1.Systemic comparison of molecular characteristics in different skin fibroblast senescent models.
Xiaokai FANG ; Shan ZHANG ; Mingyang WU ; Yang LUO ; Xingyu CHEN ; Yuan ZHOU ; Yu ZHANG ; Xiaochun LIU ; Xu YAO
Chinese Medical Journal 2025;138(17):2180-2191
BACKGROUND:
Senescent human skin primary fibroblast (FB) models have been established for studying aging-related, proliferative, and inflammatory skin diseases. The aim of this study was to compare the transcriptome characteristics of human primary dermal FBs from children and the elderly with four senescence models.
METHODS:
Human skin primary FBs were obtained from healthy children (FB-C) and elderly donors (FB-E). Senescence models were generated by ultraviolet B irradiation (FB-UVB), D-galactose stimulation (FB-D-gal), atazanavir treatment (FB-ATV), and replication exhaustion induction (FB-P30). Flow cytometry, immunofluorescence staining, real-time quantitative polymerase chain reaction, co-culturing with immune cells, and bulk RNA sequencing were used for systematic comparisons of the models.
RESULTS:
In comparison with FB-C, FB-E showed elevated expression of senescence-related genes related to the skin barrier and extracellular matrix, proinflammatory factors, chemokines, oxidative stress, and complement factors. In comparison with FB-E, FB-UVB and FB-ATV showed higher levels of senescence and expression of the genes related to the senescence-associated secretory phenotype (SASP), and their shaped immune microenvironment highly facilitated the activation of downstream immune cells, including T cells, macrophages, and natural killer cells. FB-P30 was most similar to FB-E in terms of general transcriptome features, such as FB migration and proliferation, and aging-related characteristics. FB-D-gal showed the lowest expression levels of senescence-related genes. In comparisons with the single-cell RNA sequencing results, FB-E showed almost complete simulation of the transcriptional spectrum of FBs in elderly patients with atopic dermatitis, followed by FB-P30 and FB-UVB. FB-E and FB-P30 showed higher similarity with the FBs in keloids.
CONCLUSIONS
Each senescent FB model exhibited different characteristics. In addition to showing upregulated expression of natural senescence features, FB-UVB and FB-ATV showed high expression levels of senescence-related genes, including those involved in the SASP, and FB-P30 showed the greatest similarity with FB-E. However, D-galactose-stimulated FBs did not clearly present aging characteristics.
Humans
;
Fibroblasts/drug effects*
;
Cellular Senescence/physiology*
;
Skin/metabolism*
;
Child
;
Transcriptome/genetics*
;
Aged
;
Ultraviolet Rays
;
Cells, Cultured
;
Galactose/pharmacology*
2.Association of leukocyte telomere length with the risk of digestive diseases: A large-scale cohort study.
Hongqun YANG ; Lanlan CHEN ; Yahui LIU
Chinese Medical Journal 2025;138(1):60-67
BACKGROUND:
Leukocyte telomere length (LTL) shortening, a biomarker of telomere attrition, has been linked to multiple diseases. However, the relationship between LTL and digestive diseases remains uncertain. This study aimed to investigate the association between LTL and the risk of digestive diseases.
METHODS:
A cohort analysis of over 500,000 participants from the UK Biobank (UKB) between 2006 and 2021 was conducted to estimate the associations of LTL with more than 90 common digestive diseases. LTL was quantified using multiplex quantitative polymerase chain reaction, and cases of each disease were determined according to inpatient and primary care data. Multivariable Cox proportional hazards regression analysis was used to evaluate the associations of LTL with the risk of digestive diseases. Furthermore, such associations were also evaluated after stratification by sex and ethnicity.
RESULTS:
After a mean follow-up time of 11.8 years, over 20 International Classification of Diseases, 10th Revision ( ICD-10 ) codes were showed to be associated with telomere attrition. LTL shortening is associated with an increased risk of several digestive diseases, including gastroesophageal reflux disease (K21: hazard ratio [HR] = 1.30, 95% confidence interval [95% CI]: 1.19-1.42), esophageal ulcer (K221: HR = 1.81, 95% CI: 1.22-2.71), Barrett's esophagus (K227: HR = 1.58, 95% CI: 1.14-2.17), gastritis (K29: HR = 1.39, 95% CI: 1.26-1.52), duodenal ulcer (K26: HR = 1.55, 95% CI: 1.14-2.12), functional dyspepsia (K30X: HR = 1.36, 95% CI: 1.06-1.69), non-alcoholic fatty liver disease (NAFLD) (K760: HR = 1.39, 95% CI: 1.09-1.78), liver cirrhosis (K74: HR = 4.73, 95% CI: 3.27-6.85), cholangitis (K830: HR = 2.55, 95% CI: 1.30-5.00), and hernia (K43: HR = 1.50, 95% CI: 1.17-1.94; K44: HR = 1.29, 95% CI: 1.17-1.42). The risk of rectal polyps (K621: HR = 0.77, 95% CI: 0.63-0.92) decreased per unit shortening of LTL.
CONCLUSIONS
This study suggests that LTL shortening is associated with an increased risk of most digestive diseases except for rectal polyps. These findings may provide some clues for understanding the pathogenesis of digestive diseases.
Humans
;
Male
;
Female
;
Middle Aged
;
Cohort Studies
;
Leukocytes/metabolism*
;
Telomere/genetics*
;
Proportional Hazards Models
;
Adult
;
Digestive System Diseases/genetics*
;
Aged
;
Risk Factors
;
Telomere Shortening
3.Cellular senescence in renal ischemia-reperfusion injury.
Chinese Medical Journal 2025;138(15):1794-1806
Acute kidney injury (AKI) affects more than 20% of hospitalized patients and is a significant contributor to morbidity and mortality, primarily due to ischemia-reperfusion injury (IRI), which is one of the leading causes of AKI. IRI not only exacerbates the immediate impact of AKI but also facilitates its progression to chronic kidney disease (CKD) and, in cases of preexisting CKD, to end-stage renal disease (ESRD). One of the critical pathological processes associated with IRI-AKI is cellular senescence, characterized by an irreversible arrest in the cell cycle, morphological and chromatin organization changes, altered transcriptional and metabolic profiles, and the development of a hypersecretory phenotype known as the senescence-associated secretory phenotype (SASP). The SASP amplifies senescence signals in surrounding normal cells through senescence-related pathways, contributing to tissue damage, fibrosis, and chronic inflammation. This review provides an overview of the defining features of senescent cells and explores the fundamental mechanisms underlying senescent cell generation following IRI. We elucidate the pivotal roles of cellular senescence in the transition from IRI-AKI to chronic kidney injury. Furthermore, we discuss emerging therapies targeting cellular senescence, including senolytics and senomorphics, which have shown promising results in both preclinical and clinical settings. These therapies position cellular senescence as a crucial target for the treatment of IRI in the kidneys. Additionally, advancements in single-cell sequencing technology and artificial intelligence-assisted drug screening are expected to accelerate the discovery of novel senescent biomarkers and synotherapeutics, paving the way for optimized and personalized therapeutic interventions.
Humans
;
Cellular Senescence/physiology*
;
Reperfusion Injury/pathology*
;
Acute Kidney Injury/pathology*
;
Animals
;
Kidney/metabolism*
;
Senescence-Associated Secretory Phenotype/physiology*
4.Cellular senescence in kidney diseases.
Xiaojie WANG ; Yujia LI ; Qingqing CHU ; Hang LV ; Jing LI ; Fan YI
Chinese Medical Journal 2025;138(18):2234-2242
Cellular senescence, stable cell cycle arrest that can be triggered in normal cells in response to various intrinsic and extrinsic stressors, has been highlighted as one of the most important mechanisms involved in kidney diseases. It not only serves as a fundamental biological process promoting normal organogenesis and successful wound repair but also contributes to organ dysfunction, tissue fibrosis, and the generalized aging phenotype. Moreover, senescent cells exhibit reduced regenerative capacity, which impairs renal function recovery from injuries. Importantly, senescent cells are involved in immune regulation via secreting a diverse array of proinflammatory and profibrotic factors known as senescence-associated secretory phenotype (SASP) with autocrine, paracrine, and endocrine activities. Thus, eliminating detrimental senescent cells or inhibiting SASP production holds great promise for developing innovative therapeutic strategies for kidney diseases. In this review, we summarize the current knowledge of the intricate mechanisms and hallmarks of cellular senescence in kidney diseases and emphasize novel therapeutic targets, including epigenetic regulators, G protein-coupled receptors, and lysosome-related proteins. Particularly, we highlight the recently identified senotherapeutics, which provide new therapeutic strategies for treating kidney diseases.
Humans
;
Cellular Senescence/genetics*
;
Kidney Diseases/pathology*
;
Senescence-Associated Secretory Phenotype/physiology*
;
Animals
;
Epigenesis, Genetic/physiology*
5.Mechanism of matrine against senescence in human umbilical vein endothelial cells based on network pharmacology and experimental verification.
Dian LIU ; Zi-Ping XIANG ; Ze-Sen DUAN ; Xin-Ying LIU ; Xing WANG ; Hui-Xin ZHANG ; Chao WANG
China Journal of Chinese Materia Medica 2025;50(8):2260-2269
Utilizing network pharmacology, molecular docking, and cellular experimental validation, this study delved into the therapeutic efficacy and underlying mechanisms of matrine in combating senescence. Databases were utilized to predict targets related to the anti-senescence effects of matrine, resulting in the identification of 81 intersecting targets for matrine in the treatment of senescence. A protein-protein interaction(PPI) network was constructed, and key targets were screened based on degree values. Gene Ontology(GO) function and Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway enrichment analyses were performed on the key targets to elucidate the critical pathways involved in the anti-senescence effects of matrine. Molecular docking was conducted between matrine and key targets. A senescence model was established using human umbilical vein endothelial cells(HUVECs) induced with hydrogen peroxide(H_2O_2). Following treatment with varying concentrations of matrine(0.5, 1, and 2 mmol·L~(-1)), cell viability was assessed by using the CCK-8. SA-β-galactosidase staining was employed to observe the positive rate of senescent cells. Flow cytometry was utilized to measure the apoptosis rate. Real-time quantitative PCR(RT-PCR) was utilized to measure the mRNA expression of apoptosis-related cysteine peptidase 3(CASP3), albumin(ALB), glycogen synthase kinase 3β(GSK3B), CD44 molecule(CD44), and tumor necrosis factor-α(TNF-α). Western blot was performed to detect the protein expression of tumor protein p53(p53), cyclin-dependent kinase inhibitor 1A(p21), cyclin-dependent kinase inhibitor 2A(p16), and retinoblastoma tumor suppressor protein(pRb) in the senescence signaling pathway, p38 protein kinase(p38), c-Jun N-terminal kinase(JNK), and extracellular regulated protein kinases(ERK) in the mitogen-activated protein kinase(MAPK) pathway, and phosphatidylinositol 3-kinase(PI3K) and protein kinase B(Akt) in the PI3K/Akt signaling pathway. The experimental results revealed that matrine significantly increased the viability of HUVECs(P<0.05), decreased the positive rate of senescent cells and the apoptosis rate(P<0.05), and reduced the mRNA expression levels of CASP3, ALB, GSK3B, CD44, and TNF-α(P<0.05). It also inhibited the protein expression of p53, p21, p16 and pRb in the senescence signaling pathway(P<0.05), upregulated the protein expression of p-PI3K/PI3K and p-Akt/Akt(P<0.05), and downregulated the protein expression of p-p38/p38, p-JNK/JNK, and p-ERK/ERK(P<0.05). Collectively, these findings suggest that matrine exerts an inhibitory effect on HUVECs senescence, and its mechanism involves the modulation of the senescence signaling pathway, MAPK pathway, and PI3K/Akt signaling pathway to suppress cell apoptosis and inflammation.
Humans
;
Matrines
;
Quinolizines/chemistry*
;
Alkaloids/chemistry*
;
Human Umbilical Vein Endothelial Cells/cytology*
;
Cellular Senescence/drug effects*
;
Network Pharmacology
;
Molecular Docking Simulation
;
Signal Transduction/drug effects*
;
Protein Interaction Maps/drug effects*
;
Cell Survival/drug effects*
;
Apoptosis/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
6.Effects of ROCK-siRNA transfection on Ang II-induced endothelial cell senescence and endothelial microparticles.
Kai WANG ; Yan WANG ; Tianqi CHEN ; Fang PENG ; Hui ZHOU ; Qin SHI
Chinese Journal of Cellular and Molecular Immunology 2025;41(9):778-783
Objective To investigate the effects of ROCK-siRNA transfection on endothelial cell senescence and endothelial microparticles (EMPs) induced by angiotensin II (Ang II). Methods Human umbilical vein endothelial cells (HUVECs) were treated with Ang II (1.0 μmo/L) to induce cellular senescence models, followed by transfection with ROCK-siRNA. The cells were divided into four groups: control group, model group, negative transfection control group (Ang II combined with NC-siRNA), and ROCK-siRNA transfection group (Ang II combined with ROCK-siRNA). Cellular senescence was assessed by SA-β-Gal staining. EMP levels in cell supernatants and intracellular reactive oxygen species (ROS) levels were assessed using flow cytometry. The expression levels of silenced information regulator 1(SIRT1) and p53 protein in each group were analyzed by Western blotting. Results Following ROCK-siRNA transfection, the number of senescent cells induced by Ang II was significantly reduced, accompanied by decreased CD31+ EMP levels and suppressed intracellular ROS levels. Meanwhile, the expression levels of SIRT1 were up-regulated, while the expression levels of p53 were down-regulated. Conclusion Silencing ROCK expression suppresses EMP release, reduces ROS generation, regulates the expression of SIRT1 and p53, and ultimately attenuates Ang II-induced endothelial cell senescence.
Humans
;
Angiotensin II/pharmacology*
;
Cellular Senescence/genetics*
;
Human Umbilical Vein Endothelial Cells/cytology*
;
RNA, Small Interfering/metabolism*
;
Reactive Oxygen Species/metabolism*
;
Sirtuin 1/genetics*
;
Transfection
;
Tumor Suppressor Protein p53/genetics*
;
Cell-Derived Microparticles/drug effects*
;
rho-Associated Kinases/metabolism*
;
Endothelial Cells/metabolism*
;
Cells, Cultured
7.Mechanism and significance of cell senescence induced by viral infection.
Yunchuang CHANG ; Xinna WU ; Lingli DENG ; Sanying WANG ; Genxiang MAO
Journal of Zhejiang University. Medical sciences 2025;54(1):70-80
Virus-induced senescence (VIS) is a significant biological phenomenon, which is associated with declining immune function, accelerating aging process and causing aging-related diseases. A variety of common viruses, including RNA viruses (such as SARS-CoV-2), DNA viruses (such as herpesviruses and hepatitis B virus), and prions can cause VIS in host cells. The primary mechanisms include abnormal activation of the cGAS-STING signaling pathway, DNA damage response, and potential correlations with the integrated stress response due to intracellular phase separation. Viral infection and cellular senescence influence each other: cellular senescence serves as a defense to restrict viral replication and transmission, while some viruses exploit cellular senescence to enhance their infectivity and replication. Understanding the mechanisms of VIS is conducive to the development of therapeutic strategies for viral infections and promotion of healthy aging. However, there is lack of research on therapeutic targets and drug development in this field so far. Although senolytics may be effective for anti-senescent cells therapy, their efficacy for VIS needs evidence from further clinical trials. This article reviews the research progress on the connection between viral infection and cellular senescence, to provide insights for the prevention and treatment of aging related diseases.
Humans
;
Cellular Senescence/physiology*
;
Virus Diseases/physiopathology*
;
Signal Transduction
;
Nucleotidyltransferases/metabolism*
;
DNA Damage
;
Virus Replication
;
COVID-19
;
Membrane Proteins/metabolism*
;
SARS-CoV-2
8.Hydroxysafflor Yellow A Ameliorates the Replicative Senescence of Human Umbilical Cord Mesenchymal Stem Cells by Suppressing Oxidative Stress.
Si-Yun WANG ; Qi ZHU ; Chun-Xia TAN ; Fang LU ; Tao LU
Journal of Experimental Hematology 2025;33(5):1507-1515
OBJECTIVE:
To investigate the effects and mechanisms of hydroxysafflor yellow A (HSYA) on replicative senescence in human umbilical cord mesenchymal stem cells (hUC-MSCs).
METHODS:
hUC-MSCs were cultured to construct a replicative senescence model through continuous amplification in vitro. Cells at passage 2 served as the control group, while cells at passage 10 were designated as the senescence group. The senescent cells were cultured in a culture medium containing HSYA. Cell viability was detected by the CCK-8 assay, and cell confluence was analyzed using the Incucyte S3 live-cell analysis system. The optimal concentration and time point were determined and utilized for subsequent experiments. Senescent cells were pretreated with 0.01 mg/ml HSYA, and the proportion of senescence-associated β-galactosidase (SA-β-gal) positive cells was detected to assess the senescence state. The relative telomere length was detected by qPCR. Reactive oxygen species (ROS) levels were measured using the fluorescent probe DCFH-DA. Mitochondrial membrane potential was assessed by JC-1 staining. The expression of p53, p16, p21, OCT4, and SOX2 genes was detected by qPCR. The expression of p16, p53, OCT4, and SOX2 proteins was analyzed by Western blot.
RESULTS:
HSYA significantly decreased the SA-β-gal positive staining rate, inhibited telomere attrition, reduced the ROS accumulation, increased mitochondrial membrane potential in senescent cells. Additionally, HSYA downregulated the expression of p53 and p16, and upregulated the expression of OCT4. HSYA decreased p16 protein level and increased OCT4 and SOX2 protein levels.
CONCLUSION
HSYA may ameliorate replicative senescence in hUC-MSCs by modulating the p53 and p16 signaling pathways and suppressing oxidative stress.
Humans
;
Mesenchymal Stem Cells/drug effects*
;
Cellular Senescence/drug effects*
;
Chalcone/pharmacology*
;
Oxidative Stress/drug effects*
;
Quinones/pharmacology*
;
Umbilical Cord/cytology*
;
Reactive Oxygen Species/metabolism*
;
Cells, Cultured
;
Cyclin-Dependent Kinase Inhibitor p16/metabolism*
;
Tumor Suppressor Protein p53/metabolism*
;
Membrane Potential, Mitochondrial
;
Cell Proliferation
9.Salvianolic Acid B Exerts Antiphotoaging Effect on Ultraviolet B-Irradiated Human Keratinocytes by Alleviating Oxidative Stress via SIRT1 Protein.
Qiao-Ju ZHANG ; Xi LUO ; Yu-Wen ZHENG ; Jun-Qiao ZHENG ; Xin-Ying WU ; Shu-Mei WANG ; Jun SHI
Chinese journal of integrative medicine 2025;31(11):1021-1028
OBJECTIVE:
To explore the anti-photoaging properties of salvianolic acid B (Sal B).
METHODS:
The optimal photoaging model of human immortalized keratinocytes (HaCaT cells) were constructed by expose to ultraviolet B (UVB) radiation. The cells were divided into control, model and different concentrations of Sal B groups. Cell viability was measured via cell counting kit-8. Subsequently, the levels of oxidative stress, including reactive oxygen species (ROS), hydroxyproline (Hyp), catalase (CAT), and glutathione peroxidase (GSH-Px) were detected using the relevant kits. Silent information regulator 1 (SIRT1) protein level was detected using Western blot. The binding pattern of Sal B and SIRT1 was determined via molecular docking.
RESULTS:
Sal B significantly increased the viability of UVB-irradiated HaCaT cells (P<0.05 or P<0.01). Sal B effectively scavenged the accumulation of ROS induced by UVB (P<0.05 or P<0.01). In addition, Sal B modulated oxidative stress by increasing the intracellular concentrations of Hyp and CAT and the activity of GSH-Px (P<0.05 or P<0.01). The Western blot results revealed a substantial increase in SIRT1 protein levels following Sal B administration (P<0.05). Moreover, Sal B exhibited good binding affinity toward SIRT1, with a docking energy of -7.5 kCal/mol.
CONCLUSION
Sal B could improve the repair of photodamaged cells by alleviating cellular oxidative stress and regulating the expression of SIRT1 protein.
Humans
;
Sirtuin 1/metabolism*
;
Ultraviolet Rays
;
Oxidative Stress/radiation effects*
;
Keratinocytes/metabolism*
;
Molecular Docking Simulation
;
Benzofurans/pharmacology*
;
Skin Aging/radiation effects*
;
Reactive Oxygen Species/metabolism*
;
Cell Survival/radiation effects*
;
HaCaT Cells
;
Hydroxyproline/metabolism*
;
Glutathione Peroxidase/metabolism*
;
Catalase/metabolism*
;
Depsides
10.Astragaloside IV alleviates D-GAL-induced endothelial cell senescence by promoting mitochondrial autophagy via inhibiting the PINK1/Parkin pathway.
Ming YI ; Ye LUO ; Lu WU ; Zeheng WU ; Cuiping JIANG ; Shiyu CHEN ; Xiao KE
Journal of Southern Medical University 2025;45(11):2427-2437
OBJECTIVES:
To explore the mechanism by which astragaloside IV (AS-IV) alleviates D-galactose (D-GAL)-induced senescence in human umbilical vein endothelial cells (HUVECs).
METHODS:
Cultured HUVECs were treated with D-GAL (40 g/L), AS-IV (200 μmol/L), D-GAL+AS-IV, or D-GAL+AS-IV+MTK458 (a mitochondrial autophagy agonist, 25 μmol/L) for 48 h, and the changes in cell proliferation, migration, and angiogenesis capacity were evaluated. Cell apoptosis, reactive oxygen species (ROS) levels, mitochondrial membrane potential, and expressions of autophagy-related proteins (LC3-II/LC3-I) and PINK1/Parkin pathway proteins in the treated cells were detected.
RESULTS:
AS-IV treatment significantly reduced the inhibitory effect of D-GAL on HUVEC viability, effectively alleviated D-GAL-induced impairment of tube-forming ability, and promoted angiogenesis and migration ability of the cells. AS-IV also significantly reduced the rate of D-GAL-induced HUVECs positive for senescence-associated β-galactosidase (SA-β-Gal) staining and inhibited the expression of senescence-related genes P21 and P53. AS-IV restored mitochondrial membrane potential and reduced intracellular ROS levels in D-GAL-induced HUVECs, and inhibited the fusion of autophagosomes and lysosomes to prevent the completion of autophagic flux. In HUVECs treated with both D-GAL and AS-IV, the application MTK458 significantly increased the number of yellow spots and enhanced the expressions of P21, P53, PINK1, Parkin, LC3, and Beclin proteins.
CONCLUSIONS
AS-IV alleviates D-GAL-induced endothelial cell senescence by inhibiting the PINK1/Parkin pathway to regulate mitochondrial autophagy.
Humans
;
Human Umbilical Vein Endothelial Cells/drug effects*
;
Cellular Senescence/drug effects*
;
Autophagy/drug effects*
;
Saponins/pharmacology*
;
Ubiquitin-Protein Ligases/metabolism*
;
Mitochondria/drug effects*
;
Triterpenes/pharmacology*
;
Protein Kinases/metabolism*
;
Galactose/pharmacology*
;
Reactive Oxygen Species/metabolism*
;
Signal Transduction/drug effects*
;
Cells, Cultured
;
Apoptosis/drug effects*
;
Membrane Potential, Mitochondrial
;
Cell Proliferation/drug effects*

Result Analysis
Print
Save
E-mail