1
|
He Q, Cao Y, Fan X, Li B, He Q, Zhang H. Long-term prognostic value of CRP-albumin-lymphocyte index in elderly patients with heart failure with preserved ejection fraction. Exp Gerontol 2025; 204:112744. [PMID: 40179994 DOI: 10.1016/j.exger.2025.112744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a complex and heterogeneous clinical condition characterized by the interplay of malnutrition and immune inflammation, especially in elderly patients. The CRP-Albumin-Lymphocyte (CALLY) index, a novel composite indicator reflecting immune inflammation and nutritional status, has not yet been validated as a prognostic tool in elderly patients with HFpEF. METHODS This retrospective study included 320 elderly patients hospitalized at the Air Force Medical Center from October 2016 to April 2019 due to HFpEF. Patients were stratified into the all-cause mortality and the survival groups according to follow-up outcomes. Kaplan-Meier analysis and Cox regression were performed to identify risk factors associated with poor prognosis. Additionally, we constructed and evaluated a nomogram based on the CALLY index to predict survival rates. RESULTS During the follow-up period, 137 cases (42.81 %) of patients experienced all-cause mortality. Kaplan-Meier survival curves and Cox regression analysis revealed that a lower CALLY index (HR 0.811, 95 % CI 0.714-0.921, P = 0.001) was independently associated with adverse prognosis in elderly patients with HFpEF. The nomogram incorporating the CALLY index exhibited robust predictive performance for predicting 1-year, 3-year, and 5-year survival outcomes. CONCLUSION Our findings demonstrate that the CALLY index is an independent predictor of long-term mortality in elderly patients with HFpEF. The developed nomogram incorporating the CALLY index could effectively predict survival probabilities.
Collapse
Affiliation(s)
- Qingwei He
- The Fifth School of Clinical Medicine, Air Force Clinical Medical School, Anhui Medical University, 230032 Hefei, Anhui, China; Department of Cardiology, Air Force Medical Center, Air Force Medical University, PLA, 100142 Beijing, China
| | - Yukun Cao
- Department of Cardiology, Air Force Medical Center, Air Force Medical University, PLA, 100142 Beijing, China
| | - Xingman Fan
- Department of Cardiology, Air Force Medical Center, Air Force Medical University, PLA, 100142 Beijing, China; Graduate School, Hebei North University, 075000 Zhangjiakou, Hebei, China
| | - Bowen Li
- Graduate School, Hebei North University, 075000 Zhangjiakou, Hebei, China
| | - Qiongyi He
- The Fifth School of Clinical Medicine, Air Force Clinical Medical School, Anhui Medical University, 230032 Hefei, Anhui, China
| | - Haitao Zhang
- Department of Cardiology, Air Force Medical Center, Air Force Medical University, PLA, 100142 Beijing, China.
| |
Collapse
|
2
|
Zhang W, Kong D, Zhang X, Hu L, Nian Y, Shen Z. T cell aging and exhaustion: Mechanisms and clinical implications. Clin Immunol 2025; 275:110486. [PMID: 40120658 DOI: 10.1016/j.clim.2025.110486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
T cell senescence and exhaustion represent critical aspects of adaptive immune system dysfunction, with profound implications for health and the development of disease prevention and therapeutic strategies. These processes, though distinct, are interconnected at the molecular level, leading to impaired effector functions and reduced proliferative capacity of T cells. Such impairments increase susceptibility to diseases and diminish the efficacy of vaccines and treatments. Importantly, T cell senescence and exhaustion can dynamically influence each other, particularly in the context of chronic diseases. A deeper understanding of the molecular mechanisms underlying T cell senescence and exhaustion, as well as their interplay, is essential for elucidating the pathogenesis of related diseases and restoring dysfunctional immune responses. This knowledge will pave the way for the development of targeted therapeutic interventions and strategies to enhance immune competence. This review aims to summarize the characteristics, mechanisms, and disease associations of T cell senescence and exhaustion, while also delineating the distinctions and intersections between these two states to enhance our comprehension.
Collapse
Affiliation(s)
- Weiqi Zhang
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Dejun Kong
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Xiaohan Zhang
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Lu Hu
- Tianjin Medical University First Central Clinical College, Tianjin, China.
| | - Yeqi Nian
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Science, Tianjin, China; Department of Kidney Transplant, Tianjin First Central Hospital, Tianjin, China.
| | - Zhongyang Shen
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Science, Tianjin, China.
| |
Collapse
|
3
|
Moadab A, Khorramdelazad H, Javar MTA, Nejad MSM, Mirzaie S, Hatami S, Mahdavi N, Ghaffari S, Yazdian FA. Unmasking a Paradox: Roles of the PD-1/PD-L1 Axis in Alzheimer's Disease-Associated Neuroinflammation. J Neuroimmune Pharmacol 2025; 20:46. [PMID: 40285967 DOI: 10.1007/s11481-025-10206-5] [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: 01/24/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Alzheimer's disease (AD) represents the most prevalent form of dementia, characterized by progressive cognitive impairment and chronic neuroinflammation. Immune checkpoint inhibitors (ICIs), including anti-programmed cell death (PD)-1 and anti-PD-L1, signify a revolutionary advancement in cancer treatment by preventing T-cell exhaustion; however, their therapeutic application in AD presents a conundrum. Hypothesis: Recent preclinical studies indicate that PD-1 inhibition in AD mouse models induces an interferon-gamma (IFN-γ)-mediated response, leading to increased recruitment of monocyte-derived macrophages into the brain, enhanced clearance of amyloid-beta (Aβ) plaques, and improved cognitive performance. Nonetheless, this therapeutic effect is counterbalanced by the potential for exacerbated neuroinflammation, as PD-1/PD-L1 blockade may potentiate pro-inflammatory T helper (Th)1 and Th17 responses. In this review, we critically discuss the pertinent pro-inflammatory and neuroprotective facets of T cell biology in the pathogenesis of AD, emphasizing the potential for modulation of the PD-1/PD-L1 axis to influence both Aβ clearance and the dynamics of neuroinflammatory processes. In summary, we determine that ICIs are promising tools for reducing AD pathology and improving cognition. However, it is essential to refine treatment protocols and carefully select patients to optimize neuroprotective effects while adequately considering inflammatory risks.
Collapse
Affiliation(s)
- Ali Moadab
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Mohammad Taha Akbari Javar
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Saber Mohammadian Nejad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Shahrzad Mirzaie
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sina Hatami
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Nima Mahdavi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Saeed Ghaffari
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Askari Yazdian
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
4
|
Huang S, Kang Y, Liu T, Xiong Y, Yang Z, Zhang Q. The role of immune checkpoints PD-1 and CTLA-4 in cardiovascular complications leading to heart failure. Front Immunol 2025; 16:1561968. [PMID: 40255399 PMCID: PMC12006013 DOI: 10.3389/fimmu.2025.1561968] [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: 01/16/2025] [Accepted: 02/13/2025] [Indexed: 04/22/2025] Open
Abstract
Immune checkpoints, such as PD-1 and CTLA-4, are crucial regulators of immune responses, acting as gatekeepers to balance immunity against foreign antigens and self-tolerance. These checkpoints play a key role in maintaining cardiac homeostasis by preventing immune-mediated damage to critical organs like the heart. In this study, we explored the involvement of PD-1 and CTLA-4 in cardiovascular complications, particularly atherosclerosis and myocarditis, which can lead to heart failure. We conducted a comprehensive analysis using animal models and clinical data to assess the effects of immune checkpoint inhibition on cardiac function. Our findings indicate that disruption of PD-1 and CTLA-4 pathways exacerbates myocardial inflammation, accelerates atherosclerotic plaque formation, and promotes the development of heart failure. Additionally, we observed that immune checkpoint inhibition in these models led to increased infiltration of T lymphocytes, higher levels of pro-inflammatory cytokines, and enhanced tissue damage. These results suggest that PD-1 and CTLA-4 are critical in preserving cardiac health, and their inhibition can result in severe cardiovascular toxicity. Our study emphasizes the need for careful monitoring of cardiovascular health in patients undergoing immune checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Shoulian Huang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiology, The Second People’s Hospital of Yibin, Yibin, Sichuan, China
| | - Yu Kang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Xiong
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zixuan Yang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qing Zhang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Lee DY, Noren Hooten N, O'Connell JF, Lee BY, Kim Y. The Role of Ginseng and Its Bioactive Compounds in Aging: Cells and Animal Studies. Annu Rev Food Sci Technol 2025; 16:333-354. [PMID: 39971378 DOI: 10.1146/annurev-food-111523-121753] [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] [Indexed: 02/21/2025]
Abstract
Aging is an inevitable process that is characterized by physiological deterioration and increased vulnerability to stressors. Therefore, the interest in hallmarks, mechanisms, and ways to delay or prevent aging has grown for decades. Natural plant products and their bioactive compounds have been studied as a promising strategy to overcome aging. Ginseng, a traditional herbal medicine, and its bioactive compound, the ginsenosides, have increasingly gained attention because of various pharmacological functions. This review introduces the species, useful parts, characteristics, and active components of ginseng. It primarily focuses on the bioconversion of ginsenosides through the unique steaming and drying process. More importantly, this review enumerates the antiaging mechanisms of ginseng, ginsenosides, and other bioactive compounds, highlighting their potential to extend the health span and mitigate age-related diseases based on twelve representative hallmarks of aging.
Collapse
Affiliation(s)
- Da-Yeon Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, USA;
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, Maryland, USA
| | - Jennifer F O'Connell
- Center for Scientific Review, National Institutes of Health, Bethesda, Maryland, USA
| | - Boo-Yong Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea;
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, USA;
| |
Collapse
|
6
|
Mao W, Liu X, Fan S, Zhang R, Liu M, Xiao S. Modulating oxidative stress: a reliable strategy for coping with community-acquired pneumonia in older adults. Front Med (Lausanne) 2025; 12:1549658. [PMID: 40206465 PMCID: PMC11979195 DOI: 10.3389/fmed.2025.1549658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Community-acquired pneumonia (CAP) remains one of the leading respiratory diseases worldwide. With the aging of the global population, the morbidity, criticality and mortality rates of CAP in older adults remain high every year. Modulating the signaling pathways that cause the inflammatory response and improve the immune function of patients has become the focus of reducing inflammatory damage in the lungs, especially CAP in older adults. As an important factor that causes the inflammatory response of CAP and affects the immune status of the body, oxidative stress plays an important role in the occurrence, development and treatment of CAP. Furthermore, in older adults with CAP, oxidative stress is closely associated with immune senescence, sarcopenia, frailty, aging, multimorbidity, and polypharmacy. Therefore, multiple perspectives combined with the disease characteristics of older adults with CAP were reviewed to clarify the research progress and application value of modulating oxidative stress in older adults with CAP. Clearly, there is no doubt that targeted modulation of oxidative stress benefits CAP in older adults. However, many challenges and unknowns concerning how to modulate oxidative stress for further practical clinical applications exist, and more targeted research is needed. Moreover, the limitations and challenges of modulating oxidative stress are analyzed with the aim of providing references and ideas for future clinical treatment or further research in older adults with CAP.
Collapse
Affiliation(s)
- Weixu Mao
- Department of Respiratory Medicine, The Affiliated Yongchuan Traditional Chinese Medicine Hospital of Chongqing Medical University, Chongqing, China
| | - Xuanjun Liu
- Department of General Surgery, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Senji Fan
- Department of Respiratory Medicine, The Affiliated Yongchuan Traditional Chinese Medicine Hospital of Chongqing Medical University, Chongqing, China
| | - Ruibin Zhang
- Department of Respiratory Medicine, The Affiliated Yongchuan Traditional Chinese Medicine Hospital of Chongqing Medical University, Chongqing, China
| | - Miao Liu
- Department of Respiratory Medicine, The Affiliated Yongchuan Traditional Chinese Medicine Hospital of Chongqing Medical University, Chongqing, China
| | - Shunqiong Xiao
- Department of Respiratory Medicine, The Affiliated Yongchuan Traditional Chinese Medicine Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Zeydan B, Neyal N, Nathoo N, Rangachari M, Atkinson EJ, Son J, Conway BL, Tobin WO, Keegan BM, Weinshenker BG, Kantarci K, Oh J, Kantarci OH. Effects of androgen modifying therapies on disease activity in older men with multiple sclerosis. J Neuroimmunol 2025; 403:578589. [PMID: 40139128 DOI: 10.1016/j.jneuroim.2025.578589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Anti-inflammatory properties of androgens were assessed in animal models, but only several clinical studies investigated the effects of androgen on multiple sclerosis (MS). Inflammatory activity in MS often attenuates with aging, and androgen modifying therapies (AMT), which mimic decreased androgen levels, are frequently used in older men. We aimed to investigate if the number of disease activity events would increase in older (≥55 years) male persons with MS (MPwMS) who are on AMT. METHODS MPwMS with AMT initiation ≥55 years (AMT; n = 60) and without AMT history (no-AMT; n = 80) were included from a clinical-based observational study cohort. Clinical (relapses), Radiological (new lesions), and Disease (relapses and/or new lesions) activity events were evaluated before and after the age at AMT initiation in AMT and no-AMT groups. RESULTS Age at MS onset, progressive MS rate and treatment frequencies were similar between the groups. When events before and after the age at AMT initiation (mean = 65.0 ± 7.0 years) were compared, there was a drop in the percentage of individuals with Clinical (38.5 % vs. 10 %), Radiological (46.2 % vs. 40 %) and Disease (62.8 % vs. 40 %) activity events in the no-AMT group. In the AMT group, the percentage of individuals with Clinical activity events was not as dramatically decreased (33.3 % vs. 22.2 %), Radiological activity events was increased (35.2 % vs. 46.7 %), and Disease activity events was sustained (51.9 % vs. 51.1 %). The probability of Disease activity was higher in the AMT group at 3 years of AMT initiation (40 % vs. 29 %) compared to the no-AMT group matched for disease duration, but the difference was not significant (p = 0.582). CONCLUSIONS Rather than the expected decrease in disease activity with age, MPwMS receiving AMT experienced sustained or increased disease activity, particularly at >65 years. Close clinical monitoring of MPwMS starting on these medications is necessary.
Collapse
Affiliation(s)
- Burcu Zeydan
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Radiology, Mayo Clinic, Rochester, MN, USA; Women's Health Research Center, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.
| | - Nur Neyal
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Nabeela Nathoo
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Manu Rangachari
- Centre de recherche du CHU de Québec, axe Neurosciences, Université Laval, Québec City, QC, Canada; Faculté de médecine, Université Laval, Québec City, QC, Canada
| | | | - Jiye Son
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Brittani L Conway
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - W Oliver Tobin
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - B Mark Keegan
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Brian G Weinshenker
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, University of Virginia, VA, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Women's Health Research Center, Mayo Clinic, Rochester, MN, USA
| | - Jiwon Oh
- Department of Neurology, University of Toronto, Toronto, ON, Canada
| | - Orhun H Kantarci
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
8
|
Chen L, Yao K, Fu QL. Potential immune involvement in cataract: from mechanisms to future scope of therapies. Int J Ophthalmol 2025; 18:541-548. [PMID: 40103951 PMCID: PMC11865660 DOI: 10.18240/ijo.2025.03.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/28/2024] [Indexed: 03/20/2025] Open
Abstract
The immune system is involved in many age-related pathological changes, also plays an important role in tissue regeneration after injury. But no immune involvement has been discussed regarding cataract since it is presumed that lens has no source of immune cells as an avascular zone. Latest research has challenged the longstanding view of the lens as an immune-privileged tissue, revealing the presence of resident immune cells and active immune responses within the lens. Thus, we summarized the immune involvement in maintaining lens homeostasis, which may be a deleterious role in the induction of lens opacification if inappropriately activated. Furthermore, bioengineer-based immunomodulatory therapies to fine-tune the micro immune environment within lens may be future strategies for in situ lens regeneration, as a novel treatment for cataract.
Collapse
Affiliation(s)
- Lu Chen
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, Zhejiang Province, China
| | - Ke Yao
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, Zhejiang Province, China
| | - Qiu-Li Fu
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, Zhejiang Province, China
| |
Collapse
|
9
|
Francavilla F, Intranuovo F, La Spada G, Lacivita E, Catto M, Graps EA, Altomare CD. Inflammaging and Immunosenescence in the Post-COVID Era: Small Molecules, Big Challenges. ChemMedChem 2025; 20:e202400672. [PMID: 39651728 DOI: 10.1002/cmdc.202400672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/11/2024]
Abstract
Aging naturally involves a decline in biological functions, often triggering a disequilibrium of physiological processes. A common outcome is the altered response exerted by the immune system to counteract infections, known as immunosenescence, which has been recognized as a primary cause, among others, of the so-called long-COVID syndrome. Moreover, the uncontrolled immunoreaction leads to a state of subacute, chronic inflammatory state known as inflammaging, responsible in turn for the chronicization of concomitant pathologies in a self-sustaining process. Anti-inflammatory and immunosuppressant drugs are the current choice for the therapy of inflammaging in post-COVID complications, with contrasting results. The increasing knowledge of the biochemical pathways of inflammaging led to disclose new small molecules-based therapies directed toward different biological targets involved in inflammation, immunological response, and oxidative stress. Herein, paying particular attention to recent clinical data and preclinical literature, we focus on the role of endocannabinoid system in inflammaging, and the promising therapeutic option represented by the CB2R agonists, the role of novel ligands of the formyl peptide receptor 2 and ultimately the potential of newly discovered monoamine oxidase (MAO) inhibitors with neuroprotective activity in the treatment of immunosenescence.
Collapse
Affiliation(s)
- Fabio Francavilla
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Francesca Intranuovo
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Gabriella La Spada
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Enza Lacivita
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Elisabetta Anna Graps
- ARESS Puglia - Agenzia Regionale strategica per la Salute ed il Sociale, Lungomare Nazario Sauro 33, 70121, Bari, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| |
Collapse
|
10
|
Wang L, Tang D. Immunosenescence promotes cancer development: from mechanisms to treatment strategies. Cell Commun Signal 2025; 23:128. [PMID: 40065335 PMCID: PMC11892258 DOI: 10.1186/s12964-025-02082-6] [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: 11/20/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
The body's innate immune system plays a pivotal role in identifying and eliminating cancer cells. However, as the immune system ages, its functionality can deteriorate, becoming dysfunctional, inefficient, or even inactive-a condition referred to as immunosenescence. This decline significantly increases the risk of malignancies. While the pro-cancer effects of T-cell aging have been widely explored, there remains a notable gap in the literature regarding the impact of aging on innate immune cells, such as macrophages and neutrophils. This review seeks to address this gap, with emphasis on these cell types. Furthermore, although certain cancer immunotherapies, including immune checkpoint inhibitors (ICIs), have demonstrated efficacy across a broad spectrum of cancers, elderly patients are less likely to derive clinical benefit from these treatments. In some cases, they may even experience immune-related adverse events (irAEs). While senolytic strategies have shown promise in exerting anti-cancer effects, their adverse reactions and potential off-target effects present significant challenges. This review aims to elucidate the pro-cancer effects of immunosenescence, its implications for the efficacy and safety of ICIs, and potential anti-aging treatment strategies. In addition, optimizing anti-aging therapies to minimize adverse reactions and enhance therapeutic outcomes remains a critical focus for future research endeavors.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou, People's Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University; Northern Jiangsu People's Hospital; The Yangzhou Clinical Medical College of Xuzhou Medical University; The Yangzhou School of Clinical Medicine of Dalian Medical University; The Yangzhou School of Clinical Medicine of Nanjing Medical University; Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, 225000, China.
| |
Collapse
|
11
|
von Bernhardi R, Eugenín J. Ageing-related changes in the regulation of microglia and their interaction with neurons. Neuropharmacology 2025; 265:110241. [PMID: 39617175 DOI: 10.1016/j.neuropharm.2024.110241] [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: 06/05/2024] [Revised: 09/24/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024]
Abstract
Ageing is one of the most important risk factors for chronic health conditions, including neurodegenerative diseases. Inflammation is a feature of ageing, as well as a key pathophysiological mechanism for degenerative diseases. Microglia play multiple roles in the central nervous system; their states entail a complex assemblage of responses reflecting the multiplicity of functions they fulfil both under homeostatic basal conditions and in response to stimuli. Whereas glial cells can promote neuronal homeostasis and limit neurodegeneration, age-related inflammation (i.e. inflammaging) leads to the functional impairment of microglia and astrocytes, exacerbating their response to stimuli. Thus, microglia are key mediators for age-dependent changes of the nervous system, participating in the generation of a less supportive or even hostile environment for neurons. Whereas multiple changes of ageing microglia have been described, here we will focus on the neuron-microglia regulatory crosstalk through fractalkine (CX3CL1) and CD200, and the regulatory cytokine Transforming Growth Factor β1 (TGFβ1), which is involved in immunomodulation and neuroprotection. Ageing results in a dysregulated activation of microglia, affecting neuronal survival, and function. The apparent unresponsiveness of aged microglia to regulatory signals could reflect a restriction in the mechanisms underlying their homeostatic and reactive states. The spectrum of functions, required to respond to life-long needs for brain maintenance and in response to disease, would progressively narrow, preventing microglia from maintaining their protective functions. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Universidad San Sebastian, Faculty for Odontology and Rehabilitation Sciences. Lota 2465, Providencia, Santiago, PO. 7510602, Chile.
| | - Jaime Eugenín
- Universidad de Santiago de Chile, Faculty of Chemistry and Biology, Av. Libertador Bernardo O'Higgins 3363, Santiago, PO. 7510602, Chile.
| |
Collapse
|
12
|
Yang Y, Pan M, Zhu W, Luo X, Liang X. Association between blood heavy metals exposure with uterine fibroids among American women: a cross-sectional analysis from NHANES data. BMC Womens Health 2025; 25:68. [PMID: 39966823 PMCID: PMC11834190 DOI: 10.1186/s12905-025-03596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Excessive exposure to heavy metals has been linked to various health problems, including organ damage, neurological disorders, and reproductive and developmental abnormalities. However, the relationship between heavy metals exposure and uterine fibroids remains uncertain. To explore this association, we conducted a cross-sectional study among American women. METHODS We utilized data from three cycles of the National Health and Nutrition Examination Survey (NHANES, 1999-2006) to evaluate the association between uterine fibroids and blood heavy metal levels, including lead (Pb), cadmium (Cd), and mercury (Hg). Weighted logistic regression, restricted cubic spline (RCS), Bayesian kernel machine regression (BKMR), and subgroup analyse were used to examine the potential relationships between blood heavy metals and uterine fibroids. RESULTS Of the 4502 American women studied, 542 (12.04%) had uterine fibroids. Elevated levels of all heavy metals were significantly more common in women with uterine fibroids (P < 0.001). Blood Hg levels were notably associated with uterine fibroid prevalence in the adjusted model (OR = 1.41, 95% CI: 1.06-1.89, p = 0.03). Similar patterns were partly observed for blood Pb and Cd. Age and marital status were significant interaction factors concerning Hg exposure (P for interaction < 0.05). A dose-response relationship with an inflection point at 7µmol/L was identified for Hg, and BKMR models indicated a positive association between mixed heavy metal exposure and uterine fibroid risk. CONCLUSIONS Exposure to blood heavy metals, particularly Hg, is significantly associated with an elevated risk of uterine fibroids. Further prospective studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- Yaqin Yang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Meijun Pan
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Wenyuan Zhu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Xukai Luo
- Foshan Nanhai Vocational School of Health, Foshan, 528211, China
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xuefang Liang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
| |
Collapse
|
13
|
Schaefer AS, Nibali L, Zoheir N, Moutsopoulos NM, Loos BG. Genetic risk variants implicate impaired maintenance and repair of periodontal tissues as causal for periodontitis-A synthesis of recent findings. Periodontol 2000 2025. [PMID: 39953674 DOI: 10.1111/prd.12622] [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: 05/15/2024] [Revised: 12/18/2024] [Accepted: 12/29/2024] [Indexed: 02/17/2025]
Abstract
Periodontitis is a complex inflammatory disease in which the host genome, in conjunction with extrinsic factors, determines susceptibility and progression. Genetic predisposition is the strongest risk factor in the first decades of life. As people age, chronic exposure to the periodontal microbiome puts a strain on the proper maintenance of barrier function. This review summarizes our current knowledge on genetic risk factors implicated in periodontitis, derived (i) from hypothesis-free systematic whole genome-profiling studies (genome-wide association studies [GWAS] and quantitative trait loci [QTL] mapping studies), and independently validated through further unbiased approaches; (ii) from monogenic and oligogenic forms of periodontitis; and (iii) from syndromic forms of periodontitis. The genes include, but are not limited to, SIGLEC5, PLG, ROBO2, ABCA1, PF4, and CTSC. Notably, CTSC and PLG gene mutations were also identified in non-syndromic and syndromic forms of prepubertal and early-onset periodontitis. The functions of the identified genes in this review suggest that the pathways affected by the periodontitis-associated gene variants converge in functions involved in the maintenance and repair of structural integrity of the periodontal tissues. Particularly, these genes play a role in the healing of inflamed and ulcerated periodontal tissues, including roles in fibrinolysis, extrusion of cellular debris, extracellular matrix remodeling and angiogenesis. Syndromes that include periodontitis in their phenotype indicate that neutrophils play an important role in the regulation of inflammation in the periodontium. The established genetic susceptibility genes therefore collectively provide new insights into the molecular mechanisms and plausible causal factors underlying periodontitis.
Collapse
Affiliation(s)
- Arne S Schaefer
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Luigi Nibali
- Periodontology Unit, Faculty of Dentistry, Oral and Craniofacial Sciences, Centre for Host Microbiome Interactions, King's College London, London, UK
| | - Noha Zoheir
- Periodontology Unit, Faculty of Dentistry, Oral and Craniofacial Sciences, Centre for Host Microbiome Interactions, King's College London, London, UK
| | - Niki M Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Bruno G Loos
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Alexeeva E, Dvoryakovskaya T, Fetisova A, Kriulin I, Krekhova E, Kabanova A, Labinov V, Labinova E, Kostik M. The Efficacy and Safety of Simultaneous Vaccination with Polysaccharide Conjugate Vaccines Against Pneumococcal (13-Valent Vaccine) and Haemophilus influenzae Type b Infections in Children with Juvenile Idiopathic Arthritis Without Systemic Manifestations: A Prospective Cohort Study. Vaccines (Basel) 2025; 13:177. [PMID: 40006724 PMCID: PMC11860303 DOI: 10.3390/vaccines13020177] [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: 01/04/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Immunosuppressive therapy (methotrexate and biological agents) for juvenile idiopathic arthritis (JIA) is associated with an increased risk of severe infections, higher infection rates, treatment interruptions, failure to achieve disease remission, and recurrent disease flares. Our study aimed to evaluate the safety and efficacy of simultaneous immunization with 13-valent polysaccharide conjugate vaccines (PCV13) against S. pneumoniae (SP) and Hemophilus influanzae type b infections (HibV) in children with JIA without systemic manifestations. Methods: A total of 371 non-systemic JIA patients who received 13PCV and HibV were included in this prospective cohort study. In every patient, we evaluated clinical, laboratory, anti-SP, and anti-Hib IgG antibodies before vaccination, three weeks after, and six months after, and all adverse events (AEs) were collected during the study. The number and duration of acute respiratory infection (ARI) episodes and requirements for antibacterial treatment and AE six months before and after the baseline were collected. Results: The levels of the Ig G anti-SP and anti-Hib antibodies increased in the 3 weeks after vaccination; then, anti-SP antibodies slightly decreased and anti-Hib antibodies remained increased during the whole study, as well as in a part of the patients with a protective titer. During the study, there were no patients with significant flares, and the main JIA outcomes gradually decreased during the trial. The number of patients with uveitis remained equal, as well as the part of the patients with active, low-active, and inactive uveitis. There was no significant rise in the hs-CRP or S100 protein after the vaccination. Previous or ongoing treatment with non-biological (p = 0.072) and biological (p = 0.019) disease-modified anti-rheumatic drugs affected the Hib and did not affect the anti-SP protective titer at the end of the study. Within 6 months following vaccination, the number of ARI episodes (p < 0.001) and the number of courses of antibacterial treatment (p < 0.0001) decreased twice. The median duration of ARI episodes decreased four times (p < 0.0001). Mild AEs (injection site reactions and short-term fever episodes) were found in 58 (15.6%) patients with JIA, and 1 patient (0.2%) developed an SAE. Conclusions: Simultaneous vaccination against pneumococcal and Hib infections reduces the frequency and duration of episodes of ARI, as well as the number of courses of antibacterial drugs, and does not lead to significant JIA flares. The number of reported AEs is consistent with what was expected.
Collapse
Affiliation(s)
- Ekaterina Alexeeva
- Department of Pediatric Rheumatology, National Medical Research Center of Children’s Health, Moscow 119991, Russia; (E.A.); (T.D.); (A.F.); (I.K.); (E.K.); (A.K.)
- Clinical Institute of Children’s Health Named After N.F. Filatov, Department of Pediatrics and Pediatric Rheumatology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia (E.L.)
| | - Tatyana Dvoryakovskaya
- Department of Pediatric Rheumatology, National Medical Research Center of Children’s Health, Moscow 119991, Russia; (E.A.); (T.D.); (A.F.); (I.K.); (E.K.); (A.K.)
- Clinical Institute of Children’s Health Named After N.F. Filatov, Department of Pediatrics and Pediatric Rheumatology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia (E.L.)
| | - Anna Fetisova
- Department of Pediatric Rheumatology, National Medical Research Center of Children’s Health, Moscow 119991, Russia; (E.A.); (T.D.); (A.F.); (I.K.); (E.K.); (A.K.)
| | - Ivan Kriulin
- Department of Pediatric Rheumatology, National Medical Research Center of Children’s Health, Moscow 119991, Russia; (E.A.); (T.D.); (A.F.); (I.K.); (E.K.); (A.K.)
| | - Elizaveta Krekhova
- Department of Pediatric Rheumatology, National Medical Research Center of Children’s Health, Moscow 119991, Russia; (E.A.); (T.D.); (A.F.); (I.K.); (E.K.); (A.K.)
| | - Anna Kabanova
- Department of Pediatric Rheumatology, National Medical Research Center of Children’s Health, Moscow 119991, Russia; (E.A.); (T.D.); (A.F.); (I.K.); (E.K.); (A.K.)
| | - Vladimir Labinov
- Clinical Institute of Children’s Health Named After N.F. Filatov, Department of Pediatrics and Pediatric Rheumatology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia (E.L.)
| | - Elizaveta Labinova
- Clinical Institute of Children’s Health Named After N.F. Filatov, Department of Pediatrics and Pediatric Rheumatology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia (E.L.)
| | - Mikhail Kostik
- Hospital Pediatry, Saint-Petersburg State Pediatric Medical University, Saint-Petersburg 194100, Russia
| |
Collapse
|
15
|
Li C, Yuan Y, Jia Y, Zhou Q, Wang Q, Jiang X. Cellular senescence: from homeostasis to pathological implications and therapeutic strategies. Front Immunol 2025; 16:1534263. [PMID: 39963130 PMCID: PMC11830604 DOI: 10.3389/fimmu.2025.1534263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Cellular aging is a multifactorial and intricately regulated physiological process with profound implications. The interaction between cellular senescence and cancer is complex and multifaceted, senescence can both promote and inhibit tumor progression through various mechanisms. M6A methylation modification regulates the aging process of cells and tissues by modulating senescence-related genes. In this review, we comprehensively discuss the characteristics of cellular senescence, the signaling pathways regulating senescence, the biomarkers of senescence, and the mechanisms of anti-senescence drugs. Notably, this review also delves into the complex interactions between senescence and cancer, emphasizing the dual role of the senescent microenvironment in tumor initiation, progression, and treatment. Finally, we thoroughly explore the function and mechanism of m6A methylation modification in cellular senescence, revealing its critical role in regulating gene expression and maintaining cellular homeostasis. In conclusion, this review provides a comprehensive perspective on the molecular mechanisms and biological significance of cellular senescence and offers new insights for the development of anti-senescence strategies.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Yixiao Yuan
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - YingDong Jia
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Zhou
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Xiulin Jiang
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
16
|
Li S, Wang K, Wu J, Zhu Y. The immunosenescence clock: A new method for evaluating biological age and predicting mortality risk. Ageing Res Rev 2025; 104:102653. [PMID: 39746402 DOI: 10.1016/j.arr.2024.102653] [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: 09/25/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Precisely assessing an individual's immune age is critical for developing targeted aging interventions. Although traditional methods for evaluating biological age, such as the use of cellular senescence markers and physiological indicators, have been widely applied, these methods inherently struggle to capture the full complexity of biological aging. We propose the concept of an 'immunosenescence clock' that evaluates immune system changes on the basis of changes in immune cell abundance and omics data (including transcriptome and proteome data), providing a complementary indicator for understanding age-related physiological transformations. Rather than claiming to definitively measure biological age, this approach can be divided into a biological age prediction clock and a mortality prediction clock. The main function of the biological age prediction clock is to reflect the physiological state through the transcriptome data of peripheral blood mononuclear cells (PBMCs), whereas the mortality prediction clock emphasizes the ability to identify people at high risk of mortality and disease. We hereby present nearly all of the immunosenescence clocks developed to date, as well as their functional differences. Critically, we explicitly acknowledge that no single diagnostic test can exhaustively capture the intricate changes associated with biological aging. Furthermore, as these biological functions are based on the acceleration or delay of immunosenescence, we also summarize the factors that accelerate immunosenescence and the methods for delaying it. A deep understanding of the regulatory mechanisms of immunosenescence can help establish more accurate immune-age models, providing support for personalized longevity interventions and improving quality of life in old age.
Collapse
Affiliation(s)
- Shuyu Li
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ke Wang
- Department of Breast Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingni Wu
- Department of International Healthcare Center and General Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yongliang Zhu
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
17
|
Zhou H, Zheng Z, Fan C, Zhou Z. Mechanisms and strategies of immunosenescence effects on non-small cell lung cancer (NSCLC) treatment: A comprehensive analysis and future directions. Semin Cancer Biol 2025; 109:44-66. [PMID: 39793777 DOI: 10.1016/j.semcancer.2025.01.001] [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: 09/20/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Non-small cell lung cancer (NSCLC), the most prevalent form of lung cancer, remains a leading cause of cancer-related mortality worldwide, particularly among elderly individuals. The phenomenon of immunosenescence, characterized by the progressive decline in immune cell functionality with aging, plays a pivotal role in NSCLC progression and contributes to the diminished efficacy of therapeutic interventions in older patients. Immunosenescence manifests through impaired immune surveillance, reduced cytotoxic responses, and increased chronic inflammation, collectively fostering a pro-tumorigenic microenvironment. This review provides a comprehensive analysis of the molecular, cellular, and genetic mechanisms of immunosenescence and its impact on immune surveillance and the tumor microenvironment (TME) in NSCLC. We explore how aging affects various immune cells, including T cells, B cells, NK cells, and macrophages, and how these changes compromise the immune system's ability to detect and eliminate tumor cells. Furthermore, we address the challenges posed by immunosenescence to current therapeutic strategies, particularly immunotherapy, which faces significant hurdles in elderly patients due to immune dysfunction. The review highlights emerging technologies, such as single-cell sequencing and CRISPR-Cas9, which offer new insights into immunosenescence and its potential as a therapeutic target. Finally, we outline future research directions, including strategies for rejuvenating the aging immune system and optimizing immunotherapy for older NSCLC patients, with the goal of improving treatment efficacy and survival outcomes. These efforts hold promise for the development of more effective, personalized therapies for elderly patients with NSCLC.
Collapse
Affiliation(s)
- Huatao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| |
Collapse
|
18
|
Andonian BJ, Hippensteel JA, Abuabara K, Boyle EM, Colbert JF, Devinney MJ, Faye AS, Kochar B, Lee J, Litke R, Nair D, Sattui SE, Sheshadri A, Sherman AN, Singh N, Zhang Y, LaHue SC. Inflammation and aging-related disease: A transdisciplinary inflammaging framework. GeroScience 2025; 47:515-542. [PMID: 39352664 PMCID: PMC11872841 DOI: 10.1007/s11357-024-01364-0] [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: 05/24/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Inflammaging, a state of chronic, progressive low-grade inflammation during aging, is associated with several adverse clinical outcomes, including frailty, disability, and death. Chronic inflammation is a hallmark of aging and is linked to the pathogenesis of many aging-related diseases. Anti-inflammatory therapies are also increasingly being studied as potential anti-aging treatments, and clinical trials have shown benefits in selected aging-related diseases. Despite promising advances, significant gaps remain in defining, measuring, treating, and integrating inflammaging into clinical geroscience research. The Clin-STAR Inflammation Research Interest Group was formed by a group of transdisciplinary clinician-scientists with the goal of advancing inflammaging-related clinical research and improving patient-centered care for older adults. Here, we integrate insights from nine medical subspecialties to illustrate the widespread impact of inflammaging on diseases linked to aging, highlighting the extensive opportunities for targeted interventions. We then propose a transdisciplinary approach to enhance understanding and treatment of inflammaging that aims to improve comprehensive care for our aging patients.
Collapse
Affiliation(s)
- Brian J Andonian
- Division of Rheumatology and Immunology, Duke University School of Medicine, Durham, NC, USA.
| | - Joseph A Hippensteel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katrina Abuabara
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Eileen M Boyle
- Department of Haematology, University College London Cancer Institute, London, UK
| | - James F Colbert
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael J Devinney
- Division of Critical Care, Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Adam S Faye
- Division of Gastroenterology, Department of Population Health, NYU Langone Medical Center, New York, NY, USA
| | - Bharati Kochar
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Jiha Lee
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rachel Litke
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Devika Nair
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sebastian E Sattui
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anoop Sheshadri
- Division of Nephrology, Department of Medicine, University of California, San Francisco, Nephrology Section, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | | | - Namrata Singh
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Yinan Zhang
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sara C LaHue
- Department of Neurology, School of Medicine, and the UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
19
|
Yamashita S, Hamamoto S, Furukawa J, Fujita K, Takahashi M, Miyake M, Ito N, Iwamoto H, Kohjimoto Y, Hara I. Efficacy and Safety of Nivolumab Plus Ipilimumab for Metastatic Renal Cell Carcinoma in Patients 75 Years and Older: Multicenter Retrospective Study. Cancers (Basel) 2025; 17:474. [PMID: 39941841 PMCID: PMC11816081 DOI: 10.3390/cancers17030474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES The efficacy and safety of nivolumab plus ipilimumab (NIVO + IPI) for elderly patients with metastatic renal cell carcinoma have not been reported with sufficient evidence. Our study therefore aimed to compare the efficacy and safety of NIVO + IPI between patients ≥75 years and patients <75 years with metastatic renal cell carcinoma. METHODS We retrospectively analyzed a multi-center cohort of the 156 patients that received NIVO + IPI treatment at eight institutions. Among them, 33 patients were ≥75 years old, and the remainder were <75 years old. RESULTS Patient demographics and tumor characteristics were not significantly different between the two groups except for age. The objective response rate, disease control rate, progression-free survival, or cancer-specific survival were not significantly different between the groups. However, overall survival in the patients ≥75 years was significantly shorter than that in the patients <75 years (median: 18 months vs. 46 months, p = 0.01). In addition, an age ≥75 years was shown in multivariable analysis to be a significant independent predictor of poor overall survival. Toxicity did not show any significant variation between the groups. CONCLUSIONS Although the clinical efficacy and safety of NIVO + IPI was demonstrated in patients ≥75 years old, it is suggested that the indication for NIVO + IPI in this age group should be carefully considered, taking into account patients' expected life expectancy.
Collapse
Affiliation(s)
- Shimpei Yamashita
- Department of Urology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan; (Y.K.); (I.H.)
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan;
| | - Junya Furukawa
- Department of Urology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan;
| | - Kazutoshi Fujita
- Department of Urology, Kindai University Faculty of Medicine, 377-2 Onohigashi, Osakasayama 589-0014, Japan;
| | - Masayuki Takahashi
- Department of Urology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramo-to-cho, Tokushima 770-8503, Japan;
| | - Makito Miyake
- Department of Urology, Nara Medical University, 840 Shinjo-cho, Kashihara 634-8521, Japan;
| | - Noriyuki Ito
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubara-dori, Wakayama 640-8558, Japan;
| | - Hideto Iwamoto
- Division of Urology, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan;
| | - Yasuo Kohjimoto
- Department of Urology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan; (Y.K.); (I.H.)
| | - Isao Hara
- Department of Urology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan; (Y.K.); (I.H.)
| |
Collapse
|
20
|
Gu J, Wang J, Li Y, Li L, Zou Y, Guo Y, Yu B. Global burden of bacterial skin diseases from 1990 to 2045: an analysis based on global burden disease data. Arch Dermatol Res 2025; 317:266. [PMID: 39820797 DOI: 10.1007/s00403-025-03804-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: 08/26/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Bacterial skin diseases are a category of inflammatory skin conditions caused by bacterial infections, which impose a significant global disease burden. However, they have not been well assessed or predicted on a global scale. It is necessary to update the estimates and forecast future trends of the global burden of bacterial skin diseases to evaluate the impact of past healthcare policies and to provide guidance and information for new national and international healthcare strategies. We aimed to describe the burden and trend of bacterial skin diseases and to predict the burden up to 2045. To achieve this, we employed a cross-sectional analysis based on Global Burden of Disease (GBD) data, utilizing advanced statistical models to quantify trends and forecast future burdens. Data on incidence and disability-adjusted life years (DALYs) of bacterial skin diseases were obtained from Global Burden of Disease 2021. We used average annual percent change (AAPC) by Joinpoint Regression to quantify the temporal trends. We conducted decomposition analysis to understand the contribution of aging, epidemiological changes, and population growth. Bayesian Age-Period-Cohort model was used to predict burden up to 2045. Global incidence rate of bacterial skin diseases increased from 8,988.74 per 100,000 in 1990 to 10,823.88 per 100,000, with AAPC of 0.62% (0.61 ~ 0.63%). The highest incidence rate was in low Socio-demographic Index (SDI) region and population aged < 35. The DALYs rate increased from 20.82 per 100,000 in 1990 to 25.45 per 100,000 in 2021, with AAPC of -0.11% (-0.34 ~ 0.13%). The highest increase of DALYs was in high SDI region and population aged > 85. Among the three evaluated factors of decomposition analysis, the major drivers of incident case rise were population growth, followed by epidemiological changes; the major drivers of DALYs case rise were population growth, followed by aging. The number of incidence cases has increased since 1990, reaching nearly 90 million in 2021 and expected to hit 1.2 billion in 2045. The incidence rate has also risen. Meanwhile, DALYs showed an upward trend from 1990 to 2005, peaking at 32/100,000, then a downward trend. Our findings align with our initial objectives, demonstrating a significant increase in the global incidence of bacterial skin diseases and highlighting the need for targeted prevention and treatment strategies. The variation in burden across different regions and age groups underscores the importance of tailored public health interventions. Predictive models suggest a continued rise in incidence rates and incident cases through 2045, emphasizing the urgency for action. This study provides a foundation for future research and policy development aimed at reducing the burden of bacterial skin diseases worldwide.
Collapse
Affiliation(s)
- Jiaxu Gu
- Department of Dermatology, Shenzhen Key Discipline of Dermatology, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Peking University, Shenzhen, China
| | - Jiaming Wang
- Department of Dermatology, Shenzhen Key Discipline of Dermatology, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shantou University Medical College, Shenzhen, China
| | - Yannan Li
- Faculty of Medicine, School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Lianjie Li
- Warren Skin Care Center, Phillipsburg, USA
| | - Yanfen Zou
- Department of Dermatology, Shenzhen Key Discipline of Dermatology, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yang Guo
- Department of Epidemiology and Statistics, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China.
| | - Bo Yu
- Department of Dermatology, Shenzhen Key Discipline of Dermatology, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, 518036, China.
| |
Collapse
|
21
|
Yu X, Pei W, Li B, Sun S, Li W, Wu Q. Immunosenescence, Physical Exercise, and their Implications in Tumor Immunity and Immunotherapy. Int J Biol Sci 2025; 21:910-939. [PMID: 39897036 PMCID: PMC11781184 DOI: 10.7150/ijbs.100948] [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: 07/14/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025] Open
Abstract
Aging is associated with a decline in immune function, termed immunosenescence, which compromises host defences and increases susceptibility to infections and cancer. Physical exercise is widely recognized for its myriad health benefits, including the potential to modulate the immune system. This review explores the bidirectional relationship between immunosenescence and physical exercise, focusing on their interplay in shaping antitumor immunity. We summarize the impact of aging on innate and adaptive immune cells, highlighting alterations that contribute to immunosenescence and cancer development. We further delineate the effects of exercise on immune cell function, demonstrating its potential to mitigate immunosenescence and enhance antitumor responses. We also discuss the implications of immunosenescence for the efficacy of immunotherapies, such as immune checkpoint inhibitors and adoptive T cell therapy, and explore the potential benefits of combining exercise with these interventions. Collectively, this review underscores the importance of understanding the complex relationship between immunosenescence, physical exercise, and antitumor immunity, paving the way for the development of innovative strategies to improve cancer outcomes in the aging population.
Collapse
Affiliation(s)
- Xin Yu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Wei Pei
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Wenge Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Oncology, Shanghai GoBroad Cancer Hospital, Shanghai, P. R. China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| |
Collapse
|
22
|
Li H, Zhao W, Yang F, Qiao Q, Ma S, Yang K, Song S, Wang S, Qu J, Liu GH, Bao Y, Zhang W. Immunosenescence Inventory-a multi-omics database for immune aging research. Nucleic Acids Res 2025; 53:D1047-D1054. [PMID: 39673270 PMCID: PMC11701554 DOI: 10.1093/nar/gkae1102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/08/2024] [Accepted: 11/22/2024] [Indexed: 12/16/2024] Open
Abstract
The immune system is intricately interconnected with all other bodily systems. As individuals age, the immune system undergoes changes known as immunosenescence, increasing susceptibility to disease, and contributing significantly to the morbidity and mortality observed in older populations. Immunosenescence drives systemic aging and therefore represents a key therapeutic target to extend healthy aging. In recent years, the extensive application of omics technologies has broadened our understanding of aging and immunity, necessitating a comprehensive database to encapsulate these advancements and deepen our insights into immune aging in the era of artificial intelligence. The Immunosenescence Inventory is a pioneering database designed to provide a multidimensional and integrative view of the aging immune system. By leveraging cutting-edge omics technologies and analytical tools, Immunosenescence Inventory offers a comprehensive resource for researchers to explore the intricate relationship between immunosenescence and age-related health outcomes. Furthermore, the database, which aids in the creation of diagnostic tools for immune aging conditions, is now publicly available at https://ngdc.cncb.ac.cn/iaa/home.
Collapse
Affiliation(s)
- Hao Li
- 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
| | - Wei Zhao
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
| | - Fei Yang
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
| | - Qin Qiao
- China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shuai Ma
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Kuan Yang
- China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Shuhui Song
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
| | - Si Wang
- Aging Biomarker Consortium, Beijing 100101, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Chongqing Renji Hospital, University of Chinese Academy of Sciences, Chongqing 400062, China
| | - Jing Qu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yiming Bao
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
| | - Weiqi Zhang
- 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
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
23
|
Salazar-Torres BL, González-Rocha A, Armenta-Guirado BI, Ortiz-Rodríguez MA, Muñoz-Aguirre P, Rodríguez-Ramírez S, Denova-Gutierrez E. Dietary Patterns Are Associated with Metabolic Syndrome in Mexican Older Adult Population. J Nutr Gerontol Geriatr 2025; 44:1-16. [PMID: 39773613 DOI: 10.1080/21551197.2024.2448937] [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] [Indexed: 01/11/2025]
Abstract
BACKGROUND Metabolic syndrome poses a significant public health challenge globally, particularly among older adults. Dietary patterns play a crucial role in the development and management of metabolic syndrome. However, to our knowledge, no evidence exists regarding the association between dietary patterns and metabolic syndrome among the Mexican older adult population. Thus, the objective of the present study is to assess the association between dietary patterns and metabolic syndrome in Mexican older adults. METHODS This cross-sectional study utilized data from the Mexican National Health and Nutrition Survey 2016 to investigate the relationship between dietary patterns and metabolic syndrome in 804 OA (aged ≥60 years). Dietary patterns were derived using factor analysis, and metabolic syndrome was defined based on established criteria. Logistic regression models were employed to assess associations, adjusting for potential confounders. RESULTS Three dietary patterns were identified: a "Westernized" pattern characterized by high consumption of processed meat, red meat, and saturated fats; a "Transitional" pattern high in fresh vegetables and fruits; and a "Prudent" pattern high in fish and low-fat dairy products. The "Westernized" pattern exhibited a positive association with metabolic syndrome, while the "Prudent" pattern showed a negative association. The associations remained significant after adjusting for confounders. Biological mechanisms linking the "Westernized" pattern to metabolic syndrome include chronic inflammation, endothelial dysfunction, and dyslipidemia induced by components such as processed meats and soft drinks. CONCLUSION Our study highlights the importance of dietary patterns in influencing metabolic syndrome risk among Mexican older adults. Promoting dietary patterns rich in fish, vegetables, and legumes while reducing consumption of processed meats and sugary beverages may offer significant health benefits in this population. Further prospective studies are needed to confirm these findings and inform targeted interventions for metabolic syndrome prevention and management in older adults.
Collapse
Affiliation(s)
| | - Alejandra González-Rocha
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca, México
| | | | | | - Paloma Muñoz-Aguirre
- Consejo Nacional de Humanidades Ciencias y Tecnologías/Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Sonia Rodríguez-Ramírez
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca, México
| | - Edgar Denova-Gutierrez
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca, México
| |
Collapse
|
24
|
Charisis S, Mourtzi N, Scott MR, Ntanasi E, Mamalaki E, Hatzimanolis A, Ramirez A, Lambert JC, Yannakoulia M, Kosmidis M, Dardiotis E, Hadjigeorgiou G, Sakka P, Satizabal CL, Beiser A, Yang Q, Georgakis MΚ, Seshadri S, Scarmeas N. Genetic predisposition to high circulating levels of interleukin 6 and risk for Alzheimer's disease. Discovery and replication. J Prev Alzheimers Dis 2025; 12:100018. [PMID: 39800457 DOI: 10.1016/j.tjpad.2024.100018] [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] [Indexed: 05/02/2025]
Abstract
IMPORTANCE Aging is accompanied by immune dysregulation, which has been implicated in Alzheimer's disease (AD) pathogenesis. Individuals who are genetically predisposed to elevated levels of proinflammatory mediators might be at increased risk for AD. OBJECTIVE To investigate whether genetic propensity for higher circulating levels of interleukin 6 (IL-6) is associated with AD risk. DESIGN We analyzed data from the Hellenic Longitudinal Investigation of Aging and Diet (HELIAD). Mean follow-up was 2.9 (SD, 0.8) years. Baseline assessment was from 11/2009 to 11/2016, and cognitive follow-up from 01/2013 to 07/2019. Associations of interest were also examined in the UK Biobank (UKB) for replication purposes (mean follow-up was 12.9 (SD, 2.4) years; baseline assessment was from 12/2006 to 10/2010). SETTING Population-based study. PARTICIPANTS The HELIAD sample included 622 participants ≥65 years of age without baseline dementia or amnestic mild cognitive impairment (aMCI-the prodromal stage of AD). The UKB sample included 142,637 participants ≥60 years of age without prevalent dementia. EXPOSURES Genetic predisposition to elevated circulating levels of IL-6 was estimated using a polygenic risk score (PRS), calculated based on the summary statistics of a current GWAS meta-analysis. MAIN OUTCOMES AND MEASURES AD and MCI diagnoses were based on standard clinical criteria [HELIAD], or hospital records and death registry data [UKB]. Associations with AD or aMCI incidence [HELIAD] and AD incidence [UKB] were examined with Cox regression models. RESULTS In HELIAD, mean age was 73.4 (SD, 5.0) years; 363 (58%) women. An increase in IL-6 PRS by 1 standard deviation unit (SDU) was associated with up to a 43% increase in the risk for incident AD/aMCI (HRGWAS significance threshold of 0.01, 1.43 [95%CI, 1.14 - 1.80]). In UKBB, mean age was 64.2 (SD, 2.8) years; 73,707 (52%) women. A 1 SDU increase in IL-6 PRS was associated with up to an 8% increase in the risk for incident AD (HRGWAS significance threshold of 0.2, 1.08 [95%CI, 1.04 - 1.12]). CONCLUSIONS AND RELEVANCE Genetic predisposition to higher circulating levels of IL-6 was associated with an increased risk for AD, supporting the role of IL-6-related pathways in AD pathogenesis, and suggesting that genetic predisposition to proinflammatory states might trigger or accelerate AD-related neuropathology.
Collapse
Affiliation(s)
- Sokratis Charisis
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Niki Mourtzi
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Matthew R Scott
- Department of Biostatistics, Boston University School of Public Health
| | - Eva Ntanasi
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Eirini Mamalaki
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece; Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Alexandros Hatzimanolis
- Department of Psychiatry, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Alfredo Ramirez
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA; Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, Cologne, Germany; Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liés au vieillissement, Lille, France
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Mary Kosmidis
- Lab of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larisa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| | | | - Paraskevi Sakka
- Athens Association of Alzheimer's Disease and Related Disorders, Marousi, Greece
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Alexa Beiser
- Department of Biostatistics, Boston University School of Public Health
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health
| | - Marios Κ Georgakis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, MA, USA; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece; Department of Neurology, The Gertrude H. Sergievsky Center, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| |
Collapse
|
25
|
Xiao J, Cao Y, Li X, Xu L, Wang Z, Huang Z, Mu X, Qu Y, Xu Y. Elucidation of Factors Affecting the Age-Dependent Cancer Occurrence Rates. Int J Mol Sci 2024; 26:275. [PMID: 39796131 PMCID: PMC11720044 DOI: 10.3390/ijms26010275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Cancer occurrence rates exhibit diverse age-related patterns, and understanding them may shed new and important light on the drivers of cancer evolution. This study systematically analyzes the age-dependent occurrence rates of 23 carcinoma types, focusing on their age-dependent distribution patterns, the determinants of peak occurrence ages, and the significant difference between the two genders. According to the SEER reports, these cancer types have two types of age-dependent occurrence rate (ADOR) distributions, with most having a unimodal distribution and a few having a bimodal distribution. Our modeling analyses have revealed that (1) the first type can be naturally and simply explained using two age-dependent parameters: the total number of stem cell divisions in an organ from birth to the current age and the availability levels of bloodborne growth factors specifically needed by the cancer (sub)type, and (2) for the second type, the first peak is due to viral infection, while the second peak can be explained as in (1) for each cancer type. Further analyses indicate that (i) the iron level in an organ makes the difference between the male and female cancer occurrence rates, and (ii) the levels of sex hormones are the key determinants in the onset age of multiple cancer types. This analysis deepens our understanding of the dynamics of cancer evolution shared by diverse cancer types and provides new insights that are useful for cancer prevention and therapeutic strategies, thereby addressing critical gaps in the current paradigm of oncological research.
Collapse
Affiliation(s)
- Jun Xiao
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Yangkun Cao
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| | - Xuan Li
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Long Xu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Zhihang Wang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Zhenyu Huang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Xuechen Mu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
- School of Mathematics, Jilin University, Changchun 130012, China
| | - Yinwei Qu
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Ying Xu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| |
Collapse
|
26
|
Zhang Y, Zhou J, Jin Y, Liu C, Zhou H, Sun Y, Jiang H, Gan J, Zhang C, Lu Q, Chang Y, Zhang Y, Li X, Ning S. Single-Cell and Bulk Transcriptomics Reveal the Immunosenescence Signature for Prognosis and Immunotherapy in Lung Cancer. Cancers (Basel) 2024; 17:85. [PMID: 39796714 PMCID: PMC11720133 DOI: 10.3390/cancers17010085] [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: 11/04/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Immunosenescence is the aging of the immune system, which is closely related to the development and prognosis of lung cancer. Targeting immunosenescence is considered a promising therapeutic approach. METHODS We defined an immunosenescence gene set (ISGS) and examined it across 33 TCGA tumor types and 29 GTEx normal tissues. We explored the 46,993 single cells of two lung cancer datasets. The immunosenescence risk model (ISRM) was constructed in TCGA LUAD by network analysis, immune infiltration analysis, and lasso regression and validated by survival analysis, cox regression, and nomogram in four lung cancer cohorts. The predictive ability of ISRM for drug response and immunotherapy was detected by the oncopredict algorithm and XGBoost model. RESULTS We found that senescent lung tissues were significantly enriched in ISGS and revealed the heterogeneity of immunosenescence in pan-cancer. Single-cell and bulk transcriptomics characterized the distinct immune microenvironment between old and young lung cancer. The ISGS network revealed the crucial function modules and transcription factors. Multiplatform analysis revealed specific associations between immunosenescence and the tumor progression of lung cancer. The ISRM consisted of five risk genes (CD40LG, IL7, CX3CR1, TLR3, and TLR2), which improved the prognostic stratification of lung cancer across multiple datasets. The ISRM showed robustness in immunotherapy and anti-tumor therapy. We found that lung cancer patients with a high-risk score showed worse survival and lower expression of immune checkpoints, which were resistant to immunotherapy. CONCLUSIONS Our study performed a comprehensive framework for assessing immunosenescence levels and provided insights into the role of immunosenescence in cancer prognosis and biomarker discovery.
Collapse
Affiliation(s)
- Yakun Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Jiajun Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Yitong Jin
- The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China;
| | - Chenyu Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Hanxiao Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Yue Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Han Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Jing Gan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Caiyu Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Qianyi Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Yetong Chang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| |
Collapse
|
27
|
Pangrazzi L, Meryk A. Molecular and Cellular Mechanisms of Immunosenescence: Modulation Through Interventions and Lifestyle Changes. BIOLOGY 2024; 14:17. [PMID: 39857248 PMCID: PMC11760833 DOI: 10.3390/biology14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Immunosenescence, the age-related decline in immune function, is a complex biological process with profound implications for health and longevity. This phenomenon, characterized by alterations in both innate and adaptive immunity, increases susceptibility to infections, reduces vaccine efficacy, and contributes to the development of age-related diseases. At the cellular level, immunosenescence manifests as decreased production of naive T and B cells, accumulation of memory and senescent cells, thymic involution, and dysregulated cytokine production. Recent advances in molecular biology have shed light on the underlying mechanisms of immunosenescence, including telomere attrition, epigenetic alterations, mitochondrial dysfunction, and changes in key signaling pathways such as NF-κB and mTOR. These molecular changes lead to functional impairments in various immune cell types, altering their proliferative capacity, differentiation, and effector functions. Emerging research suggests that lifestyle factors may modulate the rate and extent of immunosenescence at both cellular and molecular levels. Physical activity, nutrition, stress management, and sleep patterns have been shown to influence immune cell function, inflammatory markers, and oxidative stress in older adults. This review provides a comprehensive analysis of the molecular and cellular mechanisms underlying immunosenescence and explores how lifestyle interventions may impact these processes. We will examine the current understanding of immunosenescence at the genomic, epigenomic, and proteomic levels, and discuss how various lifestyle factors can potentially mitigate or partially reverse aspects of immune aging. By integrating recent findings from immunology, gerontology, and molecular biology, we aim to elucidate the intricate interplay between lifestyle and immune aging at the molecular level, potentially informing future strategies for maintaining immune competence in aging populations.
Collapse
Affiliation(s)
- Luca Pangrazzi
- Institute for Biomedical Aging Research, Faculty of Biology, University of Innsbruck, 6020 Innsbruck, Austria;
| | - Andreas Meryk
- Department of Pediatrics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
28
|
Lin J, Liu J, Luo Q, Zhuang J, Xiao R, Wang H, Yang X, Wei X, Cai J. Association of lymphocyte-to-C-reactive protein ratio with cerebral small vessel disease: a cross-sectional study based on dose-response analysis. Front Neurol 2024; 15:1480115. [PMID: 39777309 PMCID: PMC11704889 DOI: 10.3389/fneur.2024.1480115] [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: 08/13/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Objective We investigated the relationship between lymphocyte-to-C-reactive protein ratio (LCR) and common imaging markers of cerebral small vessel disease (CSVD). Methods Data from 835 CSVD patients were analyzed using univariate and multivariate logistic regression to determine CSVD-associated factors. Multivariate models assessed the association between LCR and CSVD, including common imaging markers. Subgroup analysis by age, sex, smoking history, hypertension, lipid levels, and other factors was conducted. The receiver operating characteristic curve analysis and 10-fold cross-validation were performed to evaluate the predictive performance of LCR. Results Lymphocyte-to-C-reactive protein ratio was independently associated with a decreased risk of CSVD (p < 0.001), indicating a protective role of LCR against CSVD. Among the imaging markers of CSVD, LCR in the highest quartile was negatively associated with moderate-to-severe white matter hyperintensities (WMH) (p = 0.002) and moderate-to-severe enlarged perivascular spaces (EPVS) (p < 0.001), but not with lacune (p > 0.05). The restrictive cubic spline analysis revealed a linear dose-response relationship between log-transformed LCR and the incidence of CSVD (P non-linear = 0.090) as well as moderate-to-severe WMH (P non-linear = 0.304), with a non-linear association with moderate and severe EPVS (P non-linear = 0.001). In the subgroup analyses, LCR remained a significant association with CSVD in most subgroups (p < 0.05). Notably, a significant correlation was observed between LCR and CSVD (p < 0.001) in the subgroups of non-smokers, those with neutrophil count ≤6.3 × 109/L, and with high-density lipoprotein cholesterol ≥1 mmol/L. No interaction effect was identified between the variables and the LCR (p > 0.1). The predictive capability of LCR for CSVD was confirmed through receiver operating characteristic curve analysis. Conclusion Lymphocyte-to-C-reactive protein ratio is an independent protective factor for CSVD and is associated with lower WMH or EPVS burden but not lacune. Inflammation is involved in CSVD pathophysiology through multiple pathways, providing potential targets for CSVD intervention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jiangping Cai
- Department of Neurology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
29
|
Teufel LU, Taks EJM, van Gemert J, Neacsu M, Föhse K, Gillard J, Diavatopoulos DA, de Jonge MI, Netea MG, Joosten LAB, Arts RJW. Interleukin 38 reduces antigen-presentation capacity and antibody production after vaccination. Vaccine 2024; 42:126396. [PMID: 39353267 DOI: 10.1016/j.vaccine.2024.126396] [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: 06/10/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
The mechanisms that underpin low vaccine responses, which can lead to inadequate protection against infection, are still partially unclear. Interleukin (IL)-38 is a member of the IL-1 family, expressed by B cells among others, that regulates inflammatory responses. A recent study shows that IL-38 suppresses plasma cell generation and antibody production upon immune activation. We hypothesis that IL-38 affects antigen-presentation capacity of innate immune cells, effecting antibody production. Here, we investigated the effect of recombinant human IL-38 on human peripheral blood mononuclear cells and myeloid-derived DCs regarding cytokine production, phagocytosis, and expression of MCH II and co-stimulatory proteins in vitro, and further relate circulating plasma IL-38 concentrations to antibody responses in a cohort of 75 females aged 18-48 vaccinated with BCG and Tdap-IPV. To this end, we found that IL-38 decreased the expression of HLA-DR, HLA-DM, and CD83 on PBMCs, and CD40 and CD86 on MDDCs. IL-38 further impaired phagocytosis capacity of monocytes. Lastly, antibody production against diphtheria toxoids up to eight months post-vaccination was negatively associated with IL-38 plasma concentrations. These data suggest that IL-38 could dampen the effectiveness of antigen-presentation and phagocytosis, and could therefore modulate the immunogenicity of some vaccine types.
Collapse
Affiliation(s)
- Lisa U Teufel
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Esther J M Taks
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jelle van Gemert
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihaela Neacsu
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Konstantin Föhse
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joshua Gillard
- Laboratory of Medical Immunology and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dimitri A Diavatopoulos
- Laboratory of Medical Immunology and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marien I de Jonge
- Laboratory of Medical Immunology and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Rob J W Arts
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
30
|
Priora M, Becciolini A, Celletti E, Di Penta M, Lo Gullo A, Paroli M, Bravi E, Andracco R, Nucera V, Ometto F, Lumetti F, Farina A, Del Medico P, Colina M, Ravagnani V, Scolieri P, Larosa M, Visalli E, Addimanda O, Vitetta R, Volpe A, Bezzi A, Girelli F, Molica Colella AB, Caccavale R, Di Donato E, Adorni G, Santilli D, Lucchini G, Arrigoni E, Sabatini E, Platè I, Mansueto N, Ianniello A, Fusaro E, Ditto MC, Bruzzese V, Camellino D, Bianchi G, Serale F, Foti R, Amato G, De Lucia F, Bosco YD, Foti R, Reta M, Fiorenza A, Rovera G, Marchetta A, Focherini MC, Mascella F, Bernardi S, Sandri G, Giuggioli D, Salvarani C, Franchina V, Molica Colella F, Ferrero G, Ariani A, Parisi S. Effectiveness and Predictors of Long-Term Treatment Response to Tofacitinib in Rheumatoid Arthritis Cohort: General Analysis and Focus on High-Cardiovascular-Risk Subgroup-A Multicenter Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1982. [PMID: 39768862 PMCID: PMC11677110 DOI: 10.3390/medicina60121982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: The treatment landscape for Rheumatoid Arthritis (RA) has evolved significantly with the introduction of Janus kinase inhibitors (JAKi), such as Tofacitinib (TOFA), which offer a new therapeutic option for patients who have failed or are intolerant to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). Safety concerns, particularly related to cardiovascular and cancer risks, prompted a need for additional investigation in real-world clinical settings. This study aimed to evaluate the long-term effectiveness and predictors of response to TOFA in two subpopulations of RA patients, categorized by differing cardiovascular risk profiles. Materials and Methods: This was a retrospective, multicenter observational study conducted as part of the BIRRA project, involving 23 Italian rheumatological referral centers. A total of 213 patients diagnosed with RA and treated with TOFA were included, with data collected on baseline demographics, clinical history, disease activity, and comorbidities. Patients were divided into high-risk and low-risk cardiovascular groups based on age (≥65 years) and the presence of at least one cardiovascular risk factor. Disease activity was assessed at baseline, 6 months, and 12 months using DAS28-ESR and DAS28-CRP. Treatment response was evaluated using intention-to-treat (ITT) and per-protocol (PP) approaches. Predictors of low disease activity (LDA) and remission were assessed through logistic regression, and clustering analyses were used to identify subgroups of patients with different therapeutic responses. Results: The study included 213 patients, with 129 classified as high-risk. For the overall cohort, patients achieving LDA and remission at 6 months were 20% and 12%, respectively, for the ITT analysis, and 29% and 14% for the PP analysis. At 12 months, 26% of patients reached LDA, and 17% achieved remission according to ITT, while for the PP analysis, these rates were 30% and 19%, respectively. No significant differences in remission or LDA rates were observed between the high-risk and low-risk groups. In the high-risk subgroup, 17% of patients reached LDA and 9% achieved remission at 6 months (ITT analysis), while these rates increased to 22% and 13%, respectively, in the PP analysis. At 12 months, 22% achieved LDA and 13% achieved remission in the ITT analysis, while 28% and 17% did so in the PP analysis. The reduction in DAS28-ESR and DAS28-CRP scores was significant (p < 0.001) across all time points for both high-risk and low-risk patients. Logistic regression analyses revealed that none of the baseline characteristics-including age, sex, comorbidities, rheumatoid factor, anti-citrullinated protein antibody (ACPA) positivity, initial disease severity, or treatment history-were significant predictors of remission or LDA at 6 or 12 months. The clustering analysis suggested that older patients, particularly those with worse baseline DAS28 scores, tended to show a less favorable response to treatment, potentially indicating impacts of age-related factors such as immunosenescence on therapeutic outcomes. Conclusions: Tofacitinib demonstrated similar effectiveness in both high- and low-risk cardiovascular subgroups of RA patients, with significant reductions in disease activity observed at both 6 and 12 months. Despite safety concerns related to cardiovascular risk, TOFA remained an effective treatment option across patient subgroups, with no significant differences in remission or LDA rates based on cardiovascular risk profiles. Age appeared to negatively impact treatment response, highlighting the role of immunosenescence in RA management. These findings support the use of TOFA as a personalized therapeutic option for RA, emphasizing the need for careful evaluation of cardiovascular and age-related risks in clinical decision-making.
Collapse
Affiliation(s)
- Marta Priora
- Rheumatology Day Hospital and Outpatient Clinic, ASL CN1, 12100 Cuneo, Italy; (M.P.); (F.S.)
| | - Andrea Becciolini
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Eleonora Celletti
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | - Myriam Di Penta
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | | | - Marino Paroli
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.P.); (R.C.)
| | - Elena Bravi
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Romina Andracco
- Rheumatology Unit, Imperia Hospital, 18100 Imperia, Italy; (R.A.); (N.M.)
| | - Valeria Nucera
- Rheumatology Outpatient Unit, ASL Novara, 28100 Novara, Italy; (V.N.); (A.I.)
| | - Francesca Ometto
- Rheumatology Outpatient Clinic, Azienda ULSS 6 Euganea, 35131 Padova, Italy;
| | - Federica Lumetti
- Rheumatology Unit, Azienda USL of Modena and AOU Policlinico of Modena, 41100 Modena, Italy;
| | - Antonella Farina
- Internal Medicine Unit, Rheumatology Outpatient Clinic, Augusto Murri Hospital, 63900 Fermo, Italy;
| | - Patrizia Del Medico
- Rheumatology Outpatient Clinic, Internal Medicine Unit, Civitanova Marche Hospital, 62012 Civitanova Marche, Italy;
| | - Matteo Colina
- Rheumatology Service, Internal Medicine Section, Department of Medicine and Oncology, Santa Maria della Scaletta Hospital, 40026 Imola, Italy;
- Alma Mater Studiorum, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Viviana Ravagnani
- Rheumatology Unit, Santa Chiara Hospital APSS—Trento, 38122 Trento, Italy;
| | - Palma Scolieri
- Rheumatology Unit, Department of Medical Specialties, Nuovo Regina Margherita Hospital, 00154 Roma, Italy; (P.S.); (V.B.)
| | - Maddalena Larosa
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Elisa Visalli
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Olga Addimanda
- Rheumatology Unit, AUSL of Bologna—Policlinico Sant’Orsola—AOU—IRCCS of Bologna, 40138 Bologna, Italy; (O.A.); (M.R.)
| | - Rosetta Vitetta
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Alessandro Volpe
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy; (A.V.); (A.M.)
| | - Alessandra Bezzi
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Francesco Girelli
- Rheumatology Unit, G.B. Morgagni—L. Pierantoni Hospital, 47121 Forli, Italy; (F.G.); (S.B.)
| | | | - Rosalba Caccavale
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.P.); (R.C.)
| | - Eleonora Di Donato
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Giuditta Adorni
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Daniele Santilli
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Gianluca Lucchini
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Eugenio Arrigoni
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Emanuela Sabatini
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | - Ilaria Platè
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Natalia Mansueto
- Rheumatology Unit, Imperia Hospital, 18100 Imperia, Italy; (R.A.); (N.M.)
| | - Aurora Ianniello
- Rheumatology Outpatient Unit, ASL Novara, 28100 Novara, Italy; (V.N.); (A.I.)
| | - Enrico Fusaro
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| | - Maria Chiara Ditto
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| | - Vincenzo Bruzzese
- Rheumatology Unit, Department of Medical Specialties, Nuovo Regina Margherita Hospital, 00154 Roma, Italy; (P.S.); (V.B.)
| | - Dario Camellino
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Gerolamo Bianchi
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Francesca Serale
- Rheumatology Day Hospital and Outpatient Clinic, ASL CN1, 12100 Cuneo, Italy; (M.P.); (F.S.)
| | - Rosario Foti
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Giorgio Amato
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Francesco De Lucia
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Ylenia Dal Bosco
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Roberta Foti
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Massimo Reta
- Rheumatology Unit, AUSL of Bologna—Policlinico Sant’Orsola—AOU—IRCCS of Bologna, 40138 Bologna, Italy; (O.A.); (M.R.)
| | - Alessia Fiorenza
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Guido Rovera
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Antonio Marchetta
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy; (A.V.); (A.M.)
| | - Maria Cristina Focherini
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Fabio Mascella
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Simone Bernardi
- Rheumatology Unit, G.B. Morgagni—L. Pierantoni Hospital, 47121 Forli, Italy; (F.G.); (S.B.)
| | - Gilda Sandri
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | - Dilia Giuggioli
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | - Carlo Salvarani
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | | | | | - Giulio Ferrero
- Unit of Diagnostic and Interventional Radiology, Santa Corona Hospital, 17027 Pietra Ligure, Italy;
| | - Alarico Ariani
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Simone Parisi
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| |
Collapse
|
31
|
Zhao CN, Jiang LQ, Musonye HA, Meng SY, He YS, Wang P, Ni J, Pan HF. Associations of accelerated biological aging and metabolic heterogeneity of obesity with rheumatoid arthritis: a prospective cohort study. Clin Rheumatol 2024; 43:3615-3623. [PMID: 39367918 DOI: 10.1007/s10067-024-07167-0] [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: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
OBJECTIVE To evaluate the associations between biological aging, metabolic heterogeneity of obesity, and rheumatoid arthritis (RA). METHODS This prospective cohort study analyzed 268,184 individuals from the UK Biobank. Biological age was estimated using phenotypic age (PhenoAge), Klemera-Doubal methods (KDM-BA), and telomere length. We calculated KDM-BA acceleration and PhenoAge acceleration after subtracting the effect of chronological age by regression residual. The metabolic heterogeneity of obesity can be evaluated by four BMI metabolic phenotypes, namely metabolically unhealthy normal weight (MUNW), metabolically healthy normal weight (MHNW), metabolically unhealthy overweight/obesity (MUOO), and metabolically healthy overweight/obesity (MHOO). Cox models were employed to estimate the associations between biological aging, metabolic heterogeneity of obesity, and RA risk. RESULTS A total of 2842 patients experienced RA during a mean follow-up time of 12.21 years. A standard deviation (SD) increase in KDM-BA acceleration and PhenoAge acceleration was associated with an increased risk of RA by 13% (hazard ratio = 1.13; 95% CI, 1.09-1.17) and 39% (HR = 1.39; 95% CI, 1.34-1.44), respectively. A SD increase in telomere length was associated with a reduced risk of RA by 5% (HR = 0.95; 95% CI, 0.91-0.98). Compared to the MHNW group, the MUOO group was associated with a 51% increase in the risk of incident RA. In the joint effect analysis, compared to the MHNW + KDM-BA younger subgroup, the HR (95% CI) for RA was 1.68 (1.48, 1.90) in the MUOO + KDM-BA older subgroup. CONCLUSION Accelerated biological aging may heighten the susceptibility to RA, particularly in individuals with obesity or metabolic dysfunction. Key Points •Accelerated biological aging increases the risk of developing RA. •Overweight/obese people with a healthy metabolism have a higher risk of RA than those with normal weight and healthy metabolism. •The BMI metabolic phenotype has a strong modifying effect on the association between KDM-BA/PhenoAge and RA risk.
Collapse
Affiliation(s)
- Chan-Na Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Ling-Qiong Jiang
- Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Harry Asena Musonye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shi-Yin Meng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Peng Wang
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
32
|
Bi J, Zeng J, Liu X, Mo C, Yao M, Zhang J, Yuan P, Jia B, Xu S. Drug delivery for age-related bone diseases: From therapeutic targets to common and emerging therapeutic strategies. Saudi Pharm J 2024; 32:102209. [PMID: 39697472 PMCID: PMC11653637 DOI: 10.1016/j.jsps.2024.102209] [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: 09/12/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
With the accumulation of knowledge on aging, people have gradually realized that among the many factors that cause individual aging, the accumulation of aging cells is an essential cause of organ degeneration and, ultimately, age-related diseases. Most cells present in the bone microenvironment gradually age over time, leading to an imbalance of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis. This imbalance contributes to age-related bone loss and the development of age-related bone diseases, such as osteoporosis. Bone aging can prolong the lifespan and delay the development of age-related diseases. Nanoparticles have controllable and stable physical and chemical properties and can precisely target different tissues and organs. By preparing multiple easily modified and biocompatible nanoparticles as different drug delivery carriers, specifically targeting various diseased tissues for controlled-release and sustained-release administration, the delivery efficiency of drugs can be significantly improved, and the toxicity and side effects of drugs can be substantially reduced, thereby improving the therapeutic effect of age-related bone diseases. In addition, other novel anti-aging strategies (such as stem cell exosomes) also have significant scientific and practical significance in anti-aging research on age-related bone diseases. This article reviews the research progress of various nano-drug-loaded particles and emerging anti-aging methods for treating age-related bone diseases, offering new insights and directions for precise targeted clinical therapies.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohao Liu
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Jing Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Lotze MT, Olejniczak SH, Skokos D. CD28 co-stimulation: novel insights and applications in cancer immunotherapy. Nat Rev Immunol 2024; 24:878-895. [PMID: 39054343 PMCID: PMC11598642 DOI: 10.1038/s41577-024-01061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Substantial progress in understanding T cell signalling, particularly with respect to T cell co-receptors such as the co-stimulatory receptor CD28, has been made in recent years. This knowledge has been instrumental in the development of innovative immunotherapies for patients with cancer, including immune checkpoint blockade antibodies, adoptive cell therapies, tumour-targeted immunostimulatory antibodies, and immunostimulatory small-molecule drugs that regulate T cell activation. Following the failed clinical trial of a CD28 superagonist antibody in 2006, targeted CD28 agonism has re-emerged as a technologically viable and clinically promising strategy for cancer immunotherapy. In this Review, we explore recent insights into the molecular functions and regulation of CD28. We describe how CD28 is central to the success of current cancer immunotherapies and examine how new questions arising from studies of CD28 as a clinical target have enhanced our understanding of its biological role and may guide the development of future therapeutic strategies in oncology.
Collapse
Affiliation(s)
- Michael T Lotze
- Department of Surgery, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Scott H Olejniczak
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | | |
Collapse
|
34
|
Almalki WH, Almujri SS. Aging, ROS, and cellular senescence: a trilogy in the progression of liver fibrosis. Biogerontology 2024; 26:10. [PMID: 39546058 DOI: 10.1007/s10522-024-10153-3] [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: 09/01/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
Ageing is an inevitable and multifaceted biological process that impacts a wide range of cellular and molecular mechanisms, leading to the development of various diseases, such as liver fibrosis. Liver fibrosis progresses to cirrhosis, which is an advanced form due to high amounts of extracellular matrix and restoration of normal liver structure with failure to repair damaged tissue and cells, marking the end of liver function and total liver failure, ultimately death. The most important factors are reactive oxygen species (ROS) and cellular senescence. Oxidative stress is defined as an impairment by ROS, which are by-products of the mitochondrial electron transport chain and other key molecular pathways that induce cell damage and can activate cellular senescence pathways. Cellular senescence is characterized by pro-inflammatory cytokines, growth factors, and proteases secreted by senescent cells, collectively known as the senescence-associated secretory phenotype (SASP). The presence of senescent cells, which disrupt tissue architecture and function and increase senescent cell production in liver tissues, contributes to fibrogenesis. Hepatic stellate cells (HSCs) are activated in response to chronic liver injury, oxidative stress, and senescence signals that drive excessive production and deposition of extracellular matrix. This review article aims to provide a comprehensive overview of the pathogenic role of ROS and cellular senescence in the aging liver and their contribution to fibrosis.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, 61421, Abha, Aseer, Saudi Arabia.
| |
Collapse
|
35
|
Manzo C, Isetta M, Castagna A. Infective agents and polymyalgia rheumatica: key discussion points emerging from a narrative review of published literature. Reumatologia 2024; 62:360-367. [PMID: 39677882 PMCID: PMC11635615 DOI: 10.5114/reum/194687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/16/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction The aetiology of polymyalgia rheumatica (PMR) is unknown. Recently, reports on cases of PMR following the coronavirus disease 2019 (COVID-19) have revived the role of infection as an aetiological or triggering factor. It is estimated that patients with PMR have manifestations of giant cell arteritis (GCA) in < 20% of cases. To date, little is known on the potential role of infectious agents in facilitating this association. Given this background, we performed a review of published literature. Our first aim was to review and discuss the relationship between PMR and infective agents. Secondly, we compared data of PMR-only patients with PMR and overlapping GCA to seek any commonalities or differences regarding the type of infectious agent in these two subgroups. Material and methods We performed a non-systematic literature search on Embase and Medline (COVID interface) with the following search terms: "polymyalgia rheumatica" AND "infections" OR "infectious agents", both MESH headings and free-text (in each language they were written). Each paper's reference list was scanned for additional publications meeting this study's aim. When papers reported data partially presented in previous articles, we referred to the most recent published data. Abstracts submitted at conferences or from non-peer-reviewed sources were not included. Polymyalgia rheumatica following vaccinations was an additional exclusion criterion. Results Several infectious agents have been held responsible for PMR. However, no definite causal link has been identified so far. According to our review, the search for a specific infectious agent, however intriguing, appears to be stagnating. Genetic background and epigenetic regulation probably play a key role. However, topical studies are lacking. Polymyalgia rheumatica as an adverse event following immunization should be kept methodologically distinct from PMR following an acute infection, as the adjuvants in the vaccine can make a significant difference. Conclusions Finally, some infectious agents are able to replicate in human arteries or have an endothelium tropism. Whilst these can theoretically trigger GCA, their role in isolated PMR seems minimal.
Collapse
Affiliation(s)
- Ciro Manzo
- Department of Internal and Geriatric Medicine, Azienda Sanitaria Locale Napoli 3 sud, Rheumatologic Outpatient Clinic, Health District No. 59, Naples, Sant’Agnello, Italy
| | - Marco Isetta
- Central and North West London NHS Trust, England
| | - Alberto Castagna
- Department of Primary Care, Health District of Soverato, Azienda Sanitaria Provinciale Catanzaro, Italy
| |
Collapse
|
36
|
Acuña-Rocha VD, Regalado-Ceballos D, Salcedo-Soto DA, Ramos-Delgado CA, Esquivel-Valerio JA, Hernandez Galarza IJ, Galarza-Delgado DA, Flores-Alvarado DE. Clinical Characteristics of Young-Onset Versus Elderly-Onset Rheumatoid Arthritis: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e74148. [PMID: 39712845 PMCID: PMC11663032 DOI: 10.7759/cureus.74148] [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] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease with a prevalence of 1%, mainly affecting women aged 25-45. It is classified by the age of onset into young-onset rheumatoid arthritis (YORA, 16-65 years) and elderly-onset rheumatoid arthritis (EORA, over 65 years), with EORA often presenting suddenly with systemic symptoms and large joint involvement due to age-related immune changes. This systematic review and meta-analysis compare the clinical and epidemiological characteristics of EORA and YORA. Observational studies were selected from PubMed, Scopus, Embase, Web of Science, and the Cochrane Central Database up to November 2023, focusing on a comparative analysis of both disease types with similar clinical progression and treatment duration limited to one month. Statistical analysis was performed in RStudio (Version 4.1.3, Posit Software, Boston, MA) using the "meta" package, applying a random effects model, inverse variance method, and Hartung-Knapp adjustment. Results for continuous variables were combined and grouped using the Cochrane formula, with medians and interquartile ranges transformed for uniformity. Four studies met the criteria. A trend was observed toward higher disease activity at diagnosis in the EORA group (mean difference (MD: 0.19, 95% CI -1.90 to 2.27), indicated by Disease Activity Score-28 (DAS28) and Simplified Disease Activity Index (SDAI) indices (MD 6.17, 95% CI -20.60 to 32.94). The EORA group also had higher Health Assessment Questionnaire (HAQ) scores (MD 0.21, 95% CI -0.03 to 0.46) and a greater number of painful (MD 1.31, 95% CI -0.86 to 3.47) and swollen joints (MD 2.35, 95% CI 0.77 to 3.92). Extra-articular manifestations, including rheumatoid nodules, lung involvement, and secondary Sjögren's syndrome, were more common in EORA patients (p < 0.004). In conclusion, the findings suggest that patients with EORA present with more intense disease activity at onset, a higher prevalence of extra-articular manifestations, greater levels of disability, and more pronounced radiographic changes. Despite these initial differences, EORA patients ultimately achieve long-term remission rates similar to those with YORA.
Collapse
Affiliation(s)
- Victor D Acuña-Rocha
- Internal Medicine, Hospital Universitario "Dr. José Eleuterio González", Monterrey, MEX
| | - Diego Regalado-Ceballos
- Rheumatology, Plataforma INVEST Medicina UANL-Ker Unit Mayo Clinic México, Universidad Autonoma de Nuevo León, Monterrey, MEX
| | | | - César A Ramos-Delgado
- Rheumatology, Plataforma INVEST Medicina UANL-Ker Unit Mayo Clinic México, Universidad Autonoma de Nuevo León, Monterrey, MEX
| | | | | | | | | |
Collapse
|
37
|
Liu F, Yao Y, Huang Y, Luo L, Wang Q, Chen B, Hu H. Gut microbiota and metabolic profile changes unveil the deterioration of alveolar bone inflammatory resorption with aging induced by D-galactose. Sci Rep 2024; 14:26135. [PMID: 39477973 PMCID: PMC11526011 DOI: 10.1038/s41598-024-75941-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
The global aging population has led to a rise in age-related health issues, such as malnutrition, metabolic disorders, and even immune decline. Among these concerns, periodontitis holds particular significance for the well-being of the elderly. This study aimed to investigate the impact of aging on inflammatory resorption of alveolar bone in mice with periodontitis, with a specific focus on alterations in the intestinal microenvironment. To achieve this, we established a D-galactose (D-gal)-induced aging mouse model with periodontitis and employed histopathological staining, oxidative stress, and inflammatory factors analyses to assess the severity of periodontitis and the health status. Additionally, the 16S rRNA sequencing and untargeted metabolomics analysis were employed to investigate alterations in the intestinal microbiota and metabolites. Our results showed that D-gal-induced aging mice with periodontitis experienced more pronounced alveolar bone inflammatory resorption and disruptions in the gut barrier, accompanied by an overall decline in physical condition. The microbial composition and structure of aged mice also underwent significant modifications, with a decreased Firmicutes/Bacteroidetes (F/B) ratio. Furthermore, metabolomics analysis demonstrated that D-gal-induced aging primarily influenced lipids and lipid-like molecules metabolism, and enrichment observed in the rheumatoid arthritis and histidine metabolism pathways. These findings provide further evidence that the aging process exacerbates age-related alveolar bone loss (ABL) through disturbances in intestinal homeostasis.
Collapse
Affiliation(s)
- Fangzhou Liu
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Yanzi Yao
- School of Stomatology, Zunyi Medical University, Zunyi, China
- Department of Stomatology, Luoyang Maternal and Child Health Hospital, Luoyang, China
| | - Yue Huang
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Liangliang Luo
- School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Qian Wang
- School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, China
| | - Bin Chen
- School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, China
| | - Huan Hu
- School of Stomatology, Zunyi Medical University, Zunyi, China.
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
38
|
Bolton C. Review of evidence linking exposure to environmental stressors and associated alterations in the dynamics of immunosenescence (ISC) with the global increase in multiple sclerosis (MS). Immun Ageing 2024; 21:73. [PMID: 39438909 PMCID: PMC11494837 DOI: 10.1186/s12979-024-00473-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
Historical survey confirms that, over the latter part of the 20th century, autoimmune-based diseases, including multiple sclerosis (MS), have shown a worldwide increase in incidence and prevalence. Analytical population studies have established that the exponential rise in MS is not solely due to improvements in diagnosis and healthcare but relates to an increase in autoimmune risk factors. Harmful environmental exposures, including non-communicable social determinants of health, anthropogens and indigenous or transmissible microbes, constitute a group of causal determinants that have been closely linked with the global rise in MS cases. Exposure to environmental stressors has profound effects on the adaptive arm of the immune system and, in particular, the associated intrinsic process of immune ageing or immunosenescence (ISC). Stressor-related disturbances to the dynamics of ISC include immune cell-linked untimely or premature (p) alterations and an accelerated replicative (ar) change. A recognised immune-associated feature of MS is pISC and current evidence supports the presence of an arISC during the disease. Moreover, collated data illustrates the immune-associated alterations that characterise pISC and arISC are inducible by environmental stressors strongly implicated in causing duplicate changes in adaptive immune cells during MS. The close relationship between exposure to environmental risk factors and the induction of pISC and arISC during MS offers a valid mechanism through which pro-immunosenescent stressors may act and contribute to the recorded increase in the global rate and number of new cases of the disease. Confirmation of alterations to the dynamics of ISC during MS provides a rational and valuable therapeutic target for the use of senolytic drugs to either prevent accumulation and enhance ablation of less efficient untimely senescent adaptive immune cells or decelerate the dysregulated process of replicative proliferation. A range of senotherapeutics are available including kinase and transcriptase inhibitors, rapalogs, flavanols and genetically-engineered T cells and the use of selective treatments to control emerging and unspecified aspects of pISC and arISC are discussed.
Collapse
|
39
|
Fu W, Wang T, Lu Y, Shi T, Yang Q. The role of lactylation in plasma cells and its impact on rheumatoid arthritis pathogenesis: insights from single-cell RNA sequencing and machine learning. Front Immunol 2024; 15:1453587. [PMID: 39421742 PMCID: PMC11484267 DOI: 10.3389/fimmu.2024.1453587] [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: 06/23/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by persistent synovitis, systemic inflammation, and autoantibody production. This study aims to explore the role of lactylation in plasma cells and its impact on RA pathogenesis. Methods We utilized single-cell RNA sequencing (scRNA-seq) data and applied bioinformatics and machine learning techniques. A total of 10,163 cells were retained for analysis after quality control. Clustering analysis identified 13 cell clusters, with plasma cells displaying the highest lactylation scores. We performed pathway enrichment analysis to examine metabolic activity, such as oxidative phosphorylation and glycolysis, in highly lactylated plasma cells. Additionally, we employed 134 machine learning algorithms to identify seven core lactylation-promoting genes and constructed a diagnostic model with an average AUC of 0.918. Results The RA lactylation score (RAlac_score) was significantly elevated in RA patients and positively correlated with immune cell infiltration and immune checkpoint molecule expression. Differential expression analysis between two plasma cell clusters revealed distinct metabolic and immunological profiles, with cluster 2 demonstrating increased immune activity and extracellular matrix interactions. qRT-PCR validation confirmed that NDUFB3, NGLY1, and SLC25A4 are highly expressed in RA. Conclusion This study highlights the critical role of lactylation in plasma cells for RA pathogenesis and identifies potential biomarkers and therapeutic targets, which may offer insights for future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Tiejun Shi
- Department of Orthopedics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Qining Yang
- Department of Orthopedics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| |
Collapse
|
40
|
Smith R, Bassand K, Dussol A, Piesse C, Duplus E, El Hadri K. A new model and precious tool to study molecular mechanisms of macrophage aging. Aging (Albany NY) 2024; 16:12697-12725. [PMID: 39373702 PMCID: PMC11501386 DOI: 10.18632/aging.206124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/06/2024] [Indexed: 10/08/2024]
Abstract
The accumulation of senescent cells, characterized by a senescence-associated secretory phenotype (SASP), contributes to chronic inflammation and age-related diseases (ARD). During aging, macrophages can adopt a senescent-like phenotype and an altered function, which promotes senescent cell accumulation. In the context of aging and ARD, controlling the resolution of the inflammatory response and preventing chronic inflammation, especially by targeting macrophages, must be a priority. Aging being a dynamic process, we developed a model of in vitro murine peritoneal macrophage aging. Our results show that macrophages cultured for 7 or 14 days exhibit a senescence-like phenotype: proliferation decrease, the levels of cyclin-dependent kinase inhibitors p16INK4A and p21CIP1 and of pro-inflammatory SASP components (MCP-1, IL-6, IL-1β, TNF-α, and MMP-9) increase, phagocytosis capacity decline and glycolytic activity is induced. In our model, chronic treatment with CB3, a thioredoxin-1 mimetic anti-inflammatory peptide, completely prevents p21CIP1 increase and enables day 14 macrophages to maintain proliferative activity.We describe a new model of macrophage aging with a senescence-like phenotype associated with inflammatory, metabolic and functional perturbations. This model is a valuable tool for characterizing macrophage aging mechanisms and developing innovative strategies with promising therapeutical purpose in limiting inflammaging and ARD.
Collapse
Affiliation(s)
- Rémy Smith
- Sorbonne Université, CNRS UMR 8256 Biological Adaptation and Ageing (B2A), INSERM U1164, Institut de Biologie Paris Seine (IBPS), Paris 75005, France
| | - Kévin Bassand
- INSERM U1148, Laboratory for Vascular and Translational Sciences (LVTS), Université Sorbonne Paris Nord, Bobigny 93000, France
| | - Ashok Dussol
- Sorbonne Université, CNRS UMR 8256 Biological Adaptation and Ageing (B2A), INSERM U1164, Institut de Biologie Paris Seine (IBPS), Paris 75005, France
| | - Christophe Piesse
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Plate-forme Ingénierie des Protéines et Synthèse Peptidique, Paris 75005, France
| | - Eric Duplus
- Sorbonne Université, CNRS UMR 8256 Biological Adaptation and Ageing (B2A), INSERM U1164, Institut de Biologie Paris Seine (IBPS), Paris 75005, France
| | - Khadija El Hadri
- Sorbonne Université, CNRS UMR 8256 Biological Adaptation and Ageing (B2A), INSERM U1164, Institut de Biologie Paris Seine (IBPS), Paris 75005, France
| |
Collapse
|
41
|
Bellanti F, Lo Buglio A, Dobrakowski M, Kasperczyk A, Kasperczyk S, Serviddio G, Vendemiale G. Adherence to Mediterranean Diet and Biomarkers of Redox Balance and Inflammation in Old Patients Hospitalized in Internal Medicine. Nutrients 2024; 16:3359. [PMID: 39408326 PMCID: PMC11478664 DOI: 10.3390/nu16193359] [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: 08/30/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES We have previously described that low adherence to the Mediterranean diet (MD) in elderly patients admitted in internal medicine wards is linked to poorer clinical outcomes. This investigation was designed to explore whether adherence to the MD is related to circulating markers of redox balance and inflammation in this clinical scenario. METHODS A cross-sectional study was performed on 306 acute old patients hospitalized in internal medicine wards. Adherence to the MD was estimated by the Italian Mediterranean Index (IMI). The circulating markers of redox balance were assessed in serum and erythrocytes and correlated with inflammatory markers across different MD adherence groups. RESULTS Compared to the patients with high adherence, those with low adherence to the MD exhibited severely impaired redox balance, as evidenced by a higher GSSG/GSH ratio and increased serum hydroxynonenal/malondialdehyde-protein adducts. No modifications were described in the expression of antioxidant enzymes in peripheral blood mononuclear cells. Patients with low adherence to the MD exhibited a higher neutrophil-to-lymphocyte ratio and markers of systemic inflammation, as well as raised levels of interleukin-6 and tumor necrosis factor, compared to those with high MD adherence. A strong association was observed between the circulating markers of redox balance and inflammation/immune response, with the highest regression coefficients found in the low adherence group. CONCLUSIONS Old patients admitted to internal medicine wards with low adherence to the MD display unfavorable profiles of the circulating markers of redox balance and inflammation. It is conceivable that such effects on redox balance can be linked to the high polyphenol content of MD. This study supports the rationale for intervention trials that attest to the effectiveness of MD as a nutritional strategy for disease prevention.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (F.B.); (A.L.B.); (G.S.)
| | - Aurelio Lo Buglio
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (F.B.); (A.L.B.); (G.S.)
| | - Michał Dobrakowski
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland; (M.D.); (A.K.); (S.K.)
| | - Aleksandra Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland; (M.D.); (A.K.); (S.K.)
| | - Sławomir Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland; (M.D.); (A.K.); (S.K.)
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (F.B.); (A.L.B.); (G.S.)
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (F.B.); (A.L.B.); (G.S.)
| |
Collapse
|
42
|
Yang J, Sun R, Pei Z. Improvement of the Immunity System Through Sports: Novel Regulatory Mechanisms for Hypertension. Rev Cardiovasc Med 2024; 25:385. [PMID: 39484112 PMCID: PMC11522756 DOI: 10.31083/j.rcm2510385] [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: 04/11/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 11/03/2024] Open
Abstract
Hypertension and its resulting target organ damage is a complex process associated with a range of physiological and molecular factors, including immune regulation. The profound effects of exercise on normal immune system function and the development and progression of hypertension are well known. This review aims to create new avenues for preventing and treating hypertension and its associated target organ damage. This narrative review emphasizes the role of exercise training in the prevention/treatment of hypertension development through immune response modulation and presents current perspectives on the available scientific evidence. Several studies have shown that exercise regulates hypertension by altering immune cells, which is partly attributable to the anti-inflammatory effects of exercise training. Regular exercise modifies immune modulation and could represent a new mechanism for regulating hypertension. Although the utilization of exercise training and the immune system in conjunction for treating and preventing hypertension is still in its early stages, current scientific literature indicates numerous potential physiological links between exercise training, the immune system, and hypertension.
Collapse
Affiliation(s)
- Jin Yang
- Department of Central Laboratory, Central Hospital of Dalian University of Technology, 116033 Dalian, Liaoning, China
| | - Rui Sun
- Emergency Department, The First Affiliated Hospital of Dalian Medical University, 116011 Dalian, Liaoning, China
| | - Zuowei Pei
- Department of Central Laboratory, Central Hospital of Dalian University of Technology, 116033 Dalian, Liaoning, China
- Department of Cardiology, Central Hospital of Dalian University of Technology, 116033 Dalian, Liaoning, China
- Faculty of Medicine, Dalian University of Technology, 116024 Dalian, Liaoning, China
| |
Collapse
|
43
|
Salminen A. Inhibitory immune checkpoints suppress the surveillance of senescent cells promoting their accumulation with aging and in age-related diseases. Biogerontology 2024; 25:749-773. [PMID: 38954358 PMCID: PMC11374851 DOI: 10.1007/s10522-024-10114-w] [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: 04/19/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The accumulation of pro-inflammatory senescent cells within tissues is a common hallmark of the aging process and many age-related diseases. This modification has been called the senescence-associated secretory phenotype (SASP) and observed in cultured cells and in cells isolated from aged tissues. Currently, there is a debate whether the accumulation of senescent cells within tissues should be attributed to increased generation of senescent cells or to a defect in their elimination from aging tissues. Emerging studies have revealed that senescent cells display an increased expression of several inhibitory immune checkpoint ligands, especially those of the programmed cell death protein-1 (PD-1) ligand-1 (PD-L1) proteins. It is known that the PD-L1 ligands, especially those of cancer cells, target the PD-1 receptor of cytotoxic CD8+ T and natural killer (NK) cells disturbing their functions, e.g., evoking a decline in their cytotoxic activity and promoting their exhaustion and even apoptosis. An increase in the level of the PD-L1 protein in senescent cells was able to suppress their immune surveillance and inhibit their elimination by cytotoxic CD8+ T and NK cells. Senescent cells are known to express ligands for several inhibitory immune checkpoint receptors, i.e., PD-1, LILRB4, NKG2A, TIM-3, and SIRPα receptors. Here, I will briefly describe those pathways and examine whether these inhibitory checkpoints could be involved in the immune evasion of senescent cells with aging and age-related diseases. It seems plausible that an enhanced inhibitory checkpoint signaling can prevent the elimination of senescent cells from tissues and thus promote the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
44
|
Liang D, Wang L, Liu S, Li S, Zhou X, Xiao Y, Zhong P, Chen Y, Wang C, Xu S, Su J, Luo Z, Ke C, Lai Y. Global Incidence of Diarrheal Diseases-An Update Using an Interpretable Predictive Model Based on XGBoost and SHAP: A Systematic Analysis. Nutrients 2024; 16:3217. [PMID: 39339819 PMCID: PMC11434730 DOI: 10.3390/nu16183217] [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: 08/21/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Diarrheal disease remains a significant public health issue, particularly affecting young children and older adults. Despite efforts to control and prevent these diseases, their incidence continues to be a global concern. Understanding the trends in diarrhea incidence and the factors influencing these trends is crucial for developing effective public health strategies. OBJECTIVE This study aimed to explore the temporal trends in diarrhea incidence and associated factors from 1990 to 2019 and to project the incidence for the period 2020-2040 at global, regional, and national levels. We aimed to identify key factors influencing these trends to inform future prevention and control strategies. METHODS The eXtreme Gradient Boosting (XGBoost) model was used to predict the incidence from 2020 to 2040 based on demographic, meteorological, water sanitation, and sanitation and hygiene indicators. SHapley Additive exPlanations (SHAP) value was performed to explain the impact of variables in the model on the incidence. Estimated annual percentage change (EAPC) was calculated to assess the temporal trends of age-standardized incidence rates (ASIRs) from 1990 to 2019 and from 2020 to 2040. RESULTS Globally, both incident cases and ASIRs of diarrhea increased between 2010 and 2019. The incident cases are expected to rise from 2020 to 2040, while the ASIRs and incidence rates are predicted to slightly decrease. During the observed (1990-2019) and predicted (2020-2040) periods, adults aged 60 years and above exhibited an upward trend in incidence rate as age increased, while children aged < 5 years consistently had the highest incident cases. The SHAP framework was applied to explain the model predictions. We identified several risk factors associated with an increased incidence of diarrhea, including age over 60 years, yearly precipitation exceeding 3000 mm, temperature above 20 °C for both maximum and minimum values, and vapor pressure deficit over 1500 Pa. A decreased incidence rate was associated with relative humidity over 60%, wind speed over 4 m/s, and populations with above 80% using safely managed drinking water services and over 40% using safely managed sanitation services. CONCLUSIONS Diarrheal diseases are still serious public health concerns, with predicted increases in the incident cases despite decreasing ASIRs globally. Children aged < 5 years remain highly susceptible to diarrheal diseases, yet the incidence rate in the older adults aged 60 plus years still warrants additional attention. Additionally, more targeted efforts to improve access to safe drinking water and sanitation services are crucial for reducing the incidence of diarrheal diseases globally.
Collapse
Affiliation(s)
- Dan Liang
- Department of Immunology and Microbiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (D.L.); (S.L.); (X.Z.); (P.Z.); (Y.C.)
| | - Li Wang
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou 510275, China;
| | - Shuang Liu
- Department of Immunology and Microbiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (D.L.); (S.L.); (X.Z.); (P.Z.); (Y.C.)
| | - Shanglin Li
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, Guangzhou 510632, China;
| | - Xing Zhou
- Department of Immunology and Microbiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (D.L.); (S.L.); (X.Z.); (P.Z.); (Y.C.)
| | - Yun Xiao
- School of Public Health, Southern Medical University, Guangzhou 510515, China;
| | - Panpan Zhong
- Department of Immunology and Microbiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (D.L.); (S.L.); (X.Z.); (P.Z.); (Y.C.)
| | - Yanxi Chen
- Department of Immunology and Microbiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (D.L.); (S.L.); (X.Z.); (P.Z.); (Y.C.)
| | - Changyi Wang
- Department of Cardiovascular and Cerebrovascular and Diabetes Prevention and Treatment, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen 518000, China; (C.W.); (S.X.)
| | - Shan Xu
- Department of Cardiovascular and Cerebrovascular and Diabetes Prevention and Treatment, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen 518000, China; (C.W.); (S.X.)
| | - Juan Su
- Guangdong Provincial Key Laboratory for Emergency Detection and Research on Pathogen of Emerging Infectious Disease, Guangdong Provincial Center for Disease Control and Prevention, Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangzhou 511430, China;
| | - Zhen Luo
- Department of Immunology and Microbiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (D.L.); (S.L.); (X.Z.); (P.Z.); (Y.C.)
| | - Changwen Ke
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou 510275, China;
- School of Public Health, Southern Medical University, Guangzhou 510515, China;
- Guangdong Provincial Key Laboratory for Emergency Detection and Research on Pathogen of Emerging Infectious Disease, Guangdong Provincial Center for Disease Control and Prevention, Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangzhou 511430, China;
| | - Yingsi Lai
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou 510275, China;
| |
Collapse
|
45
|
Gao H, Nepovimova E, Adam V, Heger Z, Valko M, Wu Q, Kuca K. Age-associated changes in innate and adaptive immunity: role of the gut microbiota. Front Immunol 2024; 15:1421062. [PMID: 39351234 PMCID: PMC11439693 DOI: 10.3389/fimmu.2024.1421062] [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: 04/21/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Aging is generally regarded as an irreversible process, and its intricate relationship with the immune system has garnered significant attention due to its profound implications for the health and well-being of the aging population. As people age, a multitude of alterations occur within the immune system, affecting both innate and adaptive immunity. In the realm of innate immunity, aging brings about changes in the number and function of various immune cells, including neutrophils, monocytes, and macrophages. Additionally, certain immune pathways, like the cGAS-STING, become activated. These alterations can potentially result in telomere damage, the disruption of cytokine signaling, and impaired recognition of pathogens. The adaptive immune system, too, undergoes a myriad of changes as age advances. These include shifts in the number, frequency, subtype, and function of T cells and B cells. Furthermore, the human gut microbiota undergoes dynamic changes as a part of the aging process. Notably, the interplay between immune changes and gut microbiota highlights the gut's role in modulating immune responses and maintaining immune homeostasis. The gut microbiota of centenarians exhibits characteristics akin to those found in young individuals, setting it apart from the microbiota observed in typical elderly individuals. This review delves into the current understanding of how aging impacts the immune system and suggests potential strategies for reversing aging through interventions in immune factors.
Collapse
Affiliation(s)
- Haoyu Gao
- College of Life Science, Yangtze University, Jingzhou, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| |
Collapse
|
46
|
Li Y, Yin H, Yuan H, Wang E, Wang C, Li H, Geng X, Zhang Y, Bai J. IL-10 deficiency aggravates cell senescence and accelerates BLM-induced pulmonary fibrosis in aged mice via PTEN/AKT/ERK pathway. BMC Pulm Med 2024; 24:443. [PMID: 39261827 PMCID: PMC11389321 DOI: 10.1186/s12890-024-03260-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is an aging-related progressive lung disorder. The aged lung undergoes functional and structural changes termed immunosenescence and inflammaging, which facilitate the occurrence of fibrosis. Interleukin-10 (IL-10) is a potent anti-inflammatory and immunoregulatory cytokine, yet it remains unclear how IL-10 deficiency-induced immunosenescence participates in the development of PF. METHODS Firstly we evaluated the susceptibility to fibrosis and IL-10 expression in aged mice. Then 13-month-old wild-type (WT) and IL-10 knockout (KO) mice were subjected to bleomycin(BLM) and analyzed senescence-related markers by PCR, western blot and immunohistochemistry staining of p16, p21, p53, as well as DHE and SA-β-gal staining. We further compared 18-month-old WT mice with 13-month-old IL-10KO mice to assess aging-associated cell senescence and inflamation infiltration in both lung and BALF. Moreover, proliferation and apoptosis of alveolar type 2 cells(AT2) were evaluated by FCM, immunofluorescence, TUNEL staining, and TEM analysis. Recombinant IL-10 (rIL-10) was also administered intratracheally to evaluate its therapeutic potential and related mechanism. For the in vitro experiments, 10-week-old naïve pramily lung fibroblasts(PLFs) were treated with the culture medium of 13-month PLFs derived from WT, IL-10KO, or IL-10KO + rIL-10 respectively, and examined the secretion of senescence-associated secretory phenotype (SASP) factors and related pathways. RESULTS The aged mice displayed increased susceptibility to fibrosis and decreased IL-10 expression. The 13-month-old IL-10KO mice exhibited significant exacerbation of cell senescence compared to their contemporary WT mice, and even more severe epithelial-mesenchymal transition (EMT) than that of 18 month WT mice. These IL-10 deficient mice showed heightened inflammatory responses and accelerated PF progression. Intratracheal administration of rIL-10 reduced lung CD45 + cell infiltration by 15%, including a 6% reduction in granulocytes and a 10% reduction in macrophages, and increased the proportion of AT2 cells by approximately 8%. Additionally, rIL-10 significantly decreased α-SMA and collagen deposition, and reduced the expression of senescence proteins p16 and p21 by 50% in these mice. In vitro analysis revealed that conditioned media from IL-10 deficient mice promoted SASP secretion and upregulated senescence genes in naïve lung fibroblasts, which was mitigated by rIL-10 treatment. Mechanistically, rIL-10 inhibited TGF-β-Smad2/3 and PTEN/PI3K/AKT/ERK pathways, thereby suppressing senescence and fibrosis-related proteins. CONCLUSIONS IL-10 deficiency in aged mice leads to accelerated cell senescence and exacerbated fibrosis, with IL-10KO-PLFs displaying increased SASP secretion. Recombinant IL-10 treatment effectively mitigates these effects, suggesting its potential as a therapeutic target for PF.
Collapse
Affiliation(s)
- Yinzhen Li
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200092, China
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Hui Yin
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200092, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, China
| | - Huixiao Yuan
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200092, China
| | - Enhao Wang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200092, China
| | - Chunmei Wang
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Hongqiang Li
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xuedi Geng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200092, China
| | - Ying Zhang
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Jianwen Bai
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
47
|
Sanchez-Garcia E, Cruz-Jentoft AJ, Ravasco P, Suominen M, Pitkälä PK. Nutritional care in older adults: are we doing everything? An expert opinion review. Curr Med Res Opin 2024; 40:1563-1576. [PMID: 39044672 DOI: 10.1080/03007995.2024.2380007] [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: 05/28/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
Malnutrition is a prevalent, yet often underdiagnosed and undertreated, condition in older adults. It is characterized by weight loss and/or reduced muscle mass due to diminished caloric intake, inflammation, and/or disease burden. In return, malnutrition can lead to diminished skeletal muscle functionality and disability, among others. Malnutrition plays a crucial role in the pathogenesis of two prevalent geriatric syndromes, namely sarcopenia and frailty. The complex interplay between malnutrition, sarcopenia, and frailty significantly impacts the older population, leading to increased morbidity, mortality, hospitalization rates, quality-of-life, and healthcare costs. Given the prognostic significance of malnutrition in geriatric care, recent guidelines emphasized the role of nutritional support in vulnerable populations. A group of vulnerable populations to malnutrition, sarcopenia, and frailty are older patients with hip fractures, cancer patients, and those with sarcopenic dysphagia. This article highlights the importance of individualized nutritional assessment and treatment in the management of vulnerable populations such as older patients with hip fractures, cancer, and those suffering from sarcopenic dysphagia. It presents practical protocols and guidelines that can be instrumental in enhancing the nutritional care of these groups, thereby improving their overall health outcomes.
Collapse
Affiliation(s)
- Elisabet Sanchez-Garcia
- Consultant in Geriatric Medicine, Mater Private Network, Cork, Ireland
- Servicio de Geriatría, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | | | - Paula Ravasco
- Universidade Católica Portuguesa, Faculty of Medicine and Centre for Interdisciplinary Research in Health, Centre for Interdisciplinary Research Egas Moniz (CiiEM), Lisbon, Portugal
| | - Merja Suominen
- Department of General Practice and Primary Health Care, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Prof Kaisu Pitkälä
- Department of General Practice and Primary Health Care, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
48
|
Koga HK, Grodstein F, Williams DR, Demeo DL, Kubzansky LD. Relations of optimism and purpose in life to immune markers in aging. J Psychosom Res 2024; 184:111851. [PMID: 38964200 DOI: 10.1016/j.jpsychores.2024.111851] [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: 01/11/2024] [Revised: 06/13/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Optimism and purpose in life are associated with improved health outcomes. More information is needed on biological mechanisms, including immunosenescence. We investigated if psychological well-being is associated with healthier immunosenescence-related measures including naïve and terminally differentiated CD4+ and CD8+ T cell percentages, CD4+:CD8+, and cytomegalovirus (CMV) IgG response. METHODS Participants were adults over age 50 from the Health and Retirement Study. Optimism was measured using the Life Orientation Test Revised. Purpose in life was assessed using the subscale from the Ryff psychological well-being measure. We examined the cross-sectional associations of optimism and purpose in life with measures of T cell subsets using linear regression and with CMV IgG using ordered logit regression, controlling for potential confounding factors. RESULTS The final analytic sample ranged from 7250 to 7870. After adjusting for sociodemographic factors, a 1-SD increment in optimism was associated with the percentage of naïve CD4+ T cells increasing by 0.6 (95%CI 0.2%, 1.0%). A 1-SD increment in purpose in life was associated with the percentage of naïve CD4+ T cells increasing by 0.9 (95%CI 0.5%, 1.3%) after adjusting for sociodemographic factors and the association was maintained after further adjustments for health conditions, depression, and health behaviors. For naïve CD8+ T cell percentages, CD4:CD8 ratios, and CMV IgG antibodies, associations were seen only in models that adjusted for age. No significant associations were seen in any models for the terminally differentiated CD4+ and CD8+ T cells. CONCLUSIONS We found associations of optimism and purpose in life with naïve CD4+ T cell percentages.
Collapse
Affiliation(s)
- Hayami K Koga
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America; Harvard Center for Population and Development Studies, Cambridge, MA, United States of America.
| | - Francine Grodstein
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States of America
| | - David R Williams
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America; Department of African and African American Studies, Department of Sociology, Harvard University, Cambridge, MA, United States of America
| | - Dawn L Demeo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Laura D Kubzansky
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| |
Collapse
|
49
|
Nakashima K, Imai T, Shiraishi A, Unose R, Goto H, Nagatomo Y, Kojima-Ishii K, Mushimoto Y, Nishiyama K, Yamamura K, Nagata H, Ishimura M, Kusuhara K, Koga Y, Sakai Y, Ohga S. The immunoreactive signature of monocyte-derived dendritic cells from patients with Down syndrome. Clin Exp Immunol 2024; 217:291-299. [PMID: 38916251 PMCID: PMC11310712 DOI: 10.1093/cei/uxae048] [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: 08/15/2023] [Revised: 04/17/2024] [Accepted: 06/24/2024] [Indexed: 06/26/2024] Open
Abstract
The clinical spectrum of Down syndrome (DS) ranges from congenital malformations to premature aging and early-onset senescence. Excessive immunoreactivity and oxidative stress are thought to accelerate the pace of aging in DS patients; however, the immunological profile remains elusive. We investigated whether peripheral blood monocyte-derived dendritic cells (MoDCs) in DS patients respond to lipopolysaccharide (LPS) distinctly from non-DS control MoDCs. Eighteen DS patients (age 2-47 years, 12 males) and 22 controls (age 4-40 years, 15 males) were enrolled. CD14-positive monocytes were immunopurified and cultured for 7 days in the presence of granulocyte-macrophage colony-stimulating factor and IL-4, yielding MoDCs in vitro. After the LPS-stimulation for 48 hours from days 7 to 9, culture supernatant cytokines were measured by multiplex cytokine bead assays, and bulk-prepared RNA from the cells was used for transcriptomic analyses. MoDCs from DS patients produced cytokines/chemokines (IL-6, IL-8, TNF-α, MCP-1, and IP-10) at significantly higher levels than those from controls in response to LPS. RNA sequencing revealed that DS-derived MoDCs differentially expressed 137 genes (74 upregulated and 63 downregulated) compared with controls. A gene enrichment analysis identified 5 genes associated with Toll-like receptor signaling (KEGG: hsa04620, P = 0.00731) and oxidative phosphorylation (hsa00190, P = 0.0173) pathways. MoDCs obtained from DS patients showed higher cytokine or chemokine responses to LPS than did control MoDCs. Gene expression profiles suggest that hyperactive Toll-like receptor and mitochondrial oxidative phosphorylation pathways configure the immunoreactive signature of MoDCs in DS patients.
Collapse
Affiliation(s)
- Kentaro Nakashima
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takashi Imai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Shiraishi
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryoko Unose
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hironori Goto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusaku Nagatomo
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kanako Kojima-Ishii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Mushimoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kei Nishiyama
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichiro Yamamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hazumu Nagata
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ishimura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Kusuhara
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuhki Koga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
50
|
Mi B, Xiong Y, Knoedler S, Alfertshofer M, Panayi AC, Wang H, Lin S, Li G, Liu G. Ageing-related bone and immunity changes: insights into the complex interplay between the skeleton and the immune system. Bone Res 2024; 12:42. [PMID: 39103328 PMCID: PMC11300832 DOI: 10.1038/s41413-024-00346-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
Ageing as a natural irreversible process inherently results in the functional deterioration of numerous organ systems and tissues, including the skeletal and immune systems. Recent studies have elucidated the intricate bidirectional interactions between these two systems. In this review, we provide a comprehensive synthesis of molecular mechanisms of cell ageing. We further discuss how age-related skeletal changes influence the immune system and the consequent impact of immune system alterations on the skeletal system. Finally, we highlight the clinical implications of these findings and propose potential strategies to promote healthy ageing and reduce pathologic deterioration of both the skeletal and immune systems.
Collapse
Affiliation(s)
- Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Michael Alfertshofer
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig - Maximilian University Munich, Munich, Germany
| | - Adriana C Panayi
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Hand-, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Haixing Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China.
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|