1
|
Zhang K, Yin Q, Ma Y, Cao M, Li L, Jin X, Leng J. Nanovaccine loaded with seno-antigen target senescent cells to improve metabolic disorders of adipose tissue and cardiac dysfunction. Hum Vaccin Immunother 2025; 21:2479229. [PMID: 40088037 PMCID: PMC11916409 DOI: 10.1080/21645515.2025.2479229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 03/17/2025] Open
Abstract
The buildup of senescent cells exacerbates metabolic disorders in adipose tissue and contributes to aging-related cardiac dysfunction. Targeted clearance of senescent cells can markedly ameliorate these aging-related diseases. Here, we developed a novel nanovaccine (GK-NaV) loaded with seno-antigen that is self-assembled from the fusion of cationic protein (K36) and seno-antigen peptide (Gpnmb). The GK-NaV could be highly engulfed by bone marrow-derived dendritic cells (BMDCs) and efficiently present antigens on the cellular surface, thereby promoting DCs maturation and activation of CD8+T cells in vitro. Following subcutaneous immunization, GK-NaV not only exhibited a noticeable antigen depot effect but also markedly activated specific cellular immune responses, enhancing the immunoreactivity and cytotoxic effects of CD8+T cells. Consequently, the targeted anti-aging immunity triggered by GK-NaV demonstrated the ability to selectively eliminate senescent adipocytes and cardiomyocytes in high-fat diet (HFD)-induced progeroid mice, leading to a significant improvement in age-related metabolic disorders in adipose tissue and cardiac dysfunction. Hence, our findings indicate that immunization with GK-NaV targeting seno-antigens may represent a promising strategy for novel senolytic therapies.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Qiliang Yin
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Yucen Ma
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Mengyu Cao
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Lingwei Li
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Xinliang Jin
- Department of General Surgery, First Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jiyan Leng
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Picos A, Seoane N, Campos-Toimil M, Viña D. Vascular senescence and aging: mechanisms, clinical implications, and therapeutic prospects. Biogerontology 2025; 26:118. [PMID: 40418230 DOI: 10.1007/s10522-025-10256-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
The aging vasculature is characterized by endothelial dysfunction, arterial stiffness, and increased susceptibility to vascular pathologies. Central to these changes is the process of cellular senescence, where endothelial and vascular smooth muscle cells lose their replicative and functional capacity and adopt a pro-inflammatory secretory phenotype. This review provides an overview of the key mechanisms underlying vascular senescence, including the p53/p21 and p16/Rb pathways, the senescence-associated secretory phenotype (SASP), and oxidative stress, examines its contribution to cardiovascular diseases in older adults, and highlights emerging therapeutic strategies aimed at delaying or reversing these age-related vascular changes. In vascular cells, DNA damage, oxidative stress, and chronic inflammation associated with aging converge to amplify senescence. Clinically, vascular senescence is linked with hypertension, atherosclerosis, and increased overall cardiovascular risk. Several interventions, ranging from senolytics to lifestyle factors, show promise in mitigating these changes; however, long-term studies are needed. Given that vascular senescence is a pivotal driver of cardiovascular pathology in aging, targeting senescent cells or their secretory phenotype may potentially offer new avenues for preventing or attenuating age-related vascular diseases. This review presents an updated and integrative overview of vascular senescence, connecting fundamental cellular mechanisms with their clinical manifestations and highlighting the most promising therapeutic interventions.
Collapse
Affiliation(s)
- Aitor Picos
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Translational Research in Neurological Diseases (ITEN), Health Research Institute of Santiago de Compostela (IDIS), USC University Hospital Complex (CHUS), SERGAS, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| | - Nuria Seoane
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Translational Research in Neurological Diseases (ITEN), Health Research Institute of Santiago de Compostela (IDIS), USC University Hospital Complex (CHUS), SERGAS, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Manuel Campos-Toimil
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Translational Research in Neurological Diseases (ITEN), Health Research Institute of Santiago de Compostela (IDIS), USC University Hospital Complex (CHUS), SERGAS, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| | - Dolores Viña
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Translational Research in Neurological Diseases (ITEN), Health Research Institute of Santiago de Compostela (IDIS), USC University Hospital Complex (CHUS), SERGAS, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| |
Collapse
|
3
|
Feng Y, Zhu Z, Zhao S, Jiang X, Zhang W, Xu Z. Bioorthogonally Activatable Photosensitizer for NIR Fluorescence Imaging-Guided Highly Selective Elimination of Senescent Tumor Cells and Chemotherapy Enhancement. Bioconjug Chem 2025; 36:1066-1078. [PMID: 40329576 DOI: 10.1021/acs.bioconjchem.5c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Chemotherapy is a primary modality in cancer treatment, but it may induce cellular senescence, which in turn triggers the release of senescence-associated secretory phenotypes (SASPs) that promote tumor growth and metastasis. To selectively identify senescent cells and mitigate their negative impact on cancer therapy, herein, we have developed a β-galactosidase (β-Gal)-activated and self-immobilizing photosensitizer CyGF-DBCO-T. This photosensitizer can be selectively activated and fluorescently label proteins in situ within senescent cells, enabling near-infrared (NIR) fluorescence imaging-guided photodynamic therapy (PDT) for the precise ablation of these cells. First, we developed an activatable NIR fluorescent probe CyGF-N3 that can specifically in situ label senescent cells. Subsequently, DBCO-T with free radicals underwent a bioorthogonal click reaction with activated CyGF-N3 in senescent cells to generate the photosensitizer CyO-DBCO-T. Under light irradiation, CyO-DBCO-T generated singlet oxygen (1O2) in situ, thereby enabling precise PDT with fluorescence guidance and photoactivation. Both CyGF-N3 and DBCO-T were encapsulated in biotinylated liposomes (CyGF-N3@LIP-B and DBCO-T@LIP-B), which enhanced their water solubility, tumor targeting, and in vivo circulation time. This promoted the accumulation of the probes in senescent tumor cells, thus enabling intense fluorescence imaging of tumor senescence regions in mice and enhancing the efficacy of PDT. This dual-module strategy, guided by fluorescence imaging for PDT, has achieved selective identification and precise ablation of senescent tumor cells in a chemotherapy-induced senescence model, effectively alleviating chemotherapy resistance and suppressing tumor growth.
Collapse
Affiliation(s)
- Yun Feng
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Zifan Zhu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Shirui Zhao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Xingyu Jiang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Wen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai 200062, China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| |
Collapse
|
4
|
Oppong R, Orru V, Marongiu M, Qian Y, Sidore C, Delitala A, Orru M, Mulas A, Piras MG, Morrell CH, Lai S, Schlessinger D, Gorospe M, Cucca F, Fiorillo E, Ding J, Lakatta EG, Scuteri A. Age-Associated Increase in Growth Differentiation Factor 15 Levels Correlates With Central Arterial Stiffness and Predicts All-Cause Mortality in a Sardinian Population Cohort. J Am Heart Assoc 2025; 14:e036253. [PMID: 40371596 DOI: 10.1161/jaha.124.036253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/08/2024] [Indexed: 05/16/2025]
Abstract
BACKGROUND Growth differentiation factor 15 (GDF-15) levels are emerging as a candidate biomarker of aging. The present study aimed to: (1) characterize the association of GDF-15 with the continuum of arterial stiffening, assessed as carotid-femoral pulse wave velocity, as age increases; (2) determine the predictive role of serum GDF-15 levels on mortality; and (3) identify genetic determinants of serum GDF-15 levels. METHODS AND RESULTS Serum levels of GDF-15 and established cardiovascular risk factors, including pulse wave velocity, were assessed in a large (4736 individual) Sardinian population. Serum levels of GDF-15, which can be reliably measured repeatedly over time, increase with age; are associated with a stiffer aorta; "mediate" a large proportion of the age-associated increase in arterial stiffness; pose risks because of their association with greater mortality; and are significantly associated with the variant rs11549407, which causes thalassemia major in homozygosity. CONCLUSIONS Because of its consistent ability to predict functional and clinical outcomes, including all-cause mortality, we conclude that GDF-15 serum levels serve as a robust biomarker for the continuum from health to the emergence of clinical disease during aging and, subsequently, to the likelihood of mortality.
Collapse
Affiliation(s)
- Richard Oppong
- National Institute on Aging - Intramural Research Program NIH Baltimore MD
| | - Valeria Orru
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | - Michele Marongiu
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | - Yong Qian
- National Institute on Aging - Intramural Research Program NIH Baltimore MD
| | - Carlo Sidore
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | - Alessandro Delitala
- Department Surgical, and Experimental Sciences University of Sassari Sassari Italy
| | - Marco Orru
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | - Maria Grazia Piras
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | | | - Sandra Lai
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | - David Schlessinger
- National Institute on Aging - Intramural Research Program NIH Baltimore MD
| | - Myriam Gorospe
- National Institute on Aging - Intramural Research Program NIH Baltimore MD
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica (IRGB) Consiglio Nazionale delle Ricerche (CNR) Lanusei (NU) Italy
| | - Jun Ding
- National Institute on Aging - Intramural Research Program NIH Baltimore MD
| | - Edward G Lakatta
- National Institute on Aging - Intramural Research Program NIH Baltimore MD
| | - Angelo Scuteri
- Internal Medicine Unit Policlinico Universitario Monserrato - Azienda Ospedaliera Universitaria (AOU) Cagliari Cagliari Italy
| |
Collapse
|
5
|
Centonze M, Aloisio Caruso E, De Nunzio V, Cofano M, Saponara I, Pinto G, Notarnicola M. The Antiaging Potential of Dietary Plant-Based Polyphenols: A Review on Their Role in Cellular Senescence Modulation. Nutrients 2025; 17:1716. [PMID: 40431456 PMCID: PMC12114605 DOI: 10.3390/nu17101716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Aging is a complex biological process characterized by a progressive decline in physiological functions and an increased risk of chronic diseases. A key mechanism of this process is cellular senescence, the permanent arrest of the cell cycle in response to stress or damage, which contributes to the accumulation of dysfunctional cells in tissues. Recent research has highlighted the role of polyphenols, bioactive compounds present in numerous plant-based foods, in positively modulating these processes. Polyphenols exert antioxidant effects, regulate gene expression and improve mitochondrial function, helping to delay cellular aging and prevent age-related diseases. In addition, some polyphenols exhibit senolytic properties, selectively eliminating senescent cells and promoting tissue regeneration. This review summarizes the current evidence on the effects of polyphenols on aging and cellular senescence, exploring the underlying molecular mechanisms and discussing their potential in nutritional strategies aimed at promoting healthy aging.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maria Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (M.C.); (E.A.C.); (V.D.N.); (M.C.); (I.S.); (G.P.)
| |
Collapse
|
6
|
Li S, Lv H, Zhang R, Li J, Chen Z, Yang N, Dai S. Aging-related alternative splicing drive neoantigen emergence revealed by transcriptome analysis of 1,255 human blood samples. FRONTIERS IN AGING 2025; 6:1575862. [PMID: 40417629 PMCID: PMC12098113 DOI: 10.3389/fragi.2025.1575862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/30/2025] [Indexed: 05/27/2025]
Abstract
This study aimed to identify age-related genes and alternative splicing (AS) events by comprehensive transcriptome analysis of 1,255 healthy blood samples from individuals aged 8-87 years. We identified 1,029 up-regulated and 1,186 down-regulated genes in older individuals, including 17 genes overlapped with known aging-associated genes, such as TFAP2A and Klotho. Gene set enrichment analysis revealed significant alterations in immunoregulatory and metabolic pathways during aging. However, many senescence-associated secretory phenotypes (SASP) involved genes did not exhibit changes in gene expression, suggesting that AS events may reveal additional age-related mechanisms. Aging also altered 6,320 AS events in 4,566 genes, impacting immune-related protein domains. The RNA-binding protein RBMS3 emerged as a key regulator of aging-specific AS events. In addition, neoantigen prediction analyses further identified potential neoantigens generated by aging-related AS events, with the HLA-C14:02 allele presenting the most neoantigenic peptides. Notably, 60 neoantigenic peptides were confirmed using proteomic data from elderly individuals, suggesting their potential as novel targets for anti-aging immunotherapy. Our study provides new insights into the role of alternative splicing in aging, highlights promising avenues for anti-aging immunotherapy.
Collapse
Affiliation(s)
- Shuhan Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Haohao Lv
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Renxin Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Jinjun Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Zhiyuan Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Naixue Yang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Shaoxing Dai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| |
Collapse
|
7
|
Chen C, Wang L. Aging and metabolic dysfunction-associated steatotic liver disease: a bidirectional relationship. Front Med 2025:10.1007/s11684-025-1133-7. [PMID: 40316793 DOI: 10.1007/s11684-025-1133-7] [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: 10/28/2024] [Accepted: 01/09/2025] [Indexed: 05/04/2025]
Abstract
In recent years, aging and cellular senescence have triggered an increased interest in corresponding research fields. Evidence shows that the complex aging process is involved in the development of many chronic liver diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). In fact, aging has a tremendous effect on the liver, leading to a gradual decline in the metabolism, detoxification and immune functions of the liver, which in turn increases the risk of liver disease. These changes can be based on the aging of liver cells (hepatocytes, liver sinusoidal endothelial cells, hepatic stellate cells, and Kupffer cells). Similarly, patients with liver diseases exhibit increases in the aging phenotype and aging cells, often manifesting as faster physical functional decline, which is closely related to the promoting effect of liver disease on aging. This review summarizes the interplay between MASLD/MASH development and aging, aiming to reveal the complex relationships that exacerbate one another. Moreover, the corresponding schemes for delaying aging or treating diseases are discussed to provide a basis for the development of effective prevention and treatment strategies in the future.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
8
|
Liang Z, Tang Q, Liang H, Liang X, Fu C, Kang W, Zhang Y, Lv P. Glucomannogalactan inhibits senescence by promoting nuclear translocation of NRF2. Int J Biol Macromol 2025; 305:141059. [PMID: 39961569 DOI: 10.1016/j.ijbiomac.2025.141059] [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: 09/19/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
For a potential resource to improve healthspan, polysaccharides present unique advantages in terms of side effects and long-term use owing to their low cytotoxicity. In this study, we demonstrate that a glucomannogalactan (PGP) derived from Pleurotus geesteranus extends the healthspan of both naturally senescent and therapy-induced senescence (TIS) mice. Daily treatment of naturally senescent mice with PGP resulted in a reduced accumulation of senescent cells and alleviation of senescence-related parameters, including metabolic dysfunction, underlying lesions in multiple organs, and oxidative damage. PGP treatment also attenuated senescence in TIS mice. Furthermore, in an in vitro model of oxidative stress-induced senescence using a human cell line, we discovered that PGP alleviated senescence by promoting the nuclear translocation of NRF2. This study suggests that PGP may extend the healthspan of senescent mice by facilitating the nuclear translocation of NRF2.
Collapse
Affiliation(s)
- Zhenhua Liang
- Cardiovascular Medical Science Center, Department of Cell Biology, the Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Qi Tang
- Hebei Key Laboratory of Forensic Medicine, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Haiyang Liang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
| | - Xuan Liang
- Cardiovascular Medical Science Center, Department of Cell Biology, the Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Chenghao Fu
- Cardiovascular Medical Science Center, Department of Cell Biology, the Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Wenyi Kang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China.
| | - Yan Zhang
- Hebei Key Laboratory of Forensic Medicine, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Hebei Food Safety Key Laboratory, Hebei Food Inspection and Research Institute, Shijiazhuang 050091, China.
| | - Pin Lv
- Cardiovascular Medical Science Center, Department of Cell Biology, the Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
9
|
Rys RN, Calcinotto A. Senescent neutrophils: a hidden role in cancer progression. Trends Cell Biol 2025; 35:399-411. [PMID: 39362804 DOI: 10.1016/j.tcb.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Neutrophils have recently received increased attention in cancer because they contribute to all stages of cancer. Neutrophils are so far considered to have a short half-life. However, a growing body of literature has shown that tumor-associated neutrophils (TANs) acquire a prolonged lifespan. This review discusses recent work surrounding the mechanisms by which neutrophils can persist in the tumor microenvironment (TME). It also highlights different scenarios for therapeutic targeting of protumorigenic neutrophils, supporting the idea that, in tumors, inhibition of neutrophil recruitment is not sufficient because these cells can persist and remain hidden from current interventions. Hence, the elimination of long-lived neutrophils should be pursued to increase the efficacy of standard therapy.
Collapse
Affiliation(s)
- Ryan N Rys
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Arianna Calcinotto
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland.
| |
Collapse
|
10
|
Zhang H, Xu Q, Jiang Z, Sun R, Wang Q, Liu S, Luan X, Campisi J, Kirkland JL, Zhang W, Sun Y. Targeting Senescence with Apigenin Improves Chemotherapeutic Efficacy and Ameliorates Age-Related Conditions in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412950. [PMID: 40265973 PMCID: PMC12120719 DOI: 10.1002/advs.202412950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/16/2025] [Indexed: 04/24/2025]
Abstract
Cellular senescence is a cell fate triggered by stressful stimuli and displays a hypersecretory feature, the senescence-associated secretory phenotype (SASP). Senescent cell burden increases with aging and contributes to age-related organ dysfunction and multiple chronic disorders. In this study, a large scale screening of a natural product library for senotherapeutic candidates is performed. Apigenin, a dietary flavonoid previously reported with antioxidant and anti-inflammatory activities, exhibits capacity for targeting senescent cells as a senomorphic agent. This compound blocks the interactions between ATM/p38MAPK and HSPA8, preventing the transition of an acute stress-associated phenotype (ASAP) toward the SASP. Mechanistically, apigenin targets peroxiredoxin 6 (PRDX6), an intracellular redox-active molecule, suppressing the iPLA2 activity of PRDX6 and disrupting downstream reactions underlying SASP development. Apigenin reduces the severity of cancer cell malignancy promoted by senescent stromal cells in culture, while restraining chemoresistance when combined with chemotherapy in anticancer regimens. In preclinical trials, apigenin improves the physical function of animals with a premature aging-like state, alleviating physical frailty and cognitive impairment. Together, the study demonstrates the feasibility of exploiting a natural compound with senomorphic capacity to achieve geroprotective effects by modulating the SASP, thus providing a baseline for future exploration of natural agents for alleviating age-related conditions.
Collapse
Affiliation(s)
- Hongwei Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203P. R. China
| | - Qixia Xu
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthChinese Academy of SciencesShanghai200031P. R. China
| | - Zhirui Jiang
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthChinese Academy of SciencesShanghai200031P. R. China
| | - Rong Sun
- Department of Discovery BiologyBioduro‐SundiaZhangjiang Hi‐Tech ParkShanghai201210P. R. China
| | - Qun Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203P. R. China
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203P. R. China
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203P. R. China
| | | | - James L. Kirkland
- Center for Advanced GerotherapeuticsCedars‐Sinai Medical CenterPacific Design CenterWest HollywoodCA90069USA
- Division of EndocrinologyDiabetes and MetabolismCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203P. R. China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di HerbsInstitute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100193P. R. China
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthChinese Academy of SciencesShanghai200031P. R. China
- Department of Medicine and VAPSHCSUniversity of WashingtonSeattleWA98195USA
| |
Collapse
|
11
|
Kim S, Gu B, So CY, Mantik KEK, Jung SH, Moon S, Park DH, Kwak HB, Cho J, Cho EJ, Lee JS, Kang JH. Cdkn1a silencing restores myoblast differentiation by inducing selective apoptosis in senescent cells. Cell Mol Biol Lett 2025; 30:53. [PMID: 40307745 PMCID: PMC12042464 DOI: 10.1186/s11658-025-00731-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Sarcopenia, characterized by a progressive loss of skeletal muscle mass and function, is associated with the accumulation of senescent muscle stem cells, which impair muscle regeneration and contributes to the decline in muscle health. Cdkn1a, which encodes p21, is a well-known marker of cellular senescence. However, it remains unclear whether p21 inhibition eliminates senescent myoblasts and restores the differentiation capacity. METHODS We performed transcriptomic analysis to identify genes related to aging-induced sarcopenia using 21 month-old Sprague-Dawley rats. To investigate the specific role of Cdkn1a gene in muscle aging, we used an in vitro model of ceramide-induced senescence in myoblasts, which was verified by the upregulation of p21 and increased senescence-associated beta-galactosidase (SA-β-gal) staining. To inhibit p21, we treated myoblasts with small interfering RNA (siRNA) targeting Cdkn1a. Using fluorescence-activated cell sorting, we separated subpopulations of cells with high or low caspase 3/7 activity. Protein expression related to myogenesis, muscle atrophy, protein synthesis, and apoptosis were quantified by western blotting. RESULTS In our transcriptomic analysis, we identified Cdkn1a as an upregulated gene in both the soleus and white gastrocnemius muscles of aged rats, among 36 commonly upregulated genes. The upregulation of Cdkn1a appears to be linked to mitochondrial dysfunction and cellular senescence, underscoring its significance in sarcopenia pathogenesis. C2-ceramide treatment effectively induced senescence, as evidenced by increased p21 expression, enhanced SA-β-gal staining, decreased myogenesis, and increased apoptosis. Knockdown of p21 in ceramide-treated myoblasts significantly reduced SA-β-gal-positive cells, restored cell proliferation, reduced the expression of senescence-associated cytokines (i.e., interleukin (IL)-6 and tumor necrosis factor (TNF)-α), and selectively induced apoptosis in the senescent cell population, demonstrating a senolytic effect. Notably, p21 inhibition also improved differentiation of myoblasts into myotubes, as indicated by increased myosin heavy chain expression and improvements in myotube diameter and fusion index. CONCLUSIONS Our data suggest that p21 inhibition selectively eliminates senescent cells while simultaneously enhancing the regenerative capacity of healthy myoblasts, which may combine to improve muscle regeneration and promote myogenesis, ultimately improving muscle health and function in aged individuals.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Pharmacology, Inha University College of Medicine, 100, Inha-Ro, Michuhol-Gu, Incheon, 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Bonsang Gu
- Department of Pharmacology, Inha University College of Medicine, 100, Inha-Ro, Michuhol-Gu, Incheon, 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon, 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| | - Chan-Young So
- Department of Pharmacology, Inha University College of Medicine, 100, Inha-Ro, Michuhol-Gu, Incheon, 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon, 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| | - Keren Esther Kristina Mantik
- Department of Pharmacology, Inha University College of Medicine, 100, Inha-Ro, Michuhol-Gu, Incheon, 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon, 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| | - Seung-Hyun Jung
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Integrated Research Center for Genomic Polymorphism, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Republic of Korea
| | - Sohee Moon
- Department of Pharmacology, Inha University College of Medicine, 100, Inha-Ro, Michuhol-Gu, Incheon, 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Dong-Ho Park
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
- Department of Kinesiology, Inha University, Incheon, Republic of Korea
| | - Hyo-Bum Kwak
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
- Department of Kinesiology, Inha University, Incheon, Republic of Korea
| | - Jinkyung Cho
- College of Sport Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Eun-Jeong Cho
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
- Department of Kinesiology, Inha University, Incheon, Republic of Korea
| | - Jae-Seon Lee
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon, 22212, Republic of Korea.
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea.
- Department of Molecular Medicine, Inha University College of Medicine, 100, Inha-Ro, Michuhol-Gu, Incheon, 22212, Republic of Korea.
| | - Ju-Hee Kang
- Department of Pharmacology, Inha University College of Medicine, 100, Inha-Ro, Michuhol-Gu, Incheon, 22212, Republic of Korea.
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon, 22212, Republic of Korea.
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea.
| |
Collapse
|
12
|
Shi W, Lin H, Di W, He C, Shen Y. Granulosa cell RNA-Seq insights into senescence and sphingolipid metabolism disorder in PCOS: aspirin as a potential therapeutic drug. Reprod Biol Endocrinol 2025; 23:61. [PMID: 40287692 PMCID: PMC12032776 DOI: 10.1186/s12958-025-01396-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a pivotal cause of anovulatory infertility and the pathogenesis remains elusive. Cellular senescence and sphingolipid metabolism disorder are closely intertwined, and both have been demonstrated present within the granulosa cells of PCOS, while research on the combined impact of senescence and sphingolipids on PCOS-related anovulation is scarce. METHODS Here, we leveraged four datasets of PCOS and executed differential gene expression analysis, engaged in WGCNA, and harnessed machine learning algorithms-including RF, SVM-RFE, and LASSO-to deeply explore the key genes that interact with senescence and sphingolipid metabolism in granulosa cells of PCOS. These key genes were subjected to further analysis to construct a diagnostic model, forecast immune cell infiltration, and identify potential agents. Additionally, within the testosterone-stimulated granulosa cells, we validated the expression of key genes, confirmed senescence and sphingolipids dysregulation, and evaluated the therapeutic efficacy of the candidate agent. RESULTS Our research pinpointed a set of genes (LYN, PLCG2, STAT5B, MMP9, and IL6R) that showed promise as biomarkers for PCOS-related anovulation and the diagnostic nomogram was developed. These biomarkers were linked to various immune cell types infiltration. In testosterone-stimulated granulosa cells, we observed increased expression of these biomarkers, accompanied by signs of senescence and changes in sphingolipids. Importantly, the potential agent aspirin displayed the ability to ameliorate these two processes. CONCLUSION This study highlighted the important value of genes associated with senescence and sphingolipids dysregulation in PCOS. Aspirin targeting senescence could be a promising therapeutic drug for addressing anovulation associated with PCOS.
Collapse
Affiliation(s)
- Weiwei Shi
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China
| | - Hao Lin
- Department of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Clinical Science and Research, Zhongda Hospital Affiliated to Southeast University, Nanjing, 21009, Jiangsu, China
| | - Wu Di
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China
| | - Cong He
- Key Laboratory of Innovative Applications of Bioresources and Functional Molecules of Jiangsu Province, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing, 210013, Jiangsu, China
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
13
|
Ma Y, Hu T, Liu N, Guo C, Xing L, Ma W, Cui Y, Chen X. Acupotomy Ameliorates KOA Related Chondrocyte Premature Senescence Through YAP/FOXD1 Pathway. J Pain Res 2025; 18:2011-2023. [PMID: 40241815 PMCID: PMC12002075 DOI: 10.2147/jpr.s475829] [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: 09/17/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose Premature senescence of chondrocytes is a typical lesion of knee osteoarthritis (KOA). Abnormal cartilage stress can inhibit the mechanosensitive Yes-associated protein (YAP) / transcription factor forkhead box D1 (FOXD1) pathway, which is related to premature senescence of chondrocytes, thereby accelerating the progression of the lesion. This study aims to investigate whether acupotomy intervention could inhibit the premature senescence of chondrocytes and protect the cartilage of KOA rabbits. Methods 18 male New Zealand rabbits were randomly divided into 3 groups (n = 6 each): control, KOA, and KOA + acupotomy (KOA+Apo). KOA, KOA+Apo rabbits were modeled by modified Videman's method for 6 weeks. After modeling, the KOA+Apo groups were subjected to acupotomy once a week for 3 weeks on the muscles around the left hind knee. The modified Lequesne MG score and passive range of motion (PROM) were used to evaluate the general condition and exercise ability of rabbits. Cartilage degeneration was detected by safranin O-fast green staining and transmission electron microscope(TEM). Type II collagen (Col-II) and aggrecan by immunohistochemistry (IHC), IL-7 and MMP-13 by Enzyme-Linked Immunosorbent Assay (ELISA), and p53, Rb1, p - YAP, YAP, FOXD1 by IHC, Western blot, or RT - PCR. Results Acupotomy effectively curbed cartilage degeneration and chondrocyte premature senescence in KOA rabbits. Mechanistically, it cut IL - 7 and MMP-13 levels, easing the inflammatory milieu and extracellular matrix degradation. It also regulated p53 and Rb1, controlling cell - cycle progression. Crucially, acupotomy upregulated the YAP/FOXD1 pathway, which, by affecting downstream genes, modulated IL - 7, MMP-13, p53, and Rb1 levels, acting as a pivotal molecular link in its regulatory effects. Conclusion Acupotomy may protect KOA rabbits' cartilage by inhibiting chondrocytes premature senescence via the YAP/FOXD1 pathway, offering a new theoretical basis for treating mechanically - induced KOA.
Collapse
Affiliation(s)
- Yunxuan Ma
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Tingyao Hu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Naigang Liu
- Department of Acupuncture-moxibustion, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Changqing Guo
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Longfei Xing
- Department of Traditional Chinese Medicine, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Weiwei Ma
- Department of Traditional Chinese Medicine, Wuzhong People ‘s Hospital, Ningxia, People’s Republic of China
| | - Yongqi Cui
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Xilin Chen
- Department of Acupuncture and Rehabilitation, The Fifth College of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
14
|
Shao J, Liu S, Chen C, Chen W, Zhu Z, Li L. Aging Impairs Implant Osseointegration Through a Novel Reactive Oxygen Species-Hypoxia-Inducible Factor 1α/p53 Axis. Tissue Eng Part A 2025. [PMID: 40171686 DOI: 10.1089/ten.tea.2024.0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025] Open
Abstract
Enhancing bone-vessel coupling to form high-quality vascular-rich peri-implant bone is crucial for improving implant prognosis in elder patients. Notably, hypoxia-inducible factor 1α (HIF1α) is known to promote osteogenesis-angiogenesis coupling; however, this effect remains to be investigated in aged bone owing to the dual effect of HIF1α in different aged organs. In this study, HIF1α inhibitor or activator was applied to aged mice and their bone mesenchymal stem cells (BMSCs) to investigate the effects and inner mechanism of HIF1α on the peri-implant osteogenesis and angiogenesis in senescent status. Cell senescence, along with osteogenic and angiogenic abilities of aged BMSCs, was detected, respectively. Meanwhile, a femur implant implantation model was constructed on aged mice, and the bone-vessel coupling of peri-implant bone was observed. Mandibular bone morphology was also detected to further provide evidence for clinical oral implantation. Furthermore, p53 expression was examined in vivo and in vitro following HIF1α intervention. A reactive oxygen species (ROS) scavenger was also adopted to further investigate the roles of ROS in the HIF1α-p53 axis. Results showed that the suppression of HIF1α alleviated senescence and osteogenesis-angiogenesis coupling of aged BMSCs, while its activation aggravated these effects. The mandible phenotype and bone-vessel coupling in aged peri-implant bone also changed accordingly upon regulation of HIF1α. Mechanistically, p53 changed in the same direction as HIF1α in vivo and in vitro. Moreover, the ROS scavenger reversed the HIF1α-p53 relationship and weakened the effect of HIF1α inhibitor on peri-implant bone improvement. In conclusion, in aged mice, highly expressed HIF1α impaired peri-implant bone-vessel coupling and implant osseointegration through p53, and accumulated ROS was a prerequisite for HIF1α to positively regulate p53. These findings provide new insights into the role of HIF1α and the ROS-HIF1α/p53 signaling axis, offering potential therapeutic targets to improve implant outcomes in elderly patients.
Collapse
Affiliation(s)
- Jingjing Shao
- State Key Laboratory of Oral Diseases &National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases &National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chenfeng Chen
- State Key Laboratory of Oral Diseases &National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wenchuan Chen
- State Key Laboratory of Oral Diseases &National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhimin Zhu
- State Key Laboratory of Oral Diseases &National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lei Li
- State Key Laboratory of Oral Diseases &National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Ge L, Yang Y, Xiao T, Gao Y, Chang W, Du F, Yu M, Zhang JV. Ovarian Endometriosis Accelerates Premature Ovarian Failure and Contributes to Osteoporosis and Cognitive Decline in Aging Mice. Int J Mol Sci 2025; 26:3313. [PMID: 40244208 PMCID: PMC11989598 DOI: 10.3390/ijms26073313] [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: 01/22/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Ovarian endometriosis (OEM) is a chronic inflammatory condition that impairs ovarian function. While its effects on ovarian reserve are well established, the long-term consequences of OEM on ovarian dysfunction, premature ovarian failure (POF), and systemic health, particularly in the context of accelerated aging, remain insufficiently studied. In this study, we employed an OEM mouse model and bulk RNA sequencing to investigate the underlying mechanisms. Our results show that OEM accelerates primordial follicle depletion and upregulates mTOR signaling pathway gene expression, along with mechanical stress and paracrine signaling via the Hippo and Myc pathways. OEM also induces irregular estrus and ovarian fibrosis in aging mice, decreases serum AMH levels, and increases FSH levels. Systemically, elevated serum IgG levels contribute to osteoporosis and cognitive decline, both linked to ovarian dysfunction and POF in OEM. These findings elucidate the mechanisms driving premature ovarian reserve depletion in OEM and highlight its broader systemic effects. This study emphasizes the importance of monitoring ovarian health and ectopic tissue to safeguard ovarian reserves and mitigate long-term risks such as osteoporosis and cognitive decline.
Collapse
Affiliation(s)
- Lei Ge
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Yali Yang
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Tianxia Xiao
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Yuqing Gao
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Wakam Chang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Feifei Du
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Ming Yu
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Jian V. Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen 518028, China
- Sino-European Center of Biomedicine and Health, Shenzhen 518000, China
| |
Collapse
|
16
|
Yuan Y, Li S, Yin L, Su Y, Guo Z, Liu Y, Blake GM, Wang L, Liu Y, Cheng X, Engelke K, Vlug AG. Decreased muscle strength and thigh muscle area assessed with CT imaging in older Chinese adults with long duration of type 2 diabetes. J Nutr Health Aging 2025; 29:100506. [PMID: 39954532 DOI: 10.1016/j.jnha.2025.100506] [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: 09/05/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE We aimed to compare muscle strength, physical performance, and muscle parameters in non-diabetic and type 2 diabetes mellitus (T2DM) older adults and to determine the association of the duration of diabetes with these outcomes. DESIGN A cross-sectional study. SETTING The China Action on Spine and Hip Status study (CASH). PARTICIPANTS 301 participants enrolled from a subcohort of CASH, of which 114 were diagnosed with well-controlled type 2 diabetes mellitus, MEASUREMENTS: We measured physical performance and muscle strength with the timed up-and-go test (TUG) and handgrip strength (HGS) and measured the area and density of the thigh, gluteus and trunk muscles (core muscles) using quantitative CT scans. RESULTS Participants with long-term (≥10 years) diabetes had a longer TUG (β coefficient: 0.64 [95% CI, 0.06, 1.22]; P = 0.030) and a lower HGS (-2.29 [-4.48, -0.10]; P = 0.041) as well as a lower muscle area of the thigh (-7.10 [-13.44, -0.76]; P = 0.028). The lower HGS among patients with long-term (≥10 years) diabetes was largely mediated by muscle area of the thigh (compared with non-diabetic controls: percentage mediated, 38.3%; P = 0.023; compared with <10 years diabetes: percentage mediated, 51.8%; P = 0.039). CONCLUSIONS Older adults with long-term (≥10 years) diabetes had lower TUG and HGS than either non-diabetic participants, and the difference in HGS was largely mediated by a decrease in muscle area of the thigh muscles. Our finding suggested that long duration of diabetes, even in well-controlled subjects, may be associated with poor physical functions.
Collapse
Affiliation(s)
- Yi Yuan
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing 100035, China; Sarcopenia Research Center, Beijing Research Institute of Traumatology and Orthopaedics, Beijing 100035, China
| | - Sidong Li
- Institute of Public Health Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Lu Yin
- Medical Research and Biometrics Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yongbin Su
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing 100035, China
| | - Zhe Guo
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing 100035, China
| | - Yandong Liu
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing 100035, China
| | - Glen M Blake
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Ling Wang
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing 100035, China; Sarcopenia Research Center, Beijing Research Institute of Traumatology and Orthopaedics, Beijing 100035, China.
| | - Yajun Liu
- Sarcopenia Research Center, Beijing Research Institute of Traumatology and Orthopaedics, Beijing 100035, China; Department of Spine Surgery, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing 100035, China.
| | - Xiaoguang Cheng
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing 100035, China
| | - Klaus Engelke
- Institute of Medical Physics, University Erlangen-Nürnberg, Erlangen 91054, Germany; Department of Medicine 3, FAU University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen 91054, Germany
| | - Annegreet G Vlug
- Center for Bone Quality, Department of Internal Medicine, Leiden University Medical Center, Leiden RC, 2300, the Netherlands
| |
Collapse
|
17
|
Hung YL, Sato A, Takino Y, Ishigami A, Machida S. Resistance training suppresses accumulation of senescent fibro-adipogenic progenitors and senescence-associated secretory phenotype in aging rat skeletal muscle. GeroScience 2025; 47:1669-1683. [PMID: 39298108 PMCID: PMC11979060 DOI: 10.1007/s11357-024-01338-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Accumulation of senescent cells in tissues contributes to multiple aging-related pathologies. Senescent fibro-adipogenic progenitors (FAPs) contribute to aging-related muscle atrophy. Resistance training can help to maintain skeletal muscle mass, improve mobility, and reduce certain health risks commonly associated with aging. We investigated, using rat model, the impact of resistance training on FAPs in aging skeletal muscle, which remains unclear. Twenty-two-month-old female rats were divided into sedentary and training groups. The training group rodents were trained to climb a ladder while bearing a load for 20 training sessions over 2 months, after which, the flexor hallucis longus muscles were collected and analyzed. Senescent cells were identified using a senescence-associated β-galactosidase stain and p21 immunohistochemistry (IHC), and FAPs were identified using platelet-derived growth factor receptor alpha IHC. The results indicate that resistance training in rats prevented aging-associated skeletal muscle atrophy and suppressed M2 polarization of macrophages. The number of senescent cells was significantly reduced in the 24-month-old training group, with most of them being FAPs. Conversely, the number of senescent FAPs increased significantly in the 24-month-old sedentary group compared with that in the 18-month-old sedentary group. The number of senescent FAPs in the 24-month-old training group decreased significantly. Resistance training also suppressed the senescence-associated secretory phenotype (SASP). The killer T cell-specific marker, CD8α, was elevated in the skeletal muscles of the aging rats following resistance training, indicating upregulation of recognition and elimination of senescent cells. Overall, resistance training suppressed the accumulation of senescent FAPs and acquisition of SASP in aging skeletal muscles.
Collapse
Affiliation(s)
- Yung-Li Hung
- Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-Ku, Tokyo, 102-0083, Japan
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| | - Ayami Sato
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Yuka Takino
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Shuichi Machida
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
- Institute of Health and Sports Science & Medicine, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| |
Collapse
|
18
|
Tilton M, Liao J, Kim C, Shaygani H, Potes MA, Cordova DJ, Kirkland JL, Miller KM. Tracing Cellular Senescence in Bone: Time-Dependent Changes in Osteocyte Cytoskeleton Mechanics and Morphology. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408517. [PMID: 40026102 PMCID: PMC11985287 DOI: 10.1002/smll.202408517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/22/2025] [Indexed: 03/04/2025]
Abstract
Aging-related bone loss significantly impacts the growing elderly population globally, leading to debilitating conditions such as osteoporosis. Senescent osteocytes play a crucial role in the aging process of bone. This longitudinal study examines the impact of continuous local and paracrine exposure to senescence-associated secretory phenotype (SASP) factors on biophysical and biomolecular markers in osteocytes. Significant cytoskeletal stiffening in irradiated (IR) osteocytes are found, accompanied by expansion of F-actin areas and a decline in dendritic integrity. These changes, correlating with alterations in pro-inflammatory cytokine levels and osteocyte-specific gene expression, support the reliability of biophysical markers for identifying senescent osteocytes. Notably, local accumulation of SASP factors have a more pronounced impact on osteocyte biophysical properties than paracrine effects, suggesting that the interplay between local and paracrine exposure can substantially influence cellular aging. This study underscores the importance of osteocyte mechanical and morphological properties as biophysical markers of senescence, highlighting their time dependence and differential effects of local and paracrine SASP exposure. Collectively, the investigation into biophysical senescence markers offers unique and reliable functional hallmarks for the non-invasive identification of senescent osteocytes, providing insights that can inform therapeutic strategies to mitigate aging-related bone loss.
Collapse
Affiliation(s)
- Maryam Tilton
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Junhan Liao
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Chanul Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hossein Shaygani
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Maria Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Domenic J. Cordova
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - James L. Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kyle M. Miller
- Department of Radiation Oncology Emory University School of Medicine Atlanta, GA 30307, USA
| |
Collapse
|
19
|
Rana KS, Marwah MK, Raja FNS, Dias I, Hindalekar YS, Al Tahan MA, Brown JE, Bellary S. The influence of senescent associated secretory phenotype on glucose homeostasis in C2C12 muscle cells: insights into potential p38 inhibitor interventions. J Recept Signal Transduct Res 2025; 45:118-127. [PMID: 40051308 DOI: 10.1080/10799893.2025.2475441] [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: 11/13/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025]
Abstract
Increased accumulation of senescent cells with aging is associated with reduced ability of insulin-target tissues to utilize glucose, resulting in increased insulin resistance and glucotoxicity. We investigated the role of the senescent-associated secretory phenotype (SASP) within C2C12, skeletal muscle cells on glucose homeostasis and if such effects could be reduced by blocking pro-inflammatory pathways. C2C12 myotubes were treated with 40% conditioned media from senescent fibroblasts. Indirect glucose uptake and glycogen content were measured. The effect of SASP on the generation of reactive oxygen species [1] and mitochondrial function was also measured. The experiments above were repeated with a p38 inhibitor. 40% SASP treatment significantly decreased glucose utilization and glycogen storage within myotubes (p < 0.0001). 40% SASP was successful in inducing oxidative stress and increased mitochondrial density, whilst reducing membrane potential following 48 h of incubation (p < 0.0001) and blocking NF-κβ, restored glucose utilization (p < 0.01) despite the presence of SASP. Co-incubation of 40% SASP with an NF-κβ inhibitor eliminates excessive ROS production and restores mitochondrial activity to levels comparable to control treatment (p < 0.0001). This study shows changes in glucose homeostasis in senescent cells is mediated through SASP, and interventions aimed at mitigating pro-inflammatory pathways can potentially improve insulin resistance.
Collapse
Affiliation(s)
- Karan S Rana
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| | - Mandeep K Marwah
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Farah N S Raja
- School of Engineering and Physical Science, Aston University, Birmingham, UK
| | - Irundika Dias
- School of Engineering and Physical Science, Aston University, Birmingham, UK
| | | | - Mohamad Anas Al Tahan
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - James E Brown
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| | - Srikanth Bellary
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| |
Collapse
|
20
|
Li GH, Li YH, Yu Q, Zhou QQ, Zhang RF, Weng CJ, Ge MX, Kong QP. Unraveling the metabolic heterogeneity and commonality in senescent cells using systems modeling. LIFE MEDICINE 2025; 4:lnaf003. [PMID: 40224297 PMCID: PMC11992571 DOI: 10.1093/lifemedi/lnaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/16/2025] [Indexed: 04/15/2025]
Abstract
Cellular senescence is a key contributor to aging and aging-related diseases, but its metabolic profiles are not well understood. Here, we performed a systematic analysis of the metabolic features of four types of cellular senescence (replication, irradiation, reactive oxygen species [ROS], and oncogene) in 12 cell lines using genome-wide metabolic modeling and meta-analysis. We discovered that replicative and ROS-induced senescence share a common metabolic signature, marked by decreased lipid metabolism and downregulated mevalonate pathway, while irradiation and oncogene-induced senescence exhibit more heterogeneity and divergence. Our genome-wide knockout simulations showed that enhancing the mevalonate pathway, by administrating mevalonate for instance, could reverse the metabolic alterations associated with senescence and human tissue aging, suggesting a potential anti-aging or lifespan-extending effect. Indeed, the experiment in Caenorhabditis elegans showed that administrating mevalonate significantly increased the lifespan. Our study provides a new insight into the metabolic landscape of cell senescence and identifies potential targets for anti-aging interventions.
Collapse
Affiliation(s)
- Gong-Hua Li
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Yu-Hong Li
- College of Biological Resources and Food Engineering, Qujing Normal University, Qujing 655000, China
| | - Qin Yu
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Qing-Qing Zhou
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Run-Feng Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Chong-Jun Weng
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Ming-Xia Ge
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
21
|
Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev 2025; 105:593-650. [PMID: 39324871 DOI: 10.1152/physrev.00044.2023] [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: 12/04/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
This review addresses oxidative stress and redox signaling in the pancreas under healthy physiological conditions as well as in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. Physiological redox homeodynamics is maintained mainly by NRF2/KEAP1, NF-κB, protein tyrosine phosphatases, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), and normal autophagy. Depletion of reduced glutathione (GSH) in the pancreas is a hallmark of acute pancreatitis and is initially accompanied by disulfide stress, which is characterized by protein cysteinylation without increased glutathione oxidation. A cross talk between oxidative stress, MAPKs, and NF-κB amplifies the inflammatory cascade, with PP2A and PGC1α as key redox regulatory nodes. In acute pancreatitis, nitration of cystathionine-β synthase causes blockade of the transsulfuration pathway leading to increased homocysteine levels, whereas p53 triggers necroptosis in the pancreas through downregulation of sulfiredoxin, PGC1α, and peroxiredoxin 3. Chronic pancreatitis exhibits oxidative distress mediated by NADPH oxidase 1 and/or CYP2E1, which promotes cell death, fibrosis, and inflammation. Oxidative stress cooperates with mutant KRAS to initiate and promote pancreatic adenocarcinoma. Mutant KRAS increases mitochondrial reactive oxygen species (ROS), which trigger acinar-to-ductal metaplasia and progression to pancreatic intraepithelial neoplasia (PanIN). ROS are maintained at a sufficient level to promote cell proliferation, while avoiding cell death or senescence through formation of NADPH and GSH and activation of NRF2, HIF-1/2α, and CREB. Redox signaling also plays a fundamental role in differentiation, proliferation, and insulin secretion of β-cells. However, ROS overproduction promotes β-cell dysfunction and apoptosis in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Hospital Clínico, Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
22
|
Wang T, Chen J, Qu B, Zhou D, Hong Z. Scutellarin Alleviates Bone Marrow Mesenchymal Stromal Cellular Senescence via the Ezh2-Nrf2 Signalling Axis in Diabetes-Induced Bone Loss. Cell Prolif 2025; 58:e13790. [PMID: 39668494 PMCID: PMC11969241 DOI: 10.1111/cpr.13790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/08/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024] Open
Abstract
Currently, there is no specific treatment for diabetes-induced osteoporosis (DOP). Our study identified diabetes-induced cellular senescence, marked by elevated activity of senescence-associated β-galactosidase. Targeting senescent cells holds promise for osteoporosis treatment. We demonstrated that scutellarin (SCU) effectively mitigated bone loss in DOP mice, and co-treatment with SCU significantly reduced diabetes-induced senescence in LepR+MSCs. Furthermore, our research highlighted the role of Nrf2 in SCU's anti-senescence effects on bone. The deletion of Nrf2 impaired SCU's ability to alleviate DOP. Mechanistically, SCU enhances Ezh2 expression and increases H3K27me3 activity at the Keap1 promoter region, leading to Keap1 repression and enhanced Nrf2-ARE signalling. Additionally, SCU notably inhibited cellular senescence and diabetes-related osteoporosis, these effects were significantly reduced in Ezh2LepRcre conditional knockout models. These findings suggest that the Ezh2-Nrf2 signalling axis is crucial for mediating SCU's beneficial effects in this context. Overall, our discoveries provide insights into the mechanisms underlying DOP and propose a potential preventive strategy for this condition.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyInstitute of Neurology and Disease, West China Hospital of Sichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology of West China Hospital, Sichuan UniversityChengduChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduChina
| | - Jiehao Chen
- Animal Laboratory Center, West China Hospital, Sichuan UniversityChengduChina
| | - Bo Qu
- Department of OrthopedicsThe First Affiliated Hospital of Chengdu Medical CollegeChengduChina
| | - Dong Zhou
- Department of NeurologyInstitute of Neurology and Disease, West China Hospital of Sichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology of West China Hospital, Sichuan UniversityChengduChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduChina
| | - Zhen Hong
- Department of NeurologyInstitute of Neurology and Disease, West China Hospital of Sichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology of West China Hospital, Sichuan UniversityChengduChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduChina
| |
Collapse
|
23
|
Li B, Ming H, Qin S, Nice EC, Dong J, Du Z, Huang C. Redox regulation: mechanisms, biology and therapeutic targets in diseases. Signal Transduct Target Ther 2025; 10:72. [PMID: 40050273 PMCID: PMC11885647 DOI: 10.1038/s41392-024-02095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/21/2024] [Indexed: 03/09/2025] Open
Abstract
Redox signaling acts as a critical mediator in the dynamic interactions between organisms and their external environment, profoundly influencing both the onset and progression of various diseases. Under physiological conditions, oxidative free radicals generated by the mitochondrial oxidative respiratory chain, endoplasmic reticulum, and NADPH oxidases can be effectively neutralized by NRF2-mediated antioxidant responses. These responses elevate the synthesis of superoxide dismutase (SOD), catalase, as well as key molecules like nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), thereby maintaining cellular redox homeostasis. Disruption of this finely tuned equilibrium is closely linked to the pathogenesis of a wide range of diseases. Recent advances have broadened our understanding of the molecular mechanisms underpinning this dysregulation, highlighting the pivotal roles of genomic instability, epigenetic modifications, protein degradation, and metabolic reprogramming. These findings provide a foundation for exploring redox regulation as a mechanistic basis for improving therapeutic strategies. While antioxidant-based therapies have shown early promise in conditions where oxidative stress plays a primary pathological role, their efficacy in diseases characterized by complex, multifactorial etiologies remains controversial. A deeper, context-specific understanding of redox signaling, particularly the roles of redox-sensitive proteins, is critical for designing targeted therapies aimed at re-establishing redox balance. Emerging small molecule inhibitors that target specific cysteine residues in redox-sensitive proteins have demonstrated promising preclinical outcomes, setting the stage for forthcoming clinical trials. In this review, we summarize our current understanding of the intricate relationship between oxidative stress and disease pathogenesis and also discuss how these insights can be leveraged to optimize therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Siyuan Qin
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jingsi Dong
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhongyan Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China.
| |
Collapse
|
24
|
Dong J, Xu L, Qu A, Hao C, Sun M, Xu C, Hu S, Kuang H. Chiral Inorganic Nanomaterial-Based Diagnosis and Treatments for Neurodegenerative Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2418723. [PMID: 39924754 DOI: 10.1002/adma.202418723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/27/2025] [Indexed: 02/11/2025]
Abstract
Chiral nanomaterials are widely investigated over recent decades due to their biocompatibility and unique chiral effects. These key properties have significantly promoted the rapid development of chiral nanomaterials in bioengineering and medicine. In this review, the basic principles of constructing chiral nanomaterials along with the latest progress in research are comprehensively summarized. Then, the application of chiral nanomaterials for the diagnosis of neurodegenerative diseases (NDDs) is systematically described. In addition, the significant potential and broad prospects of chiral nanomaterials in the treatment of NDDs are highlighted from several aspects, including the disaggregation of neurofibrils, the scavenging of reactive oxygen species, regulation of the microbial-gut-brain axis, the elimination of senescent cells, and the promotion of directed differentiation in neural stem cells. Finally, a perspective of the challenges and future development of chiral nanomaterials for the treatment of NDDs is provided.
Collapse
Affiliation(s)
- Jingqi Dong
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aihua Qu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Changlong Hao
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Shudong Hu
- Department of Radiology, Affiliated Hospital, Jiangnan University, No. 1000, Hefeng Road, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
25
|
Melis S, Trompet D, Chagin AS, Maes C. Skeletal stem and progenitor cells in bone physiology, ageing and disease. Nat Rev Endocrinol 2025; 21:135-153. [PMID: 39379711 DOI: 10.1038/s41574-024-01039-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 10/10/2024]
Abstract
Skeletal stem cells (SSCs) and related progenitors with osteogenic potential, collectively termed skeletal stem and/or progenitor cells (SSPCs), are crucial for providing osteoblasts for bone formation during homeostatic tissue turnover and fracture repair. Besides mediating normal bone physiology, they also have important roles in various metabolic bone diseases, including osteoporosis. SSPCs are of tremendous interest because they represent prime future targets for osteoanabolic therapies and bone regenerative medicine. Remarkable progress has been made in characterizing various SSC and SSPC populations in postnatal bone. SSPCs exist in the periosteum and within the bone marrow stroma, including subsets localizing around arteriolar and sinusoidal blood vessels; they can display osteogenic, chondrogenic, adipogenic and/or fibroblastic potential, and exert critical haematopoiesis-supportive functions. However, much remains to be clarified. By the current markers, bona fide SSCs are commonly contained within broader SSPC populations characterized by considerable heterogeneity and overlap, whose common versus specific functions in health and disease have not been fully unravelled. Here, we review the present knowledge of the identity, fates and relationships of SSPC populations in the postnatal bone environment, their contributions to bone maintenance, the changes observed upon ageing, and the effect of metabolic diseases such as osteoporosis and diabetes mellitus.
Collapse
Affiliation(s)
- Seppe Melis
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Andrei S Chagin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| |
Collapse
|
26
|
Lue JC, Radisky DC. From Embryogenesis to Senescence: The Role of Mammary Gland Physiology in Breast Cancer Risk. Cancers (Basel) 2025; 17:787. [PMID: 40075637 PMCID: PMC11898936 DOI: 10.3390/cancers17050787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/16/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
The mammary gland undergoes significant changes throughout a woman's life; from embryonic development to transformations after breastfeeding and during aging. These processes, while essential for normal breast physiology, can increase breast cancer risk when disrupted. This review explores three critical stages: embryonic development; postlactational involution; and age-related lobular involution (ARLI). We highlight key signaling pathways-Wnt, FGF, SHH, Notch, EGFR, and BMP-that guide embryonic development and discuss how their dysregulation can contribute to abnormal growth. For postlactational involution, we examine the two-phase process of cell death and tissue remodeling, showing how disruptions during this period, particularly postpartum, may foster a tumor-promoting environment. We also delve into ARLI and the role of cellular senescence in the aging mammary gland, focusing on the senescence-associated secretory phenotype (SASP) and its impact on inflammation and tissue remodeling. Understanding these processes provides new opportunities for breast cancer prevention and treatment strategies.
Collapse
Affiliation(s)
- Jaida C. Lue
- Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Derek C. Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
27
|
Armanville S, Tocco C, Haj Mohamad Z, Clarke D, Robitaille R, Drouin-Ouellet J. Chemically Induced Senescence Prompts Functional Changes in Human Microglia-Like Cells. J Immunol Res 2025; 2025:3214633. [PMID: 40041406 PMCID: PMC11876530 DOI: 10.1155/jimr/3214633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
In response to various stressors, cells can enter a state called cellular senescence which is characterized by irreversible cell cycle arrest and a senescence-associated secretory phenotype (SASP). The progressive accumulation of senescent glial cells in the central nervous system (CNS) with aging suggests a potential role for senescence as driver of aging and inflammation in the brain. As the main immune cell population residing in the CNS, microglia are thought to play a pivotal role in the progression of age-associated neuroinflammation. Furthermore, due to their slow turnover, microglia are highly susceptible to undergoing cellular senescence. However, current understanding of age-related changes in microglia and their impact on brain aging is limited. Due to the challenge in accessing human primary microglia and the lack of models to adequately recapitulate aging, this knowledge is predominantly limited to rodent studies. Here, we chemically induced senescence in a human immortalized microglia cell line with a cocktail of senescence-inducing molecules. We demonstrate that chemically induced senescent microglia adopt a proinflammatory phenotype, have reduced phagocytic activity, and impaired calcium activity. Our results show that chemically induced senescence can mimic features of cellular aging and can provide insight into the impact of aging and cellular senescence on human microglia.
Collapse
Affiliation(s)
- S. Armanville
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - C. Tocco
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - Z. Haj Mohamad
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - D. Clarke
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
| | - R. Robitaille
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
- Research Group on Neural Signalling and Circuits (SNC), University of Montreal, Montreal, Quebec, Canada
- Center for Interdisciplinary Research on Brain and Learning (CIRCA), Montreal, Quebec, Canada
| | - J. Drouin-Ouellet
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
- Research Group on Neural Signalling and Circuits (SNC), University of Montreal, Montreal, Quebec, Canada
- Center for Interdisciplinary Research on Brain and Learning (CIRCA), Montreal, Quebec, Canada
| |
Collapse
|
28
|
Bi Y, Qiao X, Cai Z, Zhao H, Ye R, Liu Q, Gao L, Liu Y, Liang B, Liu Y, Zhang Y, Yang Z, Wu Y, Wang H, Jia W, Zeng C, Jia C, Wu H, Xue Y, Ji G. Exosomal miR-302b rejuvenates aging mice by reversing the proliferative arrest of senescent cells. Cell Metab 2025; 37:527-541.e6. [PMID: 39818209 DOI: 10.1016/j.cmet.2024.11.013] [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: 12/07/2023] [Revised: 09/09/2024] [Accepted: 11/25/2024] [Indexed: 01/18/2025]
Abstract
Cellular senescence, a hallmark of aging, involves a stable exit from the cell cycle. Senescent cells (SnCs) are closely associated with aging and aging-related disorders, making them potential targets for anti-aging interventions. In this study, we demonstrated that human embryonic stem cell-derived exosomes (hESC-Exos) reversed senescence by restoring the proliferative capacity of SnCs in vitro. In aging mice, hESC-Exos treatment remodeled the proliferative landscape of SnCs, leading to rejuvenation, as evidenced by extended lifespan, improved physical performance, and reduced aging markers. Ago2 Clip-seq analysis identified miR-302b enriched in hESC-Exos that specifically targeted the cell cycle inhibitors Cdkn1a and Ccng2. Furthermore, miR-302b treatment reversed the proliferative arrest of SnCs in vivo, resulting in rejuvenation without safety concerns over a 24-month observation period. These findings demonstrate that exosomal miR-302b has the potential to reverse cellular senescence, offering a promising approach to mitigate senescence-related pathologies and aging.
Collapse
Affiliation(s)
- Youkun Bi
- Henan Academy of Sciences, Zhengzhou 450000, China; Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xinlong Qiao
- Yuan Sheng Biotechnology Ltd., Qingdao 266109, China
| | - Zhaokui Cai
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hailian Zhao
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Rong Ye
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qun Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin Gao
- Yuan Sheng Biotechnology Ltd., Qingdao 266109, China
| | - Yingqi Liu
- Yuan Sheng Biotechnology Ltd., Qingdao 266109, China
| | - Bo Liang
- Henan Academy of Sciences, Zhengzhou 450000, China
| | - Yixuan Liu
- Henan Academy of Sciences, Zhengzhou 450000, China
| | - Yaning Zhang
- Henan Academy of Sciences, Zhengzhou 450000, China
| | - Zhiguang Yang
- Yuan Sheng Biotechnology Ltd., Qingdao 266109, China
| | - Yanyun Wu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Huiwen Wang
- Henan Academy of Sciences, Zhengzhou 450000, China; Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Jia
- Biomedical Institute of TaishengKangyuan Ltd., Beijing 100103, China
| | | | - Ce Jia
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongjin Wu
- Boao International Hospital, Shanghai University of Traditional Chinese Medicine, Hainan 571434, China.
| | - Yuanchao Xue
- Henan Academy of Sciences, Zhengzhou 450000, China; Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Guangju Ji
- Henan Academy of Sciences, Zhengzhou 450000, China; Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
29
|
Carver CM, Rodriguez SL, Atkinson EJ, Dosch AJ, Asmussen NC, Gomez PT, Leitschuh EA, Espindola-Netto JM, Jeganathan KB, Whaley MG, Kamenecka TM, Baker DJ, Haak AJ, LeBrasseur NK, Schafer MJ. IL-23R is a senescence-linked circulating and tissue biomarker of aging. NATURE AGING 2025; 5:291-305. [PMID: 39658621 PMCID: PMC11839461 DOI: 10.1038/s43587-024-00752-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/17/2024] [Indexed: 12/12/2024]
Abstract
Cellular senescence is an aging mechanism characterized by cell cycle arrest and a senescence-associated secretory phenotype (SASP). Preclinical studies demonstrate that senolytic drugs, which target survival pathways in senescent cells, can counteract age-associated conditions that span several organs. The comparative efficacy of distinct senolytic drugs for modifying aging and senescence biomarkers in vivo has not been demonstrated. Here, we established aging- and senescence-related plasma proteins and tissue transcripts that changed in old versus young female and male mice. We investigated responsivity to acute treatment with venetoclax, navitoclax, fisetin or luteolin versus transgenic senescent cell clearance in aged p16-InkAttac mice. We discovered that age-dependent changes in plasma proteins, including IL-23R, CCL5 and CA13, were reversed by senotherapeutics, which corresponded to expression differences in tissues, particularly in the kidney. In plasma from humans across the lifespan, IL-23R increased with age. Our results reveal circulating factors as candidate mediators of senescence-associated interorgan signal transduction and translationally impactful biomarkers of systemic senescent cell burden.
Collapse
Affiliation(s)
- Chase M Carver
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Sonia L Rodriguez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth J Atkinson
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Andrew J Dosch
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Niels C Asmussen
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Paul T Gomez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Ethan A Leitschuh
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Jair M Espindola-Netto
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Karthik B Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Madison G Whaley
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, UF Scripps Institute, The Scripps Research Institute, Scripps Florida, Jupiter, FL, USA
| | - Darren J Baker
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Nathan K LeBrasseur
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
- Department of Neuroscience, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
30
|
Sheng ZH, Gong XY, Huang PP, Xu QY, Zhang WJ, Sun FB, Song KY, Zeng DC. An Innovative Anoikis Signature With Inflammatory Infiltrates in Osteoarthritis. Int J Rheum Dis 2025; 28:e70093. [PMID: 39895467 DOI: 10.1111/1756-185x.70093] [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: 09/25/2024] [Revised: 12/15/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
AIM To explore the relationship between an innovative anoikis-related gene signature and inflammatory infiltrates in patients with osteoarthritis. METHODS Gene expression profiles (GSM1248759 and GSE200843) were curated from the Gene Expression Omnibus database, followed by the construction of a protein-protein interaction network. Functional and genomic enrichment analyses were conducted using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. The CIBERSORT method was employed to investigate immune cell infiltration differences between osteoarthritic and control tissues. Additionally, the ConsensusClusterPlus package in R software was utilized to identify distinct anoikis patterns (Cluster C1 and Cluster C2) and conduct molecular biological investigations. RESULTS Analysis revealed two distinct anoikis patterns (Cluster C1 and Cluster C2), with Cluster C2 patients exhibiting varying immune cell levels compared to Cluster C1 patients. Molecular investigations identified 84 DEGs enriched in specific pathways such as adipocytokine signaling, cytokine-cytokine receptor interaction, ECM-receptor interaction, and the PPAR signaling pathway. qPCR experiments confirmed the elevated expression levels of specific genes, including SOD2, MAPK14, CEACM3, LAMB3, COL13A1, TLR3, NOTCH3, and KLF12, in the IL-1β-induced group compared with the osteoarthritis group. CONCLUSION This study highlights the role of anoikis-related genes and immune infiltration differences in osteoarthritis, enhancing our understanding of its development.
Collapse
Affiliation(s)
- Ze-Hao Sheng
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xin-Yi Gong
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Peng-Peng Huang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qi-Yu Xu
- Department of Rehabilitation Medicine, Anhui no.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Wen-Jie Zhang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feng-Bao Sun
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kai-Yi Song
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Du-Chun Zeng
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
31
|
Cho KH, Bahuguna A, Kim JE, Lee SH, Lee Y, Jeon C. A Comparative Effect of 12-Week Dietary Intervention of Policosanol (Raydel ®) and Red Yeast Rice (RYR, Kobayashi) in Managing Dyslipidemia and Organ Damage in Hyperlipidemic Zebrafish. Pharmaceuticals (Basel) 2025; 18:200. [PMID: 40006014 PMCID: PMC11859080 DOI: 10.3390/ph18020200] [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: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND A comparative 12-week dietary intervention of red yeast rice (RYR, Beni-koji, Kobayashi, Japan) and Cuban policosanol (PCO, Raydel®, Thornleigh, Australia) was assessed for dyslipidemia, antioxidant status, and vital organ functionality in hyperlipidemic zebrafish. METHODS Hyperlipidemic zebrafish were supplemented with a high-cholesterol diet (HC, final 4%, w/w) infused with either a powdered RYR tablet (final 1.0%, w/w), a PCO tablet (final 1.0%, w/w), or a combination of 0.5% (w/w) each of RYR and PCO powder for 12 weeks. Subsequently, blood and organs were collected and processed for biochemical and histological examination. RESULTS RYR and PCO consumption showed a substantial effect against HC-induced hyperlipidemia by reducing the total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C). Exclusively, PCO supplementation revealed a significant elevation in the HC-diminished high-density lipoprotein cholesterol (HDL-C). In addition, PCO supplementation showed a significant elevation in plasma ferric ion-reducing ability (FRA) and sulfhydryl content, as well as alleviating the blood glucose level of hyperlipidemic zebrafish. The most noteworthy impact, with a significant two-fold (p < 0.001) augmentation of HC-diminished plasma paraoxonase (PON) activity, was observed in response to PCO. In contrast, the RYR supplementation failed to establish curative effects against HC-disturbed plasma antioxidant variables and blood glucose levels. The histological outcome revealed a severe toxicological impact of the RYR on the liver, reflected by fatty liver changes and three-fold heightened IL-6 production compared to HC control. Contrastingly, PCO exhibited significant hepatoprotection and effectively neutralized the hepatic toxicity triggered by HC and RYR. Also, RYR showed kidney atrophy, intense ROS generation, apoptosis, and senescence. Conversely, the PCO supplementation protected the kidney from HC- and RYR-induced toxicity. Likewise, PCO supplementation notably alleviated histological alterations and oxidative stress in the brain, ovary, and testis of hyperlipidemic zebrafish. CONCLUSIONS This comparative study establishes PCO's therapeutic effect against the challenges posed by HC, while RYR emerged with serious toxicological concerns towards the liver, kidney, and other organs of hyperlipidemic zebrafish.
Collapse
Affiliation(s)
- Kyung-Hyun Cho
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
| | | | | | | | | | | |
Collapse
|
32
|
Sarad K, Jankowska U, Skupien-Rabian B, Babler A, Kramann R, Dulak J, Jaźwa-Kusior A. Senescence of endothelial cells promotes phenotypic changes in adventitial fibroblasts: possible implications for vascular aging. Mol Cell Biochem 2025; 480:1027-1043. [PMID: 38743322 PMCID: PMC11835997 DOI: 10.1007/s11010-024-05028-7] [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: 09/29/2023] [Accepted: 05/04/2024] [Indexed: 05/16/2024]
Abstract
Aging is the most important risk factor for the development of cardiovascular diseases. Senescent cells release plethora of factors commonly known as the senescence-associated secretory phenotype, which can modulate the normal function of the vascular wall. It is currently not well understood if and how endothelial cell senescence can affect adventitial niche. The aim of this study was to characterize oxidative stress-induced endothelial cells senescence and identify their paracrine effects on the primary cell type of the adventitia, the fibroblasts. Human aortic endothelial cells (HAEC) were treated with hydrogen peroxide to induce premature senescence. Mass spectrometry analysis identified several proteomic changes in senescent HAEC with top upregulated secretory protein growth differentiation factor 15 (GDF-15). Treatment of the human adventitial fibroblast cell line (hAdv cells) with conditioned medium (CM) from senescent HAEC resulted in alterations in the proteome of hAdv cells identified in mass spectrometry analysis. Majority of differentially expressed proteins in hAdv cells treated with CM from senescent HAEC were involved in the uptake and metabolism of lipoproteins, mitophagy and ferroptosis. We next analyzed if some of these changes and pathways might be regulated by GDF-15. We found that recombinant GDF-15 affected some ferroptosis-related factors (e.g. ferritin) and decreased oxidative stress in the analyzed adventitial fibroblast cell line, but it had no effect on erastin-induced cell death. Contrary, silencing of GDF-15 in hAdv cells was protective against this ferroptotic stimuli. Our findings can be of importance for potential therapeutic strategies targeting cell senescence or ferroptosis to alleviate vascular diseases.
Collapse
Affiliation(s)
- Katarzyna Sarad
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa Str. 7, 30-387, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Urszula Jankowska
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Kraków, Poland
| | - Bozena Skupien-Rabian
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Kraków, Poland
| | - Anne Babler
- Department for Renal and Hypertensive Diseases, Rheumatological and Immunological Diseases, RWTH Aachen University, Aachen, Germany
| | - Rafael Kramann
- Department for Renal and Hypertensive Diseases, Rheumatological and Immunological Diseases, RWTH Aachen University, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa Str. 7, 30-387, Krakow, Poland
| | - Agnieszka Jaźwa-Kusior
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa Str. 7, 30-387, Krakow, Poland.
| |
Collapse
|
33
|
Tilton M, Liao J, Kim C, Shaygani H, Potes MA, Cordova D, Kirkland JL, Miller KM. Tracing Cellular Senescence in Bone: Time-Dependent Changes in Osteocyte Cytoskeleton Mechanics and Morphology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.28.615585. [PMID: 39896626 PMCID: PMC11785097 DOI: 10.1101/2024.09.28.615585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Aging-related bone loss significantly impacts the growing elderly population globally, leading to debilitating conditions such as osteoporosis. Senescent osteocytes play a crucial role in the aging process of bone. This longitudinal study examines the impact of continuous local and paracrine exposure to senescence-associated secretory phenotype (SASP) factors on senescence-associated biophysical and biomolecular markers in osteocytes. We found significant cytoskeletal stiffening in irradiated osteocytes, accompanied by expansion of F-actin areas and a decline in dendritic integrity. These changes, correlating with alterations in pro-inflammatory cytokine levels and osteocyte-specific gene expression, support the reliability of biophysical markers for identifying senescent osteocytes. Notably, local accumulation of SASP factors had a more pronounced impact on osteocyte properties than paracrine effects, suggesting that the interplay between local and paracrine exposure could substantially influence cellular aging. This study underscores the importance of osteocyte mechanical and morphological properties as biophysical markers of senescence, highlighting their time-dependence and differential effects of local and paracrine SASP exposure. Collectively, our investigation into biophysical senescence markers offer unique and reliable functional hallmarks for non-invasive identification of senescent osteocytes, providing insights that could inform therapeutic strategies to mitigate aging-related bone loss.
Collapse
Affiliation(s)
- Maryam Tilton
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Junhan Liao
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Chanul Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hossein Shaygani
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Maria Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Domenic Cordova
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - James L. Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kyle M. Miller
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
34
|
Zheng Y, Kou J, Gao X, Guo J, Liu Q, Ren H, Gao T, Wang Q, Zhao Y, Wang Y, Li H, Yang L. Berberine Inhibited SASP-Related Inflammation through RXRα/PPARγ/NEDD4 Pathway in Atherosclerosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:251-283. [PMID: 39829230 DOI: 10.1142/s0192415x25500107] [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: 01/22/2025]
Abstract
The accumulation of aging cells significantly contributes to chronic inflammatory diseases such as atherosclerosis. Human carotid artery single-cell sequencing has shown that large numbers of aging foam cells are present in the plaques of human patients. Berberine (BBR) has been shown to inhibit cell senescence, however, the mechanisms involved in its treatment of atherosclerotic senescence have not yet been determined. Changes in plaque morphology and blood chemistry were observed in ApoE[Formula: see text] mice fed with a high-fat diet before and after BBR treatment. Inflammatory proteins linked to the senescence-associated secretory phenotypes (SASP) were detected in RAW264.7 and peritoneal macrophage-derived foam cells. Smart-seq analysis was used to explore the pathways associated with BBR therapy for atherosclerosis. Finally, the effect of lentivirus-mediated knockdown of RXRα in macrophages in plaques on atherosclerosis treatment with BBR was determined. We found that BBR reduced inflammation linked to SASP in atherosclerosis through the RXRα/PPARγ/NEDD4 signaling pathway. BBR increased GATA4 binding to p62, promoted ubiquitination, and inhibited SASP-associated protein production in RAW264.7 and peritoneal macrophage-derived foam cells. Mechanistically, according to the Smart-seq results, BBR activated RXRα and PPARγ, synergistically increased NEDD4 transcription levels, and promoted ubiquitination-mediated degradation of the GATA4/p62 complex. Additionally, the anti-aging impact of BBR on atherosclerosis was negated when macrophage-specific RXRα was knocked down using lentivirus (pLVCD68-shRNA RXRα) in ApoE[Formula: see text] mice. BBR activated PPARγ through RXRα-PPARγ immune complex in macrophage-derived foam cells, increased NEDD4 transcriptional activity, promoted ubiquitination of GATA4-p62 complex, and inhibited SASP-related inflammation. These findings suggest the potential of BBR as a novel approach to addressing SASP-associated inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Yinghong Zheng
- Department of Pharmacology, Tianjin Medical University 22 Qixiangtai Road, Heping District, Tianjin 300070, P. R. China
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Jiayuan Kou
- Department of Biochemistry and Molecular Biology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Xi Gao
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Jinxiang Guo
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Qian Liu
- Department of Pharmacology, Tianjin Medical University 22 Qixiangtai Road, Heping District, Tianjin 300070, P. R. China
| | - Huiwen Ren
- Department of Pharmacology, Tianjin Medical University 22 Qixiangtai Road, Heping District, Tianjin 300070, P. R. China
| | - Tielei Gao
- Department of Forensic Medicine, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Qianbing Wang
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Yajie Zhao
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Yuqin Wang
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Hong Li
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University 157 Baojian Road, Nangang District, Harbin 150081, P. R. China
- Department of Cardiology The Second Affiliated Hospital of Harbin Medical University Harbin, P. R. China
| |
Collapse
|
35
|
Shin CH, Rossi M, Mazan-Mamczarz K, Martindale JL, Munk R, Pal A, Piao Y, Fan J, De S, Abdelmohsen K, Gorospe M. Loss of HNRNPK During Cell Senescence Linked to Reduced Production of CDC20. Mol Cell Biol 2025; 45:129-141. [PMID: 39804141 DOI: 10.1080/10985549.2024.2443590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 03/09/2025] Open
Abstract
Cellular senescence is a complex biological response to sublethal damage. The RNA-binding protein HNRNPK was previously found to decrease prominently during senescence in human diploid fibroblasts. Here, analysis of the mechanisms leading to reduced HNRNPK abundance revealed that in cells undergoing senescence, HNRNPK mRNA levels declined transcriptionally and full-length HNRNPK protein was progressively lost, while the abundance of a truncated HNRNPK increased. The ensuing loss of full-length HNRNPK enhanced cell cycle arrest along with increased DNA damage. Analysis of the RNAs enriched after HNRNPK ribonucleoprotein immunoprecipitation (RIP) revealed a prominent target of HNRNPK, CDC20 mRNA, encoding a protein critical for progression through the G2/M phase of the cell division cycle. Silencing HNRNPK markedly decreased the levels of CDC20 mRNA via reduced transcription and stability of CDC20 mRNA, leading to lower CDC20 protein levels; conversely, overexpressing HNRNPK increased CDC20 production. Depletion of either HNRNPK or CDC20 impaired cell proliferation, with a concomitant reduction in the levels of CDK1, a key kinase for progression through G2/M. Given that overexpressing CDC20 in HNRNPK-silenced cells partly alleviated growth arrest, we propose that the reduction in HNRNPK levels in senescent cells contributed to inhibiting proliferation at least in part by suppressing CDC20 production.
Collapse
Affiliation(s)
- Chang Hoon Shin
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Martina Rossi
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Apala Pal
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Jinshui Fan
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Zhang F, Li W. Association between the Fatty Liver Index, Metabolic Dysfunction-Associated Steatotic Liver Disease, and the Risk of Kidney Stones. Kidney Blood Press Res 2025; 50:115-130. [PMID: 39746337 PMCID: PMC11844708 DOI: 10.1159/000543404] [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: 08/21/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025] Open
Abstract
INTRODUCTION This study aimed to investigate the potential association between the fatty liver index (FLI), metabolic dysfunction-associated steatotic liver disease (MASLD), and the risk of kidney stones using large-scale population-based data. METHODS This study employed a cross-sectional design, utilizing data from the 2007 to 2018 National Health and Nutrition Examination Survey (NHANES) database. A total of 24,342 participants were enrolled in the study, and fatty liver status was assessed by calculating the FLI. MASLD was diagnosed by FLI in conjunction with cardiometabolic criteria. Data on the history of kidney stones were obtained by self-report. We employed logistic regression models to analyze the association between FLI, MASLD, and kidney stone risk and constructed multivariable adjustment models to control for potential confounders. Furthermore, we used restricted cubic spline curve models to investigate the dose-response relationship between FLI and kidney stone risk and conducted subgroup and interaction analyses. RESULTS The study's results indicate a strong correlation between increasing FLI quartiles and a notable rise in the prevalence of kidney stones. Specifically, the risk of developing kidney stones was 1.68 times higher among participants in the highest FLI quartile compared to those in the lowest. Furthermore, patients with MASLD exhibited a 1.35-fold increased risk of developing kidney stones compared to those with non-MASLD. Subgroup analyses demonstrated that the correlation between MASLD and kidney stone risk was consistent across multiple subgroups. However, a significant interaction was observed in the subgroups of smoking status, physical activity level, and hypertension (interaction p < 0.05). The restricted cubic spline analysis did not yield a statistically significant nonlinear association between FLI and kidney stone risk. However, the study did identify inflection point values for FLI. CONCLUSION This study demonstrated an association between FLI and MASLD and the risk of kidney stones. This suggests that these conditions may be pivotal risk factors for kidney stones. Further investigation is required to elucidate these associations' underlying mechanisms and develop efficacious interventions to reduce the risk of kidney stones. Also, formulating personalized prevention and treatment strategies for different population subgroups is paramount.
Collapse
Affiliation(s)
- Fan Zhang
- Changzhou Clinical College, Xuzhou Medical University, Changzhou, China
- Department of Endocrinology, Changzhou Third People’s Hospital, Changzhou, China
- Department of Clinical Nutrition, Changzhou Third People’s Hospital, Changzhou, China
| | - Wenjian Li
- Changzhou Clinical College, Xuzhou Medical University, Changzhou, China
- Department of Urology, Changzhou Third People’s Hospital, Changzhou, China
| |
Collapse
|
37
|
Zhong L, Yang J, Syed JN, Zhang Y, Tian Y, Fu X. Alpha-Glucosidase Inhibitors in Aging and Aging-Related Diseases: Clinical Applications and Relevant Mechanisms. Aging Dis 2025:AD.2024.1477. [PMID: 39751859 DOI: 10.14336/ad.2024.1477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Aging is a complex and universal process marked by gradual functional declines at the cellular and tissue levels, often leading to a range of aging-related diseases such as diabetes, cardiovascular diseases, and cancer. Delaying the aging process can help prevent, slow down, and alleviate the severity of these various conditions, enhancing overall health and well-being. Alpha-glucosidase inhibitors (AGIs) are a class of widely used antidiabetic drugs that inhibit alpha-glucosidase in the small intestinal mucosa, delaying carbohydrate absorption and reducing postprandial hyperglycemia. Beyond their roles in diabetes treatment, AGIs have shown potential in extending lifespan and effectively treating aging-related diseases by modulating oxidative stress, gut microbiota, inflammatory responses, and nutrient-sensing pathways. This review summarizes recent advancements in the application of AGIs for preventing and treating aging and aging-related diseases, with a focus on their mechanisms and roles in these processes.
Collapse
Affiliation(s)
- Ling Zhong
- Department of Endocrinology and Metabolism, Department of Biotherapy, Laboratory of Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jielin Yang
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, ON M5S 1A1, Canada
| | - Jibran Nehal Syed
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, ON M5S 1A1, Canada
| | - Yuwei Zhang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Tian
- Department of Endocrinology and Metabolism, Department of Biotherapy, Laboratory of Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xianghui Fu
- Department of Endocrinology and Metabolism, Department of Biotherapy, Laboratory of Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
38
|
Yan Q, Zhang H, Ma Y, Sun L, Chen Z, Zhang Y, Guo W. AQP1 mediates pancreatic β cell senescence induced by metabolic stress through modulating intracellular H 2O 2 level. Free Radic Biol Med 2025; 226:171-184. [PMID: 39551452 DOI: 10.1016/j.freeradbiomed.2024.11.029] [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: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Metabolic stress-induced pancreatic β cell senescence plays a pivotal role in the type 2 diabetes progression, and yet the precise molecular mechanisms remain elusive. Through cellular experiments and bioinformatics analyses, we identified aquaporin 1(AQP1)-mediated transmembrane transport of hydrogen peroxide as a key driver of glucolipotoxicity-induced senescence in MIN6 cells. A PPI network analysis was used to cross-reference 17 differentially expressed genes associated with type 2 diabetes from three independent GEO databases with 188 stress-induced senescence-related genes from CellAge. AQP1 was revealed as a critical molecular nexus connecting diabetes, oxidative stress, and cellular senescence. AQP1 inhibition, through Bacopaside II and si-AQP1, significantly reduced critical senescence markers in MIN6 cells, demonstrated by the reversal of glucolipotoxicity-induced upregulation of p16, p21, and p-γH2A.X, activation of the senescence-associated secretory phenotype genes, and an elevated percentage of senescence-associated-β-galactosidase positive cells. These effects were primarily mediated through oxidative stress MAPK signaling pathway modulation. AQP1 inhibition is crucial in alleviating glucolipotoxicity-induced β cell senescence. It underscores its potential as a molecular target for therapeutic strategies to delay pancreatic β cell senescence by modulating antioxidant pathways during metabolic stress.
Collapse
Affiliation(s)
- Qihui Yan
- Key Laboratory of Endocrinology and Metabolism, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Haifeng Zhang
- Interventional Therapy, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yunxiao Ma
- Key Laboratory of Endocrinology and Metabolism, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lin Sun
- Key Laboratory of Endocrinology and Metabolism, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhiyue Chen
- Key Laboratory of Endocrinology and Metabolism, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yinbei Zhang
- Key Laboratory of Endocrinology and Metabolism, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Weiying Guo
- Key Laboratory of Endocrinology and Metabolism, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
39
|
Curtis EM, Miguel M, McEvoy C, Ticinesi A, Torre C, Al-Daghri N, Alokail M, Bałkowiec-Iskra E, Bruyère O, Burlet N, Cavalier E, Cerreta F, Clark P, Cherubini A, Cooper C, D'Amelio P, Fuggle N, Gregson C, Halbout P, Kanis JA, Kaufman J, Laslop A, Maggi S, Maier A, Matijevic R, McCloskey E, Ormarsdóttir S, Yerro CP, Radermecker RP, Rolland Y, Singer A, Veronese N, Rizzoli R, Reginster JY, Harvey NC. Impact of dementia and mild cognitive impairment on bone health in older people. Aging Clin Exp Res 2024; 37:5. [PMID: 39725855 PMCID: PMC11671436 DOI: 10.1007/s40520-024-02871-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 10/20/2024] [Indexed: 12/28/2024]
Abstract
Mild cognitive impairment, dementia and osteoporosis are common diseases of ageing and, with the increasingly ageing global population, are increasing in prevalence. These conditions are closely associated, with shared risk factors, common underlying biological mechanisms and potential direct causal pathways. In this review, the epidemiological and mechanistic links between mild cognitive impairment, dementia and skeletal health are explored. Discussion will focus on how changes in brain and bone signalling can underly associations between these conditions, and will consider the molecular and cellular drivers in the context of inflammation and the gut microbiome. There is a complex interplay between nutritional changes, which may precede or follow the onset of mild cognitive impairment (MCI) or dementia, and bone health. Polypharmacy is common in patients with MCI or dementia, and there are difficult prescribing decisions to be made due to the elevated risk of falls associated with many drugs used for associated problems, which can consequently increase fracture risk. Some medications prescribed for cognitive impairment may directly impact bone health. In addition, patients may have difficulty remembering medication without assistance, meaning that osteoporosis drugs may be prescribed but not taken. Cognitive impairment may be improved or delayed by physical activity and exercise, and there is evidence for the additional benefits of physical activity on falls and fractures. Research gaps and priorities with the aim of reducing the burden of osteoporosis and fractures in people with MCI or dementia will also be discussed.
Collapse
Affiliation(s)
- Elizabeth M Curtis
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK
| | - Mario Miguel
- Centro de Estudos Egas Moniz, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Claire McEvoy
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Belfast, UK
| | - Andrea Ticinesi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Azienda Ospedaliero-Universitaria Di Parma, Parma, Italy
| | - Carla Torre
- Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines of the University of Lisbon (iMED.ULisboa), Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Nasser Al-Daghri
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University, 11451, Riyadh, Kingdom of Saudi Arabia
| | - Majed Alokail
- Biochemistry Department, College of Science, KSU, Riyadh, Kingdom of Saudi Arabia
| | - Ewa Bałkowiec-Iskra
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
- The Office for Registration of Medicinal Products, Medical Devices and Biocidal Products & CHMP, SAWP, CNSWP, PCWP, ETF (European Medicines Agency) Member, Warsaw, Poland
| | - Olivier Bruyère
- Research Unit in Public Health, Epidemiology and Health Economics, University of Liège, Liège, Belgium
- Department of Physical Activity and Rehabilitation Sciences, University of Liège, Liège, Belgium
| | - Nansa Burlet
- Research Unit in Epidemiology, University of Liege, Liège, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, CIRM, University of Liège, CHU de Liège, Liège, Belgium
| | - Francesca Cerreta
- Digital Health and Geriatrics, European Medicines Agency, Amsterdam, The Netherlands
| | - Patricia Clark
- Clinical Epidemiology Unit, Hospital Infantil Federico Gómez-Facultad de Medicina, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Antonio Cherubini
- Geriatria, Accettazione Geriatrica e Centro di ricerca per l'invecchiamento, IRCCS INRCA Istituto Nazionale di Ricovero e Cura per Anziani, Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK
| | - Patrizia D'Amelio
- Department of Medicine, Service of Geriatric Medicine & Geriatric Rehabilitation, University of Lausanne Hospital, University of Lausanne, Lausanne, Switzerland
| | - Nicholas Fuggle
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK
| | - Celia Gregson
- Musculoskeletal Research Unit, Bristol Medical School, Learning and Research Building, University of Bristol, Southmead Hospital, Bristol, BS10 5NB, UK
- The Health Research Unit of Zimbabwe (THRU ZIM), The Biomedical Research and Training Institute, Harare, Zimbabwe
| | | | - John A Kanis
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
- Centre for Metabolic Bone Diseases, University of Sheffield, Sheffield, UK
| | - Jean Kaufman
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Andrea Laslop
- Scientific Office, Austrian Medicines and Medical Devices Agency, Vienna, Austria
| | | | - Andrea Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Department of Human Movement Sciences, at AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Radmila Matijevic
- Faculty of Medicine in Novi Sad, University of Novi Sad, Novi Sad, Serbia
| | - Eugene McCloskey
- Mellanby Centre for Musculoskeletal Research, Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
- MRC Versus Arthritis Centre for Integrated Research in Musculoskeletal Ageing, University of Sheffield, Sheffield, UK
| | - Sif Ormarsdóttir
- Medicine Assessment and Licencing, Icelandic Medicines Agency, Reykjavik, Iceland
| | | | - Régis P Radermecker
- Department of Diabetes, Nutrition and Metabolic Disorders, Clinical Pharmacology, University of Liege, CHU de Liège, Liège, Belgium
| | - Yves Rolland
- HealthAge, CHU Toulouse, CERPOP UMR 1295, Inserm, Université Paul Sabatier, Toulouse, France
| | - Andrea Singer
- Departments of Obstetrics & Gynecology and Medicine, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Nicola Veronese
- Department of Internal Medicine, Geriatrics Section, University of Palermo, Palermo, Italy
| | - René Rizzoli
- Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Jean-Yves Reginster
- Protein Research Chair, Biochemistry Department, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK.
- NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK.
| |
Collapse
|
40
|
Misceo D, Mocciaro G, D'Amore S, Vacca M. Diverting hepatic lipid fluxes with lifestyles revision and pharmacological interventions as a strategy to tackle steatotic liver disease (SLD) and hepatocellular carcinoma (HCC). Nutr Metab (Lond) 2024; 21:112. [PMID: 39716321 DOI: 10.1186/s12986-024-00871-3] [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: 08/22/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Steatotic liver disease (SLD) and Hepatocellular Carcinoma (HCC) are characterised by a substantial rewiring of lipid fluxes caused by systemic metabolic unbalances and/or disrupted intracellular metabolic pathways. SLD is a direct consequence of the interaction between genetic predisposition and a chronic positive energy balance affecting whole-body energy homeostasis and the function of metabolically-competent organs. In this review, we discuss how the impairment of the cross-talk between peripheral organs and the liver stalls glucose and lipid metabolism, leading to unbalances in hepatic lipid fluxes that promote hepatic fat accumulation. We also describe how prolonged metabolic stress builds up toxic lipid species in the liver, and how lipotoxicity and metabolic disturbances drive disease progression by promoting a chronic activation of wound healing, leading to fibrosis and HCC. Last, we provide a critical overview of current state of the art (pre-clinical and clinical evidence) regarding mechanisms of action and therapeutic efficacy of candidate SLD treatment options, and their potential to interfere with SLD/HCC pathophysiology by diverting lipids away from the liver therefore improving metabolic health.
Collapse
Affiliation(s)
- Davide Misceo
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Gabriele Mocciaro
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK
| | - Simona D'Amore
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Clinica Medica "G. Baccelli", "Aldo Moro" University of Bari, 70124, Bari, Italy.
| | - Michele Vacca
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK.
| |
Collapse
|
41
|
Zhang Z, Zhou C, Yu L. LEP O-GlcNAcylation inactivates NF-κB pathway by suppressing LEP protein level and thus mediates cellular senescence and osteogenic differentiation in mouse mesenchymal stem cells. BMC Mol Cell Biol 2024; 25:26. [PMID: 39695926 DOI: 10.1186/s12860-024-00523-7] [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: 05/15/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Cellular senescence is a key driver of decreased bone formation and osteoporosis. Leptin (LEP) has been implicated in cellular senescence and osteogenic differentiation. The aim of this study was to investigate the mechanisms by which LEP mediates cellular senescence and osteogenic differentiation. METHODS C3H10T1/2 cells were treated with etoposide to induce cellular senescence, which was assessed by β-galactosidase staining. Quantitative real-time PCR and western blotting were used to measure the levels of senescence markers p21 and p16, as well as osteogenic differentiation-related genes ALP, COL1A1, and RUNX2. Alkaline phosphatase (ALP) staining and alizarin red S staining were performed to evaluate osteogenic differentiation. The NF-κB pathway and O-GlcNAcylation were assessed by western blotting. RESULTS Etoposide treatment increased the number of senescent cells and the levels of p21 and p16, along with elevated LEP expression. These effects were reversed by LEP knockdown. Additionally, LEP knockdown increased ALP staining density and osteoblast mineralization nodules, as well as the mRNA and protein levels of ALP, COL1A1, and RUNX2, indicating that LEP knockdown promoted osteogenic differentiation in C3H10T1/2 cells. Mechanistically, LEP knockdown inactivated the NF-κB pathway by inhibiting the nuclear translocation of p65. Furthermore, OGT was found to promote O-GlcNAcylation of LEP at the S50 site. CONCLUSION Our findings demonstrated that O-GlcNAcylation of LEP inactivated the NF-κB pathway by reducing LEP protein levels, thereby inhibiting cellular senescence and promoting osteogenic differentiation in C3H10T1/2 cells. This study may provide a novel therapeutic target for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zhuang Zhang
- Macau University of Science and Technology, Faculty of Chinese Medicine, E205, Avenida Wai Long, Taipa, Macau, 999078, China
- The 2nd People's Hospital of Zhuhai, Zhuhai, China
| | - Chaoqing Zhou
- Department of Traumatology, The 2nd People's Hospital of Zhuhai, No.208 Yuehua Road, Zhuhai, Guangdong, 519020, China.
| | - Lili Yu
- Macau University of Science and Technology, Faculty of Chinese Medicine, E205, Avenida Wai Long, Taipa, Macau, 999078, China.
| |
Collapse
|
42
|
Tokizane K, Imai SI. Inter-organ communication is a critical machinery to regulate metabolism and aging. Trends Endocrinol Metab 2024:S1043-2760(24)00320-5. [PMID: 39694728 DOI: 10.1016/j.tem.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
Inter-organ communication (IOC) is a complex mechanism involved in maintaining metabolic homeostasis and healthy aging. Dysregulation of distinct forms of IOC is linked to metabolic derangements and age-related pathologies, implicating these processes as a potential target for therapeutic intervention to promote healthy aging. In this review, we delve into IOC mediated by hormonal signaling, circulating factors, organelle signaling, and neuronal networks and examine their roles in regulating metabolism and aging. Given the role of the hypothalamus as a high-order control center for aging and longevity, we particularly emphasize the importance of its communication with peripheral organs and pave the way for a better understanding of this critical machinery in metabolism and aging.
Collapse
Affiliation(s)
- Kyohei Tokizane
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, 63110, MO, USA; Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA.
| |
Collapse
|
43
|
Cho KH, Lee Y, Bahuguna A, Lee SH, Yang CE, Kim JE, Kwon HS. The Consumption of Beeswax Alcohol (BWA, Raydel ®) Improved Zebrafish Motion and Swimming Endurance by Protecting the Brain and Liver from Oxidative Stress Induced by 24 Weeks of Supplementation with High-Cholesterol and D-Galactose Diets: A Comparative Analysis Between BWA and Coenzyme Q 10. Antioxidants (Basel) 2024; 13:1488. [PMID: 39765817 PMCID: PMC11672924 DOI: 10.3390/antiox13121488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
The prolonged consumption of D-galactose (Gal) has been associated with severe damage in the liver and brain via exacerbation of oxidative stress, non-enzymatic glycation, and the aging process. The current study was initiated for a comparative assessment of beeswax alcohol (BWA, final 0.5% and 1.0% w/w) and coenzyme Q10 (CoQ10, final 0.5% and 1.0% w/w) against high-cholesterol (HC, final 4%, w/w) and -galactose (Gal, final 30%, w/w)-induced adverse events in zebrafish during 24 weeks of consumption. The survivability of zebrafish decreased to 82.1% due to HC+Gal exposure, but this was substantially improved (91.0%) with the consumption of 0.5% and 1.0% BWA. In contrast, no protective effect of CoQ10 consumption (1.0%) was observed on the survivability of zebrafish. Nevertheless, both BWA and CoQ10 displayed a significant (p < 0.001) preventive effect against HC+Gal-induced body weight enhancement. The HC+Gal-induced cognitive changes, marked by staggered and confused swimming behavior, and retarded swimming speed and motion patterns (restricted to the bottom of the tank), were efficiently restored by BWA. A significantly higher residence time in the upper half of the tank, 3.1-and 4.5-fold reduced latency time along with 3.5-fold and 4.1-fold higher swimming distance, was logged in the 0.5% and 1.0% BWA groups, respectively, than the zebrafish that consumed HC+Gal. In addition, BWA effectively enhanced plasma ferric ion reduction (FRA) and paraoxonase (PON) activity and alleviated the total cholesterol (TC), triglyceride (TG), and blood glucose levels disrupted by the consumption of HC+Gal. Also, the HC+Gal-alleviated plasma high-density lipoprotein-cholesterol (HDL-C) was 2.6-fold (p < 0.001) enhanced in the group that consumed 1.0% BWA, which was significantly 1.5-fold (p < 0.001) better than the effect of 1.0% CoQ10. Similarly, BWA displayed a superior impact over CoQ10 to mitigate HC+Gal-induced plasma AST and ALT levels, hepatic IL-6 production, generation of oxidized species, cellular senescence, and fatty liver changes. Moreover, BWA protects the brain against HC+Gal-induced oxidative stress, apoptosis, and myelin sheath degeneration. Conclusively, compared to CoQ10, BWA efficiently can the HC+Gal-impaired brain and liver functionality to subside and improves the dyslipidemia and cognitive behavior of zebrafish.
Collapse
Affiliation(s)
- Kyung-Hyun Cho
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
44
|
Torres G, Salladay-Perez IA, Dhingra A, Covarrubias AJ. Genetic origins, regulators, and biomarkers of cellular senescence. Trends Genet 2024; 40:1018-1031. [PMID: 39341687 PMCID: PMC11717094 DOI: 10.1016/j.tig.2024.08.007] [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: 05/02/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024]
Abstract
This review comprehensively examines the molecular biology and genetic origins of cellular senescence. We focus on various cellular stressors and pathways leading to senescence, including recent advances in the understanding of the genetic influences driving senescence, such as telomere attrition, chemotherapy-induced DNA damage, pathogens, oncogene activation, and cellular and metabolic stress. This review also highlights the complex interplay of various signaling and metabolic pathways involved in cellular senescence and provides insights into potential therapeutic targets for aging-related diseases. Furthermore, this review outlines future research directions to deepen our understanding of senescence biology and develop effective interventions targeting senescent cells (SnCs).
Collapse
Affiliation(s)
- Grasiela Torres
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ivan A Salladay-Perez
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anika Dhingra
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anthony J Covarrubias
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
45
|
Sun W, Gao Y, Wu Y, Wu W, Wang C, Chen J, Luan C, Hua M, Liu W, Gong W, Ma X. Targeted apoptosis of senescent cells by valproic acid alleviates therapy-induced cellular senescence and lung aging. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156131. [PMID: 39395326 DOI: 10.1016/j.phymed.2024.156131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/20/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Accumulation of senescent cells in tissues and their downstream effect programs have emerged as key drivers of aging and age-associated pathologies. Recent progresses in senotherapeutics indicated that either selectively killing senescent cells with senolytics or suppressing the senescence-associated secretory phenotype (SASP) secretion using senomorphics contributes to extending of the healthy lifespan and alleviating numerous age-related disorders in mice. PURPOSE However, the potential side-effects and long-term cytotoxicity of the above novel compounds have not yet been determined. Therefore, it seems to be more efficient to explore new senotherapeutical functions from approved drugs. METHODS The effects of valproic acid (VPA), a derivative of valine, in cellular senescence were evaluated by senescence-associated β galactosidase (SA-β-Gal) staining, flow cytometry and western blot (WB). The cell viability was tested using CCK-8 kits. Cell apoptosis was detected by Annexin V-EGFP/PI apoptosis detection kit. Cell autophagy was checked using GFP-RFP-LC3 ratiometric plasmid. The roles of VPA in lung aging were investigated by in vivo experiments using H&E and Masson staining, WB, as well as electronic microscope strategies. RESULTS Here we identified VPA was able to induce an over-accumulation of reactive oxygen species (ROS) (>1.5 times increasing) and apoptosis (>2 times increasing) of senescent cells. Mechanistically, VPA activated the phospholipid modifying enzyme membrane-bound O-acyltransferase domain-containing protein 1 (MBOAT1), which was repressed during senescence, then promoted mitochondrial autophagy and apoptosis. In addition, VPA was also found to alleviate therapy induced abnormal mitochondria and lung aging phenotype (>1.5 times decreasing of lung fibrosis markers and >2.5 times increasing of naïve/memory CD4+ or CD8+ T cells) in vivo. CONCLUSION Taken together, our study demonstrated that VPA was able to selectively kill senescent cells both in vitro and in vivo, and thus shedding light on new functions and novel potential application of VPA in anti-aging and anti-age-associated diseases.
Collapse
Affiliation(s)
- Wentao Sun
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China
| | - Yue Gao
- Department of Pathology, Northern Jiangsu People's Hospital, Yangzhou, PR China
| | - Yubing Wu
- Department of Thoracic Surgery, Linyi Central Hospital, Linyi, PR China
| | - Wei Wu
- Department of Science and Technology, Linyi Central Hospital, Linyi, PR China
| | - Chaofan Wang
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China
| | - JiaXiao Chen
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China
| | - Changjiao Luan
- Department of Lung, The Third People's Hospital of Yangzhou, Yangzhou, PR China
| | - Ming Hua
- Department of Intensive Care, Guannan Country District People's Hospital, Yancheng, PR China
| | - Weili Liu
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China
| | - Weijuan Gong
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China.
| | - Xingjie Ma
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China.
| |
Collapse
|
46
|
Du K, Umbaugh DS, Dutta RK, Diehl AM. A systemic effect for liver senescence. Nat Cell Biol 2024; 26:2016-2017. [PMID: 39537754 DOI: 10.1038/s41556-024-01520-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- Kuo Du
- Department of Medicine, Duke University, Durham, NC, USA
| | | | | | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
47
|
Suryadevara V, Hudgins AD, Rajesh A, Pappalardo A, Karpova A, Dey AK, Hertzel A, Agudelo A, Rocha A, Soygur B, Schilling B, Carver CM, Aguayo-Mazzucato C, Baker DJ, Bernlohr DA, Jurk D, Mangarova DB, Quardokus EM, Enninga EAL, Schmidt EL, Chen F, Duncan FE, Cambuli F, Kaur G, Kuchel GA, Lee G, Daldrup-Link HE, Martini H, Phatnani H, Al-Naggar IM, Rahman I, Nie J, Passos JF, Silverstein JC, Campisi J, Wang J, Iwasaki K, Barbosa K, Metis K, Nernekli K, Niedernhofer LJ, Ding L, Wang L, Adams LC, Ruiyang L, Doolittle ML, Teneche MG, Schafer MJ, Xu M, Hajipour M, Boroumand M, Basisty N, Sloan N, Slavov N, Kuksenko O, Robson P, Gomez PT, Vasilikos P, Adams PD, Carapeto P, Zhu Q, Ramasamy R, Perez-Lorenzo R, Fan R, Dong R, Montgomery RR, Shaikh S, Vickovic S, Yin S, Kang S, Suvakov S, Khosla S, Garovic VD, Menon V, Xu Y, Song Y, Suh Y, Dou Z, Neretti N. SenNet recommendations for detecting senescent cells in different tissues. Nat Rev Mol Cell Biol 2024; 25:1001-1023. [PMID: 38831121 PMCID: PMC11578798 DOI: 10.1038/s41580-024-00738-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2024] [Indexed: 06/05/2024]
Abstract
Once considered a tissue culture-specific phenomenon, cellular senescence has now been linked to various biological processes with both beneficial and detrimental roles in humans, rodents and other species. Much of our understanding of senescent cell biology still originates from tissue culture studies, where each cell in the culture is driven to an irreversible cell cycle arrest. By contrast, in tissues, these cells are relatively rare and difficult to characterize, and it is now established that fully differentiated, postmitotic cells can also acquire a senescence phenotype. The SenNet Biomarkers Working Group was formed to provide recommendations for the use of cellular senescence markers to identify and characterize senescent cells in tissues. Here, we provide recommendations for detecting senescent cells in different tissues based on a comprehensive analysis of existing literature reporting senescence markers in 14 tissues in mice and humans. We discuss some of the recent advances in detecting and characterizing cellular senescence, including molecular senescence signatures and morphological features, and the use of circulating markers. We aim for this work to be a valuable resource for both seasoned investigators in senescence-related studies and newcomers to the field.
Collapse
Affiliation(s)
- Vidyani Suryadevara
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Adam D Hudgins
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | - Adarsh Rajesh
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | | | - Alla Karpova
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Amit K Dey
- National Institute on Aging, NIH, Baltimore, MD, USA
| | - Ann Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Anthony Agudelo
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
- Center on the Biology of Aging, Brown University, Providence, RI, USA
| | - Azucena Rocha
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
- Center on the Biology of Aging, Brown University, Providence, RI, USA
| | - Bikem Soygur
- The Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Chase M Carver
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Cristina Aguayo-Mazzucato
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Darren J Baker
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Diana Jurk
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Dilyana B Mangarova
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Ellen M Quardokus
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | | | - Elizabeth L Schmidt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Feng Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Francesca E Duncan
- The Buck Institute for Research on Aging, Novato, CA, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Gagandeep Kaur
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Gung Lee
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Helene Martini
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Hemali Phatnani
- New York Genome Center, New York, NY, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Iman M Al-Naggar
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jia Nie
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Judith Campisi
- The Buck Institute for Research on Aging, Novato, CA, USA
| | - Julia Wang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kanako Iwasaki
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Karina Barbosa
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Kay Metis
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kerem Nernekli
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lichao Wang
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Lisa C Adams
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Liu Ruiyang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Madison L Doolittle
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Marcos G Teneche
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Ming Xu
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Mohammadjavad Hajipour
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | | | | | - Nicholas Sloan
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Nikolai Slavov
- Center on the Biology of Aging, Brown University, Providence, RI, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Department of Biology, Northeastern University, Boston, MA, USA
- Barnett Institute for Chemical and Biological Analysis, Northeastern University, Boston, MA, USA
| | - Olena Kuksenko
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| | - Paul T Gomez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Periklis Vasilikos
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Priscila Carapeto
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Quan Zhu
- Center for Epigenomics, University of California, San Diego, CA, USA
| | | | | | - Rong Fan
- Yale-Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
| | - Runze Dong
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure and Design, University of Washington, Seattle, WA, USA
| | - Ruth R Montgomery
- Yale-Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
| | - Sadiya Shaikh
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Sanja Vickovic
- New York Genome Center, New York, NY, USA
- Herbert Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Beijer Laboratory for Gene and Neuro Research, Uppsala University, Uppsala, Sweden
| | - Shanshan Yin
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Shoukai Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Sonja Suvakov
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sundeep Khosla
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Vesna D Garovic
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Translational and Computational Neuroimmunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yanxin Xu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yizhe Song
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Zhixun Dou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicola Neretti
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA.
- Center on the Biology of Aging, Brown University, Providence, RI, USA.
| |
Collapse
|
48
|
Niu Q, Li D, Zhang J, Piao Z, Xu B, Xi Y, Mohamed Kamal NNSN, Lim V, Li P, Yin Y. The new perspective of Alzheimer's Disease Research: Mechanism and therapeutic strategy of neuronal senescence. Ageing Res Rev 2024; 102:102593. [PMID: 39566741 DOI: 10.1016/j.arr.2024.102593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Alzheimer's disease (AD), commonly known as senile dementia, is a neurodegenerative disease with insidious onset and gradually worsening course. The brain is particularly sensitive to senescence, and neuronal senescence is an important risk factor for the occurrence of AD. However, the exact pathogenesis between neuronal senescence and AD has not been fully elucidated so far. Neuronal senescence is characterized by the permanent stagnation of the cell cycle, and the changes in its structure, function, and microenvironment are closely related to the pathogenesis and progression of AD. In recent years, studies such as the Aβ cascade hypothesis and Tau protein phosphorylation have provided new strategies for the therapy of AD, but due to the complexity of the etiology of AD, there are still no effective treatment measures. This article aims to deeply analyze the pathogenesis between AD and neuronal senescence, and sort out various existing therapeutic methods, to provide new ideas and references for the clinical treatment of AD.
Collapse
Affiliation(s)
- Qianqian Niu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia
| | - Danjie Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Jiayin Zhang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Zhengji Piao
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Bo Xu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Yuting Xi
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Nik Nur Syazni Nik Mohamed Kamal
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia; Dementia Multidisciplinary Research Program of IPPT (DMR-IPPT), Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia.
| | - Vuanghao Lim
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia.
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China.
| | - Yaling Yin
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
49
|
Reguero M, Reglero G, Quintela JC, Ramos-Ruiz R, Ramírez de Molina A, Gómez de Cedrón M. Silymarin-Enriched Extract from Milk Thistle Activates Thermogenesis in a Preclinical Model of High-Fat-Diet-Induced Obesity to Relieve Systemic Meta-Inflammation. Nutrients 2024; 16:4166. [PMID: 39683558 DOI: 10.3390/nu16234166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Obesity and aging are associated with the progressive loss of brown adipose tissue (BAT), an increase in visceral white adipose tissue (vWAT), and a reduction in subcutaneous white adipose tissue (sWAT). The progressive expansion of visceral obesity promotes a low grade of systemic chronic inflammation (meta-inflammation), contributing to the onset of comorbidities such as type 2 diabetes mellitus (T2DM), metabolic syndrome, and even cancer. Thus, preserving the thermogenic activity of adipose tissue and improving the metabolic flexibility of sWAT could be an effective strategy to prevent the development of metabolic chronic diseases and promote healthy aging. Precision nutrition has emerged as a complementary approach to control the metabolic alterations associated with unhealthy obesity and aging. In a previous work, we described that a silymarin-enriched extract from milk thistle (Mthistle) increased markers of browning and thermogenesis in vitro in human differentiated adipocytes (SGBS). OBJECTIVES/METHODS Therefore, this study aims to evaluate the potential of Mthistle to activate thermogenesis in a preclinical model of high-fat diet (HFD)-induced obesity (DIO). RESULTS Our results demonstrate that Mthistle increases systemic energy expenditure (EE), preserves body temperature after cold exposure, improves insulin resistance, and reduces inflammatory markers in WAT. CONCLUSIONS Based on these results, silymarin-enriched extract from Mthistle may be proposed as a nutraceutical for the management of metabolic chronic diseases and/or accelerated aging.
Collapse
Affiliation(s)
- Marina Reguero
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
- NATAC BIOTECH, Electronica 7, E28923 Madrid, Spain
| | - Guillermo Reglero
- Production and Characterization of Novel Foods Department, Institute of Food Science Research CIAL, CEI UAM + CSIC, E28049 Madrid, Spain
| | | | - Ricardo Ramos-Ruiz
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| | - Marta Gómez de Cedrón
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
- Cell Metabolism Unit, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| |
Collapse
|
50
|
Zhang H, Zhu Z, Wei W, Liu Z, Zhou H, Gong Y, Yan X, Du J, Li H, Chen L, Sheng L. Aronia melanocarpa extract extends the lifespan and health-span of Caenorhabditis elegans via mitogen-activated protein kinase 1. Food Funct 2024; 15:11020-11035. [PMID: 39450574 DOI: 10.1039/d4fo02479f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Aging is a highly complex process and one of the largest risk factors for many chronic diseases. Aronia melanocarpa (AM) is rich in bioactive phytochemicals with antioxidant, anti-inflammatory, and anticancer properties. However, little is known about its effects on aging. The objective of this study was to evaluate the effects of AM extract on lifespan and health-span using Caenorhabditis elegans as a representative model. The mechanisms of its effects were explored using transcriptomics and untargeted metabolomics. Results showed that the lifespan of C. elegans was significantly extended by 22.2% after high-dose AM treatment. AM improved the behavior and physiological functions of C. elegans by increasing the pharyngeal pumping rate, decreasing lipofuscin accumulation and the reactive oxygen species level, enhancing resistance to oxidative stress, and increasing the activities of superoxide dismutase and catalase. Transcriptome analysis showed that the pmk-1 gene (mitogen-activated protein kinase 1), which is involved in the MAPK signaling pathway, was the gene with the largest fold change after AM intervention. However, in the C. elegans pmk-1(km25) mutant, the beneficial effect of AM in improving nematode senescence disappeared. An untargeted metabolomics study showed that the levels of 4-hydroxyproline, rhamnose, and cysteine were increased after AM supplementation, and their extending effect on the lifespan and health-span of C. elegans were partly dependent on the pmk-1 gene. In conclusion, our results revealed that AM can promote the lifespan and health-span of C. elegans via the PMK-1 pathway, highlighting the potential of AM as a dietary supplement to delay aging.
Collapse
Affiliation(s)
- Huan Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zhigang Zhu
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai 201203, China.
| | - Wenjing Wei
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zekun Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Huiji Zhou
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai 201203, China.
| | - Yueling Gong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xinlei Yan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai 201203, China.
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai 201203, China.
| | - Lili Sheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|