1
|
Mssillou I, El Abdali Y, Amrati FEZ, Lim A, Khalid A, Makeen HA, Albratty M, Lefrioui Y, Louafi B, Bousta D. Unlocking the therapeutic potential of protocatechualdehyde: pharmacological applications and mechanism insights. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04267-9. [PMID: 40493248 DOI: 10.1007/s00210-025-04267-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Accepted: 05/05/2025] [Indexed: 06/12/2025]
Abstract
Protocatechualdehyde (PCA) is a phenolic aldehyde with many pharmacological features. It is composed of two hydroxyl groups attached to C3 and C4 of the benzene ring. This review aims to discover the source of PCA, discuss its pharmacological efficiency, investigate its implication in related mechanistic pathways, approve safety, and elaborate scientific foundation for future studies. This compound has been detected in various plants, especially Salviae Miltiorrhiza Bumge. which is considered as the main source. PCA has been confirmed to possess strong antioxidant activity by inhibiting free radicals in different in vivo and in vitro models. Interestingly, PCA has an important anticancer potential and is involved in various mechanistic pathways, such as apoptosis in different cancer cells, degradation of cyclin expression, cell cycle arrest, and downregulated regulator proteins. Furthermore, this molecule has a clear effect in reducing inflammation via inhibition of cyclooxygenases-2 (COX-2), tumor necrosis factor-α (TNF-α), and interleukins (IL-6, IL-1β), reduction of NLRP3, GSDMD, and caspase-1 expression, activation of the Nrf2 pathway, and suppression of the ERK/c-Fos/NFATc1 signaling axis. In addition, the antimicrobial proprieties and antibiofilm activity of PCA have been proved on Micrococcus luteus, MRSA, Ralstonia solanacearum, and Yersinia enterocolitica. Also, it is highly reported in neurological illness and cardiovascular treatment. Particularly, PCA exhibits significant anti-atherosclerosis activity, protects skin, and is valuable for endothelial function. Exploring the pharmacokinetic applications, toxicity and recent advances in researches on PCA, we have developed the scientific insights required for future research for the accurate exploitation of PCA in future pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Ibrahim Mssillou
- National Agency of Medicinal and Aromatic Plants, 34025, Taounate, Morocco.
| | - Youness El Abdali
- Laboratory of Biotechnology, Health, Agrofood and Environment (LBEAS), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, 30000, Fez, Morocco
| | - Fatima Ez-Zahra Amrati
- Laboratory of Cell Biology and Molecular Genetics (LBCGM), Department of Biology, Faculty of Sciences, Ibn Zohr University, Agadir, Souss Massa, Morocco
| | - Adrian Lim
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Asaad Khalid
- Health Research Center, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia.
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, Faculty of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia
| | - Youssra Lefrioui
- National Agency of Medicinal and Aromatic Plants, 34025, Taounate, Morocco
- Laboratory of Functional Ecology and Environmental Engineering, Faculty of Sciences and Technology, Sidi Mohamed Ben Abdellah University, 30000, Fez, Morocco
| | - Boutaina Louafi
- National Agency of Medicinal and Aromatic Plants, 34025, Taounate, Morocco
- Laboratory of Biotechnology Environment Agrofood and Health (LBEAS), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, 30000, Fez, Morocco
| | - Dalila Bousta
- National Agency of Medicinal and Aromatic Plants, 34025, Taounate, Morocco
| |
Collapse
|
2
|
Zhou TY, Guo YY, Jing QQ, Wei MY, Xu WF, Gu YC, Shao CL. Semisynthesis and biological evaluation of 17-hydroxybrevianamide N derivatives as anti-inflammatory agents by mediating NF-κB and MAPK signaling pathways. Eur J Med Chem 2025; 290:117541. [PMID: 40174263 DOI: 10.1016/j.ejmech.2025.117541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
Chronic inflammation is a trigger for many diseases that affect approximately 10-20 % of the population around the world. Herein, (±)-17-hydroxybrevianamide N (1) was isolated from the fungus Aspergillus sp. (CHNSCLM-0151) and exhibited strong inhibitory activity against nitric oxide (NO) in lipopolysaccharide (LPS)-induced RAW264.7 cell. A series of new derivatives (±)-3-(±)-29 was semisynthesized by structural modification of the imide, phenolic hydroxyl, and carbonyl groups from the natural product (±)-1. The results of anti-inflammatory activity demonstrated that (±)-4, (±)-6, (±)-9, (±)-22, (±)-23, and (±)-24 exhibited obviously NO inhibitory (P < 0.0001) in LPS-stimulated RAW264.7 cells. To further investigate the relationship between chirality and activity, the enantiomers of the above six compounds were obtained by chiral resolution. As expected, the bioactivity results indicated stereoselectivity in the anti-inflammatory effect among the different isomers. In particular, compound (+)-4S-23 inhibited NO concentration with an IC50 value of 0.5 μM, demonstrating 3-fold greater potency compared to its (R)-enantiomer, and achieving 40-fold superior potency over the positive control NG-monomethyl-l-arginine (L-NMMA). This compound demonstrated suppression of TNF-α (25.7 ± 1.5 %), IL-6 (54.5 ± 3.9 %) and IL-1β (92.9 ± 4.1 %) production at 2 μM. More importantly, mechanistic investigations revealed that (+)-4S-23 (0.2 μM) modulates the MAPK signaling pathway, specifically downregulating phosphorylation of p38, ERK, and JNK. Furthermore, (+)-4S-23 also exhibited potent inhibitory activity against the NF-κB pathway by suppressing the phosphorylation of IκB-α and blocking nuclear translocation of phosphorylated p65. Notably, these findings position (+)-4S-23 as a promising candidate for development as a novel anti-inflammatory therapeutic targeting both MAPK and NF-κB signaling nodes.
Collapse
Affiliation(s)
- Tian-Yi Zhou
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Yang-Yang Guo
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Qian-Qian Jing
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Mei-Yan Wei
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Wei-Feng Xu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China; School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, People's Republic of China.
| | - Yu-Cheng Gu
- Syngenta Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, UK
| | - Chang-Lun Shao
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China.
| |
Collapse
|
3
|
Wu J, Zhang L, Zhao Z, Liu Y, Li Z, Feng X, Zhang L, Yao X, Du J, Chen L, Zhou Z. Advancing T-cell immunotherapy for cellular senescence and disease: Mechanisms, challenges, and clinical prospects. Ageing Res Rev 2025; 109:102783. [PMID: 40412763 DOI: 10.1016/j.arr.2025.102783] [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: 03/26/2025] [Revised: 05/12/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Cellular senescence is a complex biological process with a dual role in tissue homeostasis and aging-related pathologies. Accumulation of senescent cells promotes chronic inflammation, tissue dysfunction, age-related diseases, and tumor suppression. Recent advancements in immunotherapy have positioned T cell-based approaches as precision tools for the targeted clearance of senescent cells, offering a novel avenue for anti-aging interventions. This review explores the molecular mechanisms underlying cellular senescence, focusing on its immunogenic features and interactions with T cells, including T-cell activation, antigen recognition, modulation of tumor microenvironment (TME), and immune evasion strategies. Innovations such as chimeric antigen receptor (CAR)-T cells, immune checkpoint therapies, and SASP-neutralizing approaches are highlighted as breakthrough strategies for enhancing senescent cell eradication. The integration of multi-omics and artificial intelligence is further catalyzing the development of personalized therapies to amplify immune surveillance and tissue rejuvenation. Clinically, T cell-based interventions hold promise for mitigating age-related pathologies and extending healthspan, yet challenges remain in optimizing target specificity, countering immunosuppressive niches, and overcoming immune senescence in aging populations. This review synthesizes current advances and challenges, highlighting the potential of T cell immunotherapy as a cornerstone of anti-aging medicine and emphasizing the need for interdisciplinary innovation to translate preclinical findings into transformative therapies for aging and age-related diseases.
Collapse
Affiliation(s)
- Jizhun Wu
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lu Zhang
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Zihan Zhao
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yuping Liu
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhengxing Li
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiaohang Feng
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lin Zhang
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiang Yao
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China.
| | - Zhuolong Zhou
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Biomedical Sciences, College of Medicine and Veterinary Medicine, Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
4
|
Xie G, Okuda S, Gao JY, Wu T, Jeong J, Lu KP, Zhou XZ. The Central Role of Pin1 in Age-Related Cancer Signaling Pathways. Semin Cancer Biol 2025:S1044-579X(25)00072-0. [PMID: 40412492 DOI: 10.1016/j.semcancer.2025.05.011] [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: 01/17/2025] [Revised: 04/05/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
The prolyl-isomerase Pin1 is a unique enzyme that catalyzes cis-trans isomerization of phosphorylated Ser/Thr-Pro motifs. These motifs are present in many proteins, where isomerization of the typically rigid prolyl-peptide bond can lead to conformational changes, and subsequently regulate activity, stability, or localization. The specificity of Pin1 for phosphorylated motifs allows it to serve as a master regulator of proteins after phosphorylation, adding an additional layer of regulation to intricately control cellular signaling. As such, Pin1 plays an expansive role in numerous cancer and age-related signaling pathways, and is recognized as a major driver of cancer and promising therapeutic target. In this review, we discuss the role of Pin1 in regulation of age-related cancer signaling pathways, and we highlight the early development and current landscape of Pin1 inhibitors, and the prospect of Pin1 inhibition for cancer therapy.
Collapse
Affiliation(s)
- George Xie
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Sho Okuda
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Jing-Yan Gao
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada; Department of Chemistry, Western University, London, ON N6A 5C1, Canada
| | - Timothy Wu
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Jessica Jeong
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON N6G 2V4, Canada.
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada; Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada; Lawson Health Research Institute, Western University, London, ON N6C 2R5, Canada.
| |
Collapse
|
5
|
Bae WJ, Lee SI. Activating Transcription Factor 3 (ATF3) Regulates Cellular Senescence and Osteoclastogenesis via STAT3/ERK and p65/AP-1 Pathways in Human Periodontal Ligament Cells. Int J Mol Sci 2025; 26:4959. [PMID: 40430099 PMCID: PMC12112613 DOI: 10.3390/ijms26104959] [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: 03/26/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Oral cellular aging plays a critical role in the pathogenesis of chronic periodontitis and alveolar bone resorption. Although activating transcription factor 3 (ATF3) has been implicated as a senescence-associated factor, its specific role in periodontal ligament cell (PDLC) senescence remains unclear. Human PDLCs were exposed to lipopolysaccharide (LPS, 1 μg/mL) and nicotine (5 mM) for 3 days to induce senescence. ATF3 expression was silenced using siRNA. The expression of senescence-associated secretory phenotype (SASP) factors (IFNγ, IL6, IL8, TNFα, and IL1β) and the secretion of nitric oxide (NO) and prostaglandin E2 (PGE2) were assessed by RT-PCR and immunoassay. Conditioned media (CM) from these cells were applied to mouse bone marrow macrophages (BMMs) to evaluate osteoclast differentiation and bone resorption. In addition, key signaling pathways, including STAT3, ERK, NF-κB (p65), and AP-1, were investigated by Western blotting and immunofluorescence. ATF3 knockdown markedly reduced the LPS/nicotine-induced expression of SASP factors and decreased NO and PGE2 levels. CM from ATF3-silenced PDLCs markedly inhibited osteoclast differentiation, as evidenced by reduced tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells and diminished bone resorption. Moreover, ATF3 inhibition led to a decreased RANKL/OPG ratio and attenuated the phosphorylation of STAT3 and ERK, along with the reduced nuclear translocation of p65 and AP-1 components. These findings suggest that ATF3 plays a critical role in mediating cellular senescence and osteoclastogenesis in PDLCs. Targeting ATF3 may represent a novel therapeutic strategy for managing age-related oral diseases, such as chronic periodontitis.
Collapse
Affiliation(s)
- Won-Jung Bae
- Department of Pharmacology, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
| | - Sang-Im Lee
- Department of Dental Hygiene, College of Health Science, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
6
|
Webster R, Quintana M, Yu B, Fluke S, Kafri R, Derry WB. CDK-4 regulates nucleolar size and metabolism at the cost of late-life fitness in C. elegans. Heredity (Edinb) 2025:10.1038/s41437-025-00769-7. [PMID: 40383750 DOI: 10.1038/s41437-025-00769-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
Studies on aging have centered on two molecular pathways: CDK4/6 and insulin/mTORC1. These pathways are thought to influence aging through distinct mechanisms: mTORC1 by reprogramming systemic metabolism, and CDK4 through p16-mediated senescence and inflammatory signaling (SASP). Here, we investigate the connection between aging and CDK4 in Caenorhabditis elegans, an organism lacking both p16 and SASP. Using a conditional degradation system, we demonstrate that CDK-4 inhibition in C. elegans phenocopies its aging-related functions observed in mammals. Worms with depleted CDK-4 exhibited accelerated aging phenotypes, including reduced lifespan, decreased motility, increased yolk accumulation, and earlier onset of senescence. At the physiological level, CDK4-inhibited worms show substantial metabolic shifts; including enhanced protein synthesis, elevated ATP production, and increased fat accumulation. These metabo-aging phenotypes occur independently of mTORC1, instead operating through the canonical CDK-4 effectors LIN-35 (Rb) and EFL-1 (E2F).
Collapse
Affiliation(s)
- Rachel Webster
- Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Developmental, Stem Cell and Cancer Biology Program, PEter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Maria Quintana
- Developmental, Stem Cell and Cancer Biology Program, PEter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Bin Yu
- Developmental, Stem Cell and Cancer Biology Program, PEter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stacey Fluke
- Developmental, Stem Cell and Cancer Biology Program, PEter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ran Kafri
- Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada.
- Developmental, Stem Cell and Cancer Biology Program, PEter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - W Brent Derry
- Developmental, Stem Cell and Cancer Biology Program, PEter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
7
|
Hu T, Wang T, Luo X, Hu Z. Association between the C-reactive protein-albumin-lymphocyte index and all-cause mortality in Chinese older adults: a national cohort study based on CLHLS from 2014 to 2018. Front Nutr 2025; 12:1575470. [PMID: 40444244 PMCID: PMC12119284 DOI: 10.3389/fnut.2025.1575470] [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: 02/14/2025] [Accepted: 04/25/2025] [Indexed: 06/02/2025] Open
Abstract
Background The C-reactive protein-albumin-lymphocyte (CALLY) index, a novel inflammation-immune-nutritional biomarker, has not been comprehensively evaluated for mortality risk prediction in older populations. Here, we investigate the relationship between the CALLY index and all-cause mortality in Chinese adults aged ≥ 60 years. Methods Data were obtained from the 2014 to 2018 wave of the Chinese Longitudinal Healthy Longevity Survey (CLHLS). Upon applying a natural logarithmic transformation to the CALLY index, the lnCALLY was stratified into tertiles. Kaplan-Meier analysis and the log-rank test were employed to assess the cumulative survival probability across lnCALLY-stratified older adults populations. Cox proportional hazards regression was utilized to investigate the association between lnCALLY and all-cause mortality. Receiver operating characteristic (ROC) curves and area under the curve (AUC) values were conducted to evaluate the predictive capacity of lnCALLY for all-cause mortality. Restricted cubic splines (RCS) with four knots were applied to explore the potential non-linear dose-response association of lnCALLY with all-cause mortality. Subgroup analyses and sensitivity analyses were conducted to ensure validity. Results A total of 1,738 older adults participants were included in this cohort. Over a median follow-up of 3.3 years, 580 deaths (33.3%) occurred. The multivariable Cox regression demonstrated that the highest lnCALLY tertile was associated with a 40% reduced mortality risk compared to the lowest tertile [adjusted hazard ratio (HR) = 0.60, 95% confidence interval (CI): 0.49-0.73]. Kaplan-Meier curves revealed significantly higher survival probabilities in individuals with elevated lnCALLY (P < 0.001). Time-dependent ROC analysis showed that the AUC of lnCALLY for predicting all-cause mortality at 1-, 2-, and 3-year were 0.751, 0.746, and 0.762, respectively. RCS demonstrated an approximate "L"-shaped negative correlation between lnCALLY and all-cause mortality (P overall < 0.001, P non- linearity = 0.364). Subgroup and sensitivity analyses confirmed robustness, with no significant interactions observed across demographic or clinical strata. Conclusion These findings suggest that the CALLY index serves as a practical prognostic biomarker for monitoring survival in older populations, underscoring the interplay of inflammation, immunity, and nutrition in aging-related mortality.
Collapse
Affiliation(s)
- Tian Hu
- Department of Rehabilitation Medicine, JieYang People’s Hospital, Jieyang, Guangdong, China
| | - Taotao Wang
- Department of Rehabilitation Medicine, Zhongshan People’s Hospital, Zhongshan, Guangdong, China
| | - Xiaojing Luo
- Department of Rehabilitation Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zheng Hu
- Department of Spine Surgery, JieYang People’s Hospital, Jieyang, Guangdong, China
| |
Collapse
|
8
|
Wang Y, Liu F, Tong R, He Z, Fang Q, Feng J, An H, Liu J. Predicting incident type 2 diabetes in a Japanese cohort: an 8-year analysis of the NAGALA database. Front Endocrinol (Lausanne) 2025; 16:1465032. [PMID: 40444231 PMCID: PMC12119281 DOI: 10.3389/fendo.2025.1465032] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 04/25/2025] [Indexed: 06/02/2025] Open
Abstract
Objective Using data from the NAGALA database, this retrospective cohort study set out to identify the predictive markers for incident Type 2 Diabetes Mellitus (T2DM), with a particular focus on the non-diabetic Japanese population. Methods We examined the data from a cohort of 15,464 individuals (with a male representation of 54.5% and an average age of (43.71 ± 8.90 years) sourced from the NAGALA (NAfld in the Gifu Area Longitudinal Analysis) research which was a longitudinal study at Medical Health Checkup Center of Murakami Memorial Hospital. The analysis focused on the incidence of T2DM from 2004 to 2012. Baseline demographic, anthropometric, biochemical, and lifestyle data were collected. All participants were not type 2 diabetic at baseline. The diagnosis of T2DM was confirmed by HbA1c >= 48mmol/mol, fasting plasma glucose >= 126mg/dL, or diabetes reported by oneself. Multivariate analysis was performed after univariate Cox regression analysis was used to find early determinants of T2DM incidence. The ability of individual components and a composite risk score to discriminate was assessed using Receiver Operating Characteristic (ROC) curves. Results Over an average follow-up duration of 2207.82 ± 1379.73 days, 262 patients (1.7%) had the onset of T2DM. Following the removal of confounding variables we found that age (HR=1.03,95%CI 1.01∼1.04, P=0.001), waist circumference(HR=1.05,95%CI 1.03∼1.06, P<0.001), alanine transaminase(HR=1.01,95%CI 1.00∼1.01, P=0.045), glycated hemoglobin (HbA1c) (HR=24.30,95%CI 15.69∼37.63,P<0.001), fasting plasma glucose(HR=1.10,95%CI 1.07∼1.12, P<0.001), the presence of fatty liver(HR=1.86,95%CI 1.37∼2.53, P<0.001), current smoking(HR=1.61,95%CI 1.16∼2.23, P=0.004), and heavy alcohol consumption(HR=1.79,95%CI 1.06∼2.99, P=0.028) were identified as independent risk factors for T2DM(all P < 0.05), while high-density lipoprotein cholesterol (HDL-C) exhibited a protective effect (HR=0.98,95%CI 0.97∼1.00, P=0.010). The area under the Receiver Operating Characteristic (ROC) curve for individual factors ranged from 0.53 to 0.83, with the highest value for HbA1c. A combined risk model incorporating these factors including age, waist circumference, alanine transaminase, HbA1c, fasting plasma glucose, the presence of fatty liver, current smoking, heavy alcohol consumption, 1/HDL-C achieved an AUC of 0.90 (95% CI 0.88-0.92, P < 0.001), signifying robust discriminatory ability. At a predictive probability threshold of >0.017, the model exhibited sensitivity and specificity of 0.863 and 0.828, respectively. Conclusion Current research has underscored the significance of a multifaceted approach to the prevention of T2DM, which includes early intervention targeting modifiable risk factors such as obesity, unhealthy alcohol use, and smoking, in conjunction with the monitoring of key metabolic markers like HbA1c and liver enzymes.
Collapse
Affiliation(s)
- Yangchun Wang
- Department of Endocrinology, Nanjing Meishan Hospital, Nanjing, China
| | - Fei Liu
- Department of General Surgery, Nanjing Meishan Hospital, Nanjing, China
| | - Ruixiang Tong
- Department of Endocrinology, Nanjing Meishan Hospital, Nanjing, China
| | - Zhonghua He
- Department of Endocrinology, Nanjing Meishan Hospital, Nanjing, China
| | - Qin Fang
- Department of Endocrinology, Nanjing Meishan Hospital, Nanjing, China
| | - Jie Feng
- Department of Pharmacy, Nanjing Meishan Hospital, Nanjing, China
| | - Hongliang An
- Department of Pharmacy, Nanjing Meishan Hospital, Nanjing, China
| | - Junjun Liu
- Department of Psychiatry, Nanjing Meishan Hospital, Nanjing, China
| |
Collapse
|
9
|
Hu L, Li J, Tang Z, Gong P, Chang Z, Yang C, Ma T, Jiang S, Yang C, Zhang T. How does biological age acceleration mediate the associations of obesity with cardiovascular disease? Evidence from international multi-cohort studies. Cardiovasc Diabetol 2025; 24:209. [PMID: 40369582 PMCID: PMC12079857 DOI: 10.1186/s12933-025-02770-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Recent basic biological research found that obesity accelerates biological aging and increases cardiovascular disease (CVD) risk. However, there is still a lack of real-world population evidence. This study aimed to explore the potential mediation roles of biological age acceleration in the associations between different dimensions of obesity characterization and incident CVD. METHODS This international multi-cohort study included participants aged over 45 years with 3 waves longitudinal data from China Health and Retirement Longitudinal Study (CHARLS). China Health and Nutrition Survey (CHNS) was used to develop Klemera-Doubal method-biological age (KDM-BA), and the validation analysis was performed in UK Biobank (UKB) and Hongguang Elderly Health Examination Cohort (HEHEC). Obesity indices including body mass index (BMI), waist circumference (WC), waist height ratio (WtHR), body roundness index (BRI) for body shape; Chinese visceral adiposity index (CVAI), lipid accumulation product (LAP) for visceral fat accumulation; triglyceride-glucose index (TyG) and its derivatives (TyG-BMI, TyG-WC, TyG-WtHR) for metabolic function were used to measure obesity across different dimensions. Biological age acceleration was evaluated by the classic KDM-BA acceleration (KDM-BAacc). Causal mediation analyses assessed the role of biological age acceleration in mediating obesity and incident CVD. RESULTS In CHARLS, the median follow-up period was 9.00 years, with a baseline age of 58 (52, 65) years. Obesity, KDM-BAacc, and CVD were all significantly associated with each other. For each 1-year increase in KDM-BAacc, the risk of incident stroke, heart disease and CVD increased by 68% (OR 1.68, 95% CI 1.35-2.09), 35% (OR 1.35, 95% CI 1.15-1.59), and 44% (OR 1.44, 95% CI 1.25-1.65), respectively. KDM-BAacc mediated the associations between BMI, WC, WtHR, BRI, CVAI, LAP, TyG-BMI, TyG-WC, TyG-WtHR, with CVD, with the mediation proportions ranging from 10.03 to 25.46%. However, the mediating effect was significant mostly in middle-aged individuals aged 45-65 years. Furthermore, sex differences existed in the mediation mechanisms. Biological age acceleration strongly mediated body shape indices and incident CVD in males, whereas in females, it predominantly mediated visceral fat accumulation and metabolic function dimensions with incident CVD. Similar main results were found in UKB and HEHEC. CONCLUSIONS Biological age acceleration partially mediates the relationship between obesity and incident CVD. This temporal evidence firstly validated the mediation pathway based on international cohorts, emphasizing the importance of addressing biological aging processes in population aged 45-65 years while providing sex-specific obesity intervention strategies to prevent CVD.
Collapse
Affiliation(s)
- Lin Hu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jiayuan Li
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zihuan Tang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Peng Gong
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zongqi Chang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Chen Yang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Tianpei Ma
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Shuang Jiang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Chunxia Yang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Tao Zhang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
10
|
Tong X, Wang F, Liu M. Serum sodium within the normal range and its U-shaped relationship with biological aging in U.S. adults. Front Nutr 2025; 12:1589962. [PMID: 40406152 PMCID: PMC12095086 DOI: 10.3389/fnut.2025.1589962] [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: 03/08/2025] [Accepted: 04/23/2025] [Indexed: 05/26/2025] Open
Abstract
Background This cross-sectional study evaluated the correlation between serum sodium levels (135-145 mmol/L) and biological aging in U.S. adults. Biological age, derived from multi-system biomarkers, provides a more accurate assessment of aging than chronological age. Hydration balance, reflected by serum sodium, may modulate age-related diseases and mortality, but its link to biological aging remains underexplored. Methods Using NHANES data (1999-2018), we focused on normonatremic adults (≥20 years). The final cohort included 18,301 participants. Biological age was estimated using the Klemera and Doubal method, and ∆age (biological age-chronological age) was calculated. Associations were assessed using multivariate regression, generalized additive models, and threshold analysis. Subgroup analyses were conducted for variations across different populations. Results Nonlinear analysis revealed a U-shaped relationship between serum sodium and biological age. The lowest biological age occurred at 139.3 mmol/L: each 1 mmol/L increase below this threshold was associated with a reduction of 0.10 years in biological age (95% CI: -0.15, -0.05), whereas values above it showed a 0.08-year increase (95% CI: 0.04, 0.13). For ∆age, a negative association was observed below 141.2 mmol/L, with each increase linked to a 0.07-year decrease (95% CI: -0.10, -0.04). Subgroup analyses revealed significant interactions in diabetic and smoking populations. Conclusion Maintaining serum sodium levels within an optimal range (138-142 mmol/L) may help delay biological aging. Hydration management may serve as a modifiable factor for healthy aging, particularly in high-risk groups such as individuals with diabetes or tobacco use.
Collapse
Affiliation(s)
| | | | - Mengxue Liu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
11
|
Revankar NA, Anusha S, Muthukumar SP, Negi PS. Synbiotic pineapple beverage increases life span in Caenorhabditis elegans, ameliorates cognitive impairment, and restores gut microbiome diversity in D-galactose-induced aged C57BL/6 mice. Biogerontology 2025; 26:99. [PMID: 40317421 DOI: 10.1007/s10522-025-10239-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 04/09/2025] [Indexed: 05/07/2025]
Abstract
The incidence of age-associated ailments has increased proportionately with the expansion of the aging demographic. This study aimed to evaluate the anti-aging potential of synbiotic pineapple beverage formulated with 100% pineapple juice, 1% inulin, and Lacticaseibacillus rhamnosus ATCC 53103 (10 log CFU) in Caenorhabditis elegans and D-galactose age-induced mice. The synbiotic juice-treated nematodes exhibited a 24.52% increase in their lifespan, accompanied by lower levels of reactive oxygen species and improved structural functions. In vivo studies demonstrated that synbiotic treatment positively influences age-induced mice's cerebellar function and spatial memory. Additionally, the synbiotic beverage containing 8-10 log CFU of Lacticaseibacillus rhamnosus showed a protective effect against hippocampal neuron damage. The control group displayed a higher Firmicutes/Bacteroides (F/B) ratio, whereas the significantly lower F/B ratio in the diseased groups indicated a reversal of microbial imbalance caused by D-galactose exposure. Furthermore, the consumption of synbiotic beverage mitigated telomere shortening in aged mice. The results highlight the anti-aging effects of a pineapple beverage formulated with Lacticaseibacillus rhamnosus and inulin as a synbiotic intervention. This study suggests that dietary interventions incorporating prebiotics and probiotics may serve as promising strategy for combating age-related disorders and promoting healthy aging.
Collapse
Affiliation(s)
- Neelam Anil Revankar
- Department of Fruit and Vegetable Technology, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Siddaraju Anusha
- Department of Fruit and Vegetable Technology, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Serva Peddha Muthukumar
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Pradeep Singh Negi
- Department of Fruit and Vegetable Technology, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
12
|
Mohammadi S, Ghaderi S, Fatehi F, Kalra S, Batouli SAH. Pathological Aging of Patients With Amyotrophic Lateral Sclerosis: A Preliminary Longitudinal Study. Brain Behav 2025; 15:e70484. [PMID: 40329780 PMCID: PMC12056362 DOI: 10.1002/brb3.70484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/12/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
OBJECTIVE This longitudinal study investigated pathological brain aging in amyotrophic lateral sclerosis (ALS) by evaluating disparities between chronological age and deep learning-derived brain structure age (BSA) and exploring associations with cognitive and functional decline. METHODS Ten limb-onset ALS patients (seven males) and 10 demographically matched healthy controls (HCs) underwent structural magnetic resonance imaging (sMRI) and cognitive assessments at baseline and follow-up. The BSA was estimated using the validated volBrain platform. Cognitive domains (language, verbal fluency, executive function, memory, and visuospatial skills) and global cognition (Persian adaptive Edinburgh Cognitive and Behavioral ALS Screen [ECAS] total score) were assessed along with functional status (ALSFRS-R). RESULTS ALS patients exhibited significant BSA-chronological age disparities at baseline (Δ = +7.31 years, p = 0.009) and follow-up (Δ = +8.39 years, p = 0.003), with accelerated BSA progression over time (p = 0.004). The HCs showed no such disparities (p = 0.931). Longitudinal BSA increases were correlated with executive function decline (r = -0.651, p = 0.042). Higher education predicted preserved language (r = 0.831, p = 0.003) and verbal fluency (r = 0.738, p = 0.015). ALSFRS-R decline paralleled visuospatial (r = 0.642, p = 0.045) and global cognitive deterioration (r = 0.667, p = 0.035). CONCLUSIONS ALS is characterized by accelerated structural brain aging that progresses independently of chronological age and is correlated with executive dysfunction. Education may mitigate cognitive decline, while motor functional deterioration aligns with visuospatial and global cognitive impairments. BSA has emerged as a potential biomarker for tracking pathological aging trajectories in ALS, warranting validation using larger cohorts.
Collapse
Affiliation(s)
- Sana Mohammadi
- Neuromuscular Research Center, Department of NeurologyShariati Hospital, Tehran University of Medical SciencesTehranIran
| | - Sadegh Ghaderi
- Neuromuscular Research Center, Department of NeurologyShariati Hospital, Tehran University of Medical SciencesTehranIran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of NeurologyShariati Hospital, Tehran University of Medical SciencesTehranIran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Sanjay Kalra
- Neuroscience and Mental Health InstituteUniversity of AlbertaEdmontonAlbertaCanada
- Division of Neurology, Department of MedicineUniversity of AlbertaEdmontonAlbertaCanada
| | - Seyed Amir Hossein Batouli
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| |
Collapse
|
13
|
Osburn SC, Smith ME, Wahl D, LaRocca TJ. Novel effects of reverse transcriptase inhibitor supplementation in skeletal muscle of old mice. Physiol Genomics 2025; 57:308-320. [PMID: 40062980 DOI: 10.1152/physiolgenomics.00115.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/08/2024] [Accepted: 02/15/2025] [Indexed: 04/01/2025] Open
Abstract
Aging is the primary risk factor for the development of many chronic diseases, including dementias, cardiovascular disease, and diabetes. There is significant interest in identifying novel "geroprotective" agents, including by repurposing existing drugs, but such treatments may affect organ systems differently. One current example is the nucleoside reverse transcriptase inhibitor 3TC, which has been increasingly studied as a potential gerotherapeutic. Recent data suggest that 3TC may reduce inflammation and improve cognitive function in older mice; however, the effects of 3TC on other tissues in aged animals are less well characterized. Here, we use transcriptomics (RNA-seq) and targeted metabolomics to investigate the influence of 3TC supplementation on skeletal muscle in older mice. We show that 3TC 1) does not overtly affect muscle mass or functional/health markers, 2) largely reverses age-related changes in gene expression and metabolite signatures, and 3) is potentially beneficial for mitochondrial function in old animals via increases in antioxidant enzymes and decreases in mitochondrial reactive oxygen species. Collectively, our results suggest that, in addition to its protective effects in other tissues, 3TC supplementation does not have adverse effects in aged muscle and may even protect muscle/mitochondrial health in this context.NEW & NOTEWORTHY Recent studies suggest that the nucleoside reverse transcriptase inhibitor 3TC may improve brain health and cognitive function in old mice, but its effects on other aging tissues have not been comprehensively studied. This is the first study to use a multiomics approach to investigate the effects of 3TC treatment on skeletal muscle of old mice. The results suggest that 3TC reverses age-related transcriptomic and metabolite signatures and is potentially beneficial for mitochondrial function in aged muscle.
Collapse
Affiliation(s)
- Shelby C Osburn
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Meghan E Smith
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Devin Wahl
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Thomas J LaRocca
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| |
Collapse
|
14
|
Wang YT, Moura AK, Zuo R, Roudbari K, Hu JZ, Khan SA, Wang Z, Shentu Y, Wang M, Li PL, Hao J, Zhang Y, Li X. Cardiovascular dysfunction and altered lysosomal signaling in a murine model of acid sphingomyelinase deficiency. J Mol Med (Berl) 2025; 103:599-617. [PMID: 40232391 DOI: 10.1007/s00109-025-02542-z] [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: 09/26/2024] [Revised: 03/18/2025] [Accepted: 04/02/2025] [Indexed: 04/16/2025]
Abstract
Niemann-Pick Disease (NPD) is a rare autosomal recessive lysosomal storage disorder (LSD) caused by the deficiency of acid sphingomyelinase (ASMD), which is encoded by the Smpd1 gene. ASMD impacts multiple organ systems in the body, including the cardiovascular system. This study is the first to characterize cardiac pathological changes in ASMD mice under baseline conditions, offering novel insights into the cardiac implications of NPD. Using histological analysis, biochemical assays, and echocardiography, we assessed cardiac pathological changes and function in Smpd1-/- mice compared to Smpd1+/+ littermate controls. Immunofluorescence and biochemical assays demonstrated that ASMD induced lysosomal dysfunction, as evidenced by the accumulation of lysosomal-associated membrane proteins, lysosomal protease, and autophagosomes in pericytes and cardiomyocytes. This lysosomal dysfunction was accompanied by pericytes and cardiomyocytes inflammation, characterized by increased expression of caspase1 and inflammatory cytokines, and infiltration of inflammatory cells in the cardiac tissues of Smpd1-/- mice. In addition, histological analysis revealed increased lipid deposition and cardiac steatosis, along with pericyte-to-myofibroblast transition (PMT) and interstitial fibrosis in Smpd1-/- mice. Moreover, echocardiography further demonstrated that Smpd1-/- mice developed coronary microvascular dysfunction (CMD), as evidenced by decreased coronary blood flow velocity and increased coronary arteriolar wall thickness. Additionally, these mice exhibited significant impairments in systolic and diastolic cardiac function, as shown by a reduced ejection fraction and prolonged left ventricular relaxation time constant (Tau value). These findings suggest that ASMD induces profound pathological changes and vascular dysfunction in the myocardium, potentially driven by mechanisms involving lysosomal dysfunction as well as both pericytes and cardiac inflammation. KEY MESSAGES: Lysosomal dysfunction in ASMD leads to impaired autophagic flux in cardiac pericytes ASMD causes cardiac inflammation with leukocyte and M2 macrophage infiltration Lipid buildup in the pericytes, fibroblasts and myocardium lead to cardiac steatosis Enhanced cardiac fibrosis in ASMD links to pericyte-to-myofibroblast transition ASMD results in coronary microvascular and diastolic and systolic cardiac dysfunction.
Collapse
Affiliation(s)
- Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Alexandra K Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Jenny Z Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Saher A Khan
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yangping Shentu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mi Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jiukuan Hao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA.
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204 - 5056, USA.
| |
Collapse
|
15
|
Wu F, Mu WC, Markov NT, Fuentealba M, Halaweh H, Senchyna F, Manwaring-Mueller MN, Winer DA, Furman D. Immunological biomarkers of aging. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:889-902. [PMID: 40443365 PMCID: PMC12123219 DOI: 10.1093/jimmun/vkae036] [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] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/22/2024] [Indexed: 06/02/2025]
Abstract
The immune system has long been recognized for its critical role in the elimination of pathogens and the development of autoimmune diseases, but recent evidence demonstrates that it also contributes to noncommunicable diseases associated with biological aging processes, such as cancer, cardiovascular disease, neurodegeneration, and frailty. This review examines immunological biomarkers of aging, focusing on how the immune system evolves with age and its impact on health and disease. It discusses the historical development of immunological assessments, technological advancements, and the creation of novel biomarkers and models to study immune aging. We also explore the clinical implications of immune aging, such as increased susceptibility to infectious diseases, poor vaccine responses, and a higher incidence of noncommunicable diseases. In summary, we provide a comprehensive overview of current research, highlight the clinical relevance of immune aging, and identify gaps in knowledge that require further investigation.
Collapse
Affiliation(s)
- Fei Wu
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Wei-Chieh Mu
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Nikola T Markov
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Matias Fuentealba
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Heather Halaweh
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Fiona Senchyna
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | | | - Daniel A Winer
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
- Buck Institute for Research on Aging, Novato, CA, United States
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - David Furman
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
16
|
Wei B, Wei M, Huang H, Fan T, Zhang Z, Song X. Mesenchymal Stem Cell-Derived Exosomes: A Promising Therapeutic Strategy for Age-Related Diseases. Cell Prolif 2025; 58:e13795. [PMID: 39704104 PMCID: PMC12099225 DOI: 10.1111/cpr.13795] [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: 08/28/2024] [Revised: 11/09/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
The global increase in the aging population has led to a concurrent rise in the incidence of age-related diseases, posing substantial challenges to healthcare systems and affecting the well-being of the elderly. Identifying and securing effective treatments has become an urgent priority. In this context, mesenchymal stem cell-derived exosomes (MSC-Exos) have emerged as a promising and innovative modality in the field of anti-aging medicine, offering a multifaceted therapeutic approach. MSC-Exos demonstrate significant potential due to their immunomodulatory and anti-inflammatory properties, their ability to inhibit oxidative stress, and their reparative effects on senescent tissues. These attributes make them valuable in combating a range of conditions associated with aging, such as cardiovascular diseases, neurodegeneration, skin aging, and osteoarthritis. The integration of exosomes with membrane-penetrating peptides introduces a novel strategy for the delivery of biomolecules, surmounting traditional cellular barriers and enhancing therapeutic efficacy. This review provides a comprehensive synthesis of the current understanding of MSC-Exos, underscoring their role as a novel and potent therapeutic strategy against the intricate challenges of age-related diseases.
Collapse
Affiliation(s)
- Bohua Wei
- School of PharmacyChina Medical UniversityShenyangLiaoning ProvinceChina
| | - Mengting Wei
- School of StomatologyChina Medical UniversityShenyangLiaoning ProvinceChina
| | - Haonan Huang
- China Medical UniversityShenyangLiaoning ProvinceChina
| | - Ting Fan
- Department of Computer, School of Intelligent MedicineChina Medical UniversityShenyangLiaoning ProvinceChina
| | - Zhichang Zhang
- Department of Computer, School of Intelligent MedicineChina Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaoyu Song
- The College of Basic Medical Science, Health Sciences InstituteChina Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
17
|
Yan X, Li J, Wang Q, Xu T, Wu D. Peripheral blood lymphocyte subsets of Chinese adults with liver steatosis. Sci Rep 2025; 15:14970. [PMID: 40301513 PMCID: PMC12041267 DOI: 10.1038/s41598-025-99510-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
To evaluate the effect of liver steatosis on systematic cellular immunity through the changes of peripheral blood lymphocyte subsets, we retrospectively reviewed subjects receiving lymphocyte subtyping during annual medical check-ups. Liver steatosis was detected by abdominal computerized tomography or ultrasound. Immunophenotyping of peripheral blood lymphocytes was analyzed by flow cytometry. Cell counts of lymphocyte subsets were calculated using a dual-platform method. The relationship between lymphocyte subsets and liver steatosis was analyzed using multivariate linear models. Using the database from January 2017 to December 2022, we included 5042 subjects, including 1441 participants with liver steatosis. After adjusting for age, gender, and metabolic dysfunctions, the presence of liver steatosis increased the absolute values of CD19+, CD16+56+, CD3+, CD4+, CD4+CD28+, CD4+CD45RA-, CD4+CD45RA+, CD4+CD45RA+62L+, CD8+, CD8+CD28+, CD8+DR+, and CD8+CD38+ cell. In males, T lymphocyte counts of all T subsets increased in liver steatosis. In females, the significant differences in subsets included increased CD4+CD28+, CD4+CD45RA-, CD8+, and CD8+DR+ cells. Significant decreases were revealed in functional and naïve T cells with aging. Metabolic factors such as hypertension and abnormal glucose metabolism increase CD4+, CD4+CD28+, and CD4+CD45RA subsets. Hyperlipidaemia appears not to affect T cell counts, whereas obesity has some effect on both CD4+ (β = 0.041, 95% CI 0.032-0.051) and CD8+ cells (β = 0.036, 95% CI 0.025-0.048). Liver steatosis potentially affects peripheral blood lymphocyte subsets, while age, gender, and metabolic dysfunctions are also associated with these immune alterations.
Collapse
Affiliation(s)
- Xiaxiao Yan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Li
- Department of Health Management, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiang Wang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tengda Xu
- Department of Health Management, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Dong Wu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
18
|
Ding F, Yu Y, Zhao J, Wei S, Zhang Y, Han JH, Li Z, Jiang HB, Ryu D, Cho M, Bae SJ, Park W, Ha KT, Gao B. The interplay of cellular senescence and reprogramming shapes the biological landscape of aging and cancer revealing novel therapeutic avenues. Front Cell Dev Biol 2025; 13:1593096. [PMID: 40356604 PMCID: PMC12066513 DOI: 10.3389/fcell.2025.1593096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Cellular senescence and cellular reprogramming represent two fundamentally intertwined processes that profoundly influence aging and cancer. This paper explores how the permanent cell-cycle arrest of senescent cells and the identity-resetting capacity of reprogramming jointly shape biological outcomes in later life and tumor development. We synthesize recent findings to show that senescent cells, while halting the proliferation of damaged cells, can paradoxically promote tissue dysfunction and malignancy via their secretory phenotype. Conversely, induced reprogramming of somatic cells-exemplified by Yamanaka factors-resets cellular age and epigenetic marks, offering a potential to rejuvenate aged cells. Key findings highlight shared mechanisms (e.g., DNA damage responses and epigenetic remodeling) and bidirectional crosstalk between these processes: senescence signals can facilitate neighboring cell plasticity, whereas reprogramming attempts can trigger intrinsic senescence programs as a barrier. In aging tissues, transient (partial) reprogramming has been shown to erase senescence markers and restore cell function without inducing tumorigenesis, underlining a novel strategy to combat age-related degeneration. In cancer, we discuss how therapy-induced senescence of tumor cells may induce stem-cell-like traits in some cells and drive relapse, revealing a delicate balance between tumor suppression and tumor promotion. Understanding the interplay between senescence and reprogramming is crucial for developing innovative therapies. By targeting the senescence-reprogramming axis-for instance, via senolytic drugs, SASP inhibitors, or safe reprogramming techniques-there is significant therapeutic potential to ameliorate aging-related diseases and improve cancer treatment. Our findings underscore that carefully modulating cellular senescence and rejuvenation processes could pave the way for novel regenerative and anti-cancer strategies.
Collapse
Affiliation(s)
- Fuan Ding
- Department of Vascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ying Yu
- Department of Surgery, Changchun University of Chinese Medicine, Changchun, China
| | - Jiangqi Zhao
- Department of Dermatology, The Second Hospital of Jilin University, Changchun, China
| | - Shibo Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yan Zhang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jung Ho Han
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Zhuo Li
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hong-Bo Jiang
- Department of Dermatology, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Minkyoung Cho
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Sung-Jin Bae
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan, Republic of Korea
| | - Wonyoung Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
- Research Institute for Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
- Research Institute for Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Bo Gao
- Department of Vascular Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Zhang T, Pan Y, Sawa T, Akaike T, Matsunaga T. Supersulfide donors and their therapeutic targets in inflammatory diseases. Front Immunol 2025; 16:1581385. [PMID: 40308575 PMCID: PMC12040673 DOI: 10.3389/fimmu.2025.1581385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Inflammation is one defense mechanism of the body that has multiple origins, ranging from physical agents to infectious agents including viruses and bacteria. The resolution of inflammation has emerged as a critical endogenous process that protects host tissues from prolonged or excessive inflammation, which can become chronic. Failure of the inflammation resolution is a key pathological mechanism that drives the progression of numerous inflammatory diseases. Owing to the various side effects of currently available drugs to control inflammation, novel therapeutic agents that can prevent or suppress inflammation are needed. Supersulfides are highly reactive and biologically potent molecules that function as antioxidants, redox regulators, and modulators of cell signaling. The catenation state of individual sulfur atoms endows supersulfides with unique biological activities. Great strides have recently been made in achieving a molecular understanding of these sulfur species, which participate in various physiological and pathological pathways. This review mainly focuses on the anti-inflammatory effects of supersulfides. The review starts with an overview of supersulfide biology and highlights the roles of supersulfides in both immune and inflammatory responses. The various donors used to generate supersulfides are assessed as research tools and potential therapeutic agents. Deeper understanding of the molecular and cellular bases of supersulfide-driven biology can help guide the development of innovative therapeutic strategies to prevent and treat diseases associated with various immune and inflammatory responses.
Collapse
Affiliation(s)
- Tianli Zhang
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita, Japan
| | - Yuexuan Pan
- Department of Redox Molecular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takaaki Akaike
- Department of Redox Molecular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Shimadzu × Tohoku University Supersulfides Life Science Co-creation Research Center, Sendai, Japan
| | - Tetsuro Matsunaga
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita, Japan
- Shimadzu × Tohoku University Supersulfides Life Science Co-creation Research Center, Sendai, Japan
| |
Collapse
|
20
|
Polini B, Ricardi C, Di Lupo F, Runfola M, Bacci A, Rapposelli S, Bizzarri R, Scalese M, Saponaro F, Chiellini G. Novel Thyroid Hormone Receptor-β Agonist TG68 Exerts Anti-Inflammatory, Lipid-Lowering and Anxiolytic Effects in a High-Fat Diet (HFD) Mouse Model of Obesity. Cells 2025; 14:580. [PMID: 40277905 PMCID: PMC12026167 DOI: 10.3390/cells14080580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Recent advances in drug development allowed for the identification of THRβ-selective thyromimetic TG68 as a very promising lipid lowering and anti-amyloid agent. In the current study, we first investigated the neuroprotective effects of TG68 on in vitro human models of neuroinflammation and β-amyloid neurotoxicity in order to expand our knowledge of the therapeutic potential of this novel thyromimetic. Subsequently, we examined metabolic and inflammatory profiles, along with cognitive changes, using a high-fat diet (HFD) mouse model of obesity. Our data demonstrated that TG68 was able to prevent either LPS/TNFα-induced inflammatory response or β-amyloid-induced cytotoxicity in human microglial (HMC3) cells. Next, we demonstrated that in HFD-fed mice, treatment with TG68 (10 mg/kg/day; 2 weeks) significantly reduced anxiety-like behavior in stretch-attend posture (SAP) tests while producing a 12% BW loss and a significant decrease in blood glucose and lipid levels. Notably, these data highlight a close relationship between improved serum metabolic parameters and a reduction of anxious behavior. Moreover, TG68 administration was observed to efficiently counteract HFD-altered central and peripheral expressions in mice with selected biomarkers of metabolic dysfunction, inflammation, and neurotoxicity, revealing promising neuroprotective effects. In conclusion, our work provides preliminary evidence that TG68 may represent a novel therapeutic opportunity for the treatment of interlinked diseases such as obesity and neurodegenerative diseases.
Collapse
Affiliation(s)
- Beatrice Polini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Caterina Ricardi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Francesca Di Lupo
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Massimiliano Runfola
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (M.R.); (A.B.); (S.R.)
| | - Andrea Bacci
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (M.R.); (A.B.); (S.R.)
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (M.R.); (A.B.); (S.R.)
| | - Ranieri Bizzarri
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Marco Scalese
- Institute of Clinical Physiology, Italian National Research Council, 56124 Pisa, Italy;
| | - Federica Saponaro
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Grazia Chiellini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| |
Collapse
|
21
|
Kuloglu E, Sengul I, Sengul D, Muhtaroglu A, Aslan S, Issever K, Dulger AC. Dyspepsia in nonagenarian women. Eur Geriatr Med 2025:10.1007/s41999-025-01197-w. [PMID: 40208428 DOI: 10.1007/s41999-025-01197-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE Dyspeptic complaints are common across all age groups, but limited research explores their characteristics and underlying pathologies in nonagenarians. This pioneering study aims to investigate the clinical, laboratory, and histopathological features in women nonagenarians vs. middle-aged. METHODS A cross-sectional study included 93 female nonagenarians and 90 under 65 years (control), all presenting with dyspeptic complaints to general surgery, internal medicine, and gastroenterology outpatient clinics. Both groups underwent comprehensive evaluations, including laboratory parameters, upper gastric endoscopic examinations, and histopathological assessments of gastric biopsies. Statistical analysis compared the two groups' demographic, laboratory, and histopathological findings. RESULTS Of the patients included in the study, 49.8% were under 65 years, and 50.2% were nonagenarians. The mean age of the control group was 54.8 years, while nonagenarians had a mean age of 92.7 years. Significant differences were observed in several laboratory biomarkers such as leukocytes, neutrophils, glucose, creatinine, sodium, potassium, albumin, alanine transaminase, C-reactive protein, etc. between the groups (p < 0.05). Nonagenarians were less infected with Helicobacter pylori, whereas they had a higher frequency of intestinal metaplasia. CONCLUSIONS Nonagenarians group has a higher rate of intestinal metaplasia and displasia, while a lower rate of Helicobacter pylori infection in their gastric mucosal specimens. More extensive randomized controlled trials should illuminate the possible pathophysiological mechanisms for this association.
Collapse
Affiliation(s)
- Ersin Kuloglu
- Department of Internal Medicine, Faculty of Medicine, Giresun University, Gazipasa Compound, Gazi Avenue, 28100, Giresun, Turkey.
| | - Ilker Sengul
- Division of Endocrine Surgery, Faculty of Medicine, Giresun University, Giresun, Turkey
- Department of General Surgery, Faculty of Medicine, Giresun University, Giresun, Turkey
| | - Demet Sengul
- Department of Pathology, Faculty of Medicine, Giresun University, Giresun, Turkey
| | - Ali Muhtaroglu
- Department of General Surgery, Faculty of Medicine, Giresun University, Giresun, Turkey
| | - Sefer Aslan
- Department of Internal Medicine, Faculty of Medicine, Giresun University, Gazipasa Compound, Gazi Avenue, 28100, Giresun, Turkey
| | - Kubilay Issever
- Department of Internal Medicine, Faculty of Medicine, Giresun University, Gazipasa Compound, Gazi Avenue, 28100, Giresun, Turkey
| | - Ahmet Cumhur Dulger
- Department of Internal Medicine, Faculty of Medicine, Giresun University, Gazipasa Compound, Gazi Avenue, 28100, Giresun, Turkey
- Division of Gastroenterology, Faculty of Medicine, Giresun University, Giresun, Turkey
| |
Collapse
|
22
|
Park TI, Yang AH, Kanth BK, Pack SP. Aptamers as Diagnostic and Therapeutic Agents for Aging and Age-Related Diseases. BIOSENSORS 2025; 15:232. [PMID: 40277546 PMCID: PMC12024714 DOI: 10.3390/bios15040232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025]
Abstract
In the 21st century, the demographic shift toward an aging population has posed a significant challenge, particularly with respect to age-related diseases, which constitute a major threat to human health. Accordingly, the detection, prevention, and treatment of aging and age-related diseases have become critical issues, and the introduction of novel molecular recognition elements, called aptamers, has been considered. Aptamers, a class of oligonucleotides, can bind to target molecules with high specificity. In addition, aptamers exhibit superior stability, biocompatibility, and applicability, rendering them promising tools for the diagnosis and treatment of human diseases. In this paper, we present a comprehensive overview of aptamers, systematic evolution of ligands by exponential enrichment (SELEX), biomarkers associated with aging, as well as aptamer-based diagnostic and therapeutic platforms. Finally, the limitations associated with predicting and preventing age-related conditions are discussed, along with potential solutions based on advanced technologies and theoretical approaches.
Collapse
Affiliation(s)
- Tae-In Park
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (T.-I.P.); (A.H.Y.)
| | - Ah Hyun Yang
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (T.-I.P.); (A.H.Y.)
| | - Bashistha Kumar Kanth
- Department of Food Science and Nutrition, Dong-A University, Pusan 602760, Republic of Korea;
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (T.-I.P.); (A.H.Y.)
| |
Collapse
|
23
|
Mohammadzadeh R, Mahnert A, Shinde T, Kumpitsch C, Weinberger V, Schmidt H, Moissl-Eichinger C. Age-related dynamics of predominant methanogenic archaea in the human gut microbiome. BMC Microbiol 2025; 25:193. [PMID: 40181255 PMCID: PMC11969853 DOI: 10.1186/s12866-025-03921-9] [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: 05/06/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The reciprocal relationship between aging and alterations in the gut microbiota is a subject of ongoing research. While the role of bacteria in the gut microbiome is well-documented, specific changes in the composition of methanogens during extreme aging and the impact of high methane production in general on health remain unclear. This study was designed to explore the association of predominant methanogenic archaea within the human gut and aging. METHODS Shotgun metagenomic data from the stool samples of young adults (n = 127, Age: 19-59 y), older adults (n = 86, Age: 60-99 y), and centenarians (n = 34, age: 100-109 years) were analyzed. RESULTS Our findings reveal a compelling link between age and the prevalence of high methanogen phenotype, while overall archaeal diversity diminishes. Surprisingly, the archaeal composition of methanogens in the microbiome of centenarians appears more akin to that of younger adults, showing an increase in Methanobrevibacter smithii, rather than Candidatus Methanobrevibacter intestini. Remarkably, Ca. M. intestini emerged as a central player in the stability of the archaea-bacteria network in adults, paving the way for M. smithii in older adults and centenarians. Notably, centenarians exhibit a highly complex and stable network of these two methanogens with other bacteria. The mutual exclusion between Lachnospiraceae and these methanogens throughout all age groups suggests that these archaeal communities may compensate for the age-related drop in Lachnospiraceae by co-occurring with Oscillospiraceae. CONCLUSIONS This study underscores the dynamics of archaeal microbiome in human physiology and aging. It highlights age-related shifts in methanogen composition, emphasizing the significance of both M. smithii and Ca. M. intestini and their partnership with butyrate-producing bacteria for potential enhanced health.
Collapse
Affiliation(s)
- Rokhsareh Mohammadzadeh
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Alexander Mahnert
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Tejus Shinde
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Christina Kumpitsch
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Viktoria Weinberger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Helena Schmidt
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Christine Moissl-Eichinger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria.
- BioTechMed, Graz, 8010, Austria.
| |
Collapse
|
24
|
Chu CR, Hochberg M, White D, Rodeo S, Huard J, Shapiro S, Lattermann C, Guilak F. Transformative approaches for effective clinical trials to reduce the disease burden of osteoarthritis. Semin Arthritis Rheum 2025; 71:152652. [PMID: 39970622 DOI: 10.1016/j.semarthrit.2025.152652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/19/2024] [Accepted: 01/06/2025] [Indexed: 02/21/2025]
Abstract
Osteoarthritis (OA) is a leading cause of disability and morbidity that has eluded development of effective disease modifying drugs and therapies. While established OA in the form of symptomatic radiographic disease is a recognizable final common pathway, OA development encompasses a broad spectrum of pathological changes, susceptibilities, and etiological pathways that cannot be considered a single disease process. Beginning with preclinical disease where radiographs are normal, the concept of pre-osteoarthritis (pre-OA) offers a systems-based approach to OA prevention by targeting reduction of OA risk prior to the onset of definable OA. Early OA ensues when cellular, molecular, and joint tissue changes begin to overlap that of OA, a process that can begin before the onset of definitive symptoms or radiographic changes. A myriad of pathways and crossroads of pre-OA and early OA eventually leads to poorly irreversible symptomatic radiographic OA. With increasing recognition of pre-OA and early OA markers, pathways and subtypes, opportunities arise to address these new therapeutic targets. The current status of clinical trials in OA was identified as a critical barrier to progress by the 2022 National Institute of Arthritis, Musculoskeletal, and Skin Diseases (NIAMS) Roundtable on "Cartilage Preservation and Restoration in Knee Osteoarthritis: Challenges, Gaps, and Opportunities". This manuscript summarizes the recommendations of the work group established from the Roundtable to address this issue. The work group recommends that clinical trial design and endpoints evolve to effectively evaluate new treatment approaches suitable for pre-osteoarthritis and early OA by different criteria than what has been set for symptomatic radiographic OA. While symptomatic improvement is the primary goal for palliation of irreversible established OA, important goals for treating earlier disease states include disease modification and prevention, with the potential to alter the natural history of progressive OA. Because symptoms may not correlate with structural changes in pre-OA and early OA, the primary outcomes in these trials need to match the intended mechanistic target and the therapeutic goal for the disease state being treated. The purpose of this manuscript is to transform the approach to clinical trials in OA by establishing a new benchmark of identifying critical outcomes that are appropriate for the joint disease states and subtypes of the target patient population, and the therapeutic or mechanistic target of the intervention being tested. By shifting the approach from using standardized outcomes based on established OA towards customizing clinical trials according to these principles, new precision medicine strategies to address the full spectrum of disease from pre-OA to OA can be more readily advanced into clinical practice.
Collapse
Affiliation(s)
- Constance R Chu
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway St 94061, Redwood City, CA 94063, United States.
| | - Marc Hochberg
- Departments of Medicine and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, 20742, United States
| | - Daniel White
- Department of Physical Therapy, University of Delaware, Newark, DE 19716, United States
| | - Scott Rodeo
- Hospital for Special Surgery, New York, NY 10021, United States
| | - Johnny Huard
- Steadman Clinic, Steadman Philippon Research Institute, Vail CO 81657, United States
| | - Shane Shapiro
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Christian Lattermann
- Department of Orthopaedic Surgery, Massachusetts General-Brigham Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States
| |
Collapse
|
25
|
Coperchini F, Greco A, Teliti M, Croce L, Chytiris S, Magri F, Gaetano C, Rotondi M. Inflamm-ageing: How cytokines and nutrition shape the trajectory of ageing. Cytokine Growth Factor Rev 2025; 82:31-42. [PMID: 39237438 DOI: 10.1016/j.cytogfr.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
Population ageing is increasing in prevalence in most developed countries. Ageing is the decline of functional properties at the cellular, tissue, and organ level. Biochemical changes that occur in all organisms that experience biological ageing are referred to as the "Hallmarks of ageing". Inflammation is a common denominator of the hallmarks of ageing, being mechanistically involved in most age-related health consequences. Inflamm-ageing refers to age-related changes in the inflammatory and immune systems which somehow drive the ageing process towards healthy or unhealthy ageing. Current evidences, support that, reversing the age-related pro-inflammatory status of inflamm-ageing, is able to modulate most hallmarks of ageing. Inflamm-ageing is associated with increased levels of pro-inflammatory molecules (e.g. cytokines, chemokines), ultimately producing a chronic low-grade inflammatory state typically observed in older individuals. It is commonly accepted that, the balance between pro- and anti-inflammatory cytokines/chemokines is one of the factors determining whether healthy or unhealthy ageing occurs. Malnutrition and nutritional imbalances, are highly prevalent in the elderly, playing a role in driving the balance of pro- and anti-inflammatory immunoactive molecules. In particular, malnutrition is a major risk factor for sarcopenia, a phenomenon characterized by loss of muscle mass, which is often referred to as the biological basis for frailty. Given the close relationship between malnutrition and sarcopenia, there is also evidence for a link between malnutrition and frailty. Indeed, changes in cytokine/chemokine levels in elderly patients with malnutrition were demonstrated. The demonstration that specific cytokines play a role in modulating appetite and nutrient sensing and taste reception, provided further evidence for the existence of a link between inflamm-ageing, nutrition and cytokines in shaping the trajectory of ageing. The present review will overview current evidence supporting the role of specific circulating cytokines and chemokines in the relationship between ageing, inflammation, and malnutrition.
Collapse
Affiliation(s)
- Francesca Coperchini
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy
| | - Alessia Greco
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy
| | - Marsida Teliti
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Laura Croce
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Spyridon Chytiris
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Flavia Magri
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Mario Rotondi
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy.
| |
Collapse
|
26
|
Gong H, Su WJ, Deng SL, Luo J, Du ZL, Luo Y, Lv KY, Zhu DM, Fan XT. Anti-inflammatory interventions for the treatment and prevention of depression among older adults: a systematic review and meta-analysis. Transl Psychiatry 2025; 15:114. [PMID: 40169548 PMCID: PMC11961752 DOI: 10.1038/s41398-025-03317-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 02/19/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025] Open
Abstract
Recent evidence from clinical and animal studies with anti-inflammatory agents in depression is conflicting. One possible reason is the heterogeneity of baseline inflammation levels. Since older adults are generally associated with chronic low-grade inflammation and depression is one of the most common mental disorders in this population, this meta-analysis aimed to evaluate the therapeutic and preventative effects of anti-inflammatory interventions for depression among older adults. PubMed, Cochrane Library, Embase, and PsycINFO were searched for randomized controlled trials (RCTs) up to November 18, 2024. The primary outcomes were mean change scores of depression scores and incidences of depression after treatment. Pooled standard mean differences (SMDs) and odds ratios (ORs) including 95% confidence intervals (95% CI) were calculated. Of 3116 screened articles, 31 RCTs met the inclusion criteria, with 25 studies investigating efficacy and 7 studies investigating the incidence following anti-inflammatory treatment. Anti-inflammatory interventions were statistically significantly more effective than placebo in reducing depressive scores for older adults with depression (SMD = -0.57, 95% CI = -0.98 to -0.15, p = 0.008). Sub-group analyses supported the use of omega-3 fatty acids (SMD = -0.14, 95% CI = -0.27 to -0.02, p = 0.03) and botanical drug or dietary intervention (SMD = -0.86, 95% CI = -1.58 to -0.13, p = 0.02) among older participants. While limited by substantial heterogeneity among included studies, these results reveal the moderate beneficial effects of anti-inflammatory interventions for the treatment and prevention of depression among older adults. Future high-quality RCTs are warranted to determine which anti-inflammatory interventions are most preferential for older patients with depression.
Collapse
Affiliation(s)
- Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wen-Jun Su
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai, China
| | - Shi-Long Deng
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jing Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhu-Lin Du
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ke-Yi Lv
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dong-Mei Zhu
- Department of Hospital Infection Control, Chongqing Health Center for Women and Children, Chongqing, China.
- Department of Hospital Infection Control, Women and Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiao-Tang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
27
|
Huang H, Ren J, Liu GH. Insights and Interventions in Age-Associated Inflammation. Curr Opin Genet Dev 2025; 91:102306. [PMID: 39837098 DOI: 10.1016/j.gde.2024.102306] [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: 09/24/2024] [Revised: 12/20/2024] [Accepted: 12/25/2024] [Indexed: 01/23/2025]
Abstract
Aging is a systemic, complex, and heterogeneous process characterized by a progressive decline in physiological functions, rendering it a major risk factor for various chronic diseases. Chronic inflammation has emerged as both a hallmark and a driver in this complicated process. This persistent inflammatory state arises from a spectrum of stimuli, ranging from external pathogens to internal cellular remnants, to metabolic dysregulation, and to chronic stress. Here, we examine recent mechanistic advances into the driving forces behind age-related chronic inflammation, explore promising anti-inflammatory strategies to mitigate aging, and address current challenges, proposing future directions to propel this evolving field toward translational breakthrough.
Collapse
Affiliation(s)
- Haoyan Huang
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Jie Ren
- Key Laboratory of RNA Science and Engineering, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guang-Hui Liu
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China; Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
28
|
Soni N, Kaur P, Gurjar V, Bhargava A, Tiwari R, Chouksey A, Srivastava RK, Mishra PK. Unveiling the Interconnected Dynamics of Mitochondrial Dysfunction Associated With Age-Related Cardiovascular Risk: A Cross-Sectional Pilot Study. Cureus 2025; 17:e82961. [PMID: 40416162 PMCID: PMC12103716 DOI: 10.7759/cureus.82961] [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] [Accepted: 04/25/2025] [Indexed: 05/27/2025] Open
Abstract
Aging, influenced by complex epigenetic mechanisms, significantly contributes to the progression of cardiovascular diseases (CVDs). This cross-sectional pilot study investigated mitochondrial-associated epigenetic stress responses in two age groups: Group I (18-38, n = 154), representing younger adults generally at lower risk for CVD, and Group II (39-65, n = 105), representing middle-aged and older adults with increased biological susceptibility. The age grouping was based on established physiological and cardiovascular risk transitions typically observed around age 40. To assess age-related molecular variations, we examined key mitochondrial and metabolic parameters, including mitochondrial DNA (mtDNA) damage repair capacity, mtDNA copy number (mtDNAcn), methylation status, mitochondrial dynamics (fusion/fission), telomere length, expression of respiratory complex genes, levels of pro-inflammatory cytokines, and N-terminal pro-B-type natriuretic peptide (NT-proBNP) concentrations. Our results indicated that the older group exhibited higher mtDNA methylation (r² = 0.5205, p < 0.0001), increased mtDNAcn, and elevated NT-proBNP levels, which also showed a weak positive correlation with mtDNA methylation (r² = 0.3218, p < 0.0001). Additionally, a strong negative correlation was observed between telomerase reverse transcriptase (TERT) expression and age (r² = 0.6070, p < 0.0001), suggesting a decline in telomeric maintenance with advancing age. Group II also showed altered inflammatory and telomeric profiles and a notable reduction in the expression of mitochondrial respiratory complex genes (ND6, COXI, ATPase 6 and 8), alongside increased expression of genes involved in mitochondrial stress response pathways. We employed four machine learning models - Logistic Regression, Decision Tree, Random Forest, and Support Vector Machine (SVM) - for CVD risk prediction, using selected mitochondrial and metabolic features. All models demonstrated high classification accuracy, ranging from 0.920 to 1.0, with the Random Forest model achieving the highest accuracy of 0.984. These preliminary findings highlight distinct age-related molecular signatures and illustrate the potential of combining biomarkers with machine-learning approaches to improve cardiovascular risk prediction and therapeutic targeting in aging populations.
Collapse
Affiliation(s)
- Nikita Soni
- Environmental Biotechnology, Genetics, and Molecular Biology, Indian Council of Medical Research (ICMR) - National Institute for Research in Environmental Health, Bhopal, IND
| | - Prasan Kaur
- Environmental Biotechnology, Genetics, and Molecular Biology, Indian Council of Medical Research (ICMR) - National Institute for Research in Environmental Health, Bhopal, IND
| | - Vikas Gurjar
- Environmental Biotechnology, Genetics, and Molecular Biology, Indian Council of Medical Research (ICMR) - National Institute for Research in Environmental Health, Bhopal, IND
| | - Arpit Bhargava
- Faculty of Science, Ram Krishna Dharmarth Foundation (RKDF) University, Bhopal, IND
| | - Rajnarayan Tiwari
- Epidemiology, Indian Council of Medical Research (ICMR) - National Institute for Research in Environmental Health, Bhopal, IND
| | - Apoorva Chouksey
- Environmental Biotechnology, Genetics, and Molecular Biology, Indian Council of Medical Research (ICMR) - National Institute for Research in Environmental Health, Bhopal, IND
| | - Rupesh K Srivastava
- Biotechnology, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Pradyumna K Mishra
- Environmental Biotechnology, Genetics, and Molecular Biology, Indian Council of Medical Research (ICMR) - National Institute for Research in Environmental Health, Bhopal, IND
| |
Collapse
|
29
|
Jinesh S, Özüpek B, Aditi P. Premature aging and metabolic diseases: the impact of telomere attrition. FRONTIERS IN AGING 2025; 6:1541127. [PMID: 40231186 PMCID: PMC11995884 DOI: 10.3389/fragi.2025.1541127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/03/2025] [Indexed: 04/16/2025]
Abstract
Driven by genetic and environmental factors, aging is a physiological process responsible for age-related degenerative changes in the body, cognitive decline, and impaired overall wellbeing. Notably, premature aging as well as the emergence of progeroid syndromes have posed concerns regarding chronic health conditions and comorbidities in the aging population. Accelerated telomere attrition is also implicated in metabolic dysfunction and the development of metabolic disorders. Impaired metabolic homeostasis arises secondary to age-related increases in the synthesis of free radicals, decreased oxidative capacity, impaired antioxidant defense, and disrupted energy metabolism. In particular, several cellular and molecular mechanisms of aging have been identified to decipher the influence of premature aging on metabolic diseases. These include defective DNA repair, telomere attrition, epigenetic alterations, and dysregulation of nutrient-sensing pathways. The role of telomere attrition premature aging in the pathogenesis of metabolic diseases has been largely attributed to pro-inflammatory states that promote telomere shortening, genetic mutations in the telomerase reverse transcriptase, epigenetic alteration, oxidative stress, and mitochondrial dysfunctions. Nonetheless, the therapeutic interventions focus on restoring the length of telomeres and may include treatment approaches to restore telomerase enzyme activity, promote alternative lengthening of telomeres, counter oxidative stress, and decrease the concentration of pro-inflammatory cytokines. Given the significance and robust potential of delaying telomere attrition in age-related metabolic diseases, this review aimed to explore the molecular and cellular mechanisms of aging underlying premature telomere attrition and metabolic diseases, assimilating evidence from both human and animal studies.
Collapse
Affiliation(s)
| | | | - Prerana Aditi
- Department of Medical Biochemistry, Faculty of Allied Health Sciences, Mahayogi Gorakhnath University, Gorakhpur, Uttar Pradesh, India
| |
Collapse
|
30
|
Carollo C, Sorce A, Cirafici E, Mulè G, Caimi G. Sirtuins and Resveratrol in Cardiorenal Diseases: A Narrative Review of Mechanisms and Therapeutic Potential. Nutrients 2025; 17:1212. [PMID: 40218970 PMCID: PMC11990745 DOI: 10.3390/nu17071212] [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: 03/14/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Aging is a very complex process, and it has been linked with Sirtuins. Sirtuin enzymes are a family of deacetylases that are related to caloric restriction and aging by modulating energy metabolism, genomic stability, and stress resistance. Up to now, seven sirtuins have been recognized. This narrative review aimed to analyze the literature produced between January 2005 and March 2025 to evaluate the role of sirtuins in chronic kidney disease and, as heart and kidney diseases are strictly interrelated, to explore their role in heart diseases and cardio-renal cross-talk. A reciprocal relationship between CKD and aging seems to exist since CKD may contribute to premature biological aging of different organ systems. SIRTs are involved in the pathophysiology of renal diseases; their activation can delay the progression of several renal diseases. Notably, an increasing number of studies linked SIRTs with different CVDs. SIRTs affect the production of mitochondrial reactive oxygen species (ROS) by modulating mitochondrial function. The imbalance of SIRT levels may increase the vulnerability to CVDs. SIRTs are involved in the pathophysiological mechanisms of HFpEF (heart failure with preserved ejection fraction) through different signaling pathways. Fibrosis is the linkage mechanism between the heart and kidney in the development of cardio-renal diseases. Current studies on sirtuins, resveratrol, and cardiorenal disease highlight their potential therapeutic benefits in regulating blood pressure, kidney function, lipid profiles, and inflammation, making them a promising area of investigation for improving cardiovascular and renal health outcomes. However, significant gaps remain. The limited availability of highly selective and potent sirtuin modulators hampers their clinical translation, as most existing compounds exhibit poor bioavailability and suboptimal pharmacokinetic properties.
Collapse
Affiliation(s)
- Caterina Carollo
- Department of Health Promotion, Mother and Child Care, Internal and Specialistic Medicine, University of Palermo, 90127 Palermo, Italy (E.C.); (G.M.)
| | | | | | | | | |
Collapse
|
31
|
Khoso MA, Liu H, Zhao T, Zhao W, Huang Q, Sun Z, Dinislam K, Chen C, Kong L, Zhang Y, Liu X. Impact of plant-derived antioxidants on heart aging: a mechanistic outlook. Front Pharmacol 2025; 16:1524584. [PMID: 40191425 PMCID: PMC11969199 DOI: 10.3389/fphar.2025.1524584] [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: 11/07/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Heart aging involves a complex interplay of genetic and environmental influences, leading to a gradual deterioration of cardiovascular integrity and function. Age-related physiological changes, including ventricular hypertrophy, diastolic dysfunction, myocardial fibrosis, increased arterial stiffness, and endothelial dysfunction, are influenced by key mechanisms like autophagy, inflammation, and oxidative stress. This review aims to explore the therapeutic potential of plant-derived bioactive antioxidants in mitigating heart aging. These compounds, often rich in polyphenols, flavonoids, and other phytochemicals, exhibit notable antioxidant, anti-inflammatory, and cardioprotective properties. These substances have intricate cardioprotective properties, including the ability to scavenge ROS, enhance endogenous antioxidant defenses, regulate signaling pathways, and impede fibrosis and inflammation-promoting processes. By focusing on key molecular mechanisms linked to cardiac aging, antioxidants produced from plants provide significant promise to reduce age-related cardiovascular decline and improve general heart health. Through a comprehensive analysis of preclinical and clinical studies, this work highlights the mechanisms associated with heart aging and the promising effects of plant-derived antioxidants. The findings may helpful for researchers in identifying specific molecules with therapeutic and preventive potential for aging heart.
Collapse
Affiliation(s)
- Muneer Ahmed Khoso
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Heng Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Tong Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Wenjie Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Qiang Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Zeqi Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Khuzin Dinislam
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Chen Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Lingyi Kong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| |
Collapse
|
32
|
Wang YT, Moura AK, Zuo R, Roudbari K, Hu JZ, Khan SA, Wang Z, Shentu Y, Wang M, Li PL, Hao J, Zhang Y, Li X. Cardiovascular Dysfunction and Altered Lysosomal Signaling in a Murine Model of Acid Sphingomyelinase Deficiency. RESEARCH SQUARE 2025:rs.3.rs-5154105. [PMID: 40166006 PMCID: PMC11957194 DOI: 10.21203/rs.3.rs-5154105/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Niemann-Pick Disease (NPD) is a rare autosomal recessive lysosomal storage disorder (LSD) caused by the deficiency of acid sphingomyelinase (ASMD), which is encoded by the Smpd1 gene. ASMD impacts multiple organ systems in the body, including the cardiovascular system. This study is the first to characterize cardiac pathological changes in ASMD mice under baseline conditions, offering novel insights into the cardiac implications of NPD. Using histological analysis, biochemical assays, and echocardiography, we assessed cardiac pathological changes and function in Smpd1 -/- mice compared to Smpd1 +/+ littermate controls. Immunofluorescence and biochemical assays demonstrated that ASMD induced lysosomal dysfunction, as evidenced by the accumulation of lysosomal-associated membrane proteins, lysosomal protease, and autophagosomes in pericytes and cardiomyocytes. This lysosomal dysfunction was accompanied by pericytes and cardiomyocytes inflammation, characterized by increased expression of caspase1 and inflammatory cytokines, and infiltration of inflammatory cells in the cardiac tissues of Smpd1 -/- mice. In addition, histological analysis revealed increased lipid deposition and cardiac steatosis, along with pericyte-to-myofibroblast transition (PMT) and interstitial fibrosis in Smpd1 -/- mice. Moreover, echocardiography further demonstrated that Smpd1 -/- mice developed coronary microvascular dysfunction (CMD), as evidenced by decreased coronary blood flow velocity and increased coronary arteriolar wall thickness. Additionally, these mice exhibited significant impairments in systolic and diastolic cardiac function, as shown by a reduced ejection fraction and prolonged left ventricular relaxation time constant (Tau value). These findings suggest that ASMD induces profound pathological changes and vascular dysfunction in the myocardium, potentially driven by mechanisms involving lysosomal dysfunction as well as both pericytes and cardiac inflammation.
Collapse
|
33
|
Komleva Y, Shpiliukova K, Bondar N, Salmina A, Khilazheva E, Illarioshkin S, Piradov M. Decoding brain aging trajectory: predictive discrepancies, genetic susceptibilities, and emerging therapeutic strategies. Front Aging Neurosci 2025; 17:1562453. [PMID: 40177249 PMCID: PMC11962000 DOI: 10.3389/fnagi.2025.1562453] [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: 01/17/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The global extension of human lifespan has intensified the focus on aging, yet its underlying mechanisms remain inadequately understood. The article highlights aspects of genetic susceptibility to impaired brain bioenergetics, trends in age-related gene expression related to neuroinflammation and brain senescence, and the impact of stem cell exhaustion and quiescence on accelerated brain aging. We also review the accumulation of senescent cells, mitochondrial dysfunction, and metabolic disturbances as central pathological processes in aging, emphasizing how these factors contribute to inflammation and disrupt cellular competition defining the aging trajectory. Furthermore, we discuss emerging therapeutic strategies and the future potential of integrating advanced technologies to refine aging assessments. The combination of several methods including genetic analysis, neuroimaging techniques, cognitive tests and digital twins, offer a novel approach by simulating and monitoring individual health and aging trajectories, thereby providing more accurate and personalized insights. Conclusively, the accurate estimation of brain aging trajectories is crucial for understanding and managing aging processes, potentially transforming preventive and therapeutic strategies to improve health outcomes in aging populations.
Collapse
Affiliation(s)
| | | | - Nikolai Bondar
- Research Center of Neurology, Moscow, Russia
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Elena Khilazheva
- Department of Biological Chemistry with Courses in Medical, Research Institute of Molecular Medicine and Pathobiochemistry, Pharmaceutical and Toxicological Chemistry Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Healthcare of the Russian Federation, Krasnoyarsk, Russia
| | | | | |
Collapse
|
34
|
Jana R, Das Sarma J. The crosstalk between CNS resident glial cells and peripheral immune cells is critical for age-dependent demyelination and subsequent remyelination. Biogerontology 2025; 26:74. [PMID: 40085264 DOI: 10.1007/s10522-025-10213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
White-matter diseases like multiple sclerosis begin in young adulthood. Aging, being a risk factor, contributes to the progression of these diseases and makes neurological disabilities worsen. Aging causes white matter alteration due to myelin loss, axonal degeneration, and hyperintensities, resulting in cognitive impairment and neurological disorders. Aging also negatively affects central nervous system resident glial cells and peripheral immune cells, contributing to myelin degeneration and diminished myelin renewal process. Restoration of myelin failure with aging accelerates the progression of cognitive decline. This review will mainly focus on how age-related altered functions of glial and peripheral cells will affect myelin sheath alteration and myelin restoration. This understanding can give us insights into the underlying mechanisms of demyelination and failure of remyelination with aging concerning altered glial and peripheral immune cell function and their crosstalk. Also, we will explain the therapeutic strategies to enhance the remyelination process of an aging brain to improve the cognitive health of an aging person.
Collapse
Affiliation(s)
- Rishika Jana
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.
- Departments of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
35
|
Hadzibegovic S, Nicole O, Andelkovic V, de Gannes FP, Hurtier A, Lagroye I, Bontempi B. Examining the effects of extremely low-frequency magnetic fields on cognitive functions and functional brain markers in aged mice. Sci Rep 2025; 15:8365. [PMID: 40069380 PMCID: PMC11897315 DOI: 10.1038/s41598-025-93230-y] [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: 09/21/2024] [Accepted: 03/05/2025] [Indexed: 03/15/2025] Open
Abstract
Extremely low-frequency magnetic fields (ELF-MFs) are ubiquitously present in various environments of everyday life. While surveys from the World Health Organization (WHO) have not demonstrated the existence of ELF-MF-induced harmful consequences in healthy subjects, whether older adults are more vulnerable to the effects of residential and occupational ELF-MF exposure, and therefore may be at risk, remains unsettled. Here, we explored this potential health issue by investigating, in aged mice, the effects of chronic exposure to ELF-MFs (50 Hz ELF-MF at 1 mT for 8 h/day, 5 days/week for 12 consecutive weeks) on cognitive functions and expression profile of brain markers typically associated with aggravated aging or the development of Alzheimer`s disease (AD). Sham-exposed mice showed a significant age-related decline in spatial memory functions compared to young adult mice. However, this expected pattern was neither exacerbated nor counteracted by chronic exposure to ELF-MFs. No difference in hippocampal expression of APP-695, Aβ(1-42), S100b and GFAP proteins or in the pTau/Tau ratio was observed between sham- and ELF-MF-exposed aged mice, suggesting that chronic exposure to ELF-MFs does not aggravate aging and associated neuroinflammation, or promote pathological pathways involved in the initiation of AD. Because care should be taken in extrapolating these results to older adults with various comorbidities, applying current exposure limits to existing or new sensitive ELF-MF locations is recommended.
Collapse
Affiliation(s)
- Senka Hadzibegovic
- Neurocentre Magendie, INSERM U1215, Université de Bordeaux, 33000, Bordeaux, France.
| | - Olivier Nicole
- Institut Interdisciplinaire de Neurosciences, CNRS UMR 5297, Université de Bordeaux, 33000, Bordeaux, France
| | - Vojislav Andelkovic
- Laboratoire de l'Intégration du Matériau au Système, CNRS UMR 5218, Université de Bordeaux, 33405, Talence, France and Ecole Pratique des Hautes Etudes-PSL, 75014, Paris, France
| | - Florence Poulletier de Gannes
- Laboratoire de l'Intégration du Matériau au Système, CNRS UMR 5218, Université de Bordeaux, 33405, Talence, France and Ecole Pratique des Hautes Etudes-PSL, 75014, Paris, France
| | - Annabelle Hurtier
- Laboratoire de l'Intégration du Matériau au Système, CNRS UMR 5218, Université de Bordeaux, 33405, Talence, France and Ecole Pratique des Hautes Etudes-PSL, 75014, Paris, France
| | - Isabelle Lagroye
- Laboratoire de l'Intégration du Matériau au Système, CNRS UMR 5218, Université de Bordeaux, 33405, Talence, France and Ecole Pratique des Hautes Etudes-PSL, 75014, Paris, France
| | - Bruno Bontempi
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Université de Bordeaux and Ecole Pratique des Hautes Etudes, 33000, Bordeaux, France.
| |
Collapse
|
36
|
Cordiano R, Caserta S, Minciullo PL, Allegra A, Gangemi S. Anthraquinones and Aloe Vera Extracts as Potential Modulators of Inflammaging Mechanisms: A Translational Approach from Autoimmune to Onco-Hematological Diseases. Molecules 2025; 30:1251. [PMID: 40142026 PMCID: PMC11944353 DOI: 10.3390/molecules30061251] [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: 02/06/2025] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Inflammaging is a chronic, low-grade inflammatory state that contributes to age-related diseases, including cardiovascular disorders, osteoporosis, neurodegeneration, and cancer. This process involves immunosenescence, oxidative stress, and immune aging, all of which contribute to the breakdown of immune tolerance and the onset of autoimmune disorders. Aloe vera (AV) has recently gained attention for its immunomodulatory, anti-inflammatory, and antioxidant properties. This review explores the effects of AV extracts and anthraquinones (e.g., aloe-emodin, emodin, aloin) on key inflammaging-driven mechanisms in autoimmunity. Our analysis highlights AV's ability to regulate hormone balance, autoantibody production, and cytokine/chemokine signaling (such as interleukin-1β, tumor necrosis factor-α, and interferon-γ). It modulates inflammatory pathways, including mitogen-activated protein kinases (MAPKs) and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), thereby inhibiting nuclear factor kappa-light-chain-enhancer of activated B-cell (NF-κB) activation. Additionally, AV enhances antioxidant defenses and restores immune balance by reducing Th1/Th17 subsets while promoting Th2-mediated regulation. Notably, AV also modulates inflammasome-mediated mechanisms and counteracts immunosenescence, which is driven by autophagy-related processes. These effects position AV as a potential integrative approach to mitigating inflammaging-driven autoimmunity. Furthermore, as inflammaging is increasingly recognized in onco-hematological diseases, AV-based strategies may offer novel therapeutic avenues. Future studies should focus on clinical validation, optimizing formulations, and expanding applications to broader age-related and immune-mediated disorders.
Collapse
Affiliation(s)
- Raffaele Cordiano
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.C.); (S.G.)
| | - Santino Caserta
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (A.A.)
| | - Paola Lucia Minciullo
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.C.); (S.G.)
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (A.A.)
| | - Sebastiano Gangemi
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.C.); (S.G.)
| |
Collapse
|
37
|
Rodríguez-Rodríguez S, Oviedo GR, López-de-Celis C, Bosch-Sabater J, Jovell-Fernández E, Pérez-Bellmunt A, Cuadra-Llopart L, Rodríguez-Sanz J. Stay Active, Stay Healthy: A Cross-Sectional View of the Impact of Physical Activity Levels on Health Parameters of Older Adults Institutionalized in Nursing Homes of Barcelona. Life (Basel) 2025; 15:412. [PMID: 40141756 PMCID: PMC11943667 DOI: 10.3390/life15030412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
(1) Background: Institutionalized older adults represent a vulnerable population. It is important to understand that higher levels of physical activity in older adults are associated with less risk of cardiovascular diseases, better cognition, and lower inflammaging and sarcopenia levels. The main objective was to evaluate the differences in health parameters in institutionalized older adults who perform different levels of weekly physical activity. The secondary objective was to analyze if weekly physical activity levels are a predictor of health parameters. (2) Methods: A cross-sectional observational study was conducted in nursing homes with adults over 75 years of age. A total of 76 participants was divided into three groups based on their weekly physical exercise frequency (1 day/week, 2 days/week, and 3 days/week). We measured demographic and anthropometric variables, along with cognitive level through the Mini Exam of Lobo. Handgrip strength, leg muscle strength, and power were also evaluated, and C-reactive protein levels were assessed through blood tests. Physical performance was measured using the Short Physical Performance Battery and walking speed. (3) Results: Significant differences were found in body mass index (p < 0.01; ES = 0.96), muscular strength (p < 0.01; ES = 0.70), and power (p < 0.01; ES = 1.09), Short Physical Performance Battery (p < 0.01; ES = 1.46) and walking speed (p < 0.01; ES = 0.87), cognitive function (p < 0.01; ES = 1.21), and C-reactive protein levels (p < 0.01; ES = 1.73), favoring the group who performed 3 days/week of physical activity. (4) Conclusions: Institutionalized older adults with three days per week of physical activity have greater physical and muscle function and less cognitive decline. Three days of weekly physical activity is associated with systemic inflammation and better cognitive status in institutionalized older adults.
Collapse
Affiliation(s)
- Sergi Rodríguez-Rodríguez
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Barcelona, Spain; (S.R.-R.); (J.B.-S.); (E.J.-F.); (A.P.-B.); (J.R.-S.)
- Actium Functional Anatomy Research Group, Sant Cugat del Vallès, 08195 Barcelona, Spain;
| | - Guillermo R. Oviedo
- Department of Kinesiology, Mississippi State University, Starkville, MS 38677, USA;
| | - Carlos López-de-Celis
- Actium Functional Anatomy Research Group, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Fundació Institut Universitari per a la Recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08007 Barcelona, Spain
| | - Joan Bosch-Sabater
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Barcelona, Spain; (S.R.-R.); (J.B.-S.); (E.J.-F.); (A.P.-B.); (J.R.-S.)
| | - Esther Jovell-Fernández
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Barcelona, Spain; (S.R.-R.); (J.B.-S.); (E.J.-F.); (A.P.-B.); (J.R.-S.)
- Actium Functional Anatomy Research Group, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Department of Epidemiology, Consorci Sanitari de Terrassa, 08227 Terrassa, Spain
| | - Albert Pérez-Bellmunt
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Barcelona, Spain; (S.R.-R.); (J.B.-S.); (E.J.-F.); (A.P.-B.); (J.R.-S.)
- Actium Functional Anatomy Research Group, Sant Cugat del Vallès, 08195 Barcelona, Spain;
| | - Leonor Cuadra-Llopart
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Barcelona, Spain; (S.R.-R.); (J.B.-S.); (E.J.-F.); (A.P.-B.); (J.R.-S.)
- Actium Functional Anatomy Research Group, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Department of Geriatric Medicine, Consorci Sanitari de Terrassa, 08227 Terrassa, Spain
| | - Jacobo Rodríguez-Sanz
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Barcelona, Spain; (S.R.-R.); (J.B.-S.); (E.J.-F.); (A.P.-B.); (J.R.-S.)
- Actium Functional Anatomy Research Group, Sant Cugat del Vallès, 08195 Barcelona, Spain;
| |
Collapse
|
38
|
Mahamud AGMSU, Tanvir IA, Kabir ME, Samonty I, Chowdhury MAH, Rahman MA. Gerobiotics: Exploring the Potential and Limitations of Repurposing Probiotics in Addressing Aging Hallmarks and Chronic Diseases. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10501-w. [PMID: 40029460 DOI: 10.1007/s12602-025-10501-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
As unhealthy aging continues to rise globally, there is a pressing need for effective strategies to promote healthy aging, extend health span, and address aging-related complications. Gerobiotics, an emerging concept in geroscience, offers a novel approach to repurposing selective probiotics, postbiotics, and parabiotics to modulate key aging processes and enhance systemic health. This review explores recent advancements in gerobiotics research, focusing on their role in targeting aging hallmarks, regulating longevity-associated pathways, and reducing risks of multiple age-related chronic conditions. Despite their promise, significant challenges remain, including optimizing formulations, ensuring safety and efficacy across diverse populations, and achieving successful clinical translation. Addressing these gaps through rigorous research, well-designed clinical trials, and advanced biotechnologies can establish gerobiotics as a transformative intervention for healthy aging and chronic disease prevention.
Collapse
Affiliation(s)
| | | | - Md Ehsanul Kabir
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| | - Ismam Samonty
- Department of Agricultural Chemistry, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Md Anamul Hasan Chowdhury
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Md Ashikur Rahman
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| |
Collapse
|
39
|
Gan D, Wang X, Wu X, Han S. Higher dietary intake of live microbes is inversely associated with accelerated biological aging. Food Funct 2025; 16:1809-1819. [PMID: 39927928 DOI: 10.1039/d4fo04230a] [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: 02/11/2025]
Abstract
Objective: It remains unclear whether dietary live microbe intake is associated with biological aging. Therefore, the present study aimed to investigate the associations between dietary live microbe intake and biological aging. Methods: Our study included 7719 participants aged 20 years and older from the 2007-2010 cycles of the National Health and Nutrition Examination Survey (NHANES). Participants were categorized into groups using two distinct methods. The first method grouped participants based on the live microbial level of the consumed foods, dividing them into three dietary live microbe intake groups: low, medium, and high. The second method classified participants according to the quantity of live microbe-containing foods they consumed (referred to as MedHi), with three groups: G1 (no MedHi intake), G2 (MedHi intake below the median), and G3 (MedHi intake above the median). Biological age was evaluated using the Klemera-Doubal method biological age (KDM-BA) and phenotypic age (PA). KDM-BA acceleration and PA acceleration was determined if participants' KDM-BA or PA exceeded their chronological age, respectively. Multivariable logistic regression models were conducted to explore the associations of dietary live microbe intake groups and dietary MedHi intake groups with the acceleration of KDM-BA and PA. Results: Compared with participants in the low dietary live microbe intake group, those in the high dietary live microbe intake group had a 20% (95% CI: 2% to 35%) reduced risk of KDM-BA acceleration and a 25% (95% CI: 4% to 42%) reduced risk of PA acceleration. Similarly, participants in the G3 group with higher dietary MedHi intake had a 24% (95% CI: 10% to 35%) lower risk of KDM-BA acceleration and a 29% (95% CI: 17% to 39%) lower risk of PA acceleration compared with those in the G1 group. The stratified analyses showed that the associations of dietary MedHi intake with accelerated KDM-BA and PA were not influenced by age, gender, race, smoking, alcohol consumption, physical activity, race, and history of hypertension, diabetes, and cardiovascular disease. Conclusion: Higher dietary intake of live microbes was inversely associated with accelerated biological aging. A diet rich in live microbes may help slow down the aging process.
Collapse
Affiliation(s)
- Da Gan
- Jiangxi Medicine Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Xiaoyan Wang
- Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Xuebiao Wu
- Center for Molecular Pathology, Department of Pathophysiology, Gannan Medical University, Ganzhou 341000, China
| | - Shuang Han
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
40
|
Kumasaka S, Negishi Y, Morita R, Migita M, Shima Y. Immunological role of zinc in preterm neonates. Immunol Med 2025; 48:78-88. [PMID: 39450996 DOI: 10.1080/25785826.2024.2420426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Zinc (Zn), an essential trace element, plays a significant role in fetal development and biological defense during the embryonic and neonatal periods. Therefore, exploring the kinetics of Zn related to immune disturbances in preterm neonates is important. We here performed the measurement of Zn concentration along with immunological analysis of neonates and investigated the role of Zn in the neonatal period. Serum Zn concentrations were measured immediately after birth in neonates (329 cases). Moreover, for 25 cases, the kinetics of various immune cells and cytokines were measured by flow cytometry and electrochemiluminescence. We observed that Zn levels were inversely correlated with gestational weeks. Immune cell and cytokine analysis revealed an inverse correlation between HLA-DR on monocytes and Zn levels and between inflammatory cytokine interleukin-12 and Zn levels. Furthermore, oxidative stress status was inversely correlated with Zn levels. Our results suggested that the Zn dynamics immediately after birth, which show a negative correlation with the gestational week, can provide an anti-inflammatory and anti-oxidative environment for preterm neonates. The increased Zn concentration in the blood of preterm neonates may consequently protect neonates from perinatal stress.
Collapse
Affiliation(s)
- Sakae Kumasaka
- Department of Pediatrics, Japan Red Cross Tokyo Katsushika Perinatal Center, Tokyo, Japan
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Yasuyuki Negishi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
- Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Makoto Migita
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
- Department of Pediatrics, Nippon Medical School Musashikosugi Hospital, Kanagawa, Japan
| | - Yoshio Shima
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
- Department of Neonatal Medicine, Nippon Medical School Musashikosugi Hospital, Kanagawa, Japan
| |
Collapse
|
41
|
Ng YY, Tay A. Exploring Lymph Node Stroma Ageing: Immune Implications and Future Directions. Aging Cell 2025; 24:e70000. [PMID: 39954244 PMCID: PMC11896299 DOI: 10.1111/acel.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/28/2024] [Accepted: 01/21/2025] [Indexed: 02/17/2025] Open
Abstract
Ageing is an inevitable biological process that impacts the immune system, leading to immunosenescence and inflammaging, which contribute to increased susceptibility to infections, autoimmune diseases and cancers in individuals over the age of 65. This review focuses on the ageing of lymph node stromal cells (LNSCs), which are crucial for maintaining lymph node (LN) structure and function. Age-related changes in LNs, such as fibrosis and lipomatosis, disrupt the LN architecture and reduce immune cell recruitment and function, impairing immune responses to infections and vaccinations. The review discusses the structural and functional decline of various LNSC subsets, including fibroblastic reticular cells (FRCs), lymphatic endothelial cells (LECs) and blood endothelial cells (BECs), highlighting their roles in immune cell activation and homeostasis. Potential strategies to restore aged LNSC function, such as enhancing LNSC activation during vaccination and using senotherapeutics, are explored. Outstanding questions regarding the mechanisms of LNSC ageing and how ageing of the LN stroma might impact autoimmune disorders are also addressed. This review aims to stimulate further research into the characterisation of aged LNSCs and the development of therapeutic interventions to improve immune function in the older adults.
Collapse
Affiliation(s)
- Yu Yang Ng
- Department of Biomedical EngineeringNational University of SingaporeSingapore CitySingapore
| | - Andy Tay
- Department of Biomedical EngineeringNational University of SingaporeSingapore CitySingapore
- Institute for Health Innovation & TechnologyNational University of SingaporeSingapore CitySingapore
- Tissue Engineering ProgrammeNational University of SingaporeSingapore CitySingapore
| |
Collapse
|
42
|
Chiappelli F. Protein disorder - function paradigm: Putative role in inflammation. Bioinformation 2025; 21:169-172. [PMID: 40322686 PMCID: PMC12044175 DOI: 10.6026/973206300210169] [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: 02/01/2025] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 05/08/2025] Open
Abstract
Traditional protein biochemistry defends the intimate interdependence between protein function and structure, the latter being consisting of four distinct levels: premary sstructure: viz, the sequence of its constituent amino acids linked by peptides bonds - the polypeptide chain; ssecondary structure: viz, localized folding patterns (e.g., a α-helix, β-sheets) of the polypeptide chain held by hydrogen bonds between amino acid backbones; tertiary structure: viz, three-dimensional folding of the protein held by interactions between amino acid side chains mediated by disulfide bridges, hydrogen bonds, ionic bonds and hydrophobic interactions; and quaternary structure: viz, attachment of subunits, when appropriate, by means of similar chemical interactions to form a functional protein complex. Research evidence in the last decade has described intrinsically disordered proteins (IDPs) as polypeptides that lack a well-defined three/four-dimensional structure under physiological conditions, appear structurally unstable and manifest in a dynamic set of possible conformations. IDPs are a major component of the "dark" proteome, genome protein products not yet characterized through experimental structure determination and existing homology modeling. Dark proteome in general and IDPs specifically define and characterize the novel disorder - function paradigm, which critically mediate and modulate key cellular organelles and pathways and influence physiopathological processes from aging to chronic diseases and pathogen infection. The role of the disorder-function paradigm in the immunome and the inflammasome in general and specifically in the process of chronic metabolic inflammation observed in aging - i.e., inflammAging - could be elucidated through diverse AI platforms.
Collapse
|
43
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. Harnessing Gasotransmitters to Combat Age-Related Oxidative Stress in Smooth Muscle and Endothelial Cells. Pharmaceuticals (Basel) 2025; 18:344. [PMID: 40143122 PMCID: PMC11946800 DOI: 10.3390/ph18030344] [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: 01/25/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Age-related oxidative stress is a critical factor in vascular dysfunction, contributing to hypertension and atherosclerosis. Smooth muscle cells and endothelial cells are particularly susceptible to oxidative damage, which exacerbates vascular aging through cellular senescence, chronic inflammation, and arterial stiffness. Gasotransmitters-hydrogen sulfide (H2S), nitric oxide (NO), and carbon monoxide (CO)-are emerging as promising therapeutic agents for counteracting these processes. This review synthesizes findings from recent studies focusing on the mechanisms by which H2S, NO, and CO influence vascular smooth muscle and endothelial cell function. Therapeutic strategies involving exogenous gasotransmitter delivery systems and combination therapies were analyzed. H2S enhances mitochondrial bioenergetics, scavenges ROS, and activates antioxidant pathways. NO improves endothelial function, promotes vasodilation, and inhibits platelet aggregation. CO exhibits cytoprotective and anti-inflammatory effects by modulating heme oxygenase activity and ROS production. In preclinical studies, gasotransmitter-releasing molecules (e.g., NaHS, SNAP, CORMs) and targeted delivery systems show significant promise. Synergistic effects with lifestyle modifications and antioxidant therapies further enhance their therapeutic potential. In conclusion, gasotransmitters hold significant promise as therapeutic agents to combat age-related oxidative stress in vascular cells. Their multifaceted mechanisms and innovative delivery approaches make them potential candidates for treating vascular dysfunction and promoting healthy vascular aging. Further research is needed to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
44
|
Jernigan JE, Staley HA, Baty Z, Bolen ML, Gomes BN, Holt J, Cole CL, Neighbarger NK, Dheeravath K, Merchak AR, Menees KB, Coombes SA, Tansey MG. RGS10 attenuates systemic immune dysregulation induced by chronic inflammatory stress. J Neuroinflammation 2025; 22:49. [PMID: 39994765 PMCID: PMC11852585 DOI: 10.1186/s12974-024-03322-1] [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: 11/01/2024] [Accepted: 12/10/2024] [Indexed: 02/26/2025] Open
Abstract
Regulator of G-protein signaling 10 (RGS10), a key homeostatic regulator of immune cells, has been implicated in multiple diseases associated with aging and chronic inflammation including Parkinson's Disease (PD). Interestingly, subjects with idiopathic PD display reduced levels of RGS10 in subsets of peripheral immune cells. Additionally, individuals with PD have been shown to have increased activated peripheral immune cells in cerebrospinal fluid (CSF) compared to age-matched healthy controls. However, it is unknown whether peripheral immune cells in the CSF of individuals with PD also exhibit decreased levels of RGS10. Utilizing the Michael J. Fox Foundation Parkinson's Progression Markers Initiative (PPMI) study we found that RGS10 levels are decreased in the CSF of individuals with PD compared to healthy controls and prodromal individuals. As RGS10 levels are decreased in the CSF and circulating peripheral immune cells of individuals with PD, we hypothesized that RGS10 regulates peripheral immune cell responses to chronic systemic inflammation (CSI) prior to the onset of neurodegeneration. To test this, we induced CSI for 6 weeks in C57BL6/J mice and RGS10 KO mice to assess circulating and CNS-associated immune cell responses. We found that RGS10 deficiency synergizes with CSI to induce a bias for inflammatory and cytotoxic cell populations, a reduction in antigen presentation machinery in peripheral blood immune cells, as well as in and around the brain that is most notable in males. These results highlight RGS10 as an important regulator of the systemic immune response to CSI and implicate RGS10 as a potential contributor to the development of immune dysregulation in PD.
Collapse
Affiliation(s)
- Janna E Jernigan
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Hannah A Staley
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Zachary Baty
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL, USA
| | - MacKenzie L Bolen
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Beatriz Nuñes Gomes
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jenny Holt
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Cassandra L Cole
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Noelle K Neighbarger
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kruthika Dheeravath
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Andrea R Merchak
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kelly B Menees
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Stephen A Coombes
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Malú Gámez Tansey
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
45
|
Wang L, Kuang Y, Zeng Y, Wan Z, Yang S, Li R. Association of systemic inflammation response index with latent tuberculosis infection and all-cause mortality: a cohort study from NHANES 2011-2012. Front Immunol 2025; 16:1538132. [PMID: 40046059 PMCID: PMC11880221 DOI: 10.3389/fimmu.2025.1538132] [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: 12/03/2024] [Accepted: 02/03/2025] [Indexed: 05/13/2025] Open
Abstract
Background The Systemic Inflammatory Response Index (SIRI) is a promising inflammatory marker; however, the relationship between SIRI and latent tuberculosis infection (LTBI), as well as its association with mortality rates, remains unclear. The present study aimed to explore the associations of the SIRI with LTBI and all-cause mortality. Methods We conducted a prospective cohort study using data from National Health and Nutrition Examination Survey (NHANES) cycles from 2011 to 2012. We explored the association between SIRI and LTBI prevalence using Multiple logistic regression models. We used Multivariate Cox proportional hazards model to analyze the association between SIRI and all-cause mortality. In addition, Kaplan-Meier curves, restricted cubic splines (RCS), stratified analysis, and interaction tests were performed. Results The study included a total of 4,983 adults who participated in NHANES 2011-2012. The mean follow-up period was 92.35 ± 16.82 months, and there were 525 deaths, representing a mortality rate of 10.54%. The occurrence of LTBI is significantly associated with low SIRI levels(OR=0.76, 95% CI: 0.66-0.89), after adjusting for confounders. Among LTBI patients, elevated SIRI levels are significantly correlated with an increased risk of all-cause mortality (adjusted HR = 1.48, 95% CI: 1.01-2.15). RCS revealed a linear relationship between SIRI and all-cause mortality in patients with LTBI (P =0.059[overall] and P = 0.391 [Nonlinear]). Furthermore, within the LTBI population, the area under the curve (AUC) of SIRI for all-cause mortality are 0.731 (1-year), 0.640 (3-year), and 0.634 (5-year). Conclusion The findings of this study indicate that elevated SIRI levels are independently associated with an increased risk of all-cause mortality in patients with LTBI. Notably, SIRI was significantly inversely associated with the incidence of LTBI. Therefore, SIRI may serve as an effective tool for risk stratification in adults with LTBI in the United States. Further research is needed to elucidate the underlying mechanisms and to explore any therapeutic implications of these findings.
Collapse
Affiliation(s)
- Lin Wang
- Infectious Disease Department, Jiangxi Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanchang, Jiangxi, China
| | - Yi Kuang
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Yan Zeng
- Infectious Disease Department, Jiangxi Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanchang, Jiangxi, China
| | - Zhihui Wan
- Infectious Disease Department, Jiangxi Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanchang, Jiangxi, China
| | - Sha Yang
- Infectious Disease Department, Jiangxi Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanchang, Jiangxi, China
| | - Renliang Li
- Infectious Disease Department, Jiangxi Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
46
|
Nisar A, Khan S, Pan Y, Hu L, Yang P, Gold NM, Zhou Z, Yuan S, Zi M, Mehmood SA, He Y. The Role of Hypoxia in Longevity. Aging Dis 2025:AD.2024.1630. [PMID: 39965249 DOI: 10.14336/ad.2024.1630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 02/15/2025] [Indexed: 02/20/2025] Open
Abstract
Aging is marked by a progressive decrease in physiological function and reserve capacity, which results in increased susceptibility to diseases. Understanding the mechanisms of driving aging is crucial for extending health span and promoting human longevity. Hypoxia, marked by reduced oxygen availability, has emerged as a promising area of study within aging research. This review explores recent findings on the potential of oxygen restriction to promote healthy aging and extend lifespan. While the role of hypoxia-inducible factor 1 (HIF-1) in cellular responses to hypoxia is well-established, its impact on lifespan remains complex and context-dependent. Investigations in invertebrate models suggest a role for HIF-1 in longevity, while evidence in mammalian models is limited. Hypoxia extends the lifespan independent of dietary restriction (DR), a known intervention underlying longevity. However, both hypoxia and DR converge on common downstream effectors, such as forkhead box O (FOXO) and flavin-containing monooxygenase (FMOs) to modulate the lifespan. Further work is required to elucidate the molecular mechanisms underlying hypoxia-induced longevity and optimize clinical applications. Understanding the crosstalk between HIF-1 and other longevity-associated pathways is crucial for developing interventions to enhance lifespan and healthspan. Future studies may uncover novel therapeutic strategies to promote healthy aging and longevity in human populations.
Collapse
Affiliation(s)
- Ayesha Nisar
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Sawar Khan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410083, China
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan
| | - Yongzhang Pan
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Li Hu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Pengyun Yang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Naheemat Modupeola Gold
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Zhen Zhou
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Shengjie Yuan
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Meiting Zi
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | | | - Yonghan He
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| |
Collapse
|
47
|
Izadi M, Sadri N, Abdi A, Raeis Zadeh MM, Sadatipour S, Baghdadi G, Jalaei D, Tahmasebi S. Harnessing the fundamental roles of vitamins: the potent anti-oxidants in longevity. Biogerontology 2025; 26:58. [PMID: 39920477 DOI: 10.1007/s10522-025-10202-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
Aging is a complex and heterogeneous biological process characterized by telomere attrition, genomic instability, mitochondrial dysfunction, and disruption in nutrient sensing. Besides contributing to the progression of cancer, metabolic disorders, and neurodegenerative diseases, these manifestations of aging also adversely affect organ function. It is crucial to understand these mechanisms and identify interventions to modulate them to promote healthy aging and prevent age-related diseases. Vitamins have emerged as potential modulators of aging beyond their traditional roles in health maintenance. There is an increasing body of evidence that hormetic effects of vitamins are responsible for activating cellular stress responses, repair mechanisms, and homeostatic processes when mild stress is induced by certain vitamins. It is evident from this dual role that vitamins play a significant role in preventing frailty, promoting resilience, and mitigating age-related cellular damage. Moreover, addressing vitamin deficiencies in the elderly could have a significant impact on slowing aging and extending life expectancy. A review of recent advances in the role of vitamins in delaying aging processes and promoting multiorgan health is presented in this article. The purpose of this paper is to provide a comprehensive framework for using vitamins as strategic tools for fostering longevity and vitality. It offers a fresh perspective on vitamins' role in aging research by bridging biological mechanisms and clinical opportunities.
Collapse
Affiliation(s)
- Mehran Izadi
- Department of Infectious and Tropical Diseases, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran
- Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
| | - Nariman Sadri
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran
- Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhossein Abdi
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran
- Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
- School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdi Raeis Zadeh
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran
- Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
- School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sana Sadatipour
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran
- Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
- Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ghazalnaz Baghdadi
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran
- Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
- Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Dorsa Jalaei
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran
- Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Safa Tahmasebi
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran.
- Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran.
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Araújo EM, Coelho-Júnior HJ, Sousa CV, Rosa TDS, Sousa Neto IV, Marzetti E, Franco OL, Aguiar SDS. Improving serum redox balance, inflammatory status, physical function, and cognitive ability through dual-task resistance training and detraining in nursing home residents. Exp Gerontol 2025; 200:112662. [PMID: 39701431 DOI: 10.1016/j.exger.2024.112662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/29/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVE This study investigated the effects of dual-task resistance training (RT) and detraining on physical function, cognitive capacity, lipid profile, renal function, oxidative stress markers, and chronic inflammation of institutionalized older adults. METHODS The study involved 11 older adults (83.09 ± 8.1 years) residing in a long-term care institution, spanning 42 weeks with assessments at weeks 1, 14-15, 28, and 42. The initial 12 weeks following the first assessment (weeks 2-13) served as a baseline, during which participants maintained their routine activities. A dual-task resistance training protocol was implemented from weeks 16 to 27, followed by a detraining period from weeks 29 to 41. Assessments included clinical characteristics, physical function, cognitive ability, blood samples for biochemical parameters, oxidative stress, and chronic inflammation. RESULTS Dual-task RT significantly enhanced balance (p = 0.027) and 4 m walking speed (p = 0.027) post-training compared to the baseline. It also decreased the completion time for the sit-and-stand test both post-training (p = 0.008) and post-detraining (p = 0.015) relative to baseline. Cognitive ability showed significant improvements (p < 0.05). The CAT/TBARS ratio increased significantly post-training (p < 0.001) and remained elevated post-detraining. Nitric Oxide levels increased post-training (p < 0.05) and stayed higher post-detraining. The IL-10/TNF-α ratio significantly increased post-training (p < 0.05). CONCLUSION Dual-task RT performed over 12 weeks improved physical function, cognitive capacity, muscular strength, oxidative stress markers, and chronic inflammation in institutionalized older adults. Furthermore, these benefits were sustained even after a period of detraining.
Collapse
Affiliation(s)
- Erivaldo Machado Araújo
- Postgraduate Program in Physical Education, Federal University of Mato Grosso, Cuiabá, MT, Brazil
| | - Hélio José Coelho-Júnior
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy; Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Caio Victor Sousa
- Department of Health and Human Sciences, Loyola Marymount University, Los Angeles, United States of America
| | - Thiago Dos Santos Rosa
- Graduate Program in Physical Education and Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasilia, Taguatinga, Brazil
| | - Ivo Vieira Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy; Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Octávio Luiz Franco
- Center for Proteomic and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, Brazil; S-Inova Biotech, Postgraduate Program in Biotechnology, Catholic University Dom Bosco (UCDB), Campo Grande, Brazil
| | - Samuel da Silva Aguiar
- Postgraduate Program in Physical Education, Federal University of Mato Grosso, Cuiabá, MT, Brazil; Center for Proteomic and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, Brazil.
| |
Collapse
|
49
|
Massaga C, Paul L, Kwiyukwa LP, Vianney JM, Chacha M, Raymond J. Computational analysis of Urolithin A as a potential compound for anti-inflammatory, antioxidant, and neurodegenerative pathways. Free Radic Biol Med 2025; 227:508-520. [PMID: 39643139 DOI: 10.1016/j.freeradbiomed.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/24/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Urolithin A, an active precursor derived from the metabolism of ellagitanins in rats and humans, is known for its potential health benefits, including stimulating mitophagy and promoting muscular skeletal function. While experimental studies have demonstrated Urolithin A's potential to enhance cellular health, the detailed molecular interactions through which Urolithin A exerts its effects are not fully elucidated. In this study, we investigated the anti-inflammatory, antioxidation and neuroprotective abilities of Urolithin A in selected targets using molecular docking and molecular dynamics simulation methods. Molecular docking studies revealed the strong affinity for receptors involved in inflammation activities, including human p38 MAP kinase (4DLI) with -10.1 kcal/mol interacting with SER252, LYS249, and ASP294 residues. The binding energy in the 5KIR target was -8.6 kcal/mol, interacting with GLN203 through hydrogen bond, and lastly, 1A9U with an affinity of -6.8 with no hydrogen bond formed with Urolithin A and interacts with van der Waals interactions. In oxidant targets, the influence of Urolithin was observed in 1OG5 with -7.9 kcal/mol interacting with GLN185, PHE447. For the 1M17 target, the binding affinity was -7.7 kcal/mol interacting with THR95 residue and 1ZXM target at -7.4 kcal/mol interacting with TYR36, TYR216, and LEU234 residues. The neuroprotective ability of urolithin A was observed in selected targets for acetylcholinesterase; the binding energy was -9.7 kcal/mol interacting with van der Waals and π interactions; for the 1GQR target, the binding energy was -9.9 kcal/mol interacting with van der Waals and π interactions and for β-amylase (1iyt) the binding energy was -5.5 forming hydrogen bond with SER8, GLN15 residues. Molecular Dynamics simulations at 100 ns of Urolithin A compared with reference 4DLI. The Urolithin A-4DLI complex exhibited greater stability than the reference receptor, as confirmed by RMSD, RMSF, Radius of Gyration, Hydrogen bond, and SASA analyses.
Collapse
Affiliation(s)
- Caroline Massaga
- School of Life Science and Bioengineering, The Nelson Mandela African Institution of Science and Technology, P.O. Box 447, Arusha, Tanzania.
| | - Lucas Paul
- Department of Chemistry, Dar es Salaam University College of Education, P.O. Box 2329, Dar es Salaam, Tanzania.
| | - Lucas P Kwiyukwa
- Chemistry Department, College of Natural and Applied Sciences, University of Dar es Salaam, P.O. Box 35061, Dar es Salaam, Tanzania.
| | - John-Mary Vianney
- School of Life Science and Bioengineering, The Nelson Mandela African Institution of Science and Technology, P.O. Box 447, Arusha, Tanzania.
| | - Musa Chacha
- School of Life Science and Bioengineering, The Nelson Mandela African Institution of Science and Technology, P.O. Box 447, Arusha, Tanzania.
| | - Jofrey Raymond
- School of Life Science and Bioengineering, The Nelson Mandela African Institution of Science and Technology, P.O. Box 447, Arusha, Tanzania.
| |
Collapse
|
50
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of chemokines in aging and age-related diseases. Mech Ageing Dev 2025; 223:112009. [PMID: 39631472 DOI: 10.1016/j.mad.2024.112009] [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: 06/01/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Chemokines (chemotactic cytokines) play essential roles in developmental process, immune cell trafficking, inflammation, immunity, angiogenesis, cellular homeostasis, aging, neurodegeneration, and tumorigenesis. Chemokines also modulate response to immunotherapy, and consequently influence the therapeutic outcome. The mechanisms underlying these processes are accomplished by interaction of chemokines with their cognate cell surface G protein-coupled receptors (GPCRs) and subsequent cellular signaling pathways. Chemokines play crucial role in influencing aging process and age-related diseases across various tissues and organs, primarily through inflammatory responses (inflammaging), recruitment of macrophages, and orchestrated trafficking of other immune cells. Chemokines are categorized in four distinct groups based on the position and number of the N-terminal cysteine residues; namely, the CC, CXC, CX3C, and (X)C. They mediate inflammatory responses, and thereby considerably impact aging process across multiple organ-systems. Therefore, understanding the underlying mechanisms mediated by chemokines may be of crucial importance in delaying and/or modulating the aging process and preventing age-related diseases. In this review, we highlight recent progress accomplished towards understanding the role of chemokines and their cellular signaling pathways involved in aging and age-relaed diseases of various organs. Moreover, we explore potential therapeutic strategies involving anti-chemokines and chemokine receptor antagonists aimed at reducing aging and mitigating age-related diseases. One of the modern methods in this direction involves use of chemokine receptor antagonists and anti-chemokines, which suppress the pro-inflammatory response, thereby helping in resolution of inflammation. Considering the wide-spectrum of functional involvements of chemokines in aging and associated diseases, several clinical trials are being conducted to develop therapeutic approaches using anti-chemokine and chemokine receptor antagonists to improve life span and promote healthy aging.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi 110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Akshay Bhardwaj
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad Road, Faridabad, Haryana 121001, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|