair Ziekenhuis Brussel (UZ Brussel), Belgium
| | - Patricia De Vriendt
- Frailty in Ageing (FRIA) Research Department, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), Belgium
- Artevelde Hogeschool, Ghent, Belgium
| |
Collapse
|
33
|
Livshits G, Kalinkovich A. Specialized, pro-resolving mediators as potential therapeutic agents for alleviating fibromyalgia symptomatology. PAIN MEDICINE 2021; 23:977-990. [PMID: 33565588 DOI: 10.1093/pm/pnab060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To present a hypothesis on a novel strategy in the treatment of fibromyalgia (FM). DESIGN A narrative review. SETTING FM as a disease remains a challenging concept for numerous reasons, including undefined etiopathogenesis, unclear triggers and unsuccessful treatment modalities. We hypothesize that the inflammatome, the entire set of molecules involved in inflammation, acting as a common pathophysiological instrument of gut dysbiosis, sarcopenia, and neuroinflammation, is one of the major mechanisms underlying FM pathogenesis. In this setup, dysbiosis is proposed as the primary trigger of the inflammatome, sarcopenia as the peripheral nociceptive source, and neuroinflammation as the central mechanism of pain sensitization, transmission and symptomatology of FM. Whereas neuroinflammation is highly-considered as a critical deleterious element in FM pathogenesis, the presumed pathogenic roles of sarcopenia and systemic inflammation remain controversial. Nevertheless, sarcopenia-associated processes and dysbiosis have been recently detected in FM individuals. The prevalence of pro-inflammatory factors in the cerebrospinal fluid and blood has been repeatedly observed in FM individuals, supporting an idea on the role of inflammatome in FM pathogenesis. As such, failed inflammation resolution might be one of the underlying pathogenic mechanisms. In accordance, the application of specialized, inflammation pro-resolving mediators (SPMs) seems most suitable for this goal. CONCLUSIONS The capability of various SPMs to prevent and attenuate pain has been repeatedly demonstrated in laboratory animal experiments. Since SPMs suppress inflammation in a manner that does not compromise host defense, they could be attractive and safe candidates for the alleviation of FM symptomatology, probably in combination with anti-dysbiotic medicine.
Collapse
Affiliation(s)
- Gregory Livshits
- Adelson School of Medicine, Ariel University, Ariel, Israel.,Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
34
|
Reply to Pan et al.: Whole blood metabolome analysis combined with comprehensive frailty assessment. Proc Natl Acad Sci U S A 2021; 118:2016640118. [PMID: 33443142 PMCID: PMC7817210 DOI: 10.1073/pnas.2016640118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
35
|
Whole Blood Metabolomics in Aging Research. Int J Mol Sci 2020; 22:ijms22010175. [PMID: 33375345 PMCID: PMC7796096 DOI: 10.3390/ijms22010175] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/25/2020] [Accepted: 12/25/2020] [Indexed: 02/08/2023] Open
Abstract
Diversity is observed in the wave of global aging because it is a complex biological process exhibiting individual variability. To assess aging physiologically, markers for biological aging are required in addition to the calendar age. From a metabolic perspective, the aging hypothesis includes the mitochondrial hypothesis and the calorie restriction (CR) hypothesis. In experimental models, several compounds or metabolites exert similar lifespan-extending effects, like CR. However, little is known about whether these metabolic modulations are applicable to human longevity, as human aging is greatly affected by a variety of factors, including lifestyle, genetic or epigenetic factors, exposure to stress, diet, and social environment. A comprehensive analysis of the human blood metabolome captures complex changes with individual differences. Moreover, a non-targeted analysis of the whole blood metabolome discloses unexpected aspects of human biology. By using such approaches, markers for aging or aging-relevant conditions were identified. This information should prove valuable for future diagnosis or clinical interventions in diseases relevant to aging.
Collapse
|
36
|
The potential for complex computational models of aging. Mech Ageing Dev 2020; 193:111403. [PMID: 33220267 DOI: 10.1016/j.mad.2020.111403] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/24/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022]
Abstract
The gradual accumulation of damage and dysregulation during the aging of living organisms can be quantified. Even so, the aging process is complex and has multiple interacting physiological scales - from the molecular to cellular to whole tissues. In the face of this complexity, we can significantly advance our understanding of aging with the use of computational models that simulate realistic individual trajectories of health as well as mortality. To do so, they must be systems-level models that incorporate interactions between measurable aspects of age-associated changes. To incorporate individual variability in the aging process, models must be stochastic. To be useful they should also be predictive, and so must be fit or parameterized by data from large populations of aging individuals. In this perspective, we outline where we have been, where we are, and where we hope to go with such computational models of aging. Our focus is on data-driven systems-level models, and on their great potential in aging research.
Collapse
|
37
|
Teckchandani S, Nagana Gowda GA, Raftery D, Curatolo M. Metabolomics in chronic pain research. Eur J Pain 2020; 25:313-326. [PMID: 33065770 DOI: 10.1002/ejp.1677] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/22/2020] [Accepted: 10/11/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND OBJECTIVE Metabolomics deals with the identification and quantification of small molecules (metabolites) in biological samples. As metabolite levels can reflect normal or altered metabolic pathways, their measurement provides information to improve the understanding, diagnosis and management of diseases. Despite its immense potential, metabolomics applications to pain research have been sparse. This paper describes current metabolomics techniques, reviews published human metabolomics pain research and compares successful metabolomics research in other areas of medicine with the goal of highlighting opportunities offered by metabolomics to advance pain medicine. DATABASES AND DATA TREATMENT Non-systematic review. RESULTS Our search identified 19 studies that adopted a metabolomics approach in: fibromyalgia (7), chronic widespread pain (4), other musculoskeletal pain conditions (5), neuropathic pain (1), complex regional pain syndrome (1) and pelvic pain (1). The studies used either mass spectrometry or nuclear magnetic resonance. Most are characterized by small sample sizes. Some consistency has been found for alterations in glutamate and testosterone metabolism, and metabolic imbalances caused by the gut microbiome. CONCLUSIONS Metabolomics research in chronic pain is in its infancy. Most studies are at the pilot stage. Metabolomics research has been successful in other areas of medicine. These achievements should motivate investigators to expand metabolomics research to improve the understanding of the basic mechanisms of human pain, as well as provide tools to diagnose, predict and monitor chronic pain conditions. Metabolomics research can lead to the identification of biomarkers to support the development and testing of treatments, thereby facilitating personalized pain medicine.
Collapse
Affiliation(s)
- Shweta Teckchandani
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - G A Nagana Gowda
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA.,Northwest Metabolomics Research Center, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA
| | - Daniel Raftery
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA.,Northwest Metabolomics Research Center, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA
| | - Michele Curatolo
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA.,Harborview Injury Prevention and Research Center, University of Washington, Seattle, WA, USA.,CLEAR Research Center for Musculoskeletal Disorders, University of Washington, Seattle, WA, USA
| |
Collapse
|
38
|
Omics biomarkers for frailty in older adults. Clin Chim Acta 2020; 510:363-372. [PMID: 32745578 DOI: 10.1016/j.cca.2020.07.057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/14/2022]
Abstract
Frailty is a clinical state characterized by an age-related unsteady state of the body, a decline in physiological function, and an increased vulnerability to adverse outcomes. Early diagnosis of frailty is important for improving the quality of life in older adults and promoting healthy aging. The biological mechanisms underlying frailty have been extensively studied in recent years. Combining assessment tools and biomarkers can facilitate the early diagnosis of frailty. However, there is a lack of stable and reliable frailty-related biomarkers for use in clinical practice. Advances in the multi-omics platforms have provided new information on the molecular mechanisms underlying frailty. Thus, identifying biomarkers using omics-based approaches helps explore the physiological mechanisms underlying frailty, and aids the evaluation of the risk of frailty development and progression. This article reviews the current status of frailty biomarkers from the genomics, transcriptomics, proteomics, and metabolomics perspectives.
Collapse
|
39
|
Aroke EN, Powell-Roach KL. The Metabolomics of Chronic Pain Conditions: A Systematic Review. Biol Res Nurs 2020; 22:458-471. [PMID: 32666804 DOI: 10.1177/1099800420941105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Chronic pain is a significant public health problem in the United States, affecting approximately 100 million people. Yet there is a lack of robust biomarkers for clinical use in chronic pain conditions. Downstream effects of environmental, genomic, and proteomic variations in individuals with chronic pain conditions can be identified and quantified using a metabolomic approach. AIM/DESIGN The purpose of this systematic review was to examine the literature for reports of potential metabolomic signatures associated with chronic pain conditions. METHODS We searched relevant electronic databases for published studies that used various metabolomic approaches to investigate chronic pain conditions among subjects of all ages. RESULTS Our search identified a total of 586 articles, 18 of which are included in this review. The reviewed studies used metabolomics to investigate fibromyalgia (n = 5), osteoarthritis (n = 4), migraine (n = 3), musculoskeletal pain (n = 2), and other chronic pain conditions (n = 1/condition). Results show that several known and newly identified metabolites differ in individuals with chronic pain conditions compared to those without these conditions. These include amino acids (e.g., glutamine, serine, and phenylalanine) and intermediate products (e.g., succinate, citrate, acetylcarnitine, and N-acetylornithine) of pathways that metabolize various macromolecules. CONCLUSION Though more high-quality research is needed, this review provides insights into potential biomarkers for future metabolomics studies in people with chronic pain conditions.
Collapse
Affiliation(s)
- Edwin N Aroke
- School of Nursing, University of Alabama at Birmingham, AL, USA
| | | |
Collapse
|
40
|
Sathyan S, Verghese J. Genetics of frailty: A longevity perspective. Transl Res 2020; 221:83-96. [PMID: 32289255 PMCID: PMC7729977 DOI: 10.1016/j.trsl.2020.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 12/31/2022]
Abstract
Frailty is a complex late life phenotype characterized by cumulative declines in multiple physiological systems that increases the risk for disability and mortality. The biological changes associated with aging are risk factors for frailty as well as for complex diseases; whereas longevity is assumed to be an outcome of protective biological mechanisms. Understanding the interplay between biological alterations associated with aging and protective mechanisms associated with longevity in the context of frailty may help guide development of interventions to increase healthspan and promote successful aging. The complexity of these phenotypes and relatively low heritability in studies are the main roadblocks in deciphering genetic mechanisms of these age associated conditions. We review genetic research related to frailty, and discuss the possible intertwined biology of frailty and longevity.
Collapse
Affiliation(s)
- Sanish Sathyan
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
| | - Joe Verghese
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
41
|
Frailty markers comprise blood metabolites involved in antioxidation, cognition, and mobility. Proc Natl Acad Sci U S A 2020; 117:9483-9489. [PMID: 32295884 PMCID: PMC7196897 DOI: 10.1073/pnas.1920795117] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Frailty resulting from age-related deterioration of multiple organ systems displays complex features, including cognitive dysfunction, hypomobility, and impaired daily activity. However, metabolic aspects of frailty remain unclear. We performed untargeted, comprehensive metabolomics of whole blood from 19 frail and nonfrail elderly patients. We identified 22 markers, including 15 for frailty, 6 for cognition, and 12 for hypomobility, most of which are abundant in blood. Frailty markers include 5 of 6 for cognition and 6 of 12 for hypomobility. These overlapping markers include decreased levels of metabolites related to antioxidation, nitrogen, and amino acid metabolism. Ergothioneine, an antioxidant involved in neuronal diseases, declines in frailty. Thus, we reveal essential metabolites linked to the pathogenesis of frailty, including vulnerability to oxidative stress. As human society ages globally, age-related disorders are becoming increasingly common. Due to decreasing physiological reserves and increasing organ system dysfunction associated with age, frailty affects many elderly people, compromising their ability to cope with acute stressors. Frail elderly people commonly manifest complex clinical symptoms, including cognitive dysfunction, hypomobility, and impaired daily activity, the metabolic basis of which remains poorly understood. We applied untargeted, comprehensive LC-MS metabolomic analysis to human blood from 19 frail and nonfrail elderly patients who were clinically evaluated using the Edmonton Frail Scale, the MoCA-J for cognition, and the TUG for mobility. Among 131 metabolites assayed, we identified 22 markers for frailty, cognition, and hypomobility, most of which were abundant in blood. Frailty markers included 5 of 6 markers specifically related to cognition and 6 of 12 markers associated with hypomobility. These overlapping sets of markers included metabolites related to antioxidation, muscle or nitrogen metabolism, and amino acids, most of which are decreased in frail elderly people. Five frailty-related metabolites that decreased—1,5-anhydroglucitol, acetyl-carnosine, ophthalmic acid, leucine, and isoleucine—have been previously reported as markers of aging, providing a metabolic link between human aging and frailty. Our findings clearly indicate that metabolite profiles efficiently distinguish frailty from nonfrailty. Importantly, the antioxidant ergothioneine, which decreases in frailty, is neuroprotective. Oxidative stress resulting from diminished antioxidant levels could be a key vulnerability for the pathogenesis of frailty, exacerbating illnesses related to human aging.
Collapse
|
42
|
Rockwood K, Howlett SE. Fifteen years of progress in understanding frailty and health in aging. BMC Med 2018; 16:220. [PMID: 30477486 PMCID: PMC6258409 DOI: 10.1186/s12916-018-1223-3] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 02/26/2023] Open
Abstract
The notion of frailty has evolved for more than 15 years. Although there is no consensus definition, frailty reflects a state of increased vulnerability to adverse health outcomes for individuals of the same chronological age. Two commonly used clinical tools, the frailty index and the frailty phenotype, both measure health-related deficits. The frailty index is a ratio of the number of deficits that an individual has accumulated divided by all deficits measured, whereas the phenotype specifies frailty as represented by poor performance in three of five criteria (i.e., weight loss, exhaustion, weakness, slowness, lack of activity). From human studies, animal models of both approaches have been developed and are beginning to shed light on mechanisms underlying frailty, the influence of frailty on disease expression, and new interventions to attenuate frailty. Currently, back-translation to humans is occurring. As we start to understand subcellular mechanisms involved in damage and repair as well as their response to treatment, we will begin to understand the molecular basis of aging and, thus, of frailty.
Collapse
Affiliation(s)
- Kenneth Rockwood
- Geriatric Medicine, Department of Medicine, Dalhousie University, Halifax, NS, Canada.
| | - Susan E Howlett
- Geriatric Medicine, Department of Medicine, Dalhousie University, Halifax, NS, Canada.,Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|