1
|
Yi J, Jiang C, Xia L. Mediated roles of oxidative stress and kidney function to leukocyte telomere length and prognosis in chronic kidney disease. Ren Fail 2025; 47:2464828. [PMID: 40011224 PMCID: PMC11866651 DOI: 10.1080/0886022x.2025.2464828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Few studies have focused on the correlation between leukocyte telomere length (LTL) and cancer-related mortality or identified potential factors that mediate the relationship between LTL and mortality among chronic kidney disease (CKD) patients. Our study aimed to explore the associations between LTL and all-cause and cause-specific mortality and to identify the underlying mediators. METHODS CKD patients were obtained from the National Health and Nutrition Examination Survey (NHANES) 1999-2002. Cox regression analysis and restricted cubic spline analysis were used to explore the associations between LTL and all-cause or specific-cause mortality and their nonlinear connections. Stratified analyses were executed to assess the relationships among the different subgroups. The latent mediated factors were confirmed using mediation analysis. Sensitivity analyses were used to evaluate the robustness of our findings. RESULTS Longer LTL associated with the lower risk of all-cause mortality, cardiovascular disease (CVD) and cancer-related mortality, and U-shaped relationships were detected. Patients younger than 65 years with greater LTL or who had hypertension had better prognoses. Age and history of hypertension were associated with LTL and overall mortality. In addition, estimated glomerular filtration rate (eGFR), albumin, and total bilirubin mediated the association, and the proportions of indirect effects were 7.81%, 3.77%, and 2.50%, respectively. Six sensitivity analyses confirmed the robustness of our findings. CONCLUSIONS This study revealed that LTL was a protective factor for survival among patients with CKD and emphasized the mediating roles of oxidative stress and kidney function.
Collapse
Affiliation(s)
- Jiahong Yi
- Department of VIP Region, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Chang Jiang
- Department of VIP Region, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Liangping Xia
- Department of VIP Region, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| |
Collapse
|
2
|
Zhang Y, Cui Y, Sun C, Guo J, Li M. ED-71 ameliorates OVX-induced osteoporosis by regulating calcium homeostasis and SIRT1-mediated mitochondrial function, alleviating osteoblast senescence and suppressing osteoclastogenesis. Cell Signal 2025; 131:111713. [PMID: 40049265 DOI: 10.1016/j.cellsig.2025.111713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/12/2025] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Osteoporosis arising from estrogen deficiency is characterized by oxidative stress and cellular senescence accompanied by calcium loss and disrupted bone metabolism. The paracrine interaction between osteoblasts and osteoclasts, along with the ratio of receptor activator of nuclear factor-κB ligand (RANKL) to osteoprotegerin (OPG), play a pivotal role in maintaining bone homeostasis. Eldecalcitol (ED-71), a novel active form of vitamin D, can reduce the ratio of RANKL to OPG in osteoblasts. In this study, an ovariectomized (OVX) rat model was established in vivo, and a cell model was constructed in vitro using H₂O₂ to explore the specific mechanism by which ED-71 improved the release of RANKL/OPG in senescent osteoblasts. Mitochondrial dysfunction and calcium imbalance were identified as significant factors. Under oxidative stress conditions, ED-71 alleviated endoplasmic reticulum (ER) stress by decreasing the ratio of phosphorylated protein kinase R-like ER kinase (P-PERK/PERK), and augmented the expression levels of sarcoplasmic reticulum/endoplasmic reticulum calcium ATPase 2 (SCERA2) thereby promoting calcium uptake by the ER, enhancing ER calcium influx, and effectively ameliorating calcium homeostasis between the ER and mitochondria. Consequently, it mitigates mitochondrial calcium overload and associated dysfunction. In contrast, ED-71 increased the expression of silent information regulator 1 (SIRT1) and phosphorylated AMP-activated protein kinase (P-AMPK). This alleviates mitochondrial dysfunction and promotes adenosine triphosphate (ATP). The combined effects of these two factors synergistically contribute to the improvement in osteoblast senescence.
Collapse
Affiliation(s)
- Yaoguang Zhang
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China; Center of Osteoporosis and Bone Mineral Research, Shandong University, China
| | - Yajun Cui
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Center of Osteoporosis and Bone Mineral Research, Shandong University, China
| | - Changyun Sun
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China; Center of Osteoporosis and Bone Mineral Research, Shandong University, China.
| | - Jie Guo
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China.
| | - Minqi Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China; Center of Osteoporosis and Bone Mineral Research, Shandong University, China; School of Clinical Medicine, Jining Medical University, Jining, China.; Institute of Oral Basic Research, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China
| |
Collapse
|
3
|
Xiao T, Ünal E. Remodeling, compartmentalization, and degradation: a trifecta for organelle quality control during gametogenesis. Curr Opin Genet Dev 2025; 92:102347. [PMID: 40233504 DOI: 10.1016/j.gde.2025.102347] [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: 01/15/2025] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/17/2025]
Abstract
The key to healthy offspring production lies in the accurate inheritance of components from progenitor germ cells during gametogenesis. Along with genetic material, precise regulation of organelle inheritance is vital for gamete health and embryonic development, especially in aged organisms, where organelle function declines and damage accumulates. In these cases, removing age-related organellar defects in precursor cells is crucial for successful reproduction. The single-celled organism Saccharomyces cerevisiae shares striking similarities with more complex organisms: like metazoan cells, yeast accumulate organelle damage with age, yet can still produce damage-free gametes with a reset lifespan. Recent studies show that organelles undergo significant reorganization during yeast gametogenesis, and similar remodeling occurs in metazoans, suggesting common strategies for maintaining gamete quality. This review summarizes organellar reorganization during gametogenesis in yeast and how it aids in clearing age-related cellular damage. We also explore organellar remodeling in multicellular organisms and discuss the potential mechanisms and biological benefits of meiotic organellar reshaping.
Collapse
Affiliation(s)
- Tianyao Xiao
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley 94720, USA. https://twitter.com/@XiaoTianyao
| | - Elçin Ünal
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley 94720, USA.
| |
Collapse
|
4
|
Dar NJ, Currais A, Taguchi T, Andrews N, Maher P. Cannabinol (CBN) alleviates age-related cognitive decline by improving synaptic and mitochondrial health. Redox Biol 2025; 84:103692. [PMID: 40412024 DOI: 10.1016/j.redox.2025.103692] [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: 02/21/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Age-related cognitive decline and neurodegenerative diseases, such as Alzheimer's disease, represent major global health challenges, particularly with an aging population. Mitochondrial dysfunction appears to play a central role in the pathophysiology of these conditions by driving redox dysregulation and impairing cellular energy metabolism. Despite extensive research, effective therapeutic options remain limited. Cannabinol (CBN), a cannabinoid previously identified as a potent inhibitor of oxytosis/ferroptosis through mitochondrial modulation, has demonstrated promising neuroprotective effects. In cell culture, CBN targets mitochondria, preserving mitochondrial membrane potential, enhancing antioxidant defenses and regulating bioenergetic processes. However, the in vivo therapeutic potential of CBN, particularly in aging models, has not been thoroughly explored. To address this gap, this study investigated the effects of CBN on age-associated cognitive decline and metabolic dysfunction using the SAMP8 mouse model of accelerated aging. Our results show that CBN significantly improves spatial learning and memory, with more pronounced cognitive benefits observed in female mice. These cognitive improvements are accompanied by sex-specific changes in metabolic parameters, such as enhanced oxygen consumption and energy expenditure. Mechanistically, CBN modulates key regulators of mitochondrial dynamics, including mitofusin 2 (MFN2) and dynamin-related protein 1 (DRP1), while upregulating markers of mitochondrial biogenesis including mitochondrial transcription factor A (TFAM) and translocase of outer mitochondrial membrane 20 (TOM20). Additionally, CBN upregulates key synaptic proteins involved in vesicle trafficking and postsynaptic signaling suggesting that it enhances synaptic function and neurotransmission, further reinforcing its neuroprotective effects. This study provides in vivo evidence supporting CBN's potential to mitigate age-related cognitive and metabolic dysfunction, with notable sex-specific effects, highlighting its promise for neurodegenerative diseases and cognitive decline.
Collapse
Affiliation(s)
- Nawab John Dar
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Antonio Currais
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Taketo Taguchi
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nick Andrews
- Behaviour Testing Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Pamela Maher
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
5
|
Bonetto V, Ferraresi A, Sampò S, Isidoro C. Fungal Bioactive Compounds as Emerging Therapeutic Options for Age-Related Neurodegenerative Disorders. Int J Mol Sci 2025; 26:4800. [PMID: 40429941 PMCID: PMC12111997 DOI: 10.3390/ijms26104800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/07/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Aging is a complex biological process characterized by progressive multiorgan deterioration that compromises the quality of life. Unhealthy aging often associates with cognitive decline and motor-neurological disorders including Alzheimer's disease, Parkinson's disease, and Huntington's disease. Genetic, environmental, and lifestyle factors, which include dietary habits, interact with aging and influence brain health, thus having an impact on the development of neurodegenerative disorders. In this context, fungal-derived bioactive compounds have emerged as promising neuroprotective agents due to their diverse biological properties that include antioxidative, anti-inflammatory, pro-autophagic, and neurotrophic effects. Key fungal metabolites, including polysaccharides, terpenoids, alkaloids, and phenolic compounds have been shown to modulate neuroinflammatory pathways, enhance neuronal survival, stimulate protective autophagy, and promote synaptic plasticity. Still, challenges related to their bioavailability, standardization, and clinical translation remain unresolved. Future deep research will be crucial to unlocking the full therapeutic potential of fungal-derived neuroprotective compounds. This review examines the potential therapeutic role of fungal metabolites, providing a comparative evaluation with a focus on their mechanisms of action in promoting brain health and longevity.
Collapse
Affiliation(s)
- Valentina Bonetto
- Department of Science and Technologic Innovation, Università del Piemonte Orientale, 15121 Alessandria, Italy
| | - Alessandra Ferraresi
- Department of Health Sciences, Università del Piemonte Orientale, Via Paolo Solaroli 17, 28100 Novara, Italy;
| | - Simonetta Sampò
- Department for Sustainable Development and Ecological Transition, Università del Piemonte Orientale, 13100 Vercelli, Italy;
| | - Ciro Isidoro
- Department of Health Sciences, Università del Piemonte Orientale, Via Paolo Solaroli 17, 28100 Novara, Italy;
| |
Collapse
|
6
|
Xu W, Li XJ, Zhong YS, He JQ, Xie W, Kang YK, Ying HZ, Yu CH. Structural characterizations and antiaging activities of hydrolyzed low-molecular-weight polysaccharides from Polygoni Multiflori Radix Praeparata. Carbohydr Polym 2025; 356:123381. [PMID: 40049961 DOI: 10.1016/j.carbpol.2025.123381] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 05/13/2025]
Abstract
Natural polysaccharides as the primary active components derived from herbal medicine often face challenges due to their large molecular weights, varying chemical structures and poor bioavailability, which significantly restrict their bioactive mechanism investigation and clinical applications. To improve the bioavailability and clarify the antiaging mechanism of polysaccharides from Polygoni Multiflori Radix Praeparata, the high-molecular-weight polysaccharides (PRP) were hydrolyzed into two low-molecular-weight fractions (PRP1 and PRP2) by hydrogen peroxide-ascorbic acid method. The results of structural characterization showed that they were glucans with the molecular weights of 13.43 kDa and 5.97 kDa, respectively. Compared with PRP and PRP1, PRP2 exhibited the most potent antiaging activity in D-galactose-treated T lymphocytes, attributed to its shorter chain length and lower molecular weight. Furthermore, oral administration with PRP2 not only decreased the levels of senescence-associated secretory phenotype (SASP)-related inflammatory cytokines, elevated the counts of T cells, NK cells, and macrophages in the blood, but also reduced the expressions of p16 and p21 proteins in spleen tissues of naturally aged C57BL/6J mice and two fast-aging (ERCC2+/- and TERT-/-) mice. Mechanistically, PRP2 competitively bound with Keap1 and subsequently activated Nrf2/HO-1 pathway. Therefore, PRP2 could be explored as a potential candidate for treatment of age-related diseases and overall aging.
Collapse
Affiliation(s)
- Wei Xu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Hangzhou 310053, China
| | - Xue-Jian Li
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China; Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Yu-Sen Zhong
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Jia-Qi He
- Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Wei Xie
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China; Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - You-Kun Kang
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Chen-Huan Yu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China.
| |
Collapse
|
7
|
Luo Y, Bähler J, Huang Y. The Insertion Domain of Mti2 Facilitates the Association of Mitochondrial Initiation Factors with Mitoribosomes in Schizosaccharomyces pombe. Biomolecules 2025; 15:695. [PMID: 40427588 PMCID: PMC12109253 DOI: 10.3390/biom15050695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 05/03/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Translation initiation in mitochondria involves unique mechanisms distinct from those in the cytosol or in bacteria. The Schizosaccharomyces pombe mitochondrial translation initiation factor 2 (Mti2) is the ortholog of human MTIF2, which plays a vital role in synthesizing proteins in mitochondria. Here, we investigate the insertion domain of Mti2, which stabilizes its interaction with the ribosome and is crucial for efficient translation initiation. Our results show that the insertion domain is critical for the proper folding and function of Mti2. The absence of the insertion domain disrupts cell growth and affects the expression of genes encoded by mitochondrial DNA. Additionally, we show that Mti2 physically interacts with the small subunits of mitoribosomes (mtSSU), and deletion of the insertion domain dissociates mitochondrial initiation factors from the mitoribosome, reducing the efficiency of mitochondrial translation. Altogether, these findings highlight the conserved role of the insertion domain in facilitating translation initiation in fission yeast and thus reveal shared principles of mitochondrial translation initiation in both fission yeast and humans.
Collapse
Affiliation(s)
- Ying Luo
- Jiangsu Key Laboratory for Microbes and Genomics, School of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China;
- Institute of Healthy Ageing, Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK
| | - Jürg Bähler
- Institute of Healthy Ageing, Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK
| | - Ying Huang
- Jiangsu Key Laboratory for Microbes and Genomics, School of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China;
| |
Collapse
|
8
|
Huang Y, Mao J, Li Z, Wang W, Ni Z, Cai F, Tang J, Wang W, Zhang L, Zhou L, Jiang X, Wu J, Guo Q, Rui M, Huang Z, Jiang H, Wang L, Xi K, Gu Y, Chen L. Signal Converter-Based Therapy Platform Promoting Aging Bone Healing by Improving Permeability of the Mitochondrial Membrane. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2500156. [PMID: 40289881 DOI: 10.1002/adma.202500156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/19/2025] [Indexed: 04/30/2025]
Abstract
The aging microenvironment promotes persistent inflammation and loss of intrinsic regenerative capacity. These are major obstacles to effective bone tissue repair in older adults. This study aims to explore how physical thermal stimulation can effectively delay the bone marrow mesenchymal stem cells (BMSCs) aging process. Based on this, an implantable physical signal-converter platform is designed as a therapeutic system that enables stable heat signals at the bone injury site under ultrasound stimulation (US). It is found that the therapeutic platform controllably reduces the mitochondrial outer membrane permeabilization of aging BMSCs, bidirectionally inhibiting mitochondrial reactive oxygen species and mitochondrial DNA (mtDNA) leakage. The leakage ratio of mtDNA decreases by 22.7%. This effectively mitigates the activation of the cGAS-STING pathway and its downstream NF-κB signaling induced by oxidative stress in aging BMSCs, thereby attenuating the pathological advancement of chronic inflammation. Thus, it effectively restores the metabolism and osteogenic differentiation of aging BMSCs in vitro, which is further confirmed in a rat model. In the GMPG/US group, the bone mineral density increases 2-3 times at 4 weeks in the rats femoral defect model. Therefore, this ultrasound-based signal-conversion platform provides a promising strategy for aging bone defect repair.
Collapse
Affiliation(s)
- Yiyang Huang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jiannan Mao
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiang Yin, 214400, P. R. China
| | - Ziang Li
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wenbo Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Zhengxia Ni
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Feng Cai
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jincheng Tang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Lichen Zhang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Zhou
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Xinzhao Jiang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jie Wu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Qiangqiang Guo
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Min Rui
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiang Yin, 214400, P. R. China
| | - Ziyan Huang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Haochen Jiang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Lingjun Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Kun Xi
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yong Gu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Chen
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| |
Collapse
|
9
|
Mohammed SN, Jasim MH, Mahmood SH, Saleh EN, Hashemzadeh A. The role of irisin in exercise-induced muscle and metabolic health: a narrative review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04083-1. [PMID: 40167628 DOI: 10.1007/s00210-025-04083-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Irisin, a myokine released during physical exercise, has emerged as a key mediator of muscle health and metabolic regulation. This review synthesizes current evidence on how aerobic exercise stimulates irisin release and its subsequent effects, including enhanced muscle mass, strength, and recovery. Additionally, irisin promotes the browning of white adipose tissue, improving fat metabolism and glucose regulation. These adaptations position irisin as a promising therapeutic target for preventing metabolic disorders and optimizing exercise protocols. By exploring human studies and mechanistic insights, this review underscores irisin's potential to address global health challenges, such as obesity and type 2 diabetes, while advancing strategies for personalized exercise interventions.
Collapse
Affiliation(s)
- Sumaya Nadhim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Mohannad Hamid Jasim
- Biology Department, College of Education, University of Fallujah, Fallujah, Iraq
| | | | - Eman Naji Saleh
- Department of Biology, College of Education for Pure Sciences, University of Anbar, Ramadi, Iraq
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Mesnage R. Environmental Health Is Overlooked in Longevity Research. Antioxidants (Basel) 2025; 14:421. [PMID: 40298664 PMCID: PMC12024188 DOI: 10.3390/antiox14040421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/30/2025] Open
Abstract
Aging is a multifactorial process influenced by genetic predisposition and lifestyle choices. Environmental exposures are too often overlooked. Environmental pollutants-ranging from airborne particulate matter and heavy metals to endocrine disruptors and microplastics-accelerate biological aging. Oxidative stress is a major molecular initiating event, driving inflammation and toxicity across biological levels. We detail the mechanisms by which pollutants enhance reactive oxygen species (ROS) production. This oxidative stress inflicts damage on DNA, proteins, and lipids, accelerating telomere shortening, dysregulating autophagy, and ultimately driving epigenetic age acceleration. For instance, exposure to polycyclic aromatic hydrocarbons, benzene, and pesticides has been associated with increased DNA methylation age. Early-life exposures and lifestyle factors such as tobacco and alcohol consumption further contribute to accelerated biological aging. The cumulative loss of healthy life years caused by these factors can conceivably reach between 5 and 10 years per person. Addressing pollutant-induced accelerated aging through regulatory measures, lifestyle changes, and therapeutic interventions is essential to mitigate their detrimental impacts, ultimately extending healthspan and improving quality of life in aging populations.
Collapse
Affiliation(s)
- Robin Mesnage
- Department of Nutritional Sciences, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9NH, UK;
- Buchinger Wilhelmi Clinic, Wilhelm-Beck-Straße 27, 88662 Überlingen, Germany
| |
Collapse
|
11
|
Guiotto A, Pecorelli A, Draelos ZD, Gueniche A, Yatskayer M, Nelson DB. Reversing Oxinflammation Associated with Glycative Stress and Formation of Advanced Glycation End Products with a Dietary Supplement Containing Rosemary Extract. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2025; 18:34-38. [PMID: 40135177 PMCID: PMC11932104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Objective Skin aging is accelerated by glycative stress, which promotes the accumulation of advanced glycation end products (AGEs) and impairs the extracellular matrix. A randomized, double-blinded, placebo-controlled trial evaluated a dietary supplement containing rosemary extract (BioR), demonstrating tissular and visible improvements in skin quality. The data reported herein evaluated markers associated with glycative stress and AGEs from skin biopsies and tape strips obtained following dietary supplement use. Methods Female participants (N=104), aged 40 to 65 years, with moderate-to-severe skin dullness and roughness/texture, and mild-to-moderate erythema, pore size, and uneven pigmentation were randomized to BioR (n=52) or placebo ([PLB] n=52). Capsules were taken with food over 12 weeks. Subjects (n=16, BioR; n=16, PLB) underwent 3mm punch biopsies (volar upper arm) and tape stripping (16 tape strips, each; volar forearm) at baseline and 12 weeks for analysis of 4-hydroxynonenal protein adducts (4HNE [oxidative stress marker]) and AGEs. Results Immunohistochemistry and ELISA revealed that levels of 4HNE protein adducts were significantly decreased from baseline in the BioR versus PLB group (p<0.005; biopsies) and significantly decreased from baseline in the BioR group alone (p<0.05; tape strips) at 12 weeks. Significant reductions in AGEs occurred in the BioR versus PLB group (p<0.005; biopsies) at 12 weeks. No significant changes from baseline occurred in 4HNE protein adduct levels or AGEs in the PLB group. Conclusion After 12 weeks, a dietary supplement containing rosemary extract led to significant reductions in a marker associated with oxidative stress, a component of glycation, and AGEs versus placebo in skin in addition to visible improvements in skin quality.
Collapse
Affiliation(s)
- Anna Guiotto
- Drs. Guiotto and Pecorelli are with the Department of Environmental and Prevention Sciences at the University of Ferrara in Ferrara, Italy
| | - Alessandra Pecorelli
- Drs. Guiotto and Pecorelli are with the Department of Environmental and Prevention Sciences at the University of Ferrara in Ferrara, Italy
| | - Zoe Diana Draelos
- Dr. Draelos is with Dermatology Consulting Services, PLLC, in High Point, North Carolina
| | - Audrey Gueniche
- Dr. Gueniche is with L’Oréal Research and Innovation, Development in Chevilly Larue, France
| | - Margarita Yatskayer
- Ms. Yatskayer is with L’Oréal Research and Innovation, Evaluation Intelligence in Clark, New Jersey
| | - Diane B. Nelson
- Ms. Nelson is with skinbetter science, a Dermatological Beauty brand of L'Oréal USA Inc., in Phoenix, Arizona
| |
Collapse
|
12
|
Negri S, Reyff Z, Troyano-Rodriguez E, Milan M, Ihuoma J, Tavakol S, Shi H, Patai R, Jiang R, Mohon J, Boma-Iyaye J, Ungvari Z, Csiszar A, Yabluchanskiy A, Moccia F, Tarantini S. Endothelial Colony-Forming Cells (ECFCs) in cerebrovascular aging: Focus on the pathogenesis of Vascular Cognitive Impairment and Dementia (VCID), and treatment prospects. Ageing Res Rev 2025; 104:102672. [PMID: 39884362 DOI: 10.1016/j.arr.2025.102672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
Endothelial colony-forming cells (ECFCs), a unique endothelial progenitor subset, are essential for vascular integrity and repair, providing significant regenerative potential. Recent studies highlight their role in cerebrovascular aging, particularly in the pathogenesis of vascular cognitive impairment and dementia (VCID). Aging disrupts ECFC functionality through mechanisms such as oxidative stress, chronic inflammation, and cellular senescence, leading to compromised vascular repair and reduced neurovascular resilience. ECFCs influence key cerebrovascular processes, including neurovascular coupling (NVC), blood-brain barrier (BBB) integrity, and vascular regeneration, which are critical for cognitive health. Age-related decline in ECFC quantity and functionality contributes to vascular rarefaction, diminished cerebral blood flow (CBF), and BBB permeability-processes that collectively exacerbate cognitive decline. This review delves into the multifaceted role of ECFCs in cerebrovascular aging and underscores their potential as therapeutic targets in addressing age-related vascular dysfunctions, presenting new directions for mitigating the effects of aging on brain health.
Collapse
Affiliation(s)
- Sharon Negri
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zeke Reyff
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eva Troyano-Rodriguez
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Madison Milan
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jennifer Ihuoma
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sherwin Tavakol
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Helen Shi
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roland Patai
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Raymond Jiang
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Casady School, Oklahoma City, OK, USA
| | - Jonah Mohon
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma School of Science and Mathematics, OK, USA
| | - Jed Boma-Iyaye
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma School of Science and Mathematics, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Francesco Moccia
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso 86100, Italy
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
13
|
Roato I, Visca M, Mussano F. Suppressing the Aging Phenotype of Mesenchymal Stromal Cells: Are We Ready for Clinical Translation? Biomedicines 2024; 12:2811. [PMID: 39767719 PMCID: PMC11673080 DOI: 10.3390/biomedicines12122811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are involved in the maintenance and regeneration of a large variety of tissues due to their stemness and multi-lineage differentiation capability. Harnessing these advantageous features, a flurry of clinical trials have focused on MSCs to treat different pathologies, but only few protocols have received regulatory approval so far. Among the various causes hindering MSCs' efficacy is the emergence of cellular senescence, which has been correlated with specific characteristics, such as morphological and epigenetic alterations, DNA damage, ROS production, mitochondrial dysfunction, telomere shortening, non-coding RNAs, loss of proteostasis, and a peculiar senescence-associated secretory phenotype. Several strategies have been investigated for delaying or even hopefully reverting the onset of senescence, as assessed by the senescent phenotype of MSCs. Here, the authors reviewed the most updated literature on the potential causes of senescence, with a particular emphasis on the current and future therapeutic approaches aimed at reverting senescence and/or extending the functional lifespan of stem cells.
Collapse
Affiliation(s)
- Ilaria Roato
- Department of Surgical Sciences, CIR-Dental School, University of Turin, 10126 Turin, Italy; (M.V.); (F.M.)
| | | | | |
Collapse
|
14
|
von Zglinicki T. Oxidative stress and cell senescence as drivers of ageing: Chicken and egg. Ageing Res Rev 2024; 102:102558. [PMID: 39454760 DOI: 10.1016/j.arr.2024.102558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Oxidative stress and cell senescence are both important drivers of ageing and age-associated disease and disability. In vitro, they are closely interconnected in a chicken-and-egg relationship: Not only is oxidative stress an important cause of cell senescence, but senescent cells are also sources of oxidative stress, obscuring cause-effect relationships during the ageing process. We hypothesize that cell senescence is a significant cause of tissue and systemic oxidative stress during ageing. This review aims to critically summarize the available evidence for this hypothesis. After summarizing the cellular feedback mechanisms that make oxidative stress an integral part of the senescent phenotype, it critically reviews the existing evidence for a role of senescent cells as causes of oxidative stress during mammalian ageing in vivo, focussing on results from intervention experiments. It is concluded that while the available data are in agreement with this hypothesis, they are still too scarce to support a robust conclusion.
Collapse
Affiliation(s)
- Thomas von Zglinicki
- Ageing Research Laboratories, Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Health, Newcastle University, UK.
| |
Collapse
|
15
|
Xu L, Ren W, Long Y, Yang B, Chen L, Chen W, Chen S, Cao Y, Wu D, Qu J, Li H, Yu Y, Zhang A, Wang S, Wang H, Chen T, Fan G, Li Q, Chen Z. Antisenescence Expansion of Mesenchymal Stem Cells Using Piezoelectric β-Poly(vinylidene fluoride) Film-Based Culture. ACS APPLIED MATERIALS & INTERFACES 2024; 16:63207-63224. [PMID: 39503875 DOI: 10.1021/acsami.4c12725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Regenerative therapies based on mesenchymal stem cells (MSCs) show promise in treating a wide range of disorders. However, the replicative senescence of MSCs during in vitro expansion poses a challenge to obtaining a substantial quantity of high-quality MSCs. In this investigation, a piezoelectric β-poly(vinylidene fluoride) film-based culture plate (β-CP) was developed with an antisenescence effect on cultured human umbilical cord-derived MSCs. Compared to traditional tissue culture plates (TCPs) and α-poly(vinylidene fluoride) film-based culture plates, the senescence markers of p21, p53, interleukin-6 and insulin-like growth factor-binding protein-7, stemness markers of OCT4 and NANOG, and telomere length of MSCs cultured on β-CPs were significantly improved. Additionally, MSCs at passage 18 cultured on β-CPs showed significantly better multipotency and pro-angiogenic capacities in vitro, and higher wound healing abilities in a mouse model. Mechanistically, β-CPs rejuvenated senescent MSCs by improving mitochondrial functions and mitigating oxidative and glycoxidative stresses. Overall, this study presents β-CPs as a promising approach for efficient and straightforward antisenescence expansion of MSCs while preserving their stemness, thereby holding great potential for large-scale production of MSCs for clinical application in cell therapies.
Collapse
Affiliation(s)
- Liuyue Xu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenxiang Ren
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yaoying Long
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bianlei Yang
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Chen
- Department of Hematology, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Wenlan Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Siyi Chen
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yulin Cao
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Wu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiao Qu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - He Li
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yali Yu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anyuan Zhang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shan Wang
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongxiang Wang
- Department of Hematology, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Ting Chen
- Hubei Engineering Research Center for Application of Extracellular Vesicles, Hubei University of Science and Technology, Xianning 437100, China
| | - Guifen Fan
- School of Optical and Electronic Information, Key Lab of Functional Materials for Electronic Information(B), MOE, Huazhong University of Science and Technology, Wuhan 430074, China
- Wenzhou Advanced Manufacturing Institute, Huazhong University of Science and Technology, Wenzhou 325035, China
| | - Qiubai Li
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Application of Extracellular Vesicles, Hubei University of Science and Technology, Xianning 437100, China
| | - Zhichao Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
16
|
Pollard CA, Saito ER, Burns JM, Hill JT, Jenkins TG. Considering Biomarkers of Neurodegeneration in Alzheimer's Disease: The Potential of Circulating Cell-Free DNA in Precision Neurology. J Pers Med 2024; 14:1104. [PMID: 39590596 PMCID: PMC11595805 DOI: 10.3390/jpm14111104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), are a growing public health crisis, exacerbated by an aging global population and the lack of effective early disease-modifying therapies. Early detection of neurodegenerative disorders is critical to delaying symptom onset and mitigating disease progression, but current diagnostic tools often rely on detecting pathology once clinical symptoms have emerged and significant neuronal damage has already occurred. While disease-specific biomarkers, such as amyloid-beta and tau in AD, offer precise insights, they are too limited in scope for broader neurodegeneration screening for these conditions. Conversely, general biomarkers like neurofilament light chain (NfL) provide valuable staging information but lack targeted insights. Circulating cell-free DNA (cfDNA), released during cell death, is emerging as a promising biomarker for early detection. Derived from dying cells, cfDNA can capture both general neurodegenerative signals and disease-specific insights, offering multi-layered genomic and epigenomic information. Though its clinical potential remains under investigation, advances in cfDNA detection sensitivity, standardized protocols, and reference ranges could establish cfDNA as a valuable tool for early screening. cfDNA methylation signatures, in particular, show great promise for identifying tissue-of-origin and disease-specific changes, offering a minimally invasive biomarker that could transform precision neurology. However, further research is required to address technological challenges and validate cfDNA's utility in clinical settings. Here, we review recent work assessing cfDNA as a potential early biomarker in AD. With continued advances, cfDNA could play a pivotal role in shifting care from reactive to proactive, improving diagnostic timelines and patient outcomes.
Collapse
Affiliation(s)
- Chad A. Pollard
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
- Resonant, Heber, UT 84032, USA
| | | | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS 66205, USA
| | - Jonathon T. Hill
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Timothy G. Jenkins
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
- Resonant, Heber, UT 84032, USA
| |
Collapse
|
17
|
Tauil RB, Golono PT, de Lima EP, de Alvares Goulart R, Guiguer EL, Bechara MD, Nicolau CCT, Yanaguizawa Junior JL, Fiorini AMR, Méndez-Sánchez N, Abenavoli L, Direito R, Valente VE, Laurindo LF, Barbalho SM. Metabolic-Associated Fatty Liver Disease: The Influence of Oxidative Stress, Inflammation, Mitochondrial Dysfunctions, and the Role of Polyphenols. Pharmaceuticals (Basel) 2024; 17:1354. [PMID: 39458995 PMCID: PMC11510109 DOI: 10.3390/ph17101354] [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: 09/26/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is a clinical-pathological scenario that occurs due to the accumulation of triglycerides in hepatocytes which is considered a significant cause of liver conditions and contributes to an increased risk of death worldwide. Even though the possible causes of MAFLD can involve the interaction of genetics, hormones, and nutrition, lifestyle (diet and sedentary lifestyle) is the most influential factor in developing this condition. Polyphenols comprise many natural chemical compounds that can be helpful in managing metabolic diseases. Therefore, the aim of this review was to investigate the impact of oxidative stress, inflammation, mitochondrial dysfunction, and the role of polyphenols in managing MAFLD. Some polyphenols can reverse part of the liver damage related to inflammation, oxidative stress, or mitochondrial dysfunction, and among them are anthocyanin, baicalin, catechin, curcumin, chlorogenic acid, didymin, epigallocatechin-3-gallate, luteolin, mangiferin, puerarin, punicalagin, resveratrol, and silymarin. These compounds have actions in reducing plasma liver enzymes, body mass index, waist circumference, adipose visceral indices, lipids, glycated hemoglobin, insulin resistance, and the HOMA index. They also reduce nuclear factor-KB (NF-KB), interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), blood pressure, liver fat content, steatosis index, and fibrosis. On the other hand, they can improve HDL-c, adiponectin levels, and fibrogenesis markers. These results show that polyphenols are promising in the prevention and treatment of MAFLD.
Collapse
Affiliation(s)
- Raissa Bulaty Tauil
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Paula Takano Golono
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Ricardo de Alvares Goulart
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Claudia C. T. Nicolau
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - José Luiz Yanaguizawa Junior
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Adriana M. R. Fiorini
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy;
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| | - Vitor Engrácia Valente
- Autonomic Nervous System Center, School of Philosophy and Sciences, São Paulo State University, Marília 17525-902, São Paulo, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil;
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
- Research Coordination, UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| |
Collapse
|