1
|
Ragab A, Ayman R, Salem MA, Ammar YA, Abusaif MS. Unveiling a novel pyrazolopyrimidine scaffold as a dual COX-2/5-LOX inhibitor with immunomodulatory potential: Design, synthesis, target prediction, anti-inflammatory activity, and ADME-T with docking simulation. Eur J Med Chem 2025; 290:117499. [PMID: 40101450 DOI: 10.1016/j.ejmech.2025.117499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/20/2025]
Abstract
Dual-target COX-2/5-LOX inhibitors are regarded as a rational strategy for the design of potent anti-inflammatory agents with favorable safety profiles. In this study, novel pyrazolo[1,5-a]pyrimidine derivatives were synthesized, developed, and screened for their ability to inhibit the cyclooxygenase-2 enzyme in vitro, with comparisons made to the established inhibitors Celecoxib and Meloxicam. Spectroscopic analyses confirmed the structure of the designed derivatives. The target prediction using AI was performed to identify potential targets that could be engaged through Swiss target prediction database. The SAR study was established by incorporating various substituents and nuclei into the pyrazolopyrimidine pharmacophore. The synthesized pyrazolopyrimidines exhibited IC50 values ranging from 53.32 ± 4.43 to 254.90 ± 6.45 nM, in comparison to Celecoxib (IC50 = 6.73 ± 5.69 nM) and Meloxicam (IC50 = 52.35 ± 6.66 nM). Notably, compound 5a was identified as the most active derivative, demonstrating an IC50 of 53.32 ± 4.43 nM. The three most prominent pyrazolopyrimidine derivatives, 3a, 5a, and 6a, were subsequently evaluated for their ability to inhibit the COX-1 and 5-LOX enzymes. Compounds 3a, 5a, and 6a demonstrated inhibitory activity against COX-1, with IC50 values of 476.45 ± 16.56, 757.51 ± 2.61, and 169.13 ± 5.77 nM, respectively. These derivatives 3a, 5a, and 6a showed significant selectivity index values of 7.91, 14.20, and 2.80, respectively, toward COX-2 rather than COX-1 in comparison to Meloxicam (SI = 0.75) and Celecoxib (SI = 2.35). Moreover, compound 5a exhibited 86 % inhibition compared to Zileuton's 88 %, while compounds 3a and 6a displayed inhibition rates of 84 % and 80 %, respectively, at a concentration of 100 μM. The most potent compound 5a, demonstrated the highest 5-LOX inhibitory activity, with IC50 of 2.292 ± 0.14 μM. The most promising pyrazolopyrimidine derivative 5a demonstrated a down-regulation of TNF-α and IL-6 gene expression by approximately 0.3826-fold and 0.2732-fold, respectively, when compared to Celecoxib, which induced reductions of 0.2320-fold and 0.2730-fold in these cytokines to promote apoptosis in RAW264.7 cells. Finally, in-silico ADME-T and docking simulations were conducted to predict the oral bioavailability, toxicity, and binding interactions with binding affinity.
Collapse
Affiliation(s)
- Ahmed Ragab
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt; Chemistry Department, Faculty of Science, Galala University, Galala City, 43511, Suez, Egypt.
| | - Radwa Ayman
- Department of Chemistry, Faculty of Science (Girls), Al-Azhar University, Nasr City, Cairo, Egypt.
| | - Mohamed A Salem
- Department of Chemistry, Faculty of Science and Arts, King Khalid University, Mohail, Assir, Saudi Arabia
| | - Yousry A Ammar
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Moustafa S Abusaif
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt.
| |
Collapse
|
2
|
Xue L, Lewis E, Bocharova M, Young AH, Aarsland D. Decreased neutrophil-to-lymphocyte ratio predicted cognitive improvement in late-life depression treated with vortioxetine: Findings from an eight-week randomized controlled trial. Brain Behav Immun 2025; 126:53-58. [PMID: 39921151 DOI: 10.1016/j.bbi.2025.01.029] [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: 09/14/2024] [Revised: 01/16/2025] [Accepted: 01/31/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Elevated neutrophil-to-lymphocyte ratio, a marker of inflammation, has been reported in adult and late-life depression. Vortioxetine has shown efficacy in treatment of late-life depression, yet little is known regarding its immunomodulatory role in clinical trials. METHODS This is a post-hoc analysis of an eight-week randomized controlled trial. Depressed patients aged 65 or above were treated by vortioxetine, duloxetine or placebo. 321 patients that have taken blood tests at baseline and endpoint were included in the analysis. Neutrophil-to-lymphocyte ratio (NLR) was calculated using the absolute counts of each cell type. Cognitive performance was assessed by composite score of Digit Symbol Substitution Test (DSST) and the Rey Auditory Verbal Learning Test (RAVLT) tasks, while depressive symptoms were assessed by Montgomery-Åsberg Depression Rating Scale (MADRS) and Geriatric Depression Scale (GDS). RESULTS NLR levels decreased significantly in the entire analysis set (t(320) = 2.64, p = 0.008) and in the vortioxetine group (M = -0.186, t(105) = 2.070, p = 0.041, Cohen's d = 0.20), but not in the two other groups. This decrease was not significantly different compared to placebo (F(1, 213) = 0.420, p = 0.517). Furthermore, larger NLR changes in vortioxetine arm predicted significant cognitive improvement (β = -4.03, p = 0.03), specifically regarding the DSST correct symbols (β = -1.97, p = 0.04) and RAVLT delayed recall (β = -1.87, p = 0.02) tasks. Additionally, decreased NLR significantly predicted reduced GDS score (β = 1.82, p = 0.02), yet not MADRS score. CONCLUSION Vortioxetine treatment was associated with decreased NLR levels in late-life depression, and reductions in NLR predicted improvements in cognitive function and depressive symptoms, suggesting a potential link between inflammation and clinical outcomes.
Collapse
Affiliation(s)
- Lingfeng Xue
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom.
| | - Elin Lewis
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| | - Mariia Bocharova
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| | - Allan H Young
- Centre for Affective Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| | - Dag Aarsland
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| |
Collapse
|
3
|
Huang Y, Li S, Ye W, Wang H, Su J, Gao L, Shi R, Mou X, Leng SX, Xiao C, Chen G. Viral Infections in Elderly Individuals: A Comprehensive Overview of SARS-CoV-2 and Influenza Susceptibility, Pathogenesis, and Clinical Treatment Strategies. Vaccines (Basel) 2025; 13:431. [PMID: 40333344 PMCID: PMC12031201 DOI: 10.3390/vaccines13040431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
As age increases, the immune function of elderly individuals gradually decreases, increasing their susceptibility to infectious diseases. Therefore, further research on common viral infections in the elderly population, especially severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses, is crucial for scientific progress. This review delves into the genetic structure, infection mechanisms, and impact of coinfections with these two viruses and provides a detailed analysis of the reasons for the increased susceptibility of elderly individuals to dual viral infections. We evaluated the clinical manifestations in elderly individuals following coinfections, including complications in the respiratory, gastrointestinal, nervous, and cardiovascular systems. Ultimately, we have summarized the current strategies for the prevention, diagnosis, and treatment of SARS-CoV-2 and influenza coinfections in older adults. Through these studies, we aim to reduce the risk of dual infections in elderly individuals and provide a scientific basis for the prevention, diagnosis, and treatment of age-related viral diseases, thereby improving their health status.
Collapse
Affiliation(s)
- Yanhao Huang
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), School of Medicine, Jinan University, Dongguan 523000, China;
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Shumin Li
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Wenjie Ye
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Haoyun Wang
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jun Su
- First Affiliated Hospital, Jinan University, Guangzhou 510632, China;
| | - Lijuan Gao
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Ruohu Shi
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xinyi Mou
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Sean Xiao Leng
- Johns Hopkins Center on Aging and Immune Remodeling, Division of Geriatric Medicine and Gerontology, Departments of Medicine, Molecular Microbiology and Immunology, Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Chanchan Xiao
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), School of Medicine, Jinan University, Dongguan 523000, China;
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| | - Guobing Chen
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), School of Medicine, Jinan University, Dongguan 523000, China;
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| |
Collapse
|
4
|
Guo LL, Liu HK, Cao JF, Zhang HX, Li B, Li T, Li L. Senility, defecation disorders, sleep disorders, and non-operative spinal infections: A single-center retrospective analysis. World J Orthop 2025; 16:103388. [PMID: 40290607 PMCID: PMC12019144 DOI: 10.5312/wjo.v16.i4.103388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/21/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Non-operative spinal infections (NOSI) are caused by tuberculosis, brucella, and other specific bacteria. The etiology of the disease is insidious, the onset is slow and the diagnosis and treatment are difficult. Identifying the factors associated with spinal infection and early intervention can reduce the occurrence of the disease. At present, the research mainly focuses on the accurate diagnosis and treatment of spinal infection, and there are few studies on the prevention of spinal infection. The concept of "preventive treatment of diseases" in traditional Chinese medicine may help identify the causes and reduce the occurrence of NOSI. AIM To determine the association of age, bowel movements, and sleep patterns with NOSI. METHODS Data of 69 NOSI patients and 84 healthy controls in a tertiary hospital from January 2019 to June 2024 were collected. Patients with NOSI had imaging evidence (magnetic resonance imaging) of spinal infections (including infections caused by tuberculosis, brucopathy, and other pathogens) and had no history of spinal surgery in the last 1 year were included in the analysis. Patients with spinal infection due to spinal surgery are excluded in the study. Data including age, sex, place of residence, sleeping status, and bowel movements were collected. SPSS22.0 was used for correlation analysis of all data. RESULTS The mean age of the NOSI group and the control group was 63.55 ± 14.635 years and 59.18 ± 17.111 years, respectively, without statistical difference (P = 0.096). There was also no statistically significant difference in gender between the two groups. In the NOSI group, 45 (65.22%) were over 60 years old, and 44 (63.77%) were rural residents. Compared with the control group, the NOSI group had more patients with sleep disorder and defecation disorder, accounting for 69.57% and 68.12%, respectively, with significant statistical difference (both P < 0.001). Regression analysis showed that defecation and sleep disorders were closely related to NOSI (both P < 0.001). CONCLUSION Most patients with NOSI are older and have sleep disorders and abnormal defecation.
Collapse
Affiliation(s)
- Ling-Ling Guo
- Department of Orthopaedics of Integrated Traditional Chinese and Western Medicine, Zibo Central Hospital, Zibo 255000, Shandong Province, China
| | - Hong-Kun Liu
- Department of Orthopaedics of Integrated Traditional Chinese and Western Medicine, Zibo Central Hospital, Zibo 255000, Shandong Province, China
| | - Jin-Feng Cao
- Department of Radiology, Zibo Central Hospital, Zibo 255036, Shandong Province, China
| | - Hai-Xia Zhang
- Telemedicine Consultation Center, Zibo Central Hospital, Zibo 255000, Shandong Province, China
| | - Bo Li
- Department of Orthopaedics of Integrated Traditional Chinese and Western Medicine, Zibo Central Hospital, Zibo 255000, Shandong Province, China
| | - Tong Li
- Department of Traditional Chinese Medicine, Zibo Central Hospital, Zibo 255000, Shandong Province, China
| | - Liang Li
- Department of Traditional Chinese Medicine, Zibo Central Hospital, Zibo 255000, Shandong Province, China
| |
Collapse
|
5
|
Kannan A, Jeffrey K, Misbah S, Ramasamy K. Practical guidance on the prevention and management of infection in multiple myeloma patients: A case-based approach. Blood Rev 2025:101287. [PMID: 40240231 DOI: 10.1016/j.blre.2025.101287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
The risk of infection in multiple myeloma patients is significant, due to immune dysfunction secondary to myeloma, immunosenescence and age-related comorbidities, given the elderly myeloma patient demographic. Newer treatments, despite providing unprecedented improvements in disease-control, have further elevated infection risk. This risk is so substantial that we are approaching a period where a subset of older myeloma patients may be more likely to die secondary to infectious complications imposed by redirected T-cell therapy rather than from myeloma. As a result, it is essential to provide myeloma patients with the appropriate prophylaxis and monitoring against infection. In this review, we discuss disease-related, patient-related and treatment-related reasons for the increased infection risk in myeloma patients, and how to both prevent and manage this risk through creating a dynamic, infection prevention plan that is personalised to the individual patient.
Collapse
Affiliation(s)
- A Kannan
- Medical Sciences Division, Medical Sciences Division, Academic Centre, John Radcliffe Hospital, University of Oxford, Headington OX3 9DU, United Kingdom.
| | - K Jeffrey
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK; Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford OX3 7BN, UK.
| | - S Misbah
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK.
| | - K Ramasamy
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK; Oxford Translational Myeloma Centre, NDORMS, University of Oxford, Oxford OX3 7LD, UK.
| |
Collapse
|
6
|
Jiang B, Dong YN, Xiong Y, Jiang CX, Ping J, Wu Q, Xu LJ, Shu RZ, Gao DD, Zhu SM, Ye WD, Zhang F. Global research trends in inflammaging from 2005 to 2024: a bibliometric analysis. FRONTIERS IN AGING 2025; 6:1554186. [PMID: 40276724 PMCID: PMC12018403 DOI: 10.3389/fragi.2025.1554186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025]
Abstract
Background Inflammaging, defined as chronic low-grade inflammation associated with aging, is considered a key factor in many age-related diseases. Despite growing research, comprehensive assessments of trends and focuses on this field over the past 2 decades remain lacking. Objective To comprehensively analyze literature development trends, scientific priorities, and their evolution in the field of inflammaging from 2005 to 2024 using bibliometric analysis. Methods Academic literature on inflammaging was retrieved from the Web of Science Core Collection. CiteSpace software was used as the bibliometric tool to analyze annual publication trends, contributing countries/regions, leading research institutions, primary journals, and keyword co-occurrence, including clustering and burst analysis in this field. Results The study included 1,800 eligible articles, demonstrating a consistent growth in research publications over the past 20 years. The United States and Italy were the principal contributors. The University of Bologna had the highest publication. Professor Claudio Franceschi has been a leading figure in this field. Journal analysis shows that research themes predominantly focus on molecular biology/immunology and medicine/clinical fields. Keyword analysis identifies major research hotspots as "inflammaging," "Crohn's disease," "periodontitis," "immunosenescence," "skeletal muscle," "gut microbiota," and "Parkinson's disease." Emerging term analysis indicates a shift from specific inflammatory diseases to broader aging and immune modulation studies. Conclusion This first systematic assessment of literature trends in the field of inflammaging from 2005 to 2024 reveals sustained academic growth and an increasingly deep research focus.
Collapse
Affiliation(s)
- Beier Jiang
- Department of Respiratory and Critical Care, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Yi-ni Dong
- Wenzhou Medical University, Wenzhou, China
| | - Yu Xiong
- Wenzhou Medical University, Wenzhou, China
| | - Chun-xia Jiang
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Jun Ping
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Qi Wu
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Liu-jun Xu
- Department of Respiratory and Critical Care, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Run-zhe Shu
- Department of Respiratory and Critical Care, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
- Shunxi Bio-Pharmaceutical Technology Co., LTD., Hangzhou, China
| | | | - Sheng-mei Zhu
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Wei-dong Ye
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Feng Zhang
- Department of Respiratory and Critical Care, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| |
Collapse
|
7
|
Astroth C, Shah KS, Agrawal S, Agrawal A. Weathering the Storm: How Age and Biologics Influence the COVID-19 Cytokine Surge. Pathogens 2025; 14:346. [PMID: 40333142 PMCID: PMC12030216 DOI: 10.3390/pathogens14040346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 05/09/2025] Open
Abstract
SARS-CoV-2, first identified in December 2019, caused a global pandemic, resulting in over 6.8 million deaths by March 2023. The elderly, or individuals over 65, accounted for the majority of COVID-19 deaths, with 81% of fatalities in the US in 2020 occurring in this group. Beyond mortality, aging populations are also at higher risk of long-term cardiovascular complications and acute respiratory distress syndrome (ARDS). Although these outcomes may be influenced by comorbidities common in the elderly, age has been found to be a standalone risk factor for severe COVID-19 infection. Therefore, investigating age-related factors in COVID-19 outcomes is crucial in protecting this vulnerable group. Of particular interest is the cytokine storm phenomenon, an excessive inflammatory response that contributes to severe COVID-19 symptoms, including ARDS and cardiovascular damage. Elevated levels of multiple cytokines are common in severe cases of COVID-19. We propose that changes that occur to cytokine profiles as we age may contribute to these aberrant inflammatory responses. This review specifically explored the interleukin class cytokines IL-1, IL-6, IL-17, and IL-23 and considered the potential of biologics targeting these cytokines to alleviate severe outcomes in both COVID-19 and aging individuals.
Collapse
Affiliation(s)
| | | | | | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA 92697, USA; (C.A.); (K.S.S.); (S.A.)
| |
Collapse
|
8
|
Wang YX, Zhao WX, Wang ZM, Zhao N, Li ZL, Wu ZY, Diao YP, Li YJ. Frailty impacts all-cause mortality after endovascular abdominal aortic aneurysm repair: a retrospective cohort study. J Nutr Health Aging 2025; 29:100489. [PMID: 39827699 DOI: 10.1016/j.jnha.2025.100489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/29/2024] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE This study aimed to evaluate the impact of frailty and inflammation on all-cause mortality in patients with abdominal aortic aneurysm (AAA) who underwent endovascular aneurysm repair (EVAR), and key risk factors were also explored. METHODS A retrospective analysis was conducted on 174 patients with AAA who underwent EVAR at Beijing Hospital between 2016 and 2024. Frailty was assessed using the modified five-item Frailty Index (mFI-5). Inflammation was quantified by the red cell distribution width-to-albumin ratio (RAR), a novel inflammatory marker. We examined the associations between frailty, preoperative risk factors, and mortality using Kaplan-Meier survival analysis and Cox proportional hazards models. Mediation analysis was performed to evaluate the role of RAR in the relationship between frailty and mortality. RESULTS Frailty was found to be an independent risk factor for all-cause mortality following EVAR (HR = 1.95, P = 0.048). Preoperative anemia (HR = 0.98, P = 0.032), elevated creatinine levels (HR = 1.01, P = 0.013), and prolonged operation time (HR = 1.01, P = 0.029) were also independent predictors of mortality. Kaplan-Meier survival analysis revealed significantly lower survival rates for frailty patients (P = 0.004). Additionally, RAR mediated 23.8% of the relationship between frailty and mortality (P = 0.012), underscoring its role as a key indicator of chronic inflammation. CONCLUSIONS Frailty and chronic inflammation, as measured by the innovative RAR marker, are significant contributors to mortality after EVAR. This study highlights the clinical utility of RAR in identifying high-risk AAA patients and its potential for guiding targeted preoperative interventions. Incorporating frailty assessments and inflammation monitoring into routine preoperative evaluations may improve patient outcomes by enabling personalized approaches such as nutritional optimization and inflammation control.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China; Peking University Fifth School of Clinical Medicine
| | - Wen-Xin Zhao
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zi-Mo Wang
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China; Peking University Fifth School of Clinical Medicine
| | - Ning Zhao
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhao-Long Li
- Institute of Molecular Vascular Medicine, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research DZHK, Partner Site Munich Heart Alliance, Berlin, Germany
| | - Zhi-Yuan Wu
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong-Peng Diao
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yong-Jun Li
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
9
|
Shahbaz SK, Mokhlesi A, Sadegh RK, Rahimi K, Jamialahmadi T, Butler AE, Kesharwani P, Sahebkar A. TLR/NLRP3 inflammasome signaling pathways as a main target in frailty, cachexia and sarcopenia. Tissue Cell 2025; 93:102723. [PMID: 39823704 DOI: 10.1016/j.tice.2025.102723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/28/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025]
Abstract
Mobility disability is a common condition affecting older adults, making walking and the performance of activities of daily living difficult. Frailty, cachexia and sarcopenia are related conditions that occur with advancing age and are characterized by a decline in muscle mass, strength, and functionality that negatively impacts health. Chronic low-grade inflammation is a significant factor in the onset and progression of these conditions. The toll-like receptors (TLRs) and the NLRP3 inflammasome are the pathways of signaling that regulate inflammation. These pathways can potentially be targeted therapeutically for frailty, cachexia and sarcopenia as research has shown that dysregulation of the TLR/NLRP3 inflammasome signaling pathways is linked to these conditions. Activation of TLRs with pathogen-associated molecular patterns (PAMPs or DAMPs) results in chronic inflammation and tissue damage by releasing pro-inflammatory cytokines. Additionally, NLRP3 inflammasome activation enhances the inflammatory response by promoting the production and release of interleukins (ILs), thus exacerbating the underlying inflammatory mechanisms. These pathways are activated in the advancement of disease in frail and sarcopenic individuals. Targeting these pathways may offer therapeutic options to reduce frailty, improve musculoskeletal resilience and prevent or reverse cachexia-associated muscle wasting. Modulating TLR/NLRP3 inflammasome pathways may also hold promise in slowing down the progression of sarcopenia, preserving muscle mass and enhancing overall functional ability in elderly people. The aim of this review is to investigate the signaling pathways of the TLR/NLRP3 inflammasome as a main target in frailty, cachexia and sarcopenia.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Aida Mokhlesi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; Social Determinants of Health Research Center, Research Institute for Prevention of Non-communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran; Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kimia Rahimi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Amirhossein Sahebkar
- Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Aitken RJ. Spermatozoa as harbingers of mortality: the curious link between semen quality and life expectancy. Hum Reprod 2025; 40:580-584. [PMID: 40037897 DOI: 10.1093/humrep/deaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Indexed: 03/06/2025] Open
Affiliation(s)
- Robert John Aitken
- Centre for Reproductive Science, Discipline of Biological Sciences, School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
11
|
Bracken OV, De Maeyer RPH, Akbar AN. Enhancing immunity during ageing by targeting interactions within the tissue environment. Nat Rev Drug Discov 2025; 24:300-315. [PMID: 39875569 DOI: 10.1038/s41573-024-01126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/30/2025]
Abstract
Immunity declines with age. This results in a higher risk of age-related diseases, diminished ability to respond to new infections and reduced response to vaccines. The causes of this immune dysfunction are cellular senescence, which occurs in both lymphoid and non-lymphoid tissue, and chronic, low-grade inflammation known as 'inflammageing'. In this Review article, we highlight how the processes of inflammation and senescence drive each other, leading to loss of immune function. To break this cycle, therapies are needed that target the interactions between the altered tissue environment and the immune system instead of targeting each component alone. We discuss the relative merits and drawbacks of therapies that are directed at eliminating senescent cells (senolytics) and those that inhibit inflammation (senomorphics) in the context of tissue niches. Furthermore, we discuss therapeutic strategies designed to directly boost immune cell function and improve immune surveillance in tissues.
Collapse
Affiliation(s)
| | - Roel P H De Maeyer
- Division of Medicine, University College London, London, UK
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Arne N Akbar
- Division of Medicine, University College London, London, UK.
| |
Collapse
|
12
|
Khan A, Alzahrani HA, Felemban SG, Algarni AS, Alenezi ABS, Kamal M, Rehman ZU, Asdaq SMB, Ahmed N, Alharbi BM, Alanazi BS, Imran M. Exploring TGF-β signaling in benign prostatic hyperplasia: from cellular senescence to fibrosis and therapeutic implications. Biogerontology 2025; 26:79. [PMID: 40159577 DOI: 10.1007/s10522-025-10226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
As men get older, they often develop benign prostatic hyperplasia (BPH), an enlarged prostate that is not cancerous or dangerous. Although the etiology of BPH is unknown, increasing evidence indicates that the TGF-β signaling pathway might be a key player in its pathogenesis. TGF-β is a pleiotropic cytokine involved in proliferation, differentiation, and extracellular matrix re-modeling, which are all dysregulated in BPH. Cellular senescence is primarily initiated by TGF-β--induced, irreversible growth arrest and usually limits the prostate gland's hyperplastic growth. Moreover, senescent cells generate a Senescence-Associated Secretory Phenotype (SASP), which consists of numerous proinflammatory and profibrotic factors that can worsen disease ontogeny. In addition, TGF-β is among the most fibrogenic factors. At the same time, fibrosis involves a massive accumulation of extracellular matrix proteins, which can increase tissue stiffness and a loss of normal organ functions. TGF-β-mediated fibrosis in BPH changes the mechanical properties of the prostate and surrounding tissues to contribute to lower urinary tract symptoms. This review discusses the complicated molecular signaling of TGF-β underlying changes in cellular senescence and fibrosis during BPH concerning its therapeutic potential.
Collapse
Affiliation(s)
- Abida Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, 73213, Saudi Arabia
| | - Hayat Ali Alzahrani
- Medical Laboratory Technology Department, College of Medical Applied Science, Northern Border University, Arar, Saudi Arabia
| | - Shatha Ghazi Felemban
- Medical Laboratory Sciences Department, Fakeeh College for Medical Sciences, 21461, Jeddah, Saudi Arabia
| | - Alanood Saeed Algarni
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Zia Ur Rehman
- Health Research Centre, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jazan University, P.O. Box 114, Jazan, 45142, Kingdom of Saudi Arabia
| | | | - Naveed Ahmed
- Department of Assistance Medical Sciences, Applied College, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Bashayer Mohammed Alharbi
- Department of Pharmacy, Johns Hopkins Aramco Healthcare, P.O. Box 10352, 31311, Dhahran, Eastern Province, Saudi Arabia
| | - Bander Sharqi Alanazi
- Department of Nursing Administration, Northern Area Armed Forces Hospital, 31991, Hafer AlBaten, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, 73213, Saudi Arabia.
| |
Collapse
|
13
|
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
|
14
|
Miao L, Gong C, Liao J, Xie C, Shen X, Cheng Y. Dynamic Trends of Albumin-to-C-Reactive Protein Ratio: A Prognostic Indicator in Elderly Patients with Community-Acquired Pneumonia. J Inflamm Res 2025; 18:4195-4211. [PMID: 40125077 PMCID: PMC11930240 DOI: 10.2147/jir.s512632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Background The prognostic significance of dynamic changes in the albumin-to-C-reactive protein ratio (ACR) in elderly patients with community-acquired pneumonia (CAP) has not been fully elucidated. This study aims to evaluate the utility of ACR as a dynamic biomarker for predicting 28-day mortality and enhancing risk stratification in this high-risk population. Methods A retrospective cohort study was conducted on 437 elderly CAP patients (≥65 years). Serum albumin and C-reactive protein (CRP) levels were measured at admission (T0), 24 hours (T1), and 3 days (T2) post-admission. ACR was calculated for each time point, and its prognostic value was assessed using advanced statistical methods. Results The 28-day mortality rate was 16.7%. ACR levels were consistently lower in non-survivors across all time points (P < 0.001). RCS analysis revealed a nonlinear relationship between ACR and mortality risk. Time-varying ROC analysis demonstrated that ACR consistently outperformed CRP in predicting mortality, with superior area under the curve (AUC) values at all time points. Random-effects modeling indicated minimal inter-individual variability in ACR (random effects variance: 0.030; standard deviation: 0.175). Time-varying Cox regression confirmed a strong negative association between dynamic ACR changes and mortality risk, with a C-statistic of 0.833 (P < 0.001). Conclusion Dynamic monitoring of ACR is a robust and clinically applicable tool for predicting short-term mortality in elderly CAP patients. By integrating markers of inflammation and nutritional status, ACR facilitates early identification of high-risk patients and supports personalized treatment strategies. These findings highlight the potential of ACR as a novel biomarker for improving clinical outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Lei Miao
- Department of Critical Care Medicine, The second People’s Hospital of Lianyungang, Lianyungang, 222000, People’s Republic of China
| | - Chen Gong
- Department of Geriatrics, The second People’s Hospital of Lianyungang, Lianyungang, 222000, People’s Republic of China
| | - Jingxian Liao
- Department of Geriatrics, The second People’s Hospital of Lianyungang, Lianyungang, 222000, People’s Republic of China
| | - Chunhui Xie
- Department of Geriatrics, The second People’s Hospital of Lianyungang, Lianyungang, 222000, People’s Republic of China
| | - Xiaozhu Shen
- Department of Geriatrics, The second People’s Hospital of Lianyungang, Lianyungang, 222000, People’s Republic of China
| | - Yajuan Cheng
- Department of Critical Care Medicine, The second People’s Hospital of Lianyungang, Lianyungang, 222000, People’s Republic of China
| |
Collapse
|
15
|
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
|
16
|
Salvo A, Tuttolomondo A. The Role of Olive Oil in Cardiometabolic Risk. Metabolites 2025; 15:190. [PMID: 40137153 PMCID: PMC11943877 DOI: 10.3390/metabo15030190] [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: 01/31/2025] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
Olive oil, the primary fat source in the Mediterranean diet (MedDiet), is rich in monounsaturated fatty acids (MUFA), especially oleic acid, which constitutes 70-80% of its composition. Extra-virgin olive oil (EVOO), produced by mechanically pressing olives, is the highest quality olive oil, with an intense flavor and acidity <1%. In contrast, refined olive oil (ROO), a blend of virgin and refined oils, contains fewer antioxidants and anti-inflammatory compounds. EVOO's health benefits stem largely from its MUFA content, which is linked to reduced risks of cardiovascular disease (CVD), neurodegenerative conditions, and certain cancers. Additionally, EVOO contains minor, but bioactive, components such as polyphenols, tocopherols, and phytosterols, contributing to its oxidative stability, sensory qualities, and health-promoting properties. These include polyphenols, like oleuropein, hydroxytyrosol, and tyrosol, which exhibit anti-inflammatory, cardioprotective, neuroprotective, and anticancer effects. Epidemiological studies suggest an inverse relationship between olive oil intake and CVD, with EVOO-enriched MedDiet interventions showing improved lipid profiles, reduced blood pressure, and lower cardiovascular event risk. The PREDIMED study highlights the significant role of EVOO in reducing cardiometabolic risk. This review explores the impact of EVOO's chemical components within the MedDiet framework on metabolic variables influencing cardiometabolic health.
Collapse
Affiliation(s)
- Andrea Salvo
- Internal Medicine and Stroke Care Ward, Policlinico, P. Giaccone, Piazza delle Cliniche n.2, 90127 Palermo, Italy;
- ProMISE Department, University of Palermo, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, Policlinico, P. Giaccone, Piazza delle Cliniche n.2, 90127 Palermo, Italy;
- ProMISE Department, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
17
|
Poetzsch G, Jelacic L, Dammer L, Hellmann SL, Balling M, Andrade-Navarro M, Avivi A, Shams I, Bicker A, Hankeln T. Adaptation of the Spalax galili transcriptome to hypoxia may underlie the complex phenotype featuring longevity and cancer resistance. NPJ AGING 2025; 11:16. [PMID: 40044716 PMCID: PMC11882797 DOI: 10.1038/s41514-025-00206-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
In the subterranean rodent (Nanno)spalax galili, evolutionary adaptation to hypoxia is correlated with longevity and tumor resistance. Adapted gene-regulatory networks of Spalax might pinpoint strategies to maintain health in humans. Comparing liver, kidney and spleen transcriptome data from Spalax and rat at hypoxia and normoxia, we identified differentially expressed gene pathways common to multiple organs in both species. Body-wide interspecies differences affected processes like cell death, antioxidant defense, DNA repair, energy metabolism, immune response and angiogenesis, which may play a crucial role in Spalax's adaptation to environmental hypoxia. In all organs, transcription of genes for genome stability maintenance and DNA repair was elevated in Spalax versus rat, accompanied by lower expression of aerobic energy metabolism and proinflammatory genes. These transcriptomic changes might account for the extraordinary lifespan of Spalax and its cancer resistance. The identified gene networks present candidates for further investigating the molecular basis underlying the complex Spalax phenotype.
Collapse
Affiliation(s)
- Gesa Poetzsch
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Luca Jelacic
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Leon Dammer
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Sören Lukas Hellmann
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
- Nucleic Acids Core Facility, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Michelle Balling
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
| | - Miguel Andrade-Navarro
- Computational Biology and Data Mining Group, Institute of Organismic and Molecular Evolution, Johannes Gutenberg University, Mainz, Germany
| | - Aaron Avivi
- Institute of Evolution, University of Haifa, Mount Carmel, Haifa, Israel
| | - Imad Shams
- Institute of Evolution, University of Haifa, Mount Carmel, Haifa, Israel
- Department of Evolutionary and Environmental Biology, Faculty of Natural Sciences, University of Haifa, Mount Carmel, Haifa, Israel
| | - Anne Bicker
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany
- Department of Medicine I, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Thomas Hankeln
- Molecular Genetics & Genome Analysis, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg-University, Mainz, Germany.
| |
Collapse
|
18
|
De Meo E, Portaccio E, Cortese R, Ruano L, Goretti B, Niccolai C, Patti F, Chisari C, Gallo P, Grossi P, Ghezzi A, Roscio M, Mattioli F, Stampatori C, Simone M, Viterbo RG, Bonacchi R, Rocca AM, Leveraro E, Giorgio A, De Stefano N, Filippi M, Inglese M, Amato MP. Pediatric, adult, and late onset multiple sclerosis: Cognitive phenotypes and gray matter atrophy. Ann Clin Transl Neurol 2025; 12:512-522. [PMID: 39861952 PMCID: PMC11920747 DOI: 10.1002/acn3.52291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVES We aim to investigate cognitive phenotype distribution and MRI correlates across pediatric-, elderly-, and adult-onset MS patients as a function of disease duration. METHODS In this cross-sectional study, we enrolled 1262 MS patients and 238 healthy controls, with neurological and cognitive assessments. A subset of 222 MS patients and 92 controls underwent 3T-MRI scan for brain atrophy and lesion analysis. Multinomial probabilistic models identified likelihood of belonging to cognitive phenotypes ("preserved-cognition," "mild verbal memory/semantic fluency," "mild multi-domain," "severe attention/executive," and "severe multi-domain") and experiencing MRI abnormalities based on disease duration and age at onset. RESULTS In all groups, the likelihood of "preserved-cognition" phenotype decreased, whereas "mild multi-domain" increased with longer disease duration. In pediatric- and adult-onset patients, the likelihood of "mild verbal memory/semantic fluency" phenotypes decreased with longer disease duration, and that of "severe multi-domain" increased with longer disease duration. Only in adult-onset patients, the likelihood of "severe executive/attention" phenotype increased with longer disease duration. All groups displayed escalating probabilities of cortical, thalamic, hippocampal, and deep gray matter atrophy over disease course. Compared to adult, pediatric-onset patients showed lower probability of experiencing thalamic atrophy with longer disease duration, while elderly-onset showed higher probability of experiencing cortical and hippocampal atrophy. INTERPRETATION Age at MS onset significantly influences the distribution of cognitive phenotypes and the patterns of regional gray matter atrophy throughout the disease course.
Collapse
Affiliation(s)
- Ermelinda De Meo
- Department of Neuroinflammation, Institute of NeurologyUniveristy College of LondonLondonUK
- NEUROFARBA Department, Neurosciences SectionUniversity of FlorenceFlorenceItaly
| | - Emilio Portaccio
- NEUROFARBA Department, Neurosciences SectionUniversity of FlorenceFlorenceItaly
- Azienda Ospedaliera Universitaria CareggiFlorenceItaly
| | - Rosa Cortese
- Department of Medicine, Surgery and NeuroscienceUniversity of SienaSienaItaly
| | - Luis Ruano
- EPIUnit, Instituto de Saúde Pública de Universidade do PortoPortoPortugal
- Neurology DepartmentCentro Hospitalar de Entre Douro e VougaSanta Maria da FeiraPortugal
| | - Benedetta Goretti
- Department of Neuroinflammation, Institute of NeurologyUniveristy College of LondonLondonUK
| | - Claudia Niccolai
- Department of Neuroinflammation, Institute of NeurologyUniveristy College of LondonLondonUK
- IRCCS Fondazione Don Carlo GnocchiFlorenceItaly
| | | | | | | | - Paola Grossi
- Department, ASST Crema, Neuroimmunology Center, CardiocerebrovascularCremaItaly
| | | | | | - Flavia Mattioli
- ASST Spedali Civili Brescia Neuropsychology UnitBresciaItaly
| | | | - Marta Simone
- Department of Basic Medical Sciences, Child and Adolescence Neuropsychiatry UnitNeuroscience and Sense Organs University “Aldo Moro” BariBariItaly
| | - Rosa Gemma Viterbo
- Department of Basic Medical Sciences, Child and Adolescence Neuropsychiatry UnitNeuroscience and Sense Organs University “Aldo Moro” BariBariItaly
| | | | | | - Elisa Leveraro
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child HealthUniversity of GenoaGenoaItaly
| | - Antonio Giorgio
- Department of Medicine, Surgery and NeuroscienceUniversity of SienaSienaItaly
| | - Nicola De Stefano
- Department of Medicine, Surgery and NeuroscienceUniversity of SienaSienaItaly
| | - Massimo Filippi
- Neuroimaging Research UnitIRCCS San Raffaele Scientific InstituteMilan
- Neurology UnitIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Matilde Inglese
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child HealthUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Maria Pia Amato
- NEUROFARBA Department, Neurosciences SectionUniversity of FlorenceFlorenceItaly
- Azienda Ospedaliera Universitaria CareggiFlorenceItaly
- IRCCS Fondazione Don Carlo GnocchiFlorenceItaly
| |
Collapse
|
19
|
Wang TW, Nakanishi M. Immune surveillance of senescence: potential application to age-related diseases. Trends Cell Biol 2025; 35:248-257. [PMID: 39025762 DOI: 10.1016/j.tcb.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024]
Abstract
Several lines of evidence suggest that the age-dependent accumulation of senescent cells leads to chronic tissue microinflammation, which in turn contributes to age-related pathologies. In general, senescent cells can be eliminated by the host's innate and adaptive immune surveillance system, including macrophages, NK cells, and T cells. Impaired immune surveillance leads to the accumulation of senescent cells and accelerates the aging process. Recently, senescent cells, like cancer cells, have been shown to express certain types of immune checkpoint proteins as well as non-classical immune-tolerant MHC variants, leading to immune escape from surveillance systems. Thus, immune checkpoint blockade (ICB) may be a promising strategy to enhance immune surveillance of senescence, leading to the amelioration of some age-related diseases and tissue dysfunction.
Collapse
Affiliation(s)
- Teh-Wei Wang
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
20
|
Huang S, Lu Y, Fang W, Huang Y, Li Q, Xu Z. Neurodegenerative diseases and neuroinflammation-induced apoptosis. Open Life Sci 2025; 20:20221051. [PMID: 40026360 PMCID: PMC11868719 DOI: 10.1515/biol-2022-1051] [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: 06/18/2024] [Revised: 11/30/2024] [Accepted: 12/27/2024] [Indexed: 03/05/2025] Open
Abstract
Neuroinflammation represents a critical pathway in the brain for the clearance of foreign bodies and the maintenance of homeostasis. When the neuroinflammatory process is dysregulate, such as the over-activation of microglia, which results in the excessive accumulation of free oxygen and inflammatory factors in the brain, among other factors, it can lead to an imbalance in homeostasis and the development of various diseases. Recent research has indicated that the development of numerous neurodegenerative diseases is closely associated with neuroinflammation. The pathogenesis of neuroinflammation in the brain is intricate, involving alterations in numerous genes and proteins, as well as the activation and inhibition of signaling pathways. Furthermore, excessive inflammation can result in neuronal cell apoptosis, which can further exacerbate the extent of the disease. This article presents a summary of recent studies on the relationship between neuronal apoptosis caused by excessive neuroinflammation and neurodegenerative diseases. The aim is to identify the link between the two and to provide new ideas and targets for exploring the pathogenesis, as well as the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shi Huang
- School of Clinical Medicine, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Yaxin Lu
- School of Pharmaceutical Sciences, Wannan Medical College,
241002, Wuhu, Anhui, China
| | - Wanzhen Fang
- School of Stomatology, Wannan Medical College,
241002, Wuhu, Anhui, China
| | - Yanjiao Huang
- Human Anatomy Experimental Training Center, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Qiang Li
- Human Anatomy Experimental Training Center, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Zhiliang Xu
- Department of Human Anatomy, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Basic Research and Translation of Aging-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, China
| |
Collapse
|
21
|
Kuznetsov NV, Statsenko Y, Ljubisavljevic M. An Update on Neuroaging on Earth and in Spaceflight. Int J Mol Sci 2025; 26:1738. [PMID: 40004201 PMCID: PMC11855577 DOI: 10.3390/ijms26041738] [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: 01/04/2025] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Over 400 articles on the pathophysiology of brain aging, neuroaging, and neurodegeneration were reviewed, with a focus on epigenetic mechanisms and numerous non-coding RNAs. In particular, this review the accent is on microRNAs, the discovery of whose pivotal role in gene regulation was recognized by the 2024 Nobel Prize in Physiology or Medicine. Aging is not a gradual process that can be easily modeled and described. Instead, multiple temporal processes occur during aging, and they can lead to mosaic changes that are not uniform in pace. The rate of change depends on a combination of external and internal factors and can be boosted in accelerated aging. The rate can decrease in decelerated aging due to individual structural and functional reserves created by cognitive, physical training, or pharmacological interventions. Neuroaging can be caused by genetic changes, epigenetic modifications, oxidative stress, inflammation, lifestyle, and environmental factors, which are especially noticeable in space environments where adaptive changes can trigger aging-like processes. Numerous candidate molecular biomarkers specific to neuroaging need to be validated to develop diagnostics and countermeasures.
Collapse
Affiliation(s)
- Nik V. Kuznetsov
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
| | - Yauhen Statsenko
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Milos Ljubisavljevic
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Yu P, Satyaraj E. Effect of Bovine Colostrum on Canine Immune Health. Animals (Basel) 2025; 15:185. [PMID: 39858185 PMCID: PMC11759141 DOI: 10.3390/ani15020185] [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: 11/15/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Colostrum, the first fluid secreted by the mammary glands of mammalian mothers, contains essential nutrients for the health and survival of newborns. Bovine colostrum (BC) is notable for its high concentrations of bioactive components, such as immunoglobulins and lactoferrin. Despite dogs being the world's most popular companion animals, there is limited research on their immune systems compared to humans. This summary aims to consolidate published studies that explore the immune benefits of BC, focusing specifically on its implications for dogs.
Collapse
Affiliation(s)
- Ping Yu
- Nestlé Purina Research, One Checkerboard Square, St. Louis, MO 63164, USA
| | - Ebenezer Satyaraj
- Nestlé Purina Research, One Checkerboard Square, St. Louis, MO 63164, USA
| |
Collapse
|
25
|
Li H, Wei D, Cao H, Han Y, Li L, Liu Y, Qi J, Wu X, Zhang Z. Bioinformatics-Based Exploration of the Ability of Ginkgetin to Alleviate the Senescence of Cardiomyocytes After Myocardial Infarction and Its Cardioprotective Effects. J Inflamm Res 2025; 18:301-323. [PMID: 39802510 PMCID: PMC11724673 DOI: 10.2147/jir.s491535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose Myocardial infarction (MI) is a prevalent cardiovascular disorder affecting individuals worldwide. There is a need to identify more effective therapeutic agents to minimize cardiomyocyte damage and enhance cardioprotection. Ginkgo biloba extract is extensively used to treat neurological disorders and peripheral vascular diseases. The aim of this study was to determine the protective effects and mechanisms of ginkgetin on postinfarction cardiomyocytes through bioinformatics and experimental validation. Methods Bioinformatics analysis was performed to predict the underlying biological mechanisms of ginkgetin in the treatment of MI. Next, we performed further validation through experiments. For in vivo studies, we used coronary ligation to construct an MI rat model. In vitro, oxygen and glucose deprivation (OGD) was performed to simulate ischemia in H9c2 cardiomyocytes. Results Bioinformatics analysis revealed that the key targets of ginkgetin for MI treatment were MMP2, MMP9, and VEGFA. Immune infiltration analysis revealed that ginkgetin might be involved in immune regulation by acting on the TCR signaling pathway. The results of the GO enrichment analysis revealed that ginkgetin might protect the heart by acting on the cell membrane to alleviate the senescent apoptosis of cardiomyocytes after MI. In vivo studies revealed that ginkgetin ameliorated myocardial pathological damage and cardiac decompensation after MI. It also alleviated the inflammatory infiltration and senescent apoptosis of cardiomyocytes after MI. Additionally, ginkgetin can downregulate the activation signals of the TCR signaling pathway by dephosphorylating CD3 and CD28. In vitro studies revealed that ginkgetin attenuated elevated OGD-induced cytotoxicity, increased cell viability, and alleviated OGD-induced senescent apoptosis, thus protecting cardiomyocytes. Conclusion Ginkgetin inhibits postinfarction myocardial fibrosis and cardiomyocyte hypertrophy, scavenges oxygen free radicals, decreases postinfarction limbic cell inflammatory infiltration, suppresses activation of the inflammatory-immune pathway, and delays postinfarction peripheral cells from undergoing senescent apoptosis, thus protecting the heart.
Collapse
Affiliation(s)
- Han Li
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Dongsheng Wei
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Huimin Cao
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Yelei Han
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Luzhen Li
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Yuting Liu
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Jiajie Qi
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Xinyue Wu
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Zhe Zhang
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, People’s Republic of China
| |
Collapse
|
26
|
Mapuskar KA, London B, Zacharias ZR, Houtman JC, Allen BG. Immunometabolism in the Aging Heart. J Am Heart Assoc 2025; 14:e039216. [PMID: 39719411 PMCID: PMC12054428 DOI: 10.1161/jaha.124.039216] [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: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024]
Abstract
Structural, functional, and molecular-level changes in the aging heart are influenced by a dynamic interplay between immune signaling and cellular metabolism that is referred to as immunometabolism. This review explores the crosstalk between cellular metabolic pathways including glycolysis, oxidative phosphorylation, fatty acid metabolism, and the immune processes that govern cardiac aging. With a rapidly aging population that coincides with increased cardiovascular risk and cancer incidence rates, understanding the immunometabolic underpinnings of cardiac aging provides a foundation for identifying therapeutic targets to mitigate cardiac dysfunction. Aging alters the immune environment of the heart by concomitantly driving the changes in immune cell metabolism, mitochondrial dysfunction, and redox signaling. Shifts in these metabolic pathways exacerbate inflammation and impair tissue repair, creating a vicious cycle that accelerates cardiac functional decline. Treatment with cancer therapy further complicates this landscape, as aging-associated immunometabolic disruptions augment the susceptibility to cardiotoxicity. The current review highlights therapeutic strategies that target the immunometabolic axis to alleviate cardiac aging pathologies. Interventions include modulating metabolic intermediates, improving mitochondrial function, and leveraging immune signaling pathways to restore cardiac health. Advances in immunometabolism thus hold significant potential for translating preclinical findings into therapies that improve the quality of life for the aging population and underscore the need for approaches that address the immunometabolic mechanisms of cardiac aging, providing a framework for future research.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Barry London
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Internal MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Zeb R. Zacharias
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Jon C.D. Houtman
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Microbiology and ImmunologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Bryan G. Allen
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| |
Collapse
|
27
|
Zeng Y, Buonfiglio F, Li J, Pfeiffer N, Gericke A. Mechanisms Underlying Vascular Inflammaging: Current Insights and Potential Treatment Approaches. Aging Dis 2025:AD.2024.0922. [PMID: 39812546 DOI: 10.14336/ad.2024.0922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025] Open
Abstract
Inflammaging refers to chronic, low-grade inflammation that becomes more common with age and plays a central role in the pathophysiology of various vascular diseases. Key inflammatory mediators involved in inflammaging contribute to endothelial dysfunction and accelerate the progression of atherosclerosis. In addition, specific pathological mechanisms and the role of inflammasomes have emerged as critical drivers of immune responses within the vasculature. A comprehensive understanding of these processes may lead to innovative treatment strategies that could significantly improve the management of age-related vascular diseases. Emerging therapeutic approaches, including cytokine inhibitors, senolytics, and specialized pro-resolving mediators, aim to counteract inflammaging and restore vascular health. This review seeks to provide an in-depth exploration of the molecular pathways underlying vascular inflammaging and highlight potential therapeutic interventions.
Collapse
|
28
|
Yan Y, Peng MY, Yang Y, Zhang ZB, Zhang LL, Tang L, Qin XJ, Cheng YY, Di YT, Hao XJ. Highly oxygenated ent-abietane diterpenoid lactones from Euphorbia peplus and their anti-inflammatory activity. Bioorg Chem 2025; 154:107989. [PMID: 39591686 DOI: 10.1016/j.bioorg.2024.107989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024]
Abstract
Eleven new ent-abietane diterpenoid lactones, designated euphjatrophanes H-R (1-11), were isolated from the whole plants of Euphorbia peplus, along with nine previously identified congeners (12-20). Their structures, including absolute configurations, were elucidated through a combination of NMR, HRESIMS, single-crystal X-ray diffraction, and calculations of ECD and DP4 + technologies. Notably, the absolute configurations of six compounds 1, 2, 4, 5, 6, and 7 were unambiguously determined by single-crystal X-ray diffraction analyses, conducted with Cu Kα radiation. The anti-inflammatory potential of all ent-abietane diterpenoid lactones was evaluated on macrophages. Compounds 6-9, 12-16 and 19 significantly suppressed nitric oxide production, while 10 μM of compounds 6, 9, 11 and 16 remarkably suppressed the mRNA expression of IL-6, IL-1β, and TNFα in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages. Notably, compound 6 demonstrated a dose-dependent inhibition of the levels of inflammatory mediators (IL-6, IL-1β, and TNFα). Furthermore, compound 6 effectively suppressed FOXO1 expression and reduced the phosphorylation level of NF-κB p65. These findings suggest that compound 6 might be a promising candidate for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Ying Yan
- State Key Laboratory of Functions and Applications of Medicinal Plants & College of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550014, China
| | - Ming-You Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants & College of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550014, China
| | - Ying Yang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Zhi-Bi Zhang
- Academy of Biomedical Engineering, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Long-Long Zhang
- Academy of Biomedical Engineering, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Lei Tang
- State Key Laboratory of Functions and Applications of Medicinal Plants & College of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550014, China
| | - Xu-Jie Qin
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, China.
| | - Yuan-Yuan Cheng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Ying-Tong Di
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, China.
| | - Xiao-Jiang Hao
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, China
| |
Collapse
|
29
|
Pojero F, Gervasi F. Polyphenol Treatment of Peripheral Blood Mononuclear Cells from Individuals of Different Ages. Methods Mol Biol 2025; 2857:191-221. [PMID: 39348067 DOI: 10.1007/978-1-0716-4128-6_18] [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: 10/01/2024]
Abstract
Human peripheral blood mononuclear cells (PBMCs) have been largely utilized to assess the cytotoxic, immunomodulatory, and anti-inflammatory properties of both synthetic and natural compounds. Within the latter category, polyphenols from dietary sources have been extensively analyzed. PBMCs represent a feasible in vitro model to study polyphenol hallmarks and activity according to quantitative and qualitative differences in immune responses in individuals of different age. In this chapter, we propose a method for PBMC treatment with polyphenols and analysis designed on age-dependent qualitative and quantitative variability in immune cell performance.
Collapse
Affiliation(s)
- Fanny Pojero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Francesco Gervasi
- Specialistic Oncology Laboratory Unit, A.R.N.A.S. Hospitals Civico, Di Cristina e Benfratelli, Palermo, Italy
| |
Collapse
|
30
|
Nakanishi M. Cellular senescence as a source of chronic microinflammation that promotes the aging process. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2025; 101:224-237. [PMID: 40222899 DOI: 10.2183/pjab.101.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Why and how do we age? This physiological phenomenon that we all experience remains a great mystery, largely unexplained even in this age of scientific and technological progress. Aging is a significant risk factor for numerous diseases, including cancer. However, underlying mechanisms responsible for this association remain to be elucidated. Recent findings have elucidated the significance of the accumulation of senescent cells and other inflammatory cells in organs and tissues with age, and their deleterious effects, such as the induction of inflammation in the microenvironment, as underlying factors contributing to organ dysfunction and disease development. Cellular senescence is a cellular phenomenon characterized by a permanent cessation of cell proliferation and secretion of several proinflammatory cytokines (senescence associated secretory phenotypes). Notably, the elimination of senescent cells from aging individuals has been demonstrated to alleviate age-related organ and tissue dysfunction, as well as various geriatric diseases. This review summarizes the molecular mechanisms by which senescent cells are induced and contribute to age-related diseases, as well as the technologies that ameliorate them.
Collapse
Affiliation(s)
- Makoto Nakanishi
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| |
Collapse
|
31
|
Balamurugan BS, Marimuthu MMC, Sundaram VA, Saravanan B, Chandrababu P, Chopra H, Malik T. Micro nutrients as immunomodulators in the ageing population: a focus on inflammation and autoimmunity. Immun Ageing 2024; 21:88. [PMID: 39731136 DOI: 10.1186/s12979-024-00492-7] [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: 09/19/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
Immunosenescence, the slow degradation of immune function over time that is a hallmark and driver of aging, makes older people much more likely to be killed by common infections (such as flu) than young adults, but it also contributes greatly to rates of chronic inflammation in later life. Such micro nutrients are crucial for modulating effective immune responses and their deficiencies have been associated with dysfunctional immunity in the elderly. In this review, we specifically focused on the contribution of major micro nutrients (Vitamins A, D and E, Vitamin C; Zinc and Selenium) as immunomodulators in ageing population especially related to inflame-ageing process including autoimmunity. This review will cover these hologenomic interactions, including how micro nutrients can modulate immune cell function and/or cytokine production to benefit their hosts with healthy mucous-associated immunity along with a sustainable immunologic homeostasis. For example, it points out the modulatory effects of vitamin D on both innate and adaptive immunity, with a specific focus on its ability to suppress pro-inflammatory cytokines synthesis while enhancing regulatory T-cell function. In the same context, also zinc is described as important nutrient for thymic function and T-cell differentiation but exhibits immunomodulatory functions by decreasing inflammation. In addition, the review will go over how micro nutrient deficiencies increase systemic chronic low-grade inflammation and, inflammaging as well as actually enhance autoimmune pathologies in old age. It assesses the potential role of additional targeted nutritional supplementation with micro nutrients to counteract these effects, promoting wider immune resilience in older adults. This review collates the current evidence and highlights the role of adequate micro nutrient intake on inflammation and autoimmunity during ageing, providing plausible origins for nutritional interventions to promote healthy immune aging.
Collapse
Affiliation(s)
- Bhavani Sowndharya Balamurugan
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | | | - Vickram Agaram Sundaram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India.
| | - Bharath Saravanan
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Prasanth Chandrababu
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Hitesh Chopra
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Tabarak Malik
- Department of Biomedical Sciences, Institute of Health, Jimma University, Jimma, 378, Oromia, Ethiopia.
- Division of Research & Development, Lovely Professional University, Phagwara, 144411, Punjab, India.
| |
Collapse
|
32
|
Chen R, Zou J, Chen J, Wang L, Kang R, Tang D. Immune aging and infectious diseases. Chin Med J (Engl) 2024; 137:3010-3049. [PMID: 39679477 PMCID: PMC11706578 DOI: 10.1097/cm9.0000000000003410] [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] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT The rise in global life expectancy has led to an increase in the older population, presenting significant challenges in managing infectious diseases. Aging affects the innate and adaptive immune systems, resulting in chronic low-grade inflammation (inflammaging) and immune function decline (immunosenescence). These changes would impair defense mechanisms, increase susceptibility to infections and reduce vaccine efficacy in older adults. Cellular senescence exacerbates these issues by releasing pro-inflammatory factors, further perpetuating chronic inflammation. Moreover, comorbidities, such as cardiovascular disease and diabetes, which are common in older adults, amplify immune dysfunction, while immunosuppressive medications further complicate responses to infections. This review explores the molecular and cellular mechanisms driving inflammaging and immunosenescence, focusing on genomic instability, telomere attrition, and mitochondrial dysfunction. Additionally, we discussed how aging-associated immune alterations influence responses to bacterial, viral, and parasitic infections and evaluated emerging antiaging strategies, aimed at mitigating these effects to improve health outcomes in the aging population.
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Ju Zou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Jiawang Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Ling Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
33
|
Thapa R, Ahmad Bhat A, Shahwan M, Ali H, PadmaPriya G, Bansal P, Rajotiya S, Barwal A, Siva Prasad GV, Pramanik A, Khan A, Hing Goh B, Dureja H, Kumar Singh S, Dua K, Gupta G. Proteostasis disruption and senescence in Alzheimer's disease pathways to neurodegeneration. Brain Res 2024; 1845:149202. [PMID: 39216694 DOI: 10.1016/j.brainres.2024.149202] [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: 05/23/2024] [Revised: 07/29/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Alzheimer's Disease (AD) is a progressive neurological disease associated with behavioral abnormalities, memory loss, and cognitive impairment that cause major causes of dementia in the elderly. The pathogenetic processes cause complex effects on brain function and AD progression. The proper protein homeostasis, or proteostasis, is critical for cell health. AD causes the buildup of misfolded proteins, particularly tau and amyloid-beta, to break down proteostasis, such aggregates are toxic to neurons and play a critical role in AD pathogenesis. The rise of cellular senescence is accompanied by aging, marked by irreversible cell cycle arrest and the release of pro-inflammatory proteins. Senescent cell build-up in the brains of AD patients exacerbates neuroinflammation and neuronal degeneration. These cells senescence-associated secretory phenotype (SASP) also disturbs the brain environment. When proteostasis failure and cellular senescence coalesce, a cycle is generated that compounds each other. While senescent cells contribute to proteostasis breakdown through inflammatory and degradative processes, misfolded proteins induce cellular stress and senescence. The principal aspects of the neurodegenerative processes in AD are the interaction of cellular senescence and proteostasis failure. This review explores the interconnected roles of proteostasis disruption and cellular senescence in the pathways leading to neurodegeneration in AD.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, UAE
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - G PadmaPriya
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | - Sumit Rajotiya
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Amit Barwal
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali - 140307, Punjab, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh-531162, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Abida Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia; Biofunctional Molecule Exploratory Research Group (BMEX), School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Gaurav Gupta
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, UAE; Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
34
|
Caretti M, Potenza DM, Ajalbert G, Albrecht U, Ming XF, Brenna A, Yang Z. Arginase-II gene deficiency reduces skeletal muscle aging in mice. Aging (Albany NY) 2024; 16:13563-13587. [PMID: 39670851 PMCID: PMC11723659 DOI: 10.18632/aging.206173] [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: 04/10/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024]
Abstract
Age-associated sarcopenia decreases mobility and is promoted by cell senescence, inflammation, and fibrosis. The mitochondrial enzyme arginase-II (Arg-II) plays a causal role in aging and age-associated diseases. Therefore, we aim to explore the role of Arg-II in age-associated decline of physical activity and skeletal muscle aging in a mouse model. Young (4-6 months) and old (20-24 months) wild-type (wt) mice and mice deficient in arg-ii (arg-ii-/-) of both sexes are investigated. We demonstrate a decreased physical performance of old wt mice, which is partially prevented in arg-ii-/- animals, particularly in males. The improved phenotype of arg-ii-/- mice in aging is associated with reduced sarcopenia, cellular senescence, inflammation, and fibrosis, whereas age-associated decline of microvascular endothelial cell density, satellite cell numbers, and muscle fiber types in skeletal muscle is prevented in arg-ii-/- mice. Finally, we demonstrate an increased arg-ii gene expression level in aging skeletal muscle and found Arg-II protein expression in endothelial cells and fibroblasts, but not in skeletal muscle fibers, macrophages, and satellite cells. Our results suggest that increased Arg-II in non-skeletal muscle cells promotes age-associated sarcopenia, particularly in male mice.
Collapse
Affiliation(s)
- Matteo Caretti
- Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - Duilio Michele Potenza
- Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - Guillaume Ajalbert
- Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - Urs Albrecht
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - Xiu-Fen Ming
- Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - Andrea Brenna
- Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - Zhihong Yang
- Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| |
Collapse
|
35
|
Al Qahtani M, Naghib MEDM, Alshamrani AM, Al Mazroua AM, Alayyaf AS, Ofisan SB, Kamal SM. The incidence, clinical features and outcome of urinary tract infections in geriatric patients: A prospective longitudinal study. IJID REGIONS 2024; 13:100469. [PMID: 39507391 PMCID: PMC11539122 DOI: 10.1016/j.ijregi.2024.100469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024]
Abstract
Objectives This study compares the incidence, clinical features, microbial profiles, and outcomes of urinary tract infections (UTIs) in patients aged 65 years and older versus those younger than 65 years. Methods A longitudinal cohort of 1,123 patients was divided into Group A (≥65 years, n = 560) and Group B (<65 years, n = 563) and followed for 2 years. The study analyzed UTI incidence, clinical features, microbial profiles, and outcomes, including recurrence and antibiotic resistance. Results Over 2 years, Group A had a significantly higher UTI incidence (38.0%) compared with Group B (12.8%) (P <0.0001). Complete recovery was less common in Group A (44.6%) than in Group B (70.83%), whereas recurrent UTIs and antibiotic resistance were more frequent in Group A (43.5% vs 22.2%, P <0.0001 and 11.7% vs 2.78%, P = 0.0017, respectively). Escherichia coli was the most prevalent pathogen in both groups, with Klebsiella and Pseudomonas species more common in recurrent UTIs, particularly in older patients. Risk factors for recurrent UTIs included advanced age, female sex, diabetes, immunosuppression, and renal stones. Conclusions These findings highlight the need for age-specific UTI prevention and management strategies that account for microbial resistance patterns and higher recurrence rates in older patients, addressing clinical and microbial challenges.
Collapse
Affiliation(s)
- Mohammed Al Qahtani
- Department of Internal Medicine, Prince Sattam Bin Abdulaziz University College of Medicine, Al-Kharj, Saudi Arabia
| | - Mohey El Deen Mohamed Naghib
- Department of Internal Medicine, Prince Sattam Bin Abdulaziz University College of Medicine, Al-Kharj, Saudi Arabia
| | - Abdulaziz M.M. Alshamrani
- Department of Internal Medicine, Prince Sattam Bin Abdulaziz University College of Medicine, Al-Kharj, Saudi Arabia
| | | | - Abdallah S.A. Alayyaf
- Department of Internal Medicine, Prince Sattam Bin Abdulaziz University College of Medicine, Al-Kharj, Saudi Arabia
| | - Salman Bin Ofisan
- Department of Internal Medicine, Prince Sattam Bin Abdulaziz University College of Medicine, Al-Kharj, Saudi Arabia
| | - Sanaa M. Kamal
- Department of Internal Medicine, Prince Sattam Bin Abdulaziz University College of Medicine, Al-Kharj, Saudi Arabia
| |
Collapse
|
36
|
Song G, Oh H, Jin H, Han H, Lee B. GABA Prevents Sarcopenia by Regulation of Muscle Protein Degradation and Inflammaging in 23- to 25-Month-Old Female Mice. J Cachexia Sarcopenia Muscle 2024; 15:2852-2864. [PMID: 39513373 PMCID: PMC11634462 DOI: 10.1002/jcsm.13646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Sarcopenia is the gradual decrease in skeletal muscle mass, strength and function in elderly individuals. Gamma-aminobutyric acid (GABA) is a neurotransmitter naturally produced from glutamate by the enzyme glutamic acid decarboxylase. Age-related decline in GABA is linked to age-related motor and sensory decline and seems to affect sarcopenia, yet no detailed study has been conducted. In this study, we aimed to investigate the effect of GABA on improving sarcopenia by suppressing muscle protein degradation through supplementing decreased GABA in old mice. METHODS GABA (10 or 30 mg/kg/day) was orally administered daily to young (3 months) and old (21-23 months) C57BL/6 mice for 7 weeks. The body weight and grip strength of the mice were measured weekly at the same time. After sacrificing the mice, the quadriceps and gastrocnemius muscles were excised from their hind limbs, and the spleen and serum were collected. Histological, biochemical and molecular analyses were conducted in various experiments. RESULTS The administration of GABA increased muscle strength (+41%, +70% compared to the aged mouse control group, GABA at doses of 10 or 30 mg/kg/day respectively, p < 0.05) and muscle mass (quadriceps: +28%, +46%; gastrocnemius: +12%, +19%, p < 0.05) in old mice. This increase was accompanied by a cross-sectional area (CSA) increase in the quadriceps and gastrocnemius muscle (p < 0.05). The administration of GABA increased IGF-1 levels in serum (p < 0.05), leading to the activation of muscle protein synthesis. We found that GABA inhibits sarcopenia by regulating muscle protein degradation through the activation of Akt/mTOR/FoxO3a signalling pathways. GABA also regulates inflammaging, which is a hallmark of age-related muscle atrophy. There was a significant increase in the F4/80 + CD11b + total macrophage ratio in gastrocnemius and spleen, especially the M1 macrophage ratio increased in old mice. However, GABA administration was effective in suppressing M1 macrophages (gastrocnemius: -40%, - 53%; spleen: -22%, -26%, p < 0.05). Pro-inflammatory cytokines such as TNF-α and IL-6, primarily secreted by M1 macrophages, are also decreased by treatment with GABA (TNF-α: -24%, -27%; IL-6: -45%, -59%, p < 0.05). CONCLUSIONS Together, this study demonstrates the importance of GABA in maintaining muscle and low-chronic inflammation during ageing. We suggest that GABA shows potential as a substance that can effectively address sarcopenia and enhance the overall lifespan and well-being of older individuals.
Collapse
Affiliation(s)
- Gunju Song
- Department of Food Science and Biotechnology, College of Life ScienceCHA UniversitySeongnamSouth Korea
| | - Hyun‐Ji Oh
- Department of Food Science and Biotechnology, College of Life ScienceCHA UniversitySeongnamSouth Korea
| | - Heegu Jin
- Department of Food Science and Biotechnology, College of Life ScienceCHA UniversitySeongnamSouth Korea
| | - Hyein Han
- Department of Food Science and Biotechnology, College of Life ScienceCHA UniversitySeongnamSouth Korea
| | - Boo‐Yong Lee
- Department of Food Science and Biotechnology, College of Life ScienceCHA UniversitySeongnamSouth Korea
| |
Collapse
|
37
|
Catalano A, Sacerdote C, Alvich M, Macciotta A, Milani L, Destefanis C, Gebru KT, Sodano B, Padroni L, Giraudo MT, Ciccone G, Pagano E, Boccuzzi A, Caramello V, Ricceri F. Multimorbidity and COVID-19 Outcomes in the Emergency Department: Is the Association Mediated by the Severity of the Condition at Admission? J Clin Med 2024; 13:7182. [PMID: 39685641 DOI: 10.3390/jcm13237182] [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: 10/25/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Charlson Comorbidity Index (CCI) is one of the most reliable indicators to assess the impact of multimorbidity on COVID-19-related outcomes. Moreover, the patient's clinical conditions are associated with SARS-CoV-2 outcomes. This study aimed to analyze the association between multimorbidity and COVID-19-related outcomes, evaluating whether the National Early Warning Score 2 (NEWS2) mediated these associations. Methods: Data were obtained through the platform "EPICLIN". We analyzed all patients who tested positive for COVID-19 after accessing the emergency department (ED) of San Luigi Gonzaga (Orbassano) and Molinette (Turin) hospitals from 1 March to 30 June 2020. Different outcomes were assessed: non-discharge from the ED, 30-day mortality, ICU admission/death among hospitalized patients, and length of hospitalization among surviving patients. Two subgroups of patients (<65 and 65+ years old) were analyzed using logistic regressions, Cox models, and mediation analyses. Results: There was a greater risk of not being discharged or dying among those who were younger and with CCI ≥ 2. Moreover, the higher the CCI, the longer the length of hospitalization. Considering older subjects, a greater CCI was associated with a higher risk of death. Regarding the mediation analyses, multimorbidity significantly impacted the hospitalization length and not being discharged in the younger population. Instead, in the older population, the NEWS2 played a mediation role. Conclusions: This research showed that multimorbidity is a risk factor for a worse prognosis of COVID-19. Moreover, there was a strong direct effect of CCI on not being discharged, and the NEWS2 was found to act as mediator in the association between multimorbidity and COVID-19-related outcomes.
Collapse
Affiliation(s)
- Alberto Catalano
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Carlotta Sacerdote
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
- Unit of Epidemiology, Local Health Unit of Novara, 28100 Novara, Italy
| | - Marco Alvich
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
| | - Alessandra Macciotta
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Lorenzo Milani
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
| | - Cinzia Destefanis
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
| | - Kibrom Teklay Gebru
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
| | - Barbara Sodano
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
- Department of Statistics, Computer Science, Applications, University of Florence, 50134 Florence, Italy
| | - Lisa Padroni
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
| | - Maria Teresa Giraudo
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
| | - Giovannino Ciccone
- Unit of Clinical Epidemiology, CPO, Città della Salute e della Scienza Hospital, 10126 Turin, Italy
| | - Eva Pagano
- Unit of Clinical Epidemiology, CPO, Città della Salute e della Scienza Hospital, 10126 Turin, Italy
| | - Adriana Boccuzzi
- Emergency Department and High Dependency Unit, San Luigi Gonzaga University Hospital, Orbassano, 10043 Turin, Italy
| | - Valeria Caramello
- Emergency Department and High Dependency Unit, San Luigi Gonzaga University Hospital, Orbassano, 10043 Turin, Italy
| | - Fulvio Ricceri
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy
| |
Collapse
|
38
|
Wei M, Li Q, Li S, Wang D, Wang Y. Multifaceted roles of cGAS-STING pathway in the lung cancer: from mechanisms to translation. PeerJ 2024; 12:e18559. [PMID: 39588006 PMCID: PMC11587877 DOI: 10.7717/peerj.18559] [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: 09/13/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024] Open
Abstract
Lung cancer (LC) remains one of the most prevalent and lethal malignancies globally, with a 5-year survival rate for advanced cases persistently below 10%. Despite the significant advancements in immunotherapy, a substantial proportion of patients with advanced LC fail to respond effectively to these treatments, highlighting an urgent need for novel immunotherapeutic targets. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has gained prominence as a potential target for improving LC immunotherapy due to its pivotal role in enhancing anti-tumor immune responses, augmenting tumor antigen presentation, and promoting T cell infiltration. However, emerging evidence also suggests that the cGAS-STING pathway may have pro-tumorigenic effects in the context of LC. This review aims to provide a comprehensive analysis of the cGAS-STING pathway, including its biological composition, activation mechanisms, and physiological functions, as well as its dual roles in LC and the current and emerging LC treatment strategies that target the pathway. By addressing these aspects, we intend to highlight the potential of the cGAS-STING pathway as a novel immunotherapeutic target, while also considering the challenges and future directions for its clinical application.
Collapse
Affiliation(s)
- Mingming Wei
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qingzhou Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shengrong Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Dong Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yumei Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Yu L, Liu MM, Guan MQ, Wang R, Yang XR, Zhang XM, Wei JJ, Wu SF, Gu H, Fu Q, Guo JH, Li YL. Peripheral CD4 + T cell phenotype and brain microglial activation associated with cognitive heterogeneity in aged rats. Immun Ageing 2024; 21:81. [PMID: 39543616 PMCID: PMC11562703 DOI: 10.1186/s12979-024-00486-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Cognitive decline is a critical hallmark of brain aging. Although aging is a natural process, there is significant heterogeneity in cognition levels among individuals; however, the underlying mechanisms remain uncertain. In our study, we classified aged male Sprague‒Dawley rats into aged cognition-unimpaired (AU) group and aged cognition-impaired (AI) group by using an attentional set-shifting task. The transcriptome sequencing results of medial prefrontal cortex (mPFC) demonstrated significant differences in microglial activation and inflammatory response pathways between the two groups. Specifically, compared to AU rats, AI rats exhibited a greater presence of CD86-positive microglia and major histocompatibility complex class II (MHC-II)-positive microglia, along with elevated inflammatory molecules, in mPFC. Conversely, AI rats exhibited a reduction in the percentage of microglia expressing CD200R and the anti-inflammatory molecules Arg-1 and TGF-β. Additionally, peripheral blood analysis of AI rats demonstrated elevated levels of Th17 and Th1 cells, along with proinflammatory molecules; however, decreased levels of Treg cells, along with anti-inflammatory molecules, were observed in AI rats. Our research suggested that peripheral Th17/Treg cells and central microglial activation were associated with cognitive heterogeneity in aged rats. These findings may provide a new target for healthy aging.
Collapse
Affiliation(s)
- Lian Yu
- Department of Neurology, Research Center for Neurological Diseases, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Miao-Miao Liu
- Department of Neurology, Research Center for Neurological Diseases, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Mei-Qi Guan
- Department of Pediatrics, Shanxi Medical University, Taiyuan, 030001, China
| | - Rui Wang
- Department of Neurology, Research Center for Neurological Diseases, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xiao-Rong Yang
- The Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiu-Min Zhang
- Department of Neurology, Research Center for Neurological Diseases, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jing-Jing Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, 030001, China
| | - Shu-Fen Wu
- Department of Pediatrics, Shanxi Medical University, Taiyuan, 030001, China
| | - Hong Gu
- Department of Neurology, Research Center for Neurological Diseases, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Qiang Fu
- Department of Neurology, Research Center for Neurological Diseases, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jun-Hong Guo
- Department of Neurology, Research Center for Neurological Diseases, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Yan-Li Li
- Department of Neurology, Research Center for Neurological Diseases, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
40
|
Yan H, Chen S, Gao X, Jiang Y, Liang G, Peng J, Cai S. Association Between TyG Index, Liver Steatosis and Immunosenescence in People Living with HIV. Infect Drug Resist 2024; 17:5049-5059. [PMID: 39559341 PMCID: PMC11570526 DOI: 10.2147/idr.s493140] [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: 09/02/2024] [Accepted: 11/04/2024] [Indexed: 11/20/2024] Open
Abstract
Background Metabolic disorders and immunosenescence increase the risk of complications in people living with HIV (PLWH), affecting mortality and quality of life. However, their relationship remains unclear. Methods Participants were grouped by median TyG index, and logistic regression identified baseline independent factors of a high TyG index at Week 24. The association of the TyG index for hepatic steatosis was determined using ROC curves. We also explored correlations between the TyG index and aging markers, including CD4/CD8 ratio and CD8+ T cells and evaluated health-related quality of life (HRQoL). Results A total of 203 PLWH were included in the study. We observed that PLWH in high TyG group tended to be older (P<0.001), have greater body weight (P<0.001), higher ALT levels (P=0.021), and increased low-density lipoprotein levels (P=0.001). ROC analysis revealed that TyG index was closely associated with hepatic steatosis at Week 52 (AUC=0.743) and Week 104 (AUC=0.728). Moreover, a higher TyG index was positively correlated with CD8+ T cell counts, while patients in the high TyG group had lower CD4/CD8 ratios at Week 52 and Week 104. Poorer mental health was observed in patients with CD8+ T cell counts ≥1000 and a high TyG index. Multivariate analysis further identified baseline older age (OR=1.108, P=0.002), elevated cholesterol (OR=3.407, P<0.001), and low HDL (OR=0.003, P<0.001) as factors associated with a high TyG index at Week 24. Conclusion The TyG index is closely linked to metabolic disorders and immunosenescence in PLWH. It offers a basis for personalized treatment strategies, improving physical and mental health and reducing complication risks.
Collapse
Affiliation(s)
- Haiming Yan
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Guangdong Institute of Hepatology, Guangzhou, People’s Republic of China
- Department of Infectious Diseases, The First People’s Hospital of Foshan, Foshan, People’s Republic of China
| | - Suling Chen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Guangdong Institute of Hepatology, Guangzhou, People’s Republic of China
| | - Xinrui Gao
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Guangdong Institute of Hepatology, Guangzhou, People’s Republic of China
| | - Yuanhui Jiang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Guangdong Institute of Hepatology, Guangzhou, People’s Republic of China
| | - Guangyu Liang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Guangdong Institute of Hepatology, Guangzhou, People’s Republic of China
| | - Jie Peng
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Guangdong Institute of Hepatology, Guangzhou, People’s Republic of China
| | - Shaohang Cai
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Guangdong Institute of Hepatology, Guangzhou, People’s Republic of China
| |
Collapse
|
41
|
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
|
42
|
Nogueira Silva Lima MT, Delayre-Orthez C, Howsam M, Jacolot P, Niquet-Léridon C, Okwieka A, Anton PM, Perot M, Barbezier N, Mathieu H, Ghinet A, Fradin C, Boulanger E, Jaisson S, Gillery P, Tessier FJ. Early- and life-long intake of dietary advanced glycation end-products (dAGEs) leads to transient tissue accumulation, increased gut sensitivity to inflammation, and slight changes in gut microbial diversity, without causing overt disease. Food Res Int 2024; 195:114967. [PMID: 39277266 DOI: 10.1016/j.foodres.2024.114967] [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: 05/23/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/17/2024]
Abstract
Dietary advanced glycation end-products (dAGEs) accumulate in organs and are thought to initiate chronic low-grade inflammation (CLGI), induce glycoxidative stress, drive immunosenescence, and influence gut microbiota. Part of the toxicological interest in glycation products such as dietary carboxymethyl-lysine (dCML) relies on their interaction with receptor for advanced glycation end-products (RAGE). It remains uncertain whether early or lifelong exposure to dAGEs contributes physiological changes and whether such effects are reversible or permanent. Our objective was to examine the physiological changes in Wild-Type (WT) and RAGE KO mice that were fed either a standard diet (STD - 20.8 ± 5.1 µg dCML/g) or a diet enriched with dCML (255.2 ± 44.5 µg dCML/g) from the perinatal period for up to 70 weeks. Additionally, an early age (6 weeks) diet switch (dCML→STD) was explored to determine whether potential harmful effects of dCML could be reversed. Previous dCML accumulation patterns described by our group were confirmed here, with significant RAGE-independent accumulation of dCML in kidneys, ileum and colon over the 70-week dietary intervention (respectively 3-fold, 17-fold and 20-fold increases compared with controls). Diet switching returned tissue dCML concentrations to their baseline levels. The dCML-enriched diet had no significative effect on endogenous glycation, inflammation, oxidative stress or senescence parameters. The relative expression of TNFα, VCAM1, IL6, and P16 genes were all upregulated (∼2-fold) in an age-dependent manner, most notably in the kidneys of WT animals. RAGE knockout seemed protective in this regard, diminishing age-related renal expression of TNFα. Significant increases in TNFα expression were detectable in the intestinal tract of the Switch group (∼2-fold), suggesting a higher sensitivity to inflammation perhaps related to the timing of the diet change. Minor fluctuations were observed at family level within the caecal microbiota, including Eggerthellaceae, Anaerovoracaceae and Marinifilaceae communities, indicating slight changes in composition. Despite chronic dCML consumption resulting in higher free CML levels in tissues, there were no substantial increases in parameters related to inflammageing. Age was a more important factor in inflammation status, notably in the kidneys, while the early-life dietary switch may have influenced intestinal susceptibility to inflammation. This study affirms the therapeutic potential of RAGE modulation and corroborates evidence for the disruptive effect of dietary changes occurring too early in life. Future research should prioritize the potential influence of dAGEs on disease aetiology and development, notably any exacerbating effects they may have upon existing health conditions.
Collapse
Affiliation(s)
- M T Nogueira Silva Lima
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - C Delayre-Orthez
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - M Howsam
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - P Jacolot
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - C Niquet-Léridon
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - A Okwieka
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France
| | - P M Anton
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - M Perot
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - N Barbezier
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - H Mathieu
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - A Ghinet
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France; Junia, Health and Environment, Laboratory of Sustainable Chemistry and Health, 59000 Lille, France
| | - C Fradin
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - E Boulanger
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - S Jaisson
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France; University Hospital of Reims, Laboratory of Biochemistry-Pharmacology-Toxicology, 51092 Reims, France
| | - P Gillery
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France; University Hospital of Reims, Laboratory of Biochemistry-Pharmacology-Toxicology, 51092 Reims, France
| | - F J Tessier
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France.
| |
Collapse
|
43
|
Zhang J, Guan X, Zhong X. Immunosenescence in digestive system cancers: Mechanisms, research advances, and therapeutic strategies. Semin Cancer Biol 2024; 106-107:234-250. [PMID: 39510149 DOI: 10.1016/j.semcancer.2024.10.006] [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: 08/13/2024] [Revised: 10/21/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
Increasing lifespans and external environmental factors have contributed to the increase of age-related diseases, particularly cancer. A decrease in immune surveillance and clearance of cancer cells is the result of immunosenescence, which involves the remodeling of immune organs, the changes and functional decline of immune cell subsets, in association with systemic low-grade chronic inflammation. Stem cells aging in bone marrow and thymic involution are the most important causes of immunosenescence. Senescent cancer cells promote the differentiation, recruitment, and functional upregulation of immune-suppressive cell subsets e.g. regulatory T cells (Tregs), myeloid-derived suppressor cell (MDSC), tumor-associated macrophages (TAMS) through senescence-associated secretory phenotype (SASP) further exacerbating the immunosuppressive microenvironment. For digestive system cancers, age-related damage to the intestinal mucosal barrier, the aging of gut-associated lymphoid tissue (GALT), exposure to xenobiotic stimuli throughout life, and dysbiosis make the local immune microenvironment more vulnerable. This article systematically reviews the research progress of immunosenescence and immune microenvironment in digestive system cancers, as well as the exploration of related therapy strategies, hoping to point out new directions for research in the digestive system cancers.
Collapse
Affiliation(s)
- Junyan Zhang
- Department of Surgical Oncology and General Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaojiao Guan
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Xinwen Zhong
- Department of Thoracic Surgery, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
44
|
Thiyagarajan R, Zhang L, Glover OD, Kwack KH, Ahmed S, Murray E, Yellapu NK, Bard J, Seldeen KL, Rosario SR, Troen BR, Kirkwood KL. Age-related increase of CD38 directs osteoclastogenic potential of monocytic myeloid-derived suppressor cells through mitochondrial dysfunction in male mice. Aging Cell 2024; 23:e14298. [PMID: 39180173 PMCID: PMC11561650 DOI: 10.1111/acel.14298] [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: 01/04/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/26/2024] Open
Abstract
An aged immune system undergoes substantial changes where myelopoiesis dominates within the bone marrow. Monocytic-MDSCs (M-MDSCs) have been found to play an important role in osteoclastogenesis and bone resorption. In this study, we sought to provide a more comprehensive understanding of the osteoclastogenic potential of bone marrow M-MDSCs during normal aging through transcriptomic and metabolic changes. Using young mature and aged mice, detailed immunophenotypic analyses of myeloid cells revealed that the M-MDSCs were not increased in bone marrow, however M-MDSCS were significantly expanded in peripheral tissues. Although aged mice exhibited a similar number of M-MDSCs in bone marrow, these M-MDSCs had significantly higher osteoclastogenic potential and greater demineralization activity. Intriguingly, osteoclast progenitors from aged bone marrow M-MDSCs exhibited greater mitochondrial respiration rate and glucose metabolism. Further, transcriptomic analyses revealed the upregulation of mitochondrial oxidative phosphorylation and glucose metabolism genes. Interestingly, there was 8-fold increase in Cd38 mRNA gene expression, consistent with the Mouse Aging Cell Atlas transcriptomic database, and confirmed by qRT-PCR. CD38 regulates NAD+ availability, and 78c, a small molecule inhibitor of CD38, reduced the mitochondrial oxygen consumption rate and glucose metabolism and inhibited the osteoclastogenic potential of aged mice bone marrow-derived M-MDSCs. These results indicate that the age-related increase in Cd38 expression in M-MDSCs bias the transcriptome of M-MDSCs towards osteoclastogenesis. This enhanced understanding of the mechanistic underpinnings of M-MDSCs and their osteoclastogenesis during aging could lead to new therapeutic approaches for age-related bone loss and promote healthy aging.
Collapse
Affiliation(s)
- Ramkumar Thiyagarajan
- Division of Geriatrics, Department of Internal Medicine and Landon Center on AgingUniversity of Kansas School of MedicineKansas CityKansasUSA
- Research ServiceVeteran Affairs Kansas City Healthcare SystemKansas CityMissouriUSA
| | - Lixia Zhang
- Department of Oral Biology, School of Dental MedicineUniversity at BuffaloBuffaloNew YorkUSA
| | - Omar D. Glover
- Department of Oral Biology, School of Dental MedicineUniversity at BuffaloBuffaloNew YorkUSA
| | - Kyu Hwan Kwack
- Department of Oral Biology, School of Dental MedicineUniversity at BuffaloBuffaloNew YorkUSA
- Department of Oral Microbiology, College of DentistryKyung Hee UniversitySeoulRepublic of Korea
| | - Sara Ahmed
- Department of Oral Biology, School of Dental MedicineUniversity at BuffaloBuffaloNew YorkUSA
| | - Emma Murray
- Department of Oral Biology, School of Dental MedicineUniversity at BuffaloBuffaloNew YorkUSA
| | - Nanda Kumar Yellapu
- Department of Biostatistics and Data ScienceUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Jonathan Bard
- Genomics and Bioinformatics Core, New York State Center of Excellence for Bioinformatics and Life SciencesUniversity at BuffaloBuffaloNew YorkUSA
| | - Kenneth L. Seldeen
- Division of Geriatrics, Department of Internal Medicine and Landon Center on AgingUniversity of Kansas School of MedicineKansas CityKansasUSA
- Research ServiceVeteran Affairs Kansas City Healthcare SystemKansas CityMissouriUSA
| | - Spencer R. Rosario
- Department of Biostatistics and Bioinformatics and Experimental TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Bruce R. Troen
- Division of Geriatrics, Department of Internal Medicine and Landon Center on AgingUniversity of Kansas School of MedicineKansas CityKansasUSA
- Research ServiceVeteran Affairs Kansas City Healthcare SystemKansas CityMissouriUSA
| | - Keith L. Kirkwood
- Department of Oral Biology, School of Dental MedicineUniversity at BuffaloBuffaloNew YorkUSA
- Department of Head and Neck/Plastic and Reconstructive SurgeryRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| |
Collapse
|
45
|
Ajoolabady A, Pratico D, Tang D, Zhou S, Franceschi C, Ren J. Immunosenescence and inflammaging: Mechanisms and role in diseases. Ageing Res Rev 2024; 101:102540. [PMID: 39395575 DOI: 10.1016/j.arr.2024.102540] [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: 04/28/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Age-related changes initiate a cascade of cellular and molecular alterations that lead to immune system dysfunction or abnormal activation, predisposing individuals to age-related diseases. This phenomenon, commonly referred to as immunosenescence, highlighting aging-associated progressive decline of the immune system. Moreover, mounting evidence suggests that immunosenescence contributes to a related pathological phenomenon known as inflammaging. Inflammaging refers to chronic, low-grade, and systemic inflammation associated with aging, occurring despite the absence of overt stimuli. In the body, inflammation is typically activated in response to overt stimuli such as bacterial/microbial invasion or a pathological state, however, inflammaging occurrence and its underpinning mechanisms seem to be independent and in the absence of such stimuli. Despite recent advancements in molecular characterization and the scrutiny of disease relevance, these two interconnected concepts have remained largely unexplored and unrecognized. In this comprehensive review, we aim to shed light on the mechanistic and cellular aspects of immunosenescence and inflammaging, as well as their pivotal roles in the pathogenesis of aging-related diseases, including cancer, infections, dementia, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shuqin Zhou
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Claudio Franceschi
- IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy; Department of Applied Mathematics and Laboratory of Systems Biology of Aging, Lobachevsky University, Nizhny Novgorod, Russia.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
46
|
Wu L, Lin H, Li S, Huang Y, Sun Y, Shu S, Luo T, Liang T, Lai W, Rao J, Hu Z, Peng H. Macrophage iron dyshomeostasis promotes aging-related renal fibrosis. Aging Cell 2024; 23:e14275. [PMID: 39016438 PMCID: PMC11561705 DOI: 10.1111/acel.14275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
Renal aging, marked by the accumulation of senescent cells and chronic low-grade inflammation, leads to renal interstitial fibrosis and impaired function. In this study, we investigate the role of macrophages, a key regulator of inflammation, in renal aging by analyzing kidney single-cell RNA sequencing data of C57BL/6J mice from 8 weeks to 24 months. Our findings elucidate the dynamic changes in the proportion of kidney cell types during renal aging and reveal that increased macrophage infiltration contributes to chronic low-grade inflammation, with these macrophages exhibiting senescence and activation of ferroptosis signaling. CellChat analysis indicates enhanced communications between macrophages and tubular cells during aging. Suppressing ferroptosis alleviates macrophage-mediated tubular partial epithelial-mesenchymal transition in vitro, thereby mitigating the expression of fibrosis-related genes. Using SCENIC analysis, we infer Stat1 as a key age-related transcription factor promoting iron dyshomeostasis and ferroptosis in macrophages by regulating the expression of Pcbp1, an iron chaperone protein that inhibits ferroptosis. Furthermore, through virtual screening and molecular docking from a library of anti-aging compounds, we construct a docking model targeting Pcbp1, which indicates that the natural small molecule compound Rutin can suppress macrophage senescence and ferroptosis by preserving Pcbp1. In summary, our study underscores the crucial role of macrophage iron dyshomeostasis and ferroptosis in renal aging. Our results also suggest Pcbp1 as an intervention target in aging-related renal fibrosis and highlight Rutin as a potential therapeutic agent in mitigating age-related renal chronic low-grade inflammation and fibrosis.
Collapse
Affiliation(s)
- Lingzhi Wu
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hongchun Lin
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shaomin Li
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuebo Huang
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuxiang Sun
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shuangshuang Shu
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ting Luo
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Tiantian Liang
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Weiyan Lai
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jialing Rao
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhaoyong Hu
- Nephrology Division, Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Hui Peng
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhouChina
| |
Collapse
|
47
|
Kabakibo TS, Arnold E, Padhan K, Lemieux A, Ortega-Delgado GG, Routy JP, Shoukry N, Dubé M, Kaufmann DE. Artificial antigen-presenting cell system reveals CD28's role in modulating T cell functions during human immunodeficiency virus infection. iScience 2024; 27:110947. [PMID: 39381752 PMCID: PMC11460474 DOI: 10.1016/j.isci.2024.110947] [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: 10/13/2023] [Revised: 02/16/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
T cell immune dysfunction is a prominent feature of chronic HIV infection. To evaluate non-specific dysfunction, a method involving both generic activation and T cell receptor (TCR) stimulation is necessary. We created a tunable artificial antigen-presenting cell (aAPC) system. This system consists of lipid bilayers on cytometry-compatible silica microbeads (5 μm). When only anti-CD3 is incorporated, T cell activation is limited. Introducing anti-CD28 agonists significantly elevates the cytokine expression and upregulation of activation-induced markers. CD28 co-stimulation modulates the response profile, preferentially promoting IL-2 expression relative to other cytokines. aAPCs-stimulated CD4+ and CD8+ T cells from untreated HIV-infected individuals exhibit altered effector functions and diminished CD28 dependence. These functions are skewed toward TNFα, IFNγ and CD107a, with reduced IL-2. Antiretroviral therapy partially normalizes this distorted profile in CD4+ T cells, but not in CD8+ T cells. Our findings show T cell intrinsic biases that may contribute to persistent systemic T cell dysfunction associated with HIV pathogenesis.
Collapse
Affiliation(s)
- Tayma Shaaban Kabakibo
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Université de Montréal, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Edwige Arnold
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Université de Montréal, Montréal, QC, Canada
| | - Kartika Padhan
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Audrée Lemieux
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Université de Montréal, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | | | - Jean-Pierre Routy
- Chronic Viral Illnesses Service and Division of Hematology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada
| | - Naglaa Shoukry
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Université de Montréal, Montréal, QC, Canada
- Département de Médecine, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Mathieu Dubé
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Daniel E. Kaufmann
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Université de Montréal, Montréal, QC, Canada
- Département de Médecine, Université de Montréal, Montréal, QC H2X 0A9, Canada
- Division of Infectious Diseases, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
48
|
Xiong F, Shen K, Long D, Zhou S, Ruan P, Xin Y, Xiao Y, Peng W, Yang M, Wu H, Lu Q. Quercetin ameliorates lupus symptoms by promoting the apoptosis of senescent Tfh cells via the Bcl-2 pathway. Immun Ageing 2024; 21:69. [PMID: 39407236 PMCID: PMC11476537 DOI: 10.1186/s12979-024-00474-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder that commonly affects the skin, kidneys, joints, and various other systemic tissues, with its development intricately linked to the process of immunosenescence. Quercetin (QC), a phytochemical that occurs naturally, demonstrates many different biological capabilities, such as antibacterial, antioxidant, and anti-inflammatory activities. Our investigation found that QC effectively reduced kidney damage and relieved mesenteric lymph nodes (mLNs) swelling in MRL/lpr lupus mice. Moreover, QC has been found to decrease the number of senescent follicular helper T (Tfh) cells, a pivotal kind of T cells that contribute to the progression of SLE. In vitro, QC exhibited the capacity to modulate mRNA expression levels, with the downregulation of IL-6, IL21-AS1, IL-27, BCL6, and BCL2L12, and the upregulation of FOXP1 and BIM. This modulation resulted in the suppression of Tfh cells differentiation and the enhancement of apoptosis in senescent CD4+ T cells. In addition, the HuProtTM Human Proteome Microarray revealed that QC can directly bind to BCL-2 protein and therefore promote the apoptosis of senescent CD4+ T cell. As a result, our investigative elucidate the potent inhibitory action of QC on the ontogeny of Tfh cells, along with its capacity to abrogate the immunosenescent phenotype. This positions QC as a promising therapeutic strategy for treating SLE.
Collapse
Affiliation(s)
- Feng Xiong
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Kai Shen
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Di Long
- Department of Dermatology, The Second Affiliated Hospital, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Suqing Zhou
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Pinglang Ruan
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Yue Xin
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Yuezheng Xiao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| |
Collapse
|
49
|
Shi J, Zhang M, Zhang L, Yu X, Sun L, Liu J, Zhao Y, Zheng W. Shelterin dysfunction promotes CD4+ T cell senescence in Behçet's disease. Rheumatology (Oxford) 2024; 63:2819-2827. [PMID: 38145496 DOI: 10.1093/rheumatology/kead703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/26/2023] [Accepted: 12/16/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVES To investigate the potential role of shelterin dysfunction in naïve CD4+ T cells in the pathogenesis of Behçet's disease (BD). METHODS Naïve CD4+ T cells were isolated from 40 BD patients and 40 sex- and age-matched healthy controls (HC). Senescent profiles, shelterin subunits expression, telomere length, telomerase activity and critical DNA damage response (DDR) were evaluated. Telomere repeat factor-2 (TRF2) silencing was conducted for further validation. RESULTS Compared with HC, BD patients had significantly decreased naïve CD4+ T cells, increased cell apoptosis, senescence, and productions of TNF-α and IFN-γ upon activation. Notably, BD naïve CD4+ T cells had shortened telomere, impaired telomerase activity, and expressed lower levels of shelterin subunits TRF2, TRF1- and TRF2-Interacting Nuclear Protein 2 (TIN2) and Repressor/Activator Protein 1 (RAP1). Furthermore, BD naïve CD4+ T cells exhibited significantly increased DDR, evidenced by elevated phosphorylated ataxia telangiectasia (AT) mutated (pATM), phosphorylated p53 (pp53) and p21. Finally, TRF2 silencing markedly upregulated DDR, apoptosis and proinflammatory cytokines production in HC naïve CD4+ T cells. CONCLUSION Our study demonstrated that TRF2 deficiency in BD naïve CD4+ T cells promoted cell apoptosis and senescence, leading to proinflammatory cytokines overproduction. Therefore, restoring TRF2 might be a promising therapeutic strategy for BD.
Collapse
Affiliation(s)
- Jing Shi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Menghao Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Lili Zhang
- Department of Rheumatology, Linyi People's Hospital, Linyi, Shandong, China
| | - Xin Yu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Luxi Sun
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Jinjing Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Wenjie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| |
Collapse
|
50
|
Yimam M, Horm T, O’Neal A, Chua P, Jiao P, Hong M, Jia Q. Botanical Bioflavonoid Composition from Scutellaria baicalensis- and Acacia catechu-Protected Mice against D-Galactose-Induced Immunosenescence, and Cyclophosphamide Induced Immune Suppression. Nutrients 2024; 16:3144. [PMID: 39339746 PMCID: PMC11434731 DOI: 10.3390/nu16183144] [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: 08/09/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Oxidative stress and chronic inflammation create a perpetual cycle in the elderly, where impaired immune function amplifies susceptibility to oxidative damage, and oxidative stress further weakens the immune response. This cycle is particularly detrimental to the respiratory system of the elderly, which is an easy target for constant exogenous harmful attacks during cold/flu season or under heavy air pollution. Herbal medicines that protect respiratory function are seen as safer alternatives to conventional therapies; however, there is limited availability of scientifically validated, safe, and effective natural supplements for these conditions. In this study, we evaluated a standardized bioflavonoid composition, UP446, that contains bioactives from the roots of Scutellaria baicalensis and the heartwoods of Acacia catechu as a natural and nutritional supplement for its antioxidative and immunoregulatory effects in oxidative stress-accelerated aging and chemically induced immune suppression mouse models. Immunosenescence was induced through the repeated subcutaneous inoculation of D-galactose (D-Gal) at a dose of 500 mg/kg/day in CD-1 mice. UP446 was administered orally at doses of 100 mg/kg and 200 mg/kg starting in the fifth week of immunosenescence induction. This study lasted a total of ten weeks. All mice received a quadrivalent influenza vaccine 2 weeks before termination. Whole blood, serum, spleen homogenate, and thymus tissues were processed for analysis. Cyclophosphamide (Cy)-induced immunosuppression was triggered by three consecutive injections of cyclophosphamide at 80 mg/kg/day, followed by the oral administration of UP446 for 18 days at doses of 100 mg/kg and 200 mg/kg. Blood was collected from each animal at necropsy, and serum was isolated for IgA and IgG ELISA analysis. UP446 was found to improve immune response, as evidenced by the stimulation of innate (NK cells) and adaptive immune responses (T cells and cytotoxic T cells), an increase in antioxidant capacity (glutathione peroxidase), the preservation of vital immune organs (the thymus), and a reduction in NFκB. UP446 also increased serum levels of IgA and IgG. The findings presented in this report demonstrate the antioxidative, anti-inflammatory, and immune-regulatory activities of UP446, suggesting its potential use in respiratory conditions involving immune stress due to aging, oxidative stress, and/or pathogenic challenges.
Collapse
Affiliation(s)
- Mesfin Yimam
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (P.J.); (M.H.); (Q.J.)
| | - Teresa Horm
- Department of Biology, Pacific Lutheran University, 12180 Park Ave. S, Tacoma, WA 98447, USA;
| | | | - Paola Chua
- Washington State Department of Health, 1610 NE 150th St, Shoreline, WA 98155, USA;
| | - Ping Jiao
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (P.J.); (M.H.); (Q.J.)
| | - Mei Hong
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (P.J.); (M.H.); (Q.J.)
| | - Qi Jia
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (P.J.); (M.H.); (Q.J.)
| |
Collapse
|