International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Aging, a natural yet complex process, is marked by gradual deterioration of physiological function at both the organ and organismal levels over the lifespan. Notably, disease-induced pathological alterations in aged tissues often act as accelerants, amplifying this degenerative cascade (1,2). Among the numerous hallmarks of aging, senescent cell accumulation, an essentially irreversible proliferative arrest, serves as a key driver of aging and diverse age-related illnesses (3). Cellular senescence is regarded as a fundamental stress-responsive mechanism triggered by diverse intrinsic and extrinsic cues, including viral infections, hypoxia, oxidative stress, telomere attrition, genomic instability and oncogene activation (4). Recent progress has clearly established links between cellular senescence and cardiac diseases, chronic lung diseases, types of cancer, metabolic disorders and neurodegenerative conditions (5,6). In hepatology, nevertheless, the pathobiological significance of senescence has only recently become the focus of intensive research. Untreated hepatic inflammation frequently progresses to chronic liver diseases, including liver cirrhosis and hepatocellular carcinoma (HCC), posing a significant global health burden, particularly among middle-aged and geriatric populations (7). Mechanistically, cellular senescence induces multifaceted hepatic dysfunction via senescence-associated secretory phenotype (SASP)-mediated effects. These secretory factors disrupt lobular architecture, impair multiple biochemical processes (particularly hepatic lipid homeostasis) and propagate inflammatory cascades (8). Of interest, it is reported that a category of compounds, namely senotherapeutics, are able to ameliorate the natural history of chronic liver disease by targeting senescent cells using molecular pathways associated with senescence phenotypes (9).
Importantly, cellular senescence is a fundamental biological hallmark of aging and a major risk factor for hepatic pathologies. The present review systematically analyzed the most cutting-edge advances in the pathophysiology of cellular senescence, key features of associated cellular stress responses and the susceptibility of liver-resident cells to senescence. Subsequently, due to space limitations, recent findings on cellular senescence in specific hepatic pathological contexts are summarized. Finally, the present review critically evaluated the potential of 'senolytic therapies', defined as strategies targeting cellular senescence through selective inhibition of senescence pathways (senomorphics) or elimination of senescent cells (senolytics), for treating liver diseases and highlighted both the advantages and limitations of these approaches.
One of the earliest reports on the phenomenon of cellular senescence was described in 1961 by Leonard Hayflick and Paul Moorhead (10), as the limited proliferative capacity of normal human diploid fibroblasts in vitro, which manifests as growth arrest after ~40-60 population doublings (10). Since then, cellular senescence has been increasingly recognized as a multifactorial determinant of cell fate. Extensive studies have documented that this permanent cell cycle arrest negatively affects tissue regenerative capacity and promotes conditions conducive to the onset and development of various diseases (11-13).
Cellular senescence is mainly divided into two principal mechanistic paradigms. Replicative senescence, an intrinsically regulated process, irreversibly arrests cells at the G1/S or S phase. It is initiated by telomere erosion and persistent DNA damage during repeated replication cycles, which in turn activates the ataxia-telangiectasia mutated (ATM)/ataxia-telangiectasia and RAD3-related (ATR) protein kinases (14). These kinases stabilize p53 via phosphorylation and block mouse double minute 2 (MDM2)-mediated ubiquitin degradation, enabling nuclear p53 accumulation and the subsequent p21 induction (6). As a core cell cycle inhibitor, p21 both suppresses key G1/S-phase kinases and directly binds to proliferating cell nuclear antigen, thereby inhibiting DNA synthesis and enforcing synergistic cell cycle arrest (11). Besides, ATM and ATR block the p62-dependent autophagic degradation of GATA binding protein 4, contributing to NF-κB activation (14). Various stressors, including but not limited to oxidative stress, therapy-induced damage (such as ionizing radiation and chemotherapeutic agents), genomic instability and epigenetic changes can trigger premature senescence. This process is predominantly mediated by the activation of the cyclin-dependent kinase (CDK) inhibitor 2A (also called p16INK4a) (15). Excessive p16 accumulation inhibits CDK4/6-cyclin D complex formation, inducing irreversible arrest. Mechanistically, p16 inhibits CDK4/6 kinase activity, preventing full retinoblastoma protein (pRB) phosphorylation and maintaining its hypophosphorylated state. Hypophosphorylated pRB binds and inhibits E2 promoter-binding factor, blocking the G1-to-S phase transition (16). Compared with other pathways, p16INK4a/retinoblastoma protein (Rb)-driven senescence exhibits enhanced stability. The DNA damage response (DDR) pathway functions as an upstream regulatory hub that activates p53 while indirectly modulating p16. Markedly, persistent DDR signaling also directly triggers pro-inflammatory pathways such as NF-κB and mitogen-activated protein kinase (MAPK) (17). Additionally, other senescence-associated pathways, such as the mTOR pathway and mitochondrial dysfunction-reactive oxygen species (ROS)-mediated signaling pathway, also frequently play roles in the process of organismal aging. Crucially, these pathways do not operate in isolation but synergistically drive the cellular senescence transition through extensive crosstalk.
Upon entering senescence, cells rewire their metabolic activity and exhibit distinct cytomorphological transformations (such as flattened and enlarged appearance) despite being in a state of growth arrest (18). More importantly, the secretion of a wide range of bioactive factors, including inflammatory chemokines, cytokines, growth factors, receptors/ligands, oxidative factors and extracellular matrix (ECM) remodeling proteases, constitutes a defining pathophysiological feature of cellular senescence, collectively termed as SASP (19). This pro-inflammatory and pro-apoptotic phenotype encompasses IL-1α, IL-1β, IL-6, IL-8, TGF-β, matrix metalloproteinases (MMPs), ROS, and exosomes. Critically, the SASP operates via multimodal signaling cascades. A unique 'bystander effect' occurs wherein senescence extends beyond intracellular impacts as primary senescent cells secrete bioactive signals that alter the microenvironment, thereby inducing secondary senescence in neighboring or distant non-senescent cells through distinct mechanisms (such as autocrine, paracrine and juxtacrine) (20). This transmissible state might also explain why senescent cells exert far-reaching effects even though they constitute an actual low proportion in any particular tissue. In addition, exogenous triggers (such as lipopolysaccharide) and endogenous danger signals lead to the intensification of SASP amplification (21). Notably, these senescence risk factors often interact cumulatively over time, triggering aberrant pathological cascades that ultimately induce chronic low-grade systemic inflammation, even without pathogenic processes.
Mechanistically, senescent cells present a paradoxical duality in their ability to recruit immune cells and activate immunosurveillance. For example, during acute or incipient tissue injury, senescent cells attract, anchor, and activate immune cells via the secretion of specific SASP factors and immunomodulators, facilitating transient immune activation that reinforces tissue plasticity and regeneration, thereby facilitating wound healing and tissue repair (22). However, not all senescent cells produce a pro-inflammatory SASP phenotype. For instance, in bone marrow adipose tissue, the abundance of senescent cells does not exhibit a positive correlation with the expression of proinflammatory factors (23). Furthermore, the concept of the 'Threshold Theory of Senescent Cell Burden' posits that surpassing a critical threshold in the number or percentage of senescent cells can lead to organismal or tissue dysfunction (24). It is also noteworthy that this threshold varies with individual characteristics such as age and health status. Consequently, the failure to effectively and promptly eliminate senescent cells under chronic pathogenic conditions leads to an increase in their burden beyond the tolerance limit of the tissue. This subsequently promotes the SASP to amplify aseptic inflammation and abnormal fibrotic hyperplasia, ultimately exacerbating the disease. Multiple studies have demonstrated that the SASP plays a significant role in the progression of liver disease, with its diverse cytokines exerting distinct effects (Fig. 1; Table I).
Liver tissue harbors diverse cell populations, including hepatocytes (constituting 70-85% of the total) and non-parenchymal cells such as endothelial cells, epithelial cells, hepatic stellate cells (HSCs), and Kupffer cells (KCs) (38). Cumulative evidence from in vivo and in vitro models confirms cell-type-specific senescence susceptibility across hepatic lineages. Thus, senescence of these liver-resident cells can profoundly impact the pathogenesis of inflammatory hepatic disorders (Fig. 2).
Hepatocytes, comprising a substantial proportion of the liver and orchestrating critical metabolic, synthetic and detoxification processes, exhibit age-associated genomic instability, as supported by current evidence (39). Notably, senescent hepatocytes are prevalent across diverse hepatic pathologies, suggesting their relevance as a mechanism underlying liver disease. Data from murine aging models show that, alongside reduced hepatocyte volume, polyploid and/or aneuploid senescent hepatocytes accumulate extensively in the liver (40). A study identified a hepatocyte subset with high telomerase expression; comparative RNA sequencing analysis of hepatocyte subtypes further revealed that this subpopulation enhances liver regeneration during homeostasis and injury (41). Hence, hepatocytes are relatively resistant to telomere attrition, with the causal link between telomere loss and hepatocyte senescence remaining controversial; a plausible hypothesis thus posits that hepatocyte senescence arises via telomere shortening-independent pathways. Enhanced oxidative stress caused by the imbalance of ROS induces cumulative DNA damage in senescent hepatocytes, a feature prevalent in chronic liver disease among the elderly (42). A recent work using a hepatocyte-specific MDM2 knockout mouse model showed that MDM2 inactivation resulted in p53 accumulation in the hepatocytes, followed by elevated its target gene, the cell cycle inhibitor p21, which is a key sign of senescence (43). A comprehensive study analyzing human clinical samples and in vitro models characterized senescent hepatocyte gene signatures in chronic liver disease. It revealed upregulated expression of key senescence markers, including SASP components, p53, and cell cycle regulators (such as cyclin D), in both cirrhotic liver tissues and experimentally induced senescent HepG2 cells (44).
HSCs, normally quiescent under physiological conditions, are primarily recognized for activating in response to liver injury. Single-cell sequencing has revealed that activated HSCs display marked cellular heterogeneity, with distinct subpopulations exhibiting proliferative, inflammatory, or fibrogenic phenotypes tailored to specific functions (45). Notably, a subset of activated HSCs is considered an important source of the pro-fibrotic and inflammatory effects of the SASP. TGF-β, a core SASP factor, is a master regulator of HSC activation, driving their transdifferentiation into myofibroblasts (46). These transformed cells proliferate and secrete ECM components, excessive collagen deposition (such as Col1a1) and dysregulated MMPs, at the site of injury, which alter the structure of the liver and continue to impair its cellular function (47). Importantly, persistent HSC proliferation, combined with impaired senescence mechanisms creates a permissive microenvironment that drives fibrotic progression and even hepatocellular carcinogenesis. Additional inducers of senescence in activated HSCs have been identified, including increased susceptibility to DNA damage. Moreover, IL-22 upregulation induces HSC senescence while inhibiting α-smooth muscle actin expression. Transcriptomic analyses have comprehensively characterized HSCs senescence in human and rodent models, with findings showing that senescent HSCs originate from activated HSCs (48). HSC senescence is largely absent in healthy livers, but increases with disease progression, these senescent cells interact closely with recruited immune cells, possibly in response to SASP mediators.
LSECs, a distinctive endothelial population in the liver, are regarded as key regulators of hepatic homeostasis (49). With aging, these cells exhibit marked phenotypic dysregulation, characterized by ultrastructural and functional changes. LSECs exhibit pseudocapillarization, an ultrastructural alteration involving reduced fenestration number and size, thereby disrupting the physiological filtration barrier, a phenomenon validated across species including humans, murine models and non-human primates (50,51). This, combined with hepatic aging, contributes to increased mitochondrial oxidative stress, reduced intrahepatic nitric oxide availability, upregulated p16 expression and a moderate pro-inflammatory state, such as enhanced expression of IL-1, IL-6 and TNF-α at the mRNA level, as shown in aged rats and human samples (52). In addition, senescent LSECs exhibit increased expression of cell adhesion markers [such as intercellular adhesion molecule-1 (ICAM-1)], leading to an accumulation of substantial leukocyte and reduced hepatic blood flow. This impairs the liver's ability to balance fibrosis and regeneration, indicating an age-related hepatic inflammatory response (49). Research on LSECs of rats has furthermore shown that aged rats exhibit more pronounced reductions in LSEC fenestrations and impaired injury recovery capacity compared with young counterparts, according to the model of CCl4- and phenobarbital-induced cirrhosis (53).
KCs, the resident liver macrophages of the hepatic sinusoids, constitute 80-90% of the body's total macrophage population. These cells serve as sentinels of hepatic innate and adaptive immunity, and their distribution altering during aging (54). Single-cell transcriptomic profiling has revealed that compared with younger counterparts, hepatic macrophages in aged mice exhibit a heightened pro-inflammatory (M1) phenotype, with markedly upregulated expression of pro-inflammatory cytokine genes such as IL-6, TNF-α and IL-1β (55). Therefore, KCs play a pivotal role in hepatic inflammatory aging, where the M1/M2 polarization ratio reflects age-related functional alterations in macrophages. Aging also attenuates macrophage functions, including the phagocytic clearance of extracellular pathogens and antigen-presenting capacity, which impairs bacterial killing and inflammation resolution (56). This phenomenon explains why, despite similar inflammatory responses in young and aged individuals, the capacity for inflammation resolution is markedly impaired in the elderly, resulting in persistent chronic inflammation. Mechanistically, liver macrophages promote bystander cell senescence through TGF-β1 signaling, thereby accelerating aging-related pathologies (such as fibrosis) during hepatic injury (57). Fontana et al (58) revealed that under identical high-fat diet (HFD) conditions, aged mice exhibit exacerbated hepatocellular injury and inflammatory responses compared with young controls. This age-dependent susceptibility is associated with heightened M1-polarized macrophage infiltration both in hepatic and adipose tissues. Macrophage-targeted therapies may effectively reduce susceptibility to age-associated liver diseases.
The human liver, owing to its unique anatomical location, is constantly exposed to numerous xenobiotic and metabolic stressors, including dietary metabolites, bacterial products, pathogens, carcinogens and other substances involved in detoxification and metabolic processes (59). To combat these challenges, specialized hepatic cell populations initiate elaborate stress-responsive pathways, and the liver, renowned for robust regenerative capacity, achieves full recovery even after partial resection or severe acute injury. Nevertheless, cumulative anatomical and functional hepatic alterations driven by aging, disease, or chronic injury lead to an age-related decline in the diverse mechanisms critical for preserving cellular homeostasis (Fig. 3).
Biochemical studies have consistently established the liver as a master metabolic regulator, orchestrating systemic energy balance through hepatic lipid and glucose homeostasis, steroid biosynthesis/degradation and insulin signaling (60). Under homeostatic conditions, cellular ATP is primarily generated from energy substrates (glucose, fatty acids and amino acids) via core biochemical pathways including glycolysis, the tricarboxylic acid cycle, oxidative phosphorylation and amino acid catabolism (61). It is now recognized that dysregulation of these metabolic pathways critically drives chronic liver disease pathogenesis. Single-cell RNA sequencing and proteomic studies have revealed remarkable heterogeneity and plasticity in liver-resident cells. Metabolic reprogramming and bioenergetic shifts within distinct hepatic cell populations drive complex physiological and pathological processes, initiating and sustaining tissue damage while promoting fibrogenesis (62,63). Critically, it remains uncertain whether metabolic dysregulation is a cause or consequence of age-related hepatic pathologies.
It is well established that during liver disease progression, hepatocellular damage and chronic low-grade inflammation induce metabolic reprogramming, which is characterized by dysregulated hepatic metabolism and ectopic lipid deposition (64,65). Growing evidence highlights the pivotal role of aerobic glycolysis in this pathological process. First, aerobic glycolysis promotes a pro-inflammatory state in immune cells, perpetuating hepatic inflammation and injury (66). Second, enhanced aerobic glycolysis sustains HCC progression by facilitating proliferation, metastasis and chemoresistance (67). The pivotal regulatory enzyme pyruvate kinase M2 (PKM2) functions as a central metabolic switch, mechanistically reflecting the Warburg effect observed in hepatocarcinogenesis (65). Experimental and clinical evidence demonstrates that while PKM2 expression is scarcely detectable in healthy livers, its levels are upregulated in HSCs during fibrosis, as well as in KCs and Th17 cells in nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) (68,69). Notably, PKM2 expression increases in precancerous cirrhotic livers and is strongly associated with elevated HCC risk. These findings collectively indicate that glycolytic reprogramming acts as a critical driver of HCC precursor lesions and tumor microenvironment remodeling. Intriguingly, glycolysis is augmented during both replicative and oncogene-induced senescence (70). Mechanistically, disturbances in lipid metabolism, amino acid dysregulation, and impaired mitochondrial energy production synergistically promote inflammation and oxidative stress; pathological features that have been validated in the progression of fibrogenesis (71).
Mitochondria function not only as cellular energy hubs but also as potential triggers of chronic inflammation and cell necrosis (72). With aging, mitochondrial function in senescent cells undergoes complex changes due to intertwined mechanisms, including structural damage (such as swelling, loss of cristae definition and inner membrane disruption), accumulation of mitochondrial DNA (mtDNA) deletions and mutations, and electron transport chain (ETC) impairment (73,74). These alterations also increase ROS production, which can damage cellular components through oxidative stress. Importantly, mitochondria are the most abundant organelles in liver tissue and their dysfunction is a common feature of both hepatic metabolic impairment and disease progression (75). Consistent with this, accumulating evidence confirms that mitochondrial dysregulation contributes to the pathogenesis of age-related hepatic disorders and their progression (76,77). These contain metabolic associated fatty liver disease (MAFLD), hepatic fibrosis, cirrhosis and HCC, among others.
Consistent with the microprotein humanin, plasma levels of another mtDNA-encoded microprotein, MOTS-c, also declined with age. Notably, exercise promotes the production of its metabolite 5-aminoimidazole-4-carboxamide-1-β-4-r ibofuranoside, which acts as an endogenous AMPK agonist and protects against age-dependent and HFD-induced insulin resistance, including in MAFLD (78,79). Mitochondrial dysregulation also drives lipofuscin accumulation and increased lipid deposits. Furthermore, lipid-derived aldehydes are also altered in senescent cells. Given these observations, while a strong link between cellular aging and lipid accumulation has been hypothesized, lipid metabolism within senescent microenvironments remains poorly understood. A recent study demonstrated that both HFD and aging induce mitochondrial dysfunction, reduce lipid oxidation, and cause liver damage (80). Specifically, rats with HFD intake or aging show reduced mitochondrial uncoupling, and decreased superoxide dismutase (SOD) activity is accompanied by increased ROS production, which impairs energy substrate utilization. In this scenario, hepatic lipid accumulation is substantial, and increased pro-inflammatory cytokine release from adipose tissue exacerbates liver damage, worsening with age and occurring earlier in HFD-fed models. Aldehyde dehydrogenase2 (ALDH2) is a mitochondrial enzyme that reduces cellular oxidative stress and prevents associated damage. ALDH2 gene mutation or knockout reduces mitochondrial enzyme activity and mitophagy, increases ROS production, and promotes the development of liver fibrosis-related diseases (81).
Adding to its significance, generally, diminished mitophagy is assumed to be a hallmark of liver diseases. Impaired mitophagy has been observed in most cases of chronic alcohol consumption (82). Indeed, the influence of alcohol on mitophagy depends on exposure duration and dose, with acute high-dose intake inducing excessive mitophagy activating PINK1/Parkin or BNIP3-mediated pathways to drive alcoholic hepatitis progression (83). Another study found that autophagy machinery defects in LSECs from NASH patients upregulate inflammatory pathways (including CCL2, CCL5, TNFα, IL-6 and Tgfb1 expression), confirmed in vitro, and inadequate endothelial autophagy exacerbates liver inflammation to drive disease progression (84). Paradoxically, existing reports are conflicting: For example, autophagy acts as a tumor suppressor, as Beclin 1 disruption accelerates hepatitis B virus-induced premalignant lesions and increases malignancy risk, whereas it can promote established tumors by driving metastasis and drug resistance in later stages (85).
Owing to its central role in metabolism and detoxification, the liver exhibits greater susceptibility to oxidative stress than other organs (86). To counteract such insults, hepatic antioxidant defense systems, comprising enzymatic and non-enzymatic components including SOD, glutathione peroxidase (GPx), catalase (CAT) and glutathione reductase (GR), protect the liver microenvironment from oxidative damage while maintaining physiological function (87). However, chronic exposure to physiological and pathological stressors elevates ROS beyond homeostatic thresholds, triggering oxidative damage to DNA, lipids and proteins. This cascade subsequently activates senescence-associated signaling pathways, culminating in irreversible cell cycle arrest (88). Consequently, oxidative stress represents a critical driver of age-related hepatic injury progression.
Proteomic profiling analysis revealed that in the liver of aged Wistar rats, the defense capacity against oxidative stress, microsomal fatty acid oxidation and peroxisomal function are impaired, accompanied by marked downregulation of antioxidant genes (Cyp2C11, Sod2 and Fmo3) (89). Similarly, long-term alcohol consumption markedly upregulated CYP2E1 gene expression via the CB1R-ERγ-FGF23 axis, inducing hepatic oxidative stress and accelerating the progression of alcoholic liver disease (ALD) (90). During aging, excess ROS markedly disrupt the ECM, primarily by impairing MMP activity. This interaction accelerates ECM breakdown and stiffening, dysregulates MMP activity, and enhances collagen cross-linking and deposition in the liver. Collectively, these alterations remodel the hepatic architecture and drive a pro-fibrotic microenvironment conducive to liver fibrosis. Furthermore, reduced levels of the critical anti-aging factor sirtuin (SIRT)1, potentially due to repression by CCAAT/enhancer-binding-protein/histone deacetylase 1 (C/EBPβ-HDAC1) complexes, may contribute to accelerated liver aging (91). Additionally, the age-related decline in the hepatic NAD+/NADH ratio further impairs SIRT1 function.
Proteostasis, a delicate dynamic equilibrium among protein synthesis, folding, trafficking and degradation, involves quality control mechanisms whose disruption triggers systemic network failure (92). For instance, cellular stress or tissue damage often leads to misfolded and dysfunctional proteins, which gradually form aggregates. Misfolded proteins are degraded by endoplasmic reticulum (ER)-dependent mechanisms, most prominently the unfolded protein response (UPR) and the heat shock response. Impaired proteostasis is considered hallmark of aging, with research showing that proteostatic capacity declines with age, independent of underlying disease (93,94). The mRNA and protein levels of molecular chaperones, which are essential for protein folding, are markedly reduced with aging. Meanwhile, more and more evidence indicates that UPR-regulated protein degradation critically affects chronic liver disease. For instance, ethanol or HFD could induce hepatic ER stress and activate the UPR, causing severe hepatic oxidative damage, inflammation and apoptosis (95). In addition, heat shock proteins (HSPs) facilitate ER signaling in response to stress stimuli. Analyses showed that the induction levels of HSP70, HSP27 and HSP90 in hepatocytes of aged rats exhibit a marked downward trend (51). Heat shock factor 1 (HSF1), a molecular trigger and transcription factor, controls the heat shock response, which is activated during abnormal cellular events. A recent study shows that HSF1 may increase the accumulation of collagen and promote liver fibrosis, whereas inhibition of this factor delays or attenuates fibrotic progression by suppressing the expression of profibrotic markers and cell proliferation (96). Young individuals counteract protein misfolding caused by diverse stressors through intact chaperone responses and proteolytic systems. By contrast, elderly individuals exhibit depleted proteostatic capacity, leading to impaired adaptive defense and repair under oxidative, inflammatory, or other pathological conditions, thereby accelerating disease progression (97,98).
Epigenetics, a reversible heritable mechanism that regulates gene expression without altering the DNA sequence, represents a key mediator of the complex aging process in response to environmental cues (99). Epigenetic events include DNA methylation, histone modification, chromatin remodeling and non-coding RNA regulation. The human DNA methylation landscape changes with chronological aging: Cytosine-phosphate-guanine (CpG) sites in promoter regions typically undergo hypermethylation, while other genomic CpG sites exhibit hypomethylation (100). This signature is linked to the age-associated progression of fibrosis, cirrhosis and even HCC, as validated across diverse cohorts of liver disease patients (101). RNA-seq and DNA methylation array analyses of liver tissues and peripheral blood leukocytes from HCC patients identified DNA methylation as a regulator of chronic liver disease, mediating tumorigenesis, shaping HCC transcriptional landscapes and influencing disease outcomes (102).
The activation or silencing of gene expression by post-translational modifications of histones is bound up with the aging process, with histone acetylation being the most well-documented. Accumulating evidence indicates that alterations in histone acetyltransferase and histone deacetylases affect cellular transformation and hepatic metabolism, thereby disrupting hepatic function and causing liver injury (103,104). For instance, acetylation of histone 3 and histone 4 is a well-recognized alteration in MAFLD (105). Acetylation of H3K9 and H3K18 activates the transcription of key pro-inflammatory genes (such as TNFα, CCL2 and components of the NF-κB pathway), further triggering or exacerbating the inflammatory response in MAFLD (106). SIRTs, nicotinamide adenine dinucleotide (NAD)-dependent class III HDACs that adapt to metabolic stress, are inactivated, thereby driving the induction of a senescent phenotype. These observations confirm that SIRT1 and SIRT3 are essential for redox homeostasis and hepatic lipid metabolism. Notably, MAFLD patients and individuals with alcohol consumption exhibit reduced hepatic SIRT1 and natural SIRT1 activator supplementation protects against these liver diseases (107). MacroH2A1, one of the molecular targets of SIRT1, exists in two isoforms: MacroH2A1.1 and macroH2A1.2 and is involved in cellular senescence and tumorigenic processes. The enhanced expression of the SIRT1-binding isoform macroH2A1.1 protects hepatocytes from lipid accumulation (108). Forkhead box O (FOXO) is a key transcription factor implicated in aging and is regulated by NAD+/SIRT1, promoting the hypothesis that the SIRT1/FOXO axis is a potential target for countering premature cellular senescence and hepatic pathology. In addition, perturbed SIRT3 function in mice is associated with MASLD-like abnormalities and SIRT3 knockout mice are prone to metabolic dysfunction-associated steatohepatitis (MASH) development. Beyond these mechanisms, in vivo metabolic tracing has demonstrated that histone methylation and phosphorylation in intrahepatic cells also contribute to the development of ALD (109).
Non-coding RNAs (ncRNAs) constitute a diverse class of regulatory molecules that have emerged as epigenetic factors affecting aging, including microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs) (110). Their ubiquitous presence in the tissues and biological fluids of different species under diverse pathological conditions underscores their significant role in liver biology. Among the mechanisms regulating fibrogenesis, miRNA-mediated pathways, which operate through senescence-associated regulatory protein control, have been the most extensively studied. In MAFLD and HCC, for instance, miR-21 overexpression increased p53 activity (a protein that normally suppresses cell cycle proteins and lipogenesis) (111). Notably, alongside increased miR-221 expression in hepatitis C virus (HCV) infection, miR-221 is elevated in MASH-associated fibrosis (112). Besides, expression levels of miR-19a, miR-19b, miR-122, miR-125b, miR-192 and miR-375 are increased in MAFLD individuals (113). Fortunately, unlike DNA mutations, these regulatory and often reversible changes enable the design of new anti-aging therapies.
The past decade has seen a surge in rigorously designed clinical and mechanistic studies exploring the involvement of cellular senescence in the pathogenesis of diverse hepatic diseases. This surge is driven by the recognition that cellular senescence is a stress adaptation. At the macroscopic level, the prevalence of numerous liver diseases, such as MAFLD, increases with age and advanced liver diseases, such as HCC, are more common in older compared with younger individuals (114). In addition, cellular senescence drives progressive decline in cell viability, liver function and tissue regeneration, accelerating the progression of diverse diseases. Although the detailed mechanisms and biological functions, such as determining whether cellular senescence is a causative factor or an epiphenomenon of pathological phenotypes, remain incompletely elucidated, substantial progress has been made in this field. Subsequent sections open with a concise overview of clinical entities underpinning disease pathophysiology, proceeding to discuss experimental evidence that links cellular senescence to liver cell subtypes impacted by chronic hepatic inflammation (Fig. 4).
Hepatitis B virus (HBV), a member of the Hepadnaviridae family, causes liver damage through acute and chronic infections. The latter accounts for the vast majority of severe disease burden (115). While HBV infection rates are highest in infancy and childhood, chronic progression occurs in only ~5% of adults infected later in life due to reduced exposure risk. By contrast, elderly patients exhibit markedly higher rates of progression to chronic hepatitis B (CHB) following disease onset, compared with <5% in younger individuals (116). An outbreak investigation of acute HBV in a Japanese nursing home revealed nearly 60% chronicity among patients aged >65 years (117). Moreover, advanced age is a risk factor not only for cirrhosis progression but also for HCC development. This phenomenon may be attributed to an age-related decline in pathogen-specific immune responses, warranting further research to elucidate the underlying pathophysiology and immunological mechanisms in this population.
Age-dependent telomere attrition serves as a robust indicator of the cumulative effect of cellular aging on inflammation in HBV infection. Available evidence indicates that peripheral blood mononuclear cells lose ~50 bp of telomeric DNA per year on average, however, this loss is likely to increase during chronic viral infections (118). Notably, in CHB, hepatocytes exhibit elevated p21 expression and G1 phase arrest, indicating the presence of p21-mediated G1 arrest and all indicative of a strong senescent response. For example, telomere length measurements in HBV, performed in hepatocytes, showed shorter telomeres in both liver biopsy tissue and peripheral blood lymphocytes of HBV-infected patients (119). Consistent with this, another study identified fewer and shorter telomeres in CHB patients compared with matched controls (120), whereas both symptomatic and asymptomatic HBV-positive individuals exhibited elevated serum telomerase activity, probably countering telomere shortening-induced cellular senescence (121). Extensive hepatocyte telomere shortening aligns with accelerated aging, telomere length is comparable to that of individuals with normal aging of 10 years and can be extended to 15 years in the case of more severe fibrosis in this pathological context, which helps to explain, at least in part, the pathological features of accelerated aging (122).
The mechanistic crosstalk between telomere attrition and DDR is well-established and underpinned by persistent DDR activation that, when surpassing cellular repair capacity ultimately triggers premature senescence. Existing data demonstrate that aged liver samples exhibit aberrant DNA methylation at multiple loci, this pattern of epigenetic alteration is further exacerbated in CHB. An earlier study revealed more pronounced DNA damage in peripheral lymphocytes from HBV patients (123). As aforementioned, the pathology of cellular senescence is involved in the phenotypic alterations of infected cells, a notion that at least partially justifies some conflicting findings. For instance, although most HBV-encoded proteins drive aging or enhance hepatocarcinogenesis risk, HBx represents an exception: Methylated HBx downregulates p16INK4a and p21Waf1/Cip1, inactivates Rb phosphorylation and thereby enables escape from cellular senescence (124). Notably, another study on the interaction between HBV and cellular senescence in patients with CHB, in which HBx is not involved as a key player (125).
Beyond this, early-life HBV exposure induces premature immunosenescence. Specifically, viral-mediated inhibition of topoisomerase I induces topological DNA damage and telomere attrition in CD4+ and CD8+ T cells. Epidemiological studies on HBV-endemic individuals carrying the HBV-specific T-cell marker TCR Vβ12, revealed lower CD3+ and CD4+ T-cell counts, indicating impaired immune function from CHB (126). Mechanistic studies demonstrated that in a mouse model of HBV persistence-induced systemic tolerance, hepatic CD4+ T cells continuously produce IFN-γ, which stimulate CXCL9 secretion by KCs to facilitate CXCR3-dependent retention of antiviral CD4+ T cells, ultimately driving their apoptotic clearance (127).
Age-related immunosenescence and immune checkpoint dysfunction profoundly influence HBV pathogenesis. Under physiological conditions, harmful senescent cells are cleared by recruited immune cells, such as monocytes and macrophages. During aging, nevertheless, immune surveillance and host defense mechanisms become widely suppressed and rendered ineffective in HBV, resulting in insufficient clearance of senescent and precancerous cells, a process that appears to play an important role in carcinogenesis (126,128). In vivo and in vitro findings indicate elevated SASP levels in chronic HBV infection, with excessive secretion of its components (growth factors, interleukins and MMPs) promoting HBV-associated HCC development, an effect exacerbated by aging (129). Compared with HBV-negative HCC cells, HBV-positive cell lines showed increased MMP-9 expression, reduced tissue inhibitor of metalloproteinase expression and higher invasive potential (130). Similarly, HBsAg-positive patients have higher peripheral blood TNF-α and IL-6 levels (131). Of note, consistent with these findings, a clinical study conducted by Rosenberg et al (132) found that donor-derived T cells from older donors exhibit reduced hepatitis B surface antigen-specific proliferative response, while younger donors show stronger responses.
MAFLD, previously termed as NAFLD, is characterized by hepatic steatosis and affects up to one-third of the global population. While the age of MAFLD onset has decreased in recent years, its prevalence, risk of hepatic/extrahepatic complications and rates of all-cause and disease-specific mortality increase with advancing age (133). Moreover, geriatric patients harbor more MAFLD risk factors, including hyperlipidemia, obesity, diabetes and hypertension and exhibit more severe biochemical, hematological and histological alterations than younger cohorts. A cross-sectional analysis of adult participants found that elderly patients with MAFLD had a higher likelihood of MASH and advanced fibrosis compared with younger adults (134). The detrimental effect of aging on hepatic homeostasis manifests through cellular-level structural damage, progressive decline in liver function and metabolic dysregulation. Extensive research indicates that MAFLD-associated pathological changes, including abnormal hepatocyte morphology, lipofuscin accumulation and ECM deposition, are associated with accelerated liver aging, potentially driving age-related hepatic steatosis. Notably, Gao et al (135) demonstrated that lipid accumulation directly exacerbates hepatic aging. These findings suggest that the pathogenesis of MAFLD may be intrinsically linked to biological aging. However, whether cellular senescence initiates or results from hepatic metabolic dysfunction and inflammation remains unresolved.
Clinical and preclinical evidence supports a link between cellular senescence and MAFLD. Senescent hepatocytes are more abundant in patients with MAFLD and, as observed in other chronic liver diseases, their senescence levels correlate with the severity of MAFLD, as confirmed by liver biopsies in these patients (136). Several markers of senescence, such as SA-β-Gal, p16, p21 and p53, are elevated in individuals with MAFLD/MASH (137). Similarly, rodent studies demonstrate that HFD reduces the phosphorylation of Rb by upregulating p16 and p21, inducing hepatocyte cell cycle arrest, activating senescence pathways, and ultimately promoting MAFLD (138). Analogously, in p53-null mice, inhibition of p66Shc signaling reduces hepatic lipid peroxidation and hepatocyte apoptosis, delaying nutritional steatohepatitis development (139). Meanwhile, a similar effect occurs after the systemic clearance of p16-expressing cells. DNA methylation additionally serves as a key mechanism: In high-fructose/high-cholesterol-fed mice, its patterns alter liver lipid gene expression (upregulating lipid genes and downregulating proliferation/transcription genes) and specific histone modifications induce proliferation arrest (140). Other markers of senescence (genomic instability and DNA damage), are also directly associated with the progression of MAFLD.
Of great importance, immune system aging is closely linked to MAFLD/MASH via proinflammatory factor secretion. The local microenvironment and the aging immune cell population engage in a vicious cycle, the resultant inflammatory milieu triggers and amplifies microenvironmental responses, thereby exacerbating hepatic dysfunction. A recent pivotal study identified that marked hepatic T-cell senescence and exhaustion drive metabolic liver disease progression in humans (141). Specifically, in contrast to normal individuals, patients with MASH show upregulated expression of senescence- and exhaustion-related genes in hepatic CD4+ and CD8+ T cells, along with increased expression of IL-2, IL-15 and IL-18 in these cells. Hepatic macrophage accumulation relies on CCL2-CCR2-mediated chemotaxis: M1 activation (marker CD86) is associated with MAFLD severity in humans, and older mice show more pronounced M1 infiltration in both liver and adipose tissue (142).
ALD is a globally prevalent condition driven by chronic excessive alcohol consumption, posing a serious health threat to ~75 million individuals (143). Alcohol metabolites generated during hepatic processing produce toxic substances that trigger hepatocyte inflammation and severe liver damage via inflammatory cascades involving cytokines, chemokines and ROS (144). Evidence indicates higher rates of binge drinking among middle-aged and older adults, with the risk of alcohol abuse being further elevated by age-related metabolic changes, smoking, or illicit drug use. Chronic low-grade inflammation in older adults, characterized by elevated circulating pro-inflammatory cytokines and tissue inflammatory cell infiltration, exacerbates ALD progression (145). Concurrently, emerging evidence demonstrates that ALD accelerates cellular senescence. Murine ALD models show increased senescence-associated biomarkers, such as miR-34, p16, p21 and p53, along with elevated SASP factors (TGF-β1, PAI-1 and CCL2) (146). Additionally, ALD patients exhibit significant hepatic telomere shortening, accompanied by upregulation of telomere-binding protein genes, which suggests a compensatory response to DNA damage (147).
Notably, immunosenescence plays a central role in ALD pathogenesis through senescence-associated regulatory mechanisms. Following chronic ethanol exposure, activated immune cells trigger cytokine and chemokine production, initiating a pro-inflammatory cascade (148). Current evidence indicates that most KCs polarize predominantly toward pro-inflammatory M1 phenotypes, releasing abundant cytokines including IL-1β, IL-18, TNF-α, IL-12 and IL-23. These mediators induce potent innate immune responses and critically contribute to alcohol-induced liver injury. By contrast, activated M2 macrophages increase in number and secrete abundant anti-inflammatory factors during tissue repair following injury (149). Nevertheless, macrophage polarization remains vaguely defined and controversial due to the inherent plasticity of these cells and their context-dependent responses to pathological signals. Notably, Wan et al (150) demonstrated that M2-polarized macrophages induce hepatocyte senescence via IL-6 secretion, accelerating alcohol-induced senescence both in vivo and in vitro Extensive evidence indicates that chronic ethanol exposure alters T-cell immunophenotypes in mice and humans. Liver biopsy specimens from patients with moderate-to-severe ALD reveal significant CD3+ T cell infiltration and activation (including CD4+ and CD8+ subsets) (151) Compared with controls, T cells from alcohol-fed mice produce elevated levels of IFN-γ and IL-4. A pronounced Th1-dominant response promotes ALD progression through IL-2, TNF-α, and IFN-γ production, while Th17 cells exacerbate liver damage via IL-17 secretion. Notably, Th17 cell populations expand with aging, and Th17-derived IL-17 enhances SASP production, thereby amplifying the inflammatory cascade (152).
Liver fibrogenesis constitutes a highly integrated, dynamic process involving molecular, cellular and tissue-level changes. This process is primarily characterized by excessive ECM deposition, driving extensive disruption of hepatic architecture and functional integrity (153). Organ fibrosis represents a hallmark manifestation of chronic inflammatory disease progression, accounting for ~45% of global all-cause mortality. Accordingly, hepatic fibrosis development critically determines both disease prognosis and patient quality of life (154). The aging global population and the rising prevalence of fibrosis-predisposing conditions (including viral hepatitis, MAFLD, ALD, primary sclerosing cholangitis and autoimmune hepatitis) are driving the growing burden of liver fibrosis. Regardless of etiology, clinical severity escalates substantially with fibrotic septa formation and progression toward cirrhosis, leading to increased mortality and HCC incidence. Globally, liver diseases account for 1.5-4% of annual deaths (155,156). Experimental and clinical studies have demonstrated that matrix remodeling and partial architectural restoration can occur even in advanced fibrosis/cirrhosis, suggesting this stage represents a potential therapeutic window in chronic liver disease progression (157). Thus, targeted investigations of the underlying pathophysiological mechanisms are needed.
Fundamentally, fibrosis is not a unidirectional process but rather a dynamic interplay of factors including persistent inflammation, dysregulated ECM remodeling, and myofibroblast formation and proliferation during tissue wound healing and fibrogenesis (158). Aging populations exhibit increased susceptibility to irreversible fibrotic damage due to a diminished wound-healing capacity and impaired fibrosis resolution. Substantial emerging evidence indicates that established biomarkers of cellular senescence in liver tissue (such as miR-378, miR-18b-3p and the lncRNA-ATB/miR-200a/β-catenin axis) are elevated in patients with liver fibrosis and cirrhosis, underscoring the role of senescence in the pathogenesis of these disorders (159). The SASP drives fibrosis by establishing a pro-inflammatory and pro-fibrotic microenvironment. Wijayasiri et al (160) found that HSCs exposed to conditioned media from senescent HepG2 cells exhibited activation and secreted abundant inflammatory and fibrotic mediators. HSC activation represents a key pro-fibrotic consequence of the SASP. Furthermore, during fibrotic progression, activated HSCs can undergo spontaneous senescence. TGF-β key component acts as a potent activator of HSCs, driving their transformation into myofibroblast-like cells and promoting excessive ECM protein production. Notably, immune defenses are compromised in aging patients with fibrosis, increasing susceptibility to inflammation and infection-related complications (161). The recruitment of abundant immune cells (neutrophils and macrophages) to the liver via elevated pro-inflammatory SASP factors, including IL-6, IL-8 and TNF-α, perpetuates cycles of oxidative stress, inflammation and fibrosis, a phenomenon that intensifies with aging. Single-cell transcriptome analyses have revealed expanded populations of CD4+ T cells, CD8+ T cells and γδ T cells in fibrotic liver tissue, with further increases observed in cirrhosis (162). Typically, Th1 and Th17 cells accelerate fibrotic progression by acting on hepatocytes, KCs and HSCs through pro-inflammatory mediators. By contrast, substantial evidence indicates that Treg cells exert antifibrotic effects, partly mediated by IL-10-dependent immunosuppression (163). In brief, the convergence of inflammation, immune cells mobilization, and activation of HSCs and hepatocytes drives both liver fibrosis and aging-related pathology.
Additional links between cellular senescence and fibrosis include telomere attrition and mitochondrial dysfunction. Dysregulation of the mitochondrial ETC drives oxidative stress, damaging cellular components and promoting fibrogenesis. Besides, the age-related decline in mitochondrial function predisposes the liver to heightened stress sensitivity and fibrosis susceptibility. In accordance with this, substantial release of mtDNA and mitochondria-derived damage-associated molecular patterns has been observed in both human patients and CCl4-induced mouse models with significant liver fibrosis, directly implicating mitochondrial dysfunction in fibrotic pathogenesis (164,165). Given the age-dependent prevalence of hepatic fibrosis/cirrhosis and the established role of telomere attrition as a biomarker of biological aging, replicative senescence is likely to contribute to fibrosis/cirrhosis pathogenesis. Calado et al (166) demonstrated that telomerase abnormalities are associated with hematological disorders and severe liver diseases characterized by fibrosis and inflammation. In line with this, a cross-sectional study revealed an inverse correlation between telomere length and cirrhosis severity, while mutations in telomerase-related genes were identified as cirrhosis risk factors (167,168). Of great importance, emerging evidence positions telomere attrition as a genetic determinant of cirrhosis susceptibility (169).
HCC, as the predominant form of primary liver cancer, accounts for ~90% of hepatic malignancies. Its development is associated with multiple risk factors, including chronic alcohol consumption, persistent viral infections (such as HBV/HCV), chemical exposures, and metabolic disorders (170). Regardless of etiology, hepatocarcinogenesis typically involves recurrent cycles of hepatocellular injury, inflammation and necrosis, processes exhibiting marked molecular and cellular heterogeneity and irreversibility (171). According to GLOBOCAN data, the global incidence of liver cancer reached 906,000 new cases with 830,000 mortalities annually, reflecting rapidly rising rates that impose a substantial global health burden (172). Epidemiological studies consistently demonstrate an age-dependent increase in HCC incidence, with markedly higher rates in individuals over 75 years (173). Notably, older adults may develop HCC even in the absence of fibrosis or cirrhosis, probably due to age-related physiological and metabolic alterations, underscoring the direct role of aging in hepatocarcinogenesis (174).
HCC predominantly arises from persistent viral infections or sterile inflammation These pathological states impair telomerase activity, accelerate telomere shortening, induce hepatocyte malignant transformation and foster a pro-tumorigenic microenvironment. Crucially, senescent cells act as key initiators of this process, serving as chronic inflammatory foci throughout the natural course of the disease (175). Senescent hepatocytes disrupt the local microenvironment through sustained secretion of proinflammatory mediators. These cell populations engage in a self-perpetuating cycle with their microenvironment: Inflammatory signaling induces senescence, which further amplifies microenvironmental inflammation and exacerbates hepatic dysfunction. For example, Li et al (176) revealed that the activation and senescence of HSCs were observed in both human and murine liver tumors, concomitant with SASP secretion. Similarly, in diethylnitrosamine (DEN)/CCl4 -induced HCC models, senescent hepatocytes upregulate SASP secretion (especially IL-8) during hepatocarcinogenesis and a similar phenomenon is observed in human HCC tissues (177). Furthermore, cellular senescence in this context can be prematurely induced by diverse stressors or naturally by replicative exhaustion. Research from Ho et al (178) found that aged mice with senescent β-catenin-deficient hepatocytes developed an inflammatory microenvironment that promoted HCC progression. Intriguingly, as aforementioned, the pathological role of cellular senescence is also environment-dependent, which partly explains some conflicting findings. Senescence is proposed to function as a physiological tumor-suppressive mechanism by recruiting immune cells and inhibiting the progression from benign to malignant lesions in early disease. Therefore, its complex role in cancer needs to be studied under specific conditions.
Indeed, age-related immune dysregulation and immunosenescence profoundly affect the long-term HCC prognosis. As HCC progresses to advanced stages, multiple hepatic cell types, such as immune cells, hepatocytes and endothelial cells, acquire senescence-associated phenotypes, driving systemic failure of antitumor defenses in HCC patients. This process is age-exacerbated, driven by inflammatory cascade signaling and amplified SASP production. RNA sequencing of treatment-naïve HCC tumor has revealed increased Tregs and exhausted CD8+ T cell accumulation (179). Comparative analyses further showed elevated levels of Tregs and CD25+ CD4+ T cells in the peripheral blood of HCC patients compared to healthy individuals, with higher serum immunosuppressive cytokines (TGF-β1 and IL-10) (180). Paradoxically, a meta-analysis showed that higher infiltration of neutrophils and Tregs were correlated with improved overall and disease-free survival, suggesting context-dependent immune roles (181). Notably, tumor-associated macrophages within the tumor microenvironment display aberrant activation. Another study found that M2 macrophages, along with Tregs, Th2, and Th17 cells, increase with HCC progression, and produce elevated levels of IL-6, VEGF, Arg1 and IDO, thereby promoting immune evasion and tumor progression (182).
Fundamentally, transient cellular senescence is a physiological process that supports tissue homeostasis and embryonic development by promoting acute tissue repair and limiting its further deterioration. By contrast, the chronic persistence of senescent cells has the opposite effect, their progressive accumulation in the organism impairs tissue and organ function, ultimately driving aging and age-related diseases. In the hepatic system, growing evidence highlights the central role of cellular senescence in the initiation, progression, and exacerbation of liver disease (183). Consequently, senescence-targeted interventions (senescence therapies), can serve as potential treatments to improve liver status and extend healthy lifespan during aging. Surprisingly, the effectiveness of using senolytics to eliminate senescent cells from tissues to delay illness has been elucidated. Up to now, senotherapeutic strategies primarily include senolytics (drugs that selectively clear senescent cells) and senomorphics (agents that inhibit SASP), as discussed below (Tables II and III; Fig. 5).
Senolytics, which encompass small molecules, peptides and antibodies, selectively eliminate senescent cells while sparing normal proliferating cells. These agents induce apoptosis in senescent cells by targeting key survival pathways known as senescent cell anti-apoptotic pathways, including p53, p21 and PI3K/Akt signaling, BCL-2 family members, HSP-90 and other targets (21). Recent systematic screening approaches have identified numerous senolytic compounds with therapeutic potential.
The first-generation senolytic combination of dasatinib and quercetin (D+Q) induces more potent apoptosis and ameliorates age-related pathologies compared with either compound administered alone. Preclinical studies demonstrate that D+Q alleviates multiple age-related conditions in mice, including insulin resistance, hepatic steatosis, pulmonary fibrosis and chronic kidney disease (184,185). Mechanistic studies in aged mice have revealed that D+Q triggers apoptosis in senescent hepatic progenitor cells, effectively reducing senescence-associated phenotypes including reduced SA-β-gal activity and clearance of p16INK4a-positive cells in the liver (186). Additional evidence confirms that this senolytic combination attenuates hepatic steatosis and ameliorates MAFLD pathology in HFD-induced models (187). Conditional knockout of fructose-1,6-bisphosphatase 1 (FBP1), a key tumor-suppressive metabolic enzyme, induces hepatic metabolic dysregulation in mice. This promotes HSC senescence, which accelerates hepatocarcinogenesis via SASP mechanisms. Li et al (176) demonstrated that intermittent D+Q treatment selectively eliminated senescent HSCs in hepatocyte-specific FBP1-deficient mice, thereby halting HCC progression. Moreover, D+Q reduced HCC-associated gene expression in Sod1−/− mice and markedly lowered HCC incidence (188). While this senolytic regimen shows therapeutic potential, its precise mechanisms require further exploration. Paradoxically, another preclinical study reported that D+Q exacerbated liver histopathology and tumorigenesis, accelerating disease progression without effectively clearing senescent cells (189). Given the exclusively preclinical evidence regarding D+Q and liver interactions, rigorous studies are warranted to evaluate therapeutic efficacy, while human safety profiles require thorough assessment.
The BCL-2 family proteins, including BCL-2, BCL-XL, and BCL-W, serve as key regulators of cellular survival and death. These proteins are upregulated during senescence across diverse cell types, establishing them as prime targets for senolytic therapies (190). ABT-263 (navitoclax), a pan-inhibitor of BCL-2 family proteins, suppresses tumor metastasis and recurrence by eliminating chemotherapy-induced senescent cancer cells and hepatocytes within the tumor microenvironment. Preclinically, its demonstrated dose-dependent pro-apoptotic effects against HCC in vitro have now been validated in a phase I clinical trial (NCT01364051) involving HCC patients (191,192). Gold et al (193) recently established that ABT-263 clears senescent stellate cells and hepatocytes in ALD mice, concomitantly attenuating hepatic triglyceride deposition. Ex vivo analyses confirmed caspase-3-mediated apoptosis in acetaldehyde-induced senescent hepatocytes following ABT-263 treatment. ABT-737, a precursor to ABT-263, exhibits senolytic activity in experimental models. In murine models, ABT-737 synergistically enhances sorafenib-induced apoptosis in hepatoma cells, with the combination therapy demonstrating superior suppression of xenograft tumor growth compared with sorafenib monotherapy (194). However, ABT-737 lacks oral bioavailability and exhibits low aqueous solubility. Structural optimization subsequently yielded its derivative ABT-263, which was developed as an orally bioavailable BCL-2 inhibitor (195). Targeted inhibition of BCL-XL may mitigate side effects (such as thrombocytopenia and neutropenia) associated with pan-BCL-2 inhibition while preserving therapeutic efficacy (196). Indeed, BCL-XL inhibitors A-1331852 and A-1155463 demonstrate senolytic effects in senescent HUVECs and human lung fibroblasts (196). Although Cucarull et al (197) reported that A-1331852 restored regorafenib efficacy (a second-line HCC therapeutic) in both xenograft models and in vitro systems, BCL-XL inhibitors remain underexplored for liver pathologies. Further studies are needed to evaluate their safety and efficacy profiles. Overall, BCL-2 family inhibition represents a mechanism-based strategy for senolytic discovery, however, their associated side effects hinder clinical translation.
Research has revealed that forkhead box protein O4 (FOXO4) regulates senescent cell viability by binding and sequestering nuclear p53, thereby inhibiting apoptosis. Based on this mechanism, a retro-inverso peptide, FOXO4-D-retro-inverso (FOXO4-DRI), was developed to disrupt FOXO4-p53 interaction and selectively induces apoptosis in senescent cells (198). Furthermore, FOXO4-DRI conferred protection against doxorubicin-induced accelerated senescence and hepatic damage in mice, suggesting its potential as a therapeutic strategy against senescence-driven liver pathologies (199). However, the short in vivo half-life of FOXO4-DRI necessitates development of stabilization approaches or alternative p53-activating strategies for effective senescent cell clearance.
As HSP90 is a cancer-promoting protein that regulates apoptosis, proliferation and angiogenesis, its inhibition has emerged as a validated strategy for cancer chemotherapy (200). Several HSP90 inhibitors, including 17-DMAG (alvespimycin), 17-AAG (tanespimycin), ganetespib and geldanamycin, exhibit senolytic effects across multiple senescent cell types through distinct, cell-type-specific mechanisms. For example, 17-DMAG suppresses HCC cell proliferation in a time- and concentration-dependent manner, probably by modulating cyclin D1, p53 and NF-κB protein levels (201). Mechanistically, studies indicate that 17-DMAG induces apoptosis in senescent cells, partially through disruption of the HSP90-AKT interaction and impairment of AKT activation (202-204). In both chronic and acute models of alcoholic liver injury, 17-DMAG attenuated oxidative stress and modulated inflammatory responses (205). Another HSP90 inhibitor, ganetespib (STA-9090), was well-tolerated in a phase I dose-escalation trial involving patients with advanced HCC who had progressed after at least one line of systemic therapy, though it did not confer significant clinical benefit (206). However, subsequent preclinical studies revealed that the ganetespib-sorafenib combination synergistically enhanced cytotoxicity by disrupting HIF-1α stability, increasing p62 accumulation, and inhibiting autophagy, thereby augmenting sorafenib monotherapy efficacy (207). Collectively, these findings indicate that sustained research is essential to maximize the clinical translation potential of this therapeutic combination. Critically, precise identification of the dominant senolytic mechanisms in each disease context is essential, as these mechanisms vary markedly across cell types.
Several natural products and their derivatives, including fisetin, piperlongumine and matrine, also possess senolytic properties. For example, fisetin acts through multimodal mechanisms by targeting key regulators such as the PI3K/AKT, BCL-2, p53 and NF-κB signaling pathways (208). In a Lieber-DeCarli ethanol diet-induced ALD mouse model, fisetin alleviated age-related pathologies and improved the local tissue microenvironment in vivo via anti-inflammatory and anti-fibrotic mechanisms (209). Sundarraj et al (210) discovered that fisetin dose-dependently reduced autophagic flux by activating the PI3K/AKT/mTOR signaling and modulating the AMPK pathway, thereby showing therapeutic efficacy against HCC. Piperlongumine, a natural amide alkaloid from long pepper, selectively induces senescent cell death by increasing ROS production and inhibiting PI3K/AKT and NF-κB pathways. It also exerts hepatoprotective and anti-fibrotic effects in bile duct ligation (BDL)-induced liver fibrosis model (211). Due to its biological characteristics, the precise senolytic mechanism of piperlongumine remains unclear. Researchers have developed several piperlongumine analogues (compounds 47-49) through a series of structural modifications. These analogs exhibit enhanced senolytic activity and modulate multiple antioxidant enzyme expressions (212). Nonetheless, most natural compounds exert multi-target, multi-pathway effects, making it challenging to attribute their therapeutic benefits exclusively to senescent cell clearance. Moreover, since senescent cells exert context-dependent beneficial effects, selectively targeting the SASP may represent a safer and more effective anti-aging strategy.
While completely halting physiological aging remains unrealistic, modulating pathways associated with premature senescence represents a promising therapeutic strategy to counteract accelerated aging in liver disease. Senomorphics inhibit detrimental effects of SASP without inducing senescent cell death. In general, these agents suppress SASP expression by targeting key signaling pathways, including mTOR, NF-κB, SIRT1/SIRT3 and JAK. thereby inhibiting cellular senescence (221). While less targeted than senolytics, this approach represents viable intervention strategy.
Originally developed as an anti-fungal agent, rapamycin (sirolimus) ranks among the best-established senomorphic compounds for modulating aging and suppressing the SASP. In multiple animal models, rapamycin has been found to extend lifespan not only in worms and drosophila, but also to increase both median and maximum lifespan in mice when administered later in life (222,223). Evidence confirms that rapamycin effectively suppresses SASP markers, reduces cellular senescence and thereby ameliorates age-related functional decline or delays tumorigenesis in rodent models (224). Mechanistically, rapamycin primarily inhibits mammalian target of rapamycin complex 1 (mTORC1), although chronic administration also attenuates mTORC2 signaling, while potently blocking the translation of membrane-bound IL-1α. This reduction in IL-1α suppresses NF-κB transcriptional activity, consequently inhibiting secretion of most SASP components (225). Notably, the mTOR signaling pathway has expanded markedly beyond coordinating cellular growth and metabolism in response to nutrient availability, and now includes the regulation of aging processes, particularly cellular senescence. Additional mechanisms by which rapamycin may modulate aging, such as nuclear factor erythroid 2-related factor 2 (Nrf2) pathway activation, are under investigation. Lee et al (226) investigated the therapeutic effects of rapamycin in transgenic mice with HCC induced by activated HrasG12V and p53 suppression. A low-dose of rapamycin effectively prevented HCC initiation but failed to inhibit established tumor growth. This stage-specific efficacy was mechanistically linked to the expansion of CD4+ Tregs and suppression of mTOR downstream targets (including 4E-BP1 and S6K1). Similarly, oral administration of rapamycin effectively inhibits activated hepatic mTOR in ALD mice and inhibits thioacetamide-induced liver fibrosis (227,228). In HFD-induced mice, rapamycin suppresses pro-inflammatory NFκB signaling by increasing p65 and IκBα interaction (229). Regrettably, chronic mTORC1 inhibition via long-term rapamycin administration increased hepatic inflammation and liver tumor incidence (230). Besides, rapamycin and its analogs exhibit various side effects (including pneumonitis, immunosuppression, kidney toxicity and thrombocytopenia and impaired wound healing) complicating long-term therapeutic use (231) Developing next-generation rapamycin analogs may overcome these adverse effects while improving potency and pharmacokinetic profiles.
Metformin, the first drug evaluated in the Targeting Aging with Metformin trial, modulates multiple aging pathways including inflammatory regulation, oxidative stress defense and proteostasis. For example, metformin reduces SA-β-gal activity and downregulates senescence markers across diverse senescent cell types (232). It protects against liver injury in acute viral hepatitis by regulating mTORC1 signaling and mitochondrial function to control effector T-cell activation. Metformin is also well-tolerated in NAFLD patients, though its effects on senescence phenotypes remain unassessed in these contexts (233,234). Metformin activates the adenosine monophosphate-activated protein kinase (AMPK) pathway, demonstrably preventing age-related loss of hepatic sinusoidal fenestration through chronic treatment in young mice and reversing this via acute treatment in aged mice, as shown in LSECs (235). It could be hypothesized that the effect of metformin on LSECs may contribute to the beneficial outcomes of AMPK agonists for liver fibrosis treatment. In vitro studies reveal that metformin suppresses HepG2 cell proliferation by inducing cell cycle arrest at the G0/G1 phase, as evidenced by elevated KLF6/p21 protein levels and enhanced AMPK activation (236). This provides a basis for metformin's therapeutic potential in HCC. Corresponding clinical data reveal that prediagnostic metformin use markedly prolonged overall survival in older patients with HCC and type 2 diabetes (237). However, while metformin regulates senescence, its broader effects against age-related pathologies likely involve both senescence-dependent and -independent mechanisms.
SIRT1 expression declines with age, yet its increased expression attenuates cellular senescence and prolongs longevity across species. Resveratrol, a natural SIRT1 activator, exerts therapeutic effects against obesity, tumorigenesis and age-related organ dysfunction (238). As a senomorphic and antioxidant agent, resveratrol prevents cellular senescence by activating telomerase via the PI3K-Akt pathway (239). Additionally, it suppresses SASP factors by inhibiting NF-κB signaling while upregulating the antioxidant Nrf2 pathway. In inorganic mercury-induced liver fibrosis, resveratrol activates the SIRT1/PGC-1α pathway, alleviating oxidative stress and suppressing hepatic stellate cell activation (240). Across multiple human HCC cell lines, resveratrol inhibits viability, proliferation, invasion and migration in a time- and dose-dependent manner by activating p53 and suppressing the PI3K-Akt signaling (241,242). At high concentrations, however, resveratrol exerts pro-oxidant effects inducing growth arrest, senescence, or apoptosis (243). While resveratrol extends organismal lifespan, its murine effects are context-dependent, markedly prolonging survival in HFD models but not in standard-diet controls (244). Besides, the poor bioavailability and instability of resveratrol necessitate developing structurally distinct SIRT-activating compounds. SRT1720, which is 1,000-fold more potent than resveratrol, extends lifespan in HFD mice (22). It demonstrates therapeutic efficacy against both MAFLD and ALD, though its mechanisms remain incompletely characterized (245,246).
NAD+, an upstream regulator of SIRT1, modulates both the SIRT1 and NF-κB pathways. These pathways antagonize each other in inflammatory and metabolic processes and critically regulate pathogenic mechanisms in MAFLD progression (247). The senomorphic NAD+ precursor NAM improves metabolic function in ALD mice by inhibiting cellular senescence and suppressing SASP production, while reducing liver injury markers (193). Although NAM shows promising therapeutic potential, its adverse effect profile and optimal dosage require careful evaluation. Further trials are needed to establish safe regimens and assess long-term patient outcomes.
The past decades have witnessed considerable advances in the fundamental role of cellular senescence in health and various illnesses (255). Accumulating evidence identifies cellular senescence as a primary driver of inflammatory liver diseases. Global laboratories and clinical translational investigators are exploring whether senescence-targeted interventions can delay or reverse cellular senescence to restore hepatic physiological homeostasis. As detailed in the present study, aging and SASP-mediated intrinsic hepatic cellular senescence are vital risk factor in the pathogenesis of chronic inflammatory liver diseases. Current understanding holds that inflammation-related triggers, including viral infection, alcohol and HFD, induce metabolic reprogramming, mitochondrial dysfunction, oxidative stress, loss of proteostasis and epigenetic changes, driving cellular senescence in endothelial and immune cells (256). Subsequently, senescence in these cells will promote senescence in adjacent normal cells via SASP secretion, disrupting the hepatic microenvironment, and inducing liver inflammation and tissue remodeling (257). This self-perpetuating pathological cycle accelerates progression to severe liver diseases (such as cirrhosis and HCC). Hence, anti-senescence strategies to curb this complex process may help prevent the onset and progression of chronic liver diseases.
At present, despite exponential growth in efforts to target senescent cells and SASP in normal physiological and disease-specific models, limitations remain, particularly regarding disease-specific effects and differential aging susceptibility among individual hepatic cell types. For instance, while the detrimental effects of hepatic senescence are well established, questions remain about how individual induced senescent cells drive distinct clinical outcomes and whether senescent cell clearance is feasible across all liver disease contexts. Thus, further research is warranted in linking the pathogenesis of different types of liver disease to specific senescence pathways, which could provide theoretical guidance for the precise design of antisenescence drugs. Additionally, senescence intervention data in liver disease remain largely preclinical (animal/in vitro), leaving a critical clinical trial gap; urgent trials are needed to assess targeted therapy efficacy and safety in patients and translate findings to practice. Notably, a number of senotherapeutics have only been tested at a single concentration in preclinical settings. Future preclinical and clinical investigations should focus on optimizing dose, administration route, and potential combination therapies to minimize adverse effects and improve treatment outcomes. Overall, advances in novel technologies are poised to drive progress in developing clinically viable therapies to prevent or mitigate cellular senescence.
Not applicable.
JZ provided supervision and guidance for this manuscript. YX performed literature collection, drafted the manuscript and prepared figures and tables. JZ supervised the project and critically revised the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
The present study was supported by Shanghai Science and Technology Innovation Action Plan Medical Innovation Research Special Project (grant nos. 23S21900100 and 24Y12801102).
|
Rossiello F, Jurk D, Passos JF and d'Adda di Fagagna F: Telomere dysfunction in ageing and age-related diseases. Nat Cell Biol. 24:135–147. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
de Magalhães JP: Cellular senescence in normal physiology. Science. 384:1300–1301. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kowald A, Passos JF and Kirkwood TBL: On the evolution of cellular senescence. Aging Cell. 19:e132702020. View Article : Google Scholar : PubMed/NCBI | |
|
Varela-Eirín M and Demaria M: Cellular senescence. Curr Biol. 32:R448–R452. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Afsar B and Afsar RE: Hypertension and cellular senescence. Biogerontology. 24:457–478. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Roger L, Tomas F and Gire V: Mechanisms and regulation of cellular senescence. Int J Mol Sci. 22:131732021. View Article : Google Scholar : PubMed/NCBI | |
|
Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E and Kamath PS: Global burden of liver disease: 2023 Update. J Hepatol. 79:516–537. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng Q, Gong Y, Zhu N, Shi Y, Zhang C and Qin L: Lipids and lipid metabolism in cellular senescence: Emerging targets for age-related diseases. Ageing Res Rev. 97:1022942024. View Article : Google Scholar : PubMed/NCBI | |
|
Park J and Shin DW: Senotherapeutics and their molecular mechanism for improving aging. Biomol Ther (Seoul). 30:490–500. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Hayflick L and Moorhead PS: The serial cultivation of human diploid cell strains. Exp Cell Res. 25:585–621. 1961. View Article : Google Scholar : PubMed/NCBI | |
|
Reimann M, Lee S and Schmitt CA: Cellular senescence: Neither irreversible nor reversible. J Exp Med. 221:e202321362024. View Article : Google Scholar : PubMed/NCBI | |
|
Ahmad A, Braden A, Khan S, Xiao J and Khan MM: Crosstalk between the DNA damage response and cellular senescence drives aging and age-related diseases. Semin Immunopathol. 46:102024. View Article : Google Scholar : PubMed/NCBI | |
|
Basilicata MG, Sommella E, Scisciola L, Tortorella G, Malavolta M, Giordani C, Barbieri M, Campiglia P and Paolisso G: Multi-omics strategies to decode the molecular landscape of cellular senescence. Ageing Res Rev. 111:1028242025. View Article : Google Scholar : PubMed/NCBI | |
|
Tchkonia T, Zhu Y, van Deursen J, Campisi J and Kirkland JL: Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 123:966–972. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gasek NS, Kuchel GA, Kirkland JL and Xu M: Strategies for targeting senescent cells in human disease. Nat Aging. 1:870–879. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Safwan-Zaiter H, Wagner N and Wagner KD: P16INK4A-More than a senescence marker. Life (Basel). 12:13322022.PubMed/NCBI | |
|
Ou HL and Schumacher B: DNA damage responses and p53 in the aging process. Blood. 131:488–495. 2018. View Article : Google Scholar | |
|
Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, et al: Cellular senescence: Defining a path forward. Cell. 179:813–827. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang B, Han J, Elisseeff JH and Demaria M: The senescence-associated secretory phenotype and its physiological and pathological implications. Nat Rev Mol Cell Biol. 25:958–978. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Admasu TD, Rae M and Stolzing A: Dissecting primary and secondary senescence to enable new senotherapeutic strategies. Ageing Res Re. 70:1014122021. View Article : Google Scholar | |
|
Wissler Gerdes EO, Zhu Y, Tchkonia T and Kirkland JL: Discovery, development, and future application of senolytics: Theories and predictions. FEBS J. 287:2418–2427. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Pitcher LE, Yousefzadeh MJ, Niedernhofer LJ, Robbins PD and Zhu Y: Cellular senescence: A key therapeutic target in aging and diseases. J Clin Invest. 132:e1584502022. View Article : Google Scholar : PubMed/NCBI | |
|
Espinosa De Ycaza AE, Søndergaard E, Morgan-Bathke M, Carranza Leon BG, Lytle KA, Ramos P, Kirkland JL, Tchkonia T and Jensen MD: Senescent cells in human adipose tissue: A cross-sectional study. Obesity (Silver Spring). 29:1320–1327. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kirkland JL and Tchkonia T: Senolytic drugs: From discovery to translation. J Intern Med. 288:518–536. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Naseem S, Hussain T and Manzoor S: Interleukin-6: A promising cytokine to support liver regeneration and adaptive immunity in liver pathologies. Cytokine Growth Factor Rev. 39:36–45. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Widjaja AA, Chothani SP and Cook SA: Different roles of interleukin 6 and interleukin 11 in the liver: Implications for therapy. Hum Vaccin Immunother. 16:2357–2362. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yamagishi R, Kamachi F, Nakamura M, Yamazaki S, Kamiya T, Takasugi M, Cheng Y, Nonaka Y, Yukawa-Muto Y, Thuy LTT, et al: Gasdermin D-mediated release of IL-33 from senescent hepatic stellate cells promotes obesity-associated hepatocellular carcinoma. Sci Immunol. 7:eabl72092022. View Article : Google Scholar : PubMed/NCBI | |
|
Luciano-Mateo F, Cabré N, Fernández-Arroyo S, Baiges-Gaya G, Hernández-Aguilera A, Rodríguez-Tomàs E, Muñoz-Pinedo C, Menéndez JA, Camps J and Joven J: Chemokine C-C motif ligand 2 overexpression drives tissue-specific metabolic responses in the liver and muscle of mice. Sci Rep. 10:119542020. View Article : Google Scholar : PubMed/NCBI | |
|
Xu L, Chen Y, Nagashimada M, Ni Y, Zhuge F, Chen G, Li H, Pan T, Yamashita T, Mukaida N, et al: CC chemokine ligand 3 deficiency ameliorates diet-induced steatohepatitis by regulating liver macrophage recruitment and M1/M2 status in mice. Metabolism. 125:1549142021. View Article : Google Scholar : PubMed/NCBI | |
|
Yu X, Chen Y, Cui L, Yang K, Wang X, Lei L, Zhang Y, Kong X, Lao W, Li Z, et al: CXCL8, CXCL9, CXCL10, and CXCL11 as biomarkers of liver injury caused by chronic hepatitis B. Front Microbiol. 13:10529172022. View Article : Google Scholar : PubMed/NCBI | |
|
Queck A, Bode H, Uschner FE, Brol MJ, Graf C, Schulz M, Jansen C, Praktiknjo M, Schierwagen R, Klein S, et al: Systemic MCP-1 levels derive mainly from injured liver and are associated with complications in cirrhosis. Front Immunol. 11:3542020. View Article : Google Scholar : PubMed/NCBI | |
|
Yan J, Hu B, Shi W, Wang X, Shen J, Chen Y, Huang H and Jin L: Gli2-regulated activation of hepatic stellate cells and liver fibrosis by TGF-β signaling. Am J Physiol Gastrointest Liver Physiol. 320:G720–G728. 2021. View Article : Google Scholar | |
|
Borkham-Kamphorst E and Weiskirchen R: The PDGF system and its antagonists in liver fibrosis. Cytokine Growth Factor Rev. 28:53–61. 2016. View Article : Google Scholar | |
|
Campana L, Esser H, Huch M and Forbes S: Liver regeneration and inflammation: From fundamental science to clinical applications. Nat Rev Mol Cell Biol. 22:608–624. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wójcik M, Ramadori P, Blaschke M, Sultan S, Khan S, Malik IA, Naz N, Martius G, Ramadori G and Schultze FC: Immunodetection of cyclooxygenase-2 (COX-2) is restricted to tissue macrophages in normal rat liver and to recruited mononuclear phagocytes in liver injury and cholangiocarcinoma. Histochem Cell Biol. 137:217–233. 2012. View Article : Google Scholar : | |
|
Lichtinghagen R, Michels D, Haberkorn CI, Arndt B, Bahr M, Flemming P, Manns MP and Boeker KH: Matrix metalloproteinase (MMP)-2, MMP-7, and tissue inhibitor of metalloproteinase-1 are closely related to the fibroproliferative process in the liver during chronic hepatitis C. J Hepatol. 34:239–247. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Thiele M, Johansen S, Gudmann NS, Madsen B, Kjaergaard M, Nielsen MJ, Leeming DJ, Jacobsen S, Bendtsen F, Møller S, et al: Progressive alcohol-related liver fibrosis is characterised by imbalanced collagen formation and degradation. Aliment Pharmacol Ther. 54:1070–1080. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Morio B, Panthu B, Bassot A and Rieusset J: Role of mitochondria in liver metabolic health and diseases. Cell Calcium. 94:1023362021. View Article : Google Scholar : PubMed/NCBI | |
|
Matsumoto T, Wakefield L and Grompe M: The significance of polyploid hepatocytes during aging process. Cell Mol Gastroenterol Hepatol. 11:1347–1349. 2021. View Article : Google Scholar : | |
|
Bu W, Sun X, Xue X, Geng S, Yang T, Zhang J, Li Y, Feng C, Liu Q, Zhang X, et al: Early onset of pathological polyploidization and cellular senescence in hepatocytes lacking RAD51 creates a pro-fibrotic and pro-tumorigenic inflammatory microenvironment. Hepatology. 81:491–508. 2025. View Article : Google Scholar | |
|
Lin S, Nascimento EM, Gajera CR, Chen L, Neuhöfer P, Garbuzov A, Wang S and Artandi SE: Distributed hepatocytes expressing telomerase repopulate the liver in homeostasis and injury. Nature. 556:244–248. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Seo E, Kang H, Choi H, Choi W and Jun HS: Reactive oxygen species-induced changes in glucose and lipid metabolism contribute to the accumulation of cholesterol in the liver during aging. Aging Cell. 18:e128952019. View Article : Google Scholar : PubMed/NCBI | |
|
Kiourtis C, Terradas-Terradas M, Gee LM, May S, Georgakopoulou A, Collins AL, O'Sullivan ED, Baird DP, Hassan M, Shaw R, et al: Hepatocellular senescence induces multi-organ senescence and dysfunction via TGFβ. Nat Cell Biol. 26:2075–2083. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Aravinthan A, Shannon N, Heaney J, Hoare M, Marshall A and Alexander GJ: The senescent hepatocyte gene signature in chronic liver disease. Exp Gerontol. 60:37–45. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Trivedi P, Wang S and Friedman SL: The power of plasticity-metabolic regulation of hepatic stellate cells. Cell Metab. 33:242–257. 2021. View Article : Google Scholar | |
|
Dewidar B, Meyer C, Dooley S and Meindl-Beinker AN: TGF-β in hepatic stellate cell activation and liver fibrogenesis-updated 2019. Cells. 8:14192019. View Article : Google Scholar | |
|
Yang F, Li H, Li Y, Hao Y, Wang C, Jia P, Chen X, Ma S and Xiao Z: Crosstalk between hepatic stellate cells and surrounding cells in hepatic fibrosis. Int Immunopharmacol. 99:1080512021. View Article : Google Scholar : PubMed/NCBI | |
|
Yashaswini CN, Qin T, Bhattacharya D, Amor C, Lowe S, Lujambio A, Wang S and Friedman SL: Phenotypes and ontogeny of senescent hepatic stellate cells in metabolic dysfunction-associated steatohepatitis. J Hepatol. 81:207–217. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
McConnell MJ, Kostallari E, Ibrahim S and Iwakiri Y: The evolving role of liver sinusoidal endothelial cells in liver health and disease. Hepatology. 78:649–669. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wan Y, Li X, Slevin E, Harrison K, Li T, Zhang Y, Klaunig JE, Wu C, Shetty AK, Dong XC and Meng F: Endothelial dysfunction in pathological processes of chronic liver disease during aging. FASEB J. 36:e221252022. View Article : Google Scholar | |
|
Hunt NJ, Kang SWS, Lockwood GP, Le Couteur DG and Cogger VC: Hallmarks of aging in the liver. Comput Struct Biotechnol J. 17:1151–1161. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Maeso-Díaz R, Ortega-Ribera M, Fernández-Iglesias A, Hide D, Muñoz L, Hessheimer AJ, Vila S, Francés R, Fondevila C, Albillos A, et al: Effects of aging on liver microcirculatory function and sinusoidal phenotype. Aging Cell. 17:e128292018. View Article : Google Scholar : PubMed/NCBI | |
|
Maeso-Díaz R, Ortega-Ribera M, Lafoz E, Lozano JJ, Baiges A, Francés R, Albillos A, Peralta C, García-Pagán JC, Bosch J, et al: Aging influences hepatic microvascular biology and liver fibrosis in advanced chronic liver disease. Aging Dis. 10:684–698. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Li P, He K, Li J, Liu Z and Gong J: The role of Kupffer cells in hepatic diseases. Mol Immunol. 85:222–229. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yang W, Kim DM, Jiang W, Ai W, Pan Q, Rahman S, Cai JJ, Brashear WA, Sun Y and Guo S: Suppression of FOXO1 attenuates inflamm-aging and improves liver function during aging. Aging Cell. 22:e139682023. View Article : Google Scholar : PubMed/NCBI | |
|
Yao J, Li Y and Wang H: The roles of myeloid cells in aging-related liver diseases. Int J Biol Sci. 19:1564–1578. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bird TG, Müller M, Boulter L, Vincent DF, Ridgway RA, Lopez-Guadamillas E, Lu WY, Jamieson T, Govaere O, Campbell AD, et al: TGFβ inhibition restores a regenerative response in acute liver injury by suppressing paracrine senescence. Sci Transl Med. 10:eaan12302018. View Article : Google Scholar | |
|
Fontana L, Zhao E, Amir M, Dong H, Tanaka K and Czaja MJ: Aging promotes the development of diet-induced murine steatohepatitis but not steatosis. Hepatology. 57:995–1004. 2013. View Article : Google Scholar | |
|
Trefts E, Gannon M and Wasserman DH: The liver. Curr Biol. 27:R1147–R1151. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Sanders FWB and Griffin JL: De novo lipogenesis in the liver in health and disease: More than just a shunting yard for glucose. Biol Rev Camb Philos Soc. 91:452–468. 2016. View Article : Google Scholar : | |
|
Ghosh-Choudhary S, Liu J and Finkel T: Metabolic regulation of cell fate and function. Trends Cell Biol. 30:201–212. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Andrews TS, Atif J, Liu JC, Perciani CT, Ma XZ, Thoeni C, Slyper M, Eraslan G, Segerstolpe A, Manuel J, et al: Single-cell, single-nucleus, and spatial RNA sequencing of the human liver identifies cholangiocyte and mesenchymal heterogeneity. Hepatol Commun. 6:821–840. 2022. View Article : Google Scholar | |
|
Horn P and Tacke F: Metabolic reprogramming in liver fibrosis. Cell Metab. 36:1439–1455. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Foglia B, Beltrà M, Sutti S and Cannito S: Metabolic reprogramming of HCC: A new microenvironment for immune responses. Int J Mol Sci. 24:74632023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Q, Zhang X, Qi J, Tian X, Dovjak E, Zhang J, Du H, Zhang N, Zhao J, Zhang Y, et al: Comprehensive profiling of lipid metabolic reprogramming expands precision medicine for HCC. Hepatology. 81:1164–1180. 2025. View Article : Google Scholar | |
|
Qu H, Liu J, Zhang D, Xie R, Wang L and Hong: Glycolysis in chronic liver diseases: Mechanistic insights and therapeutic opportunities. Cells. 12:19302023. View Article : Google Scholar : PubMed/NCBI | |
|
Lin J, Rao D, Zhang M and Gao Q: Metabolic reprogramming in the tumor microenvironment of liver cancer. J Hematol Oncol. 17:62024. View Article : Google Scholar : PubMed/NCBI | |
|
Moreno-Fernandez ME, Giles DA, Oates JR, Chan CC, Damen MSMA, Doll JR, Stankiewicz TE, Chen X, Chetal K, Karns R, et al: PKM2-dependent metabolic skewing of hepatic Th17 cells regulates pathogenesis of non-alcoholic fatty liver disease. Cell Metab. 33:1187–1204.e9. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Rao J, Wang H, Ni M, Wang Z, Wang Z, Wei S, Liu M, Wang P, Qiu J, Zhang L, et al: FSTL1 promotes liver fibrosis by reprogramming macrophage function through modulating the intracellular function of PKM2. Gut. 71:2539–2550. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Martin DE, Torrance BL, Haynes L and Bartley JM: Targeting aging: Lessons learned from immunometabolism and cellular senescence. Front Immunol. 12:7147422021. View Article : Google Scholar : PubMed/NCBI | |
|
Semenovich DS, Andrianova NV, Zorova LD, Pevzner IB, Abramicheva PA, Elchaninov AV, Markova OV, Petrukhina AS, Zorov DB and Plotnikov EY: Fibrosis development linked to alterations in glucose and energy metabolism and prooxidant-antioxidant balance in experimental models of liver injury. Antioxidants (Basel). 12:16042023. View Article : Google Scholar : PubMed/NCBI | |
|
Harrington JS, Ryter SW, Plataki M, Price DR and Choi AMK: Mitochondria in health, disease, and aging. Physiol Rev. 103:2349–2422. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Guo Y, Guan T, Shafiq K, Yu Q, Jiao X, Na D, Li M, Zhang G and Kong J: Mitochondrial dysfunction in aging. Ageing Res Rev. 88:1019552023. View Article : Google Scholar : PubMed/NCBI | |
|
Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM and Sinclair DA: Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol. 18:243–258. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Mansouri A, Gattolliat CH and Asselah T: Mitochondrial dysfunction and signaling in chronic liver diseases. Gastroenterology. 155:629–647. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Tian XL, Li JQ, Wu DS, Li Q and Chen B: Mitochondrial dysfunction affects hepatic immune and metabolic remodeling in patients with hepatitis B virus-related acute-on-chronic liver failure. World J Gastroenterol. 30:881–900. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang L, Shao Y and Chen Y: Mitochondrial dysfunction and mitochondrion-targeted therapeutics in liver diseases. J Drug Target. 29:1080–1093. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Reynolds JC, Lai RW, Woodhead JST, Joly JH, Mitchell CJ, Cameron-Smith D, Lu R, Cohen P, Graham NA, Benayoun BA, et al: MOTS-c is an exercise-induced mitochondrial-encoded regulator of age-dependent physical decline and muscle homeostasis. Nat Commun. 12:4702021. View Article : Google Scholar : PubMed/NCBI | |
|
López-Otín C, Blasco MA, Partridge L, Serrano M and Kroemer G: Hallmarks of aging: An expanding universe. Cell. 186:243–278. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cavaliere G, Catapano A, Trinchese G, Cimmino F, Menale C, Petrella L and Mollica MP: Crosstalk between adipose tissue and hepatic mitochondria in the development of the inflammation and liver injury during ageing in high-fat diet fed rats. Int J Mol Sci. 24:29672023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W, Wang C, Xu H and Gao Y: Aldehyde dehydrogenase, liver disease and cancer. Int J Biol Sci. 16:921–934. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Thoudam T, Gao H, Jiang Y, Huda N, Yang Z, Ma J and Liangpunsakul S: Mitochondrial quality control in alcohol-associated liver disease. Hepatol Commun. 8:e05342024. View Article : Google Scholar : PubMed/NCBI | |
|
Eid N, Ito Y, Horibe A and Otsuki Y: Ethanol-induced mitophagy in liver is associated with activation of the PINK1-Parkin pathway triggered by oxidative DNA damage. Histol Histopathol. 31:1143–1159. 2016.PubMed/NCBI | |
|
Hammoutene A, Biquard L, Lasselin J, Kheloufi M, Tanguy M, Vion AC, Mérian J, Colnot N, Loyer X, Tedgui A, et al: A defect in endothelial autophagy occurs in patients with non-alcoholic steatohepatitis and promotes inflammation and fibrosis. J Hepatol. 72:528–538. 2020. View Article : Google Scholar | |
|
Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, et al: Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. 112:1809–1820. 2023. View Article : Google Scholar | |
|
Cichoż-Lach H and Michalak A: Oxidative stress as a crucial factor in liver diseases. World J Gastroenterol. 20:8082–8091. 2014. View Article : Google Scholar | |
|
Lee J, Kim J, Lee R, Lee E, Choi TG, Lee AS, Yoon YI, Park GC, Namgoong JM, Lee SG and Tak E: Therapeutic strategies for liver diseases based on redox control systems. Biomed Pharmacother. 156:1137642022. View Article : Google Scholar : PubMed/NCBI | |
|
Hajam YA, Rani R, Ganie SY, Sheikh TA, Javaid D, Qadri SS, Pramodh S, Alsulimani A, Alkhanani MF, Harakeh S, et al: Oxidative stress in human pathology and aging: Molecular mechanisms and perspectives. Cells. 11:5522022. View Article : Google Scholar : PubMed/NCBI | |
|
Bárcena B, Salamanca A, Pintado C, Mazuecos L, Villar M, Moltó E, Bonzón-Kulichenko E, Vázquez J, Andrés A and Gallardo N: Aging induces hepatic oxidative stress and nuclear proteomic remodeling in liver from wistar rats. Antioxidants (Basel). 10:15352021. View Article : Google Scholar : PubMed/NCBI | |
|
Jung YS, Radhakrishnan K, Hammad S, Müller S, Müller J, Noh JR, Kim J, Lee IK, Cho SJ, Kim DK, et al: ERRγ-inducible FGF23 promotes alcoholic liver injury through enhancing CYP2E1 mediated hepatic oxidative stress. Redox Biol. 71:1031072024. View Article : Google Scholar | |
|
Jin J, Iakova P, Jiang Y, Medrano EE and Timchenko NA: The reduction of SIRT1 in livers of old mice leads to impaired body homeostasis and to inhibition of liver proliferation. Hepatology. 54:989–998. 2021. View Article : Google Scholar | |
|
Gressler AE, Leng H, Zinecker H and Simon AK: Proteostasis in T cell aging. Semin Immunol. 70:1018382023. View Article : Google Scholar : PubMed/NCBI | |
|
Hipp MS, Kasturi P and Hartl FU: The proteostasis network and its decline in ageing. Nat Rev Mol Cell Biol. 20:421–435. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Klaips CL, Jayaraj GG and Hartl FU: Pathways of cellular proteostasis in aging and disease. J Cell Biol. 217:51–63. 2018. View Article : Google Scholar : | |
|
Xia SW, Wang ZM, Sun SM, Su Y, Li ZH, Shao JJ, Tan SZ, Chen AP, Wang SJ, Zhang ZL and Zheng SZ: Endoplasmic reticulum stress and protein degradation in chronic liver disease. Pharmacol Res. 161:1052182020. View Article : Google Scholar : PubMed/NCBI | |
|
Wei S, Wang Q, Zhou H, Qiu J, Li C, Shi C, Zhou S, Liu R and Lu L: miR-455-3p alleviates hepatic stellate cell activation and liver fibrosis by suppressing HSF1 expression. Mol Ther Nucleic Acids. 16:758–769. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Tsao FHC, Barnes JN, Amessoudji A, Li Z and Meyer KC: Aging-related and gender specific albumin misfolding in Alzheimer's disease. J Alzheimers Dis Rep. 4:67–77. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Cuanalo-Contreras K, Schulz J, Mukherjee A, Park KW, Armijo E and Soto C: Extensive accumulation of misfolded protein aggregates during natural aging and senescence. Front Aging Neurosci. 14:10901092023. View Article : Google Scholar : PubMed/NCBI | |
|
Recillas-Targa F: Cancer epigenetics: An overview. Arch Med Res. 53:732–740. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Saul D and Kosinsky RL: Epigenetics of aging and aging-associated diseases. Int J Mol Sci. 22:4012021. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy SK, Yang H, Moylan CA, Pang H, Dellinger A, Abdelmalek MF, Garrett ME, Ashley-Koch A, Suzuki A, Tillmann HL, et al: Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology. 145:1076–1087. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gallon J, Coto-Llerena M, Ercan C, Bianco G, Paradiso V, Nuciforo S, Taha-Melitz S, Meier MA, Boldanova T, Pérez-Del-Pulgar S, et al: Epigenetic priming in chronic liver disease impacts the transcriptional and genetic landscapes of hepatocellular carcinoma. Mol Oncol. 16:665–682. 2022. View Article : Google Scholar : | |
|
Liu YR, Wang JQ, Huang ZG, Chen RN, Cao X, Zhu DC, Yu HX, Wang XR, Zhou HY, Xia Q and Li J: Histone deacetylase-2: A potential regulator and therapeutic target in liver disease (review). Int J Mol Med. 48:1312021. View Article : Google Scholar : | |
|
Joanna F, van Grunsven LA, Mathieu V, Sarah S, Sarah D, Karin V, Tamara V and Vera R: Histone deacetylase inhibition and the regulation of cell growth with particular reference to liver pathobiology. J Cell Mol Med. 13:2990–3005. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Morral N, Liu S, Conteh AM, Chu X, Wang Y, Dong XC, Liu Y, Linnemann AK and Wan J: Aberrant gene expression induced by a high fat diet is linked to H3K9 acetylation in the promoter-proximal region. Biochim Biophys Acta Gene Regul Mech. 1864:1946912021. View Article : Google Scholar : PubMed/NCBI | |
|
Sun C, Fan JG and Qiao L: Potential epigenetic mechanism in non-alcoholic fatty liver disease. Int J Mol Sci. 16:5161–5179. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Suter MA, Chen A, Burdine MS, Choudhury M, Harris RA, Lane RH, Friedman JE, Grove KL, Tackett AJ and Aagaard KM: A maternal high-fat diet modulates fetal SIRT1 histone and protein deacetylase activity in nonhuman primates. FASEB J. 26:5106–5114. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Pazienza V, Borghesan M, Mazza T, Sheedfar F, Panebianco C, Williams R, Mazzoccoli G, Andriulli A, Nakanishi T and Vinciguerra M: SIRT1-metabolite binding histone macroH2A1.1 protects hepatocytes against lipid accumulation. Aging (Albany NY). 6:35–47. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Kriss CL, Gregory-Lott E, Storey AJ, Tackett AJ, Wahls WP and Stevens SM Jr: In vivo metabolic tracing demonstrates the site-specific contribution of hepatic ethanol metabolism to histone acetylation. Alcohol Clin Exp Res. 42:1909–1923. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao Y, Hu Y and Liu S: Non-coding RNAs: A promising target for early metastasis intervention. Chin Med J (Engl). 136:2538–2550. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Dongiovanni P, Meroni M, Longo M, Fargion S and Fracanzani AL: miRNA signature in NAFLD: A turning point for a non-invasive diagnosis. Int J Mol Sci. 19:39662018. View Article : Google Scholar : PubMed/NCBI | |
|
Markovic J, Sharma AD and Balakrishnan A: MicroRNA-221: A fine tuner and potential biomarker of chronic liver injury. Cells. 9:17672020. View Article : Google Scholar : PubMed/NCBI | |
|
Sodum N, Kumar G, Bojja SL, Kumar N and Rao CM: Epigenetics in NAFLD/NASH: Targets and therapy. Pharmacol Res. 167:1054842021. View Article : Google Scholar : PubMed/NCBI | |
|
Maeso-Díaz R and Gracia-Sancho J: Aging and chronic liver disease. Semin Liver Dis. 40:373–384. 2020. View Article : Google Scholar | |
|
GBD 2019 Hepatitis B Collaborators: Global, regional, and national burden of hepatitis B, 1990-2019: A systematic analysis for the global burden of disease study 2019. Lancet Gastroenterol Hepatol. 7:796–829. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Z, Lin C, Mao X, Guo C, Suo C, Zhu D, Jiang W, Li Y, Fan J, Song C, et al: Changing prevalence of chronic hepatitis B virus infection in China between 1973 and 2021: A systematic literature review and meta-analysis of 3740 studies and 231 million people. Gut. 72:2354–2363. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Kondo Y, Tsukada K, Takeuchi T, Mitsui T, Iwano K, Masuko K, Itoh T, Tokita H, Okamoto H, Tsuda F, et al: High carrier rate after hepatitis B virus infection in the elderly. Hepatology. 18:768–774. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Bellon M and Nicot C: Telomere dynamics in immune senescence and exhaustion triggered by chronic viral infection. Viruses. 9:2892017. View Article : Google Scholar : PubMed/NCBI | |
|
Feng W, Yu D, Li B, Luo OY, Xu T, Cao Y and Ding Y: Paired assessment of liver telomere lengths in hepatocellular cancer is a reliable predictor of disease persistence. Biosci Rep. 37:BSR201606212017. View Article : Google Scholar : PubMed/NCBI | |
|
Tachtatzis PM, Marshall A, Arvinthan A, Verma S, Penrhyn-Lowe S, Mela M, Scarpini C, Davies SE, Coleman N and Alexander GJ: Chronic hepatitis B virus infection: The relation between hepatitis b antigen expression, telomere length, senescence, inflammation and fibrosis. PLoS One. 10:e01275112015. View Article : Google Scholar : PubMed/NCBI | |
|
Adelakun AA, Adediji IO, Idowu OJ, Jegede TF, Oluremi AS, Adepoju PO and Olaniyan OA: Prognostic significance of serum telomerase activity in the monitoring of hepatitis B viral infection. J Immunoassay Immunochem. 43:299–307. 2022. View Article : Google Scholar | |
|
Barnard A, Moch A and Saab S: Relationship between telomere maintenance and liver disease. Gut Liver. 13:11–15. 2019. View Article : Google Scholar : | |
|
Bolukbas C, Bolukbas FF, Kocyigit A, Aslan M, Selek S, Bitiren M and Ulukanligil M: Relationship between levels of DNA damage in lymphocytes and histopathological severity of chronic hepatitis C and various clinical forms of hepatitis B. J Gastroenterol Hepatol. 21:610–616. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Cao W, Wang S, Zhang L, Li X, Zhang Z, Xie Y and Li M: Epigenetic modification of hepatitis B virus infection and related hepatocellular carcinoma. Virulence. 15:24212312024. View Article : Google Scholar : PubMed/NCBI | |
|
Idrissi ME, Hachem H, Koering C, Merle P, Thénoz M, Mortreux F and Wattel E: HBx triggers either cellular senescence or cell proliferation depending on cellular phenotype. J Viral Hepat. 23:130–138. 2016. View Article : Google Scholar | |
|
Mastrodomenico M, Muselli M, Provvidenti L, Scatigna M, Bianchi S and Fabiani L: Long-term immune protection against HBV: Associated factors and determinants. Hum Vaccin Immunother. 17:2268–2272. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng Z, Li L, Chen Y, Wei H, Sun R and Tian Z: Interferon-γ facilitates hepatic antiviral T cell retention for the maintenance of liver-induced systemic tolerance. J Exp Med. 213:1079–1093. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Borgia M, Dal Bo M and Toffoli G: Role of virus-related chronic inflammation and mechanisms of cancer immune-suppression in pathogenesis and progression of hepatocellular carcinoma. Cancers (Basel). 13:43872021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Yang F, Yang Y, Hu Y, Liu W, Huang C, Li S and Chen Z: HBV facilitated hepatocellular carcinoma cells proliferation by up-regulating angiogenin expression through IL-6. Cell Physiol Biochem. 46:461–470. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JR and Kim CH: Association of a high activity of matrix metalloproteinase-9 to low levels of tissue inhibitors of metal-loproteinase-1 and -3 in human hepatitis B-viral hepatoma cells. Int J Biochem Cell Biol. 36:2293–2306. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Bekçibaşı M, Deveci Ö, Oğuz A, Bozkurt F, Dayan S and Çelen MK: Serum TNF-α, IL-1β, and IL-6 levels in chronic HBV-infected patients. Int J Clin Pract. 75:e142922021. View Article : Google Scholar | |
|
Rosenberg C, Bovin NV, Bram LV, Flyvbjerg E, Erlandsen M, Vorup-Jensen T and Petersen E: Age is an important determinant in humoral and T cell responses to immunization with hepatitis B surface antigen. Hum Vaccin Immunother. 9:1466–1476. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Eslam M, Sanyal AJ and George J; International Consensus Panel: MAFLD: A consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology. 158:1999–2014.e1. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Noureddin M, Yates KP, Vaughn IA, Neuschwander-Tetri BA, Sanyal AJ, McCullough A, Merriman R, Hameed B, Doo E, Kleiner DE, et al: Clinical and histological determinants of nonalcoholic steatohepatitis and advanced fibrosis in elderly patients. Hepatology. 58:1644–1654. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gao Y, Zhang W, Zeng LQ, Bai H, Li J, Zhou J, Zhou GY, Fang CW, Wang F and Qin XJ: Exercise and dietary intervention ameliorate high-fat diet-induced NAFLD and liver aging by inducing lipophagy. Redox Biol. 36:1016352020. View Article : Google Scholar : PubMed/NCBI | |
|
Dabravolski SA, Bezsonov EE and Orekhov AN: The role of mitochondria dysfunction and hepatic senescence in NAFLD development and progression. Biomed Pharmacother. 142:1120412021. View Article : Google Scholar : PubMed/NCBI | |
|
Baboota RK, Rawshani A, Bonnet L, Li X, Yang H, Mardinoglu A, Tchkonia T, Kirkland JL, Hoffmann A, Dietrich A, et al: BMP4 and Gremlin 1 regulate hepatic cell senescence during clinical progression of NAFLD/NASH. Nat Metab. 4:1007–1021. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Meijnikman AS, Herrema H, Scheithauer TPM, Kroon J, Nieuwdorp M and Groen AK: Evaluating causality of cellular senescence in non-alcoholic fatty liver disease. JHEP Rep. 3:1003012021. View Article : Google Scholar : PubMed/NCBI | |
|
Tomita K, Teratani T, Suzuki T, Oshikawa T, Yokoyama H, Shimamura K, Nishiyama K, Mataki N, Irie R, Minamino T, et al: p53/p66Shc-mediated signaling contributes to the progression of non-alcoholic steatohepatitis in humans and mice. J Hepatol. 57:837–843. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Bi H, Zhou B, Yang J, Lu Y, Mao F and Song Y: Whole-genome DNA methylation and gene expression profiling in the livers of mice with nonalcoholic steatohepatitis. Life Sci. 329:1219512023. View Article : Google Scholar : PubMed/NCBI | |
|
Sim BC, Kang YE, You SK, Lee SE, Nga HT, Lee HY, Nguyen TL, Moon JS, Tian J, Jang HJ, et al: Hepatic T-cell senescence and exhaustion are implicated in the progression of fatty liver disease in patients with type 2 diabetes and mouse model with nonalcoholic steatohepatitis. Cell Death Dis. 14:6182023. View Article : Google Scholar : PubMed/NCBI | |
|
Kazankov K, Møller HJ, Lange A, Birkebaek NH, Holland-Fischer P, Solvig J, Hørlyck A, Kristensen K, Rittig S, Handberg A, et al: The macrophage activation marker sCD163 is associated with changes in NAFLD and metabolic profile during lifestyle intervention in obese children. Pediatr Obes. 10:226–233. 2015. View Article : Google Scholar | |
|
Hernández-Évole H, Jiménez-Esquivel N, Pose E and Bataller R: Alcohol-associated liver disease: Epidemiology and management. Ann Hepatol. 29:1011622024. View Article : Google Scholar | |
|
Mackowiak B, Fu Y, Maccioni L and Gao B: Alcohol-associated liver disease. J Clin Invest. 134:e1763452024. View Article : Google Scholar : PubMed/NCBI | |
|
Åberg F, Jiang ZG, Cortez-Pinto H and Männistö V: Alcohol-associated liver disease-global epidemiology. Hepatology. 80:1307–1322. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wan Y, McDaniel K, Wu N, Ramos-Lorenzo S, Glaser T, Venter J, Francis H, Kennedy L, Sato K, Zhou T, et al: Regulation of cellular senescence by miR-34a in alcoholic liver injury. Am J Pathol. 187:2788–2798. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Huda N, Kusumanchi P, Perez K, Jiang Y, Skill NJ, Sun Z, Ma J, Yang Z and Liangpunsakul S: Telomere length in patients with alcohol-associated liver disease: A brief report. J Investig Med. 70:1438–1441. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
So-Armah K, Freiberg M, Cheng D, Lim JK, Gnatienko N, Patts G, Doyle M, Fuster D, Lioznov D, Krupitsky E and Samet J: Liver fibrosis and accelerated immune dysfunction (immunosenescence) among HIV-infected Russians with heavy alcohol consumption-an observational cross-sectional study. BMC Gastroenterol. 20:12019. View Article : Google Scholar | |
|
Dou L, Shi X, He X and Gao Y: Macrophage phenotype and function in liver disorder. Front Immunol. 10:31122020. View Article : Google Scholar : PubMed/NCBI | |
|
Wan J, Benkdane M, Alons E, Lotersztajn S and Pavoine C: M2 kupffer cells promote hepatocyte senescence: An IL-6-dependent protective mechanism against alcoholic liver disease. Am J Pathol. 184:1763–1772. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Chedid A, Mendenhall CL, Moritz TE, French SW, Chen TS, Morgan TR, Roselle GA, Nemchausky BA, Tamburro CH, Schiff ER, et al: Cell-mediated hepatic injury in alcoholic liver disease. Veterans affairs cooperative study group 275. Gastroenterology. 105:254–266. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Suh YG and Jeong WI: Hepatic stellate cells and innate immunity in alcoholic liver disease. World J Gastroenterol. 17:2543–2551. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Parola M and Pinzani M: Liver fibrosis: Pathophysiology, pathogenetic targets and clinical issues. Mol Aspects Med. 65:37–55. 2019. View Article : Google Scholar | |
|
Roehlen N, Crouchet E and Baumert TF: Liver fibrosis: Mechanistic concepts and therapeutic perspectives. Cells. 9:8752020. View Article : Google Scholar : PubMed/NCBI | |
|
Feng F, Wu J, Chi Q, Wang S, Liu W, Yang L, Song G, Pan L, Xu K and Wang C: Lactylome analysis unveils lactylation-dependent mechanisms of stemness remodeling in the liver cancer stem cells. Adv Sci (Weinh). 11:e24059752024. View Article : Google Scholar : PubMed/NCBI | |
|
Cheemerla S and Balakrishnan M: Global epidemiology of chronic liver disease. Clin Liver Dis (Hoboken). 17:365–370. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Smith A, Baumgartner K and Bositis C: Cirrhosis: Diagnosis and management. Am Fam Physician. 100:759–770. 2019.PubMed/NCBI | |
|
Weiskirchen R, Weiskirchen S and Tacke F: Organ and tissue fibrosis: Molecular signals, cellular mechanisms and translational implications. Mol Aspects Med. 65:2–15. 2019. View Article : Google Scholar | |
|
Ghafouri-Fard S, Abak A, Talebi SF, Shoorei H, Branicki W, Taheri M and Akbari Dilmaghani N: Role of miRNA and lncRNAs in organ fibrosis and aging. Biomed Pharmacother. 143:1121322021. View Article : Google Scholar : PubMed/NCBI | |
|
Wijayasiri P, Astbury S, Kaye P, Oakley F, Alexander GJ, Kendall TJ and Aravinthan AD: Role of hepatocyte senescence in the activation of hepatic stellate cells and liver fibrosis progression. Cells. 11:22212022. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Adeniji NT, Fan W, Kunimoto K and Török NJ: Non-alcoholic fatty liver disease and liver fibrosis during aging. Aging Dis. 13:1239–1251. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Y, Zhang H, Yao Y, Zhang X, Guan Y and Zheng F: CD4+ T cell activation and inflammation in NASH-related fibrosis. Front Immunol. 13:9674102022. View Article : Google Scholar | |
|
Ajith A, Merimi M, Arki MK, Hossein-Khannazer N, Najar M, Vosough M, Sokal EM and Najimi M: Immune regulation and therapeutic application of T regulatory cells in liver diseases. Front Immunol. 15:13710892024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu NN, Wang L, Wang L, Xu X, Lopaschuk GD, Zhang Y and Ren J: Site-specific ubiquitination of VDAC1 restricts its oligomerization and mitochondrial DNA release in liver fibrosis. Exp Mol Med. 55:269–280. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
An P, Wei LL, Zhao S, Sverdlov DY, Vaid KA, Miyamoto M, Kuramitsu K, Lai M and Popov YV: Hepatocyte mitochondria-derived danger signals directly activate hepatic stellate cells and drive progression of liver fibrosis. Nat Commun. 11:23622020. View Article : Google Scholar : PubMed/NCBI | |
|
Calado RT, Regal JA, Kleiner DE, Schrump DS, Peterson NR, Pons V, Chanock SJ, Lansdorp PM and Young NS: A spectrum of severe familial liver disorders associate with telomerase mutations. PLoS One. 4:e79262009. View Article : Google Scholar : PubMed/NCBI | |
|
Laish I, Mari A, Mannasse B, Hadary R, Konikoff FM, Amiel A and Kitay-Cohen Y: Telomere length, aggregates, and capture in cirrhosis. Isr Med Assoc J. 20:295–299. 2018.PubMed/NCBI | |
|
Kim D, Li AA and Ahmed A: Leucocyte telomere shortening is associated with nonalcoholic fatty liver disease-related advanced fibrosis. Liver Int. 38:1839–1848. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Carulli L: Telomere shortening as genetic risk factor of liver cirrhosis. World J Gastroenterol. 21:379–383. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Nagaraju GP, Dariya B, Kasa P, Peela S and El-Rayes BF: Epigenetics in hepatocellular carcinoma. Semin Cancer Biol. 86:622–632. 2022. View Article : Google Scholar | |
|
Foglia B, Turato C and Cannito S: Hepatocellular carcinoma: Latest research in pathogenesis, detection and treatment. Int J Mol Sci. 24:122242023. View Article : Google Scholar : PubMed/NCBI | |
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI | |
|
Sagnelli E, Macera M, Russo A, Coppola N and Sagnelli C: Epidemiological and etiological variations in hepatocellular carcinoma. Infection. 48:7–17. 2020. View Article : Google Scholar | |
|
Poon RT, Cheung TT, Kwok PC, Lee AS, Li TW, Loke KL, Chan SL, Cheung MT, Lai TW, Cheung CC, et al: Hong Kong consensus recommendations on the management of hepatocellular carcinoma. Liver Cancer. 4:51–69. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Liu P, Tang Q, Chen M, Chen W, Lu Y, Liu Z and He Z: Hepatocellular senescence: Immunosurveillance and future senescence-induced therapy in hepatocellular carcinoma. Front Oncol. 10:5899082020. View Article : Google Scholar : PubMed/NCBI | |
|
Li F, Huangyang P, Burrows M, Guo K, Riscal R, Godfrey J, Lee KE, Lin N, Lee P, Blair IA, et al: FBP1 loss disrupts liver metabolism and promotes tumorigenesis through a hepatic stellate cell senescence secretome. Nat Cell Biol. 22:728–739. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Y, Yang X, Meng Y, Shao C, Liao J, Li F, Li R, Jing Y and Huang A: The hepatic senescence-associated secretory phenotype promotes hepatocarcinogenesis through Bcl3-dependent activation of macrophages. Cell Biosci. 11:1732021. View Article : Google Scholar : PubMed/NCBI | |
|
Ho TC, Wang EY, Yeh KH, Jeng YM, Horng JH, Wu LL, Chen YT, Huang HC, Hsu CL, Chen PJ, et al: Complement C1q mediates the expansion of periportal hepatic progenitor cells in senescence-associated inflammatory liver. Proc Natl Acad Sci USA. 117:6717–6725. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, Kang B, Hu R, Huang JY, Zhang Q, et al: Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell. 169:1342–1356.e16. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou W, Deng J, Chen Q, Li R, Xu X, Guan Y, Li W, Xiong X, Li H, Li J and Cai X: Expression of CD4+CD25+CD127Low regulatory T cells and cytokines in peripheral blood of patients with primary liver carcinoma. Int J Med Sci. 17:712–719. 2020. View Article : Google Scholar : | |
|
Schoenberg MB, Li X, Li X, Han Y, Hao J, Miksch RC, Koch D, Börner N, Beger NT, Bucher JN, et al: The predictive value of tumor infiltrating leukocytes in hepatocellular carcinoma: A systematic review and meta-analysis. Eur J Surg Oncol. 47:2561–2570. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang CY, Liu S and Yang M: Regulatory T cells and their associated factors in hepatocellular carcinoma development and therapy. World J Gastroenterol. 28:3346–3358. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Cai Q, Zhu M, Rong J, Feng X and Wang K: Exploring the double-edged role of cellular senescence in chronic liver disease for new treatment approaches. Life Sci. 373:1236782025. View Article : Google Scholar : PubMed/NCBI | |
|
Guo X, Wen S, Wang J, Zeng X, Yu H, Chen Y, Zhu X and Xu L: Senolytic combination of dasatinib and quercetin attenuates renal damage in diabetic kidney disease. Phytomedicine. 130:1557052024. View Article : Google Scholar : PubMed/NCBI | |
|
Islam MT, Tuday E, Allen S, Kim J, Trott DW, Holland WL, Donato AJ and Lesniewski LA: Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue inflammation, and ameliorate metabolic function in old age. Aging Cell. 22:e137672023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, et al: The Achilles' heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell. 14:644–658. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, Day CP, Burt A, Palmer A, Anstee QM, et al: Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 8:156912017. View Article : Google Scholar : PubMed/NCBI | |
|
Thadathil N, Selvarani R, Mohammed S, Nicklas EH, Tran AL, Kamal M, Luo W, Brown JL, Lawrence MM, Borowik AK, et al: Senolytic treatment reduces cell senescence and necroptosis in Sod1 knockout mice that is associated with reduced inflammation and hepatocellular carcinoma. Aging Cell. 21:e136762022. View Article : Google Scholar : PubMed/NCBI | |
|
Raffaele M, Kovacovicova K, Frohlich J, Lo Re O, Giallongo S, Oben JA, Faldyna M, Leva L, Giannone AG, Cabibi D and Vinciguerra M: Mild exacerbation of obesity- and age-dependent liver disease progression by senolytic cocktail dasatinib + quercetin. Cell Commun Signal. 19:442021. View Article : Google Scholar | |
|
Fan Y, Cheng J, Zeng H and Shao L: Senescent cell depletion through targeting BCL-family proteins and mitochondria. Front Physiol. 11:5936302020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao X, Ogunwobi OO and Liu C: Survivin inhibition is critical for Bcl-2 inhibitor-induced apoptosis in hepatocellular carcinoma cells. PLoS One. 6:e219802011. View Article : Google Scholar : PubMed/NCBI | |
|
Emiloju OE, Yin J, Koubek E, Reid JM, Borad MJ, Lou Y, Seetharam M, Edelman MJ, Sausville EA, Jiang Y, et al: Phase 1 trial of navitoclax and sorafenib in patients with relapsed or refractory solid tumors with hepatocellular carcinoma expansion cohort. Invest New Drugs. 42:127–135. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gold NM, Ding Q, Yang Y, Pu S, Cao W, Ge X, Yang P, Okeke MN, Nisar A, Pan Y, et al: Therapeutic potential of nicotinamide and ABT263 in alcohol-associated liver disease through targeting cellular senescence. MedComm (2020). 6:e700862025. View Article : Google Scholar : PubMed/NCBI | |
|
Hikita H, Takehara T, Shimizu S, Kodama T, Shigekawa M, Iwase K, Hosui A, Miyagi T, Tatsumi T, Ishida H, et al: The Bcl-xL inhibitor, ABT-737, efficiently induces apoptosis and suppresses growth of hepatoma cells in combination with sorafenib. Hepatology. 52:1310–1321. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Tse C, Shoemaker AR, Adickes J, Anderson MG, Chen J, Jin S, Johnson EF, Marsh KC, Mitten MJ, Nimmer P, et al: ABT-263: A potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 68:3421–3428. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Leverson JD, Phillips DC, Mitten MJ, Boghaert ER, Diaz D, Tahir SK, Belmont LD, Nimmer P, Xiao Y, Ma XM, et al: Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy. Sci Transl Med. 7:279ra402015. View Article : Google Scholar : PubMed/NCBI | |
|
Cucarull B, Tutusaus A, Subías M, Stefanovic M, Hernáez-Alsina T, Boix L, Reig M, García de Frutos P, Marí M, Colell A, et al: Regorafenib alteration of the BCL-xL/MCL-1 ratio provides a therapeutic opportunity for BH3-mimetics in hepatocellular carcinoma models. Cancers (Basel). 12:3322020. View Article : Google Scholar : PubMed/NCBI | |
|
Bourgeois B and Madl T: Regulation of cellular senescence via the FOXO4-p53 axis. FEBS Lett. 592:2083–2097. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang C, Xie Y, Chen H, Lv L, Yao J, Zhang M, Xia K, Feng X, Li Y, Liang X, et al: FOXO4-DRI alleviates age-related testosterone secretion insufficiency by targeting senescent Leydig cells in aged mice. Aging (Albany NY). 12:1272–1284. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Dutta Gupta S, Bommaka MK and Banerjee A: Inhibiting protein-protein interactions of Hsp90 as a novel approach for targeting cancer. Eur J Med Chem. 178:48–63. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Leng AM, Liu T, Yang J, Cui JF, Li XH, Zhu YN, Xiong T, Zhang G and Chen Y: The apoptotic effect and associated signalling of HSP90 inhibitor 17-DMAG in hepatocellular carcinoma cells. Cell Biol Int. 36:893–899. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Hu D, Mo X, Luo J, Wang F, Huang C, Xie H and Jin L: 17-DMAG ameliorates neuroinflammation and BBB disruption via SOX5 mediated PI3K/Akt pathway after intracerebral hemorrhage in rats. Int Immunopharmacol. 123:1106982023. View Article : Google Scholar : PubMed/NCBI | |
|
Saber S, El-Fattah EEA, Abdelhamid AM, Mourad AAE, Hamouda MAM, Elrabat A, Zakaria S, Haleem AA, Mohamed SZ, Elgharabawy RM, et al: Innovative challenge for the inhibition of hepatocellular carcinoma progression by combined targeting of HSP90 and STAT3/HIF-1α signaling. Biomed Pharmacother. 158:1141962023. View Article : Google Scholar | |
|
Li L, Wang L, You QD and Xu XL: Heat shock protein 90 inhibitors: An update on achievements, challenges, and future directions. J Med Chem. 63:1798–1822. 2020. View Article : Google Scholar | |
|
Ambade A, Catalano D, Lim A, Kopoyan A, Shaffer SA and Mandrekar P: Inhibition of heat shock protein 90 alleviates steatosis and macrophage activation in murine alcoholic liver injury. J Hepatol. 61:903–911. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Goyal L, Wadlow RC, Blaszkowsky LS, Wolpin BM, Abrams TA, McCleary NJ, Sheehan S, Sundaram E, Karol MD, Chen J and Zhu AX: A phase I and pharmacokinetic study of ganetespib (STA-9090) in advanced hepatocellular carcinoma. Invest New Drugs. 33:128–137. 2015. View Article : Google Scholar | |
|
Saber S, Hasan AM, Mohammed OA, Saleh LA, Hashish AA, Alamri MMS, Al-Ameer AY, Alfaifi J, Senbel A, Aboregela AM, et al: Ganetespib (STA-9090) augments sorafenib efficacy via necroptosis induction in hepatocellular carcinoma: Implications from preclinical data for a novel therapeutic approach. Biomed Pharmacother. 164:1149182023. View Article : Google Scholar : PubMed/NCBI | |
|
Syed DN, Adhami VM, Khan N, Khan MI and Mukhtar H: Exploring the molecular targets of dietary flavonoid fisetin in cancer. Semin Cancer Biol. 40-41:130–140. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou ZS, Kong CF, Sun JR, Qu XK, Sun JH and Sun AT: Fisetin ameliorates alcohol-induced liver injury through regulating SIRT1 and SphK1 pathway. Am J Chin Med. 50:2171–2184. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sundarraj K, Raghunath A, Panneerselvam L and Perumal E: Fisetin inhibits autophagy in HepG2 cells via PI3K/Akt/mTOR and AMPK pathway. Nutr Cancer. 73:2502–2514. 2021. View Article : Google Scholar | |
|
Chilvery S, Bansod S, Saifi MA and Godugu C: Piperlongumine attenuates bile duct ligation-induced liver fibrosis in mice via inhibition of TGF-β1/Smad and EMT pathways. Int Immunopharmacol. 88:1069092020. View Article : Google Scholar | |
|
Liu X, Wang Y, Zhang X, Gao Z, Zhang S, Shi P, Zhang X, Song L, Hendrickson H, Zhou D and Zheng G: Senolytic activity of piperlongumine analogues: Synthesis and biological evaluation. Bioorg Med Chem. 26:3925–3938. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yakubo S, Abe H, Li Y, Kudo M, Kimura A, Wakabayashi T, Watanabe Y, Kimura N, Setsu T, Yokoo T, et al: Dasatinib and quercetin as senolytic drugs improve fat deposition and exhibit antifibrotic effects in the medaka metabolic dysfunction-associated steatotic liver disease model. Diseases. 12:3172024. View Article : Google Scholar : PubMed/NCBI | |
|
Song P, Duan JL, Ding J, Liu JJ, Fang ZQ, Xu H, Li ZW, Du W, Xu M, Ling YW, et al: Cellular senescence primes liver fibrosis regression through Notch-EZH2. MedComm (2020). 4:e3462023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang G, Zhan Y, Wang H and Li W: ABT-263 sensitizes TRAIL-resistant hepatocarcinoma cells by downregulating the Bcl-2 family of anti-apoptotic protein. Cancer Chemother Pharmacol. 69:799–805. 2012. View Article : Google Scholar | |
|
Ma B, Ju A, Zhang S, An Q, Xu S, Liu J, Yu L, Fu Y and Luo Y: Albumosomes formed by cytoplasmic pre-folding albumin maintain mitochondrial homeostasis and inhibit nonalcoholic fatty liver disease. Signal Transduct Target Ther. 8:2292023. View Article : Google Scholar : PubMed/NCBI | |
|
Abu-Elsaad NM, Serrya MS, El-Karef AM and Ibrahim TM: The heat shock protein 90 inhibitor, 17-AAG, attenuates thioacetamide induced liver fibrosis in mice. Pharmacol Rep. 68:275–282. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Abdelhamid AM, Saber S, Hamad RS, Abdel-Reheim MA, Ellethy AT, Amer MM, Abdel-Hamed MR, Mohamed EA, Ahmed SS, Elsisi HA, et al: STA-9090 in combination with a statin exerts enhanced protective effects in rats fed a high-fat diet and exposed to diethylnitrosamine and thioacetamide. Front Pharmacol. 15:14548292024. View Article : Google Scholar : PubMed/NCBI | |
|
Augello G, Emma MR, Cusimano A, Azzolina A, Mongiovì S, Puleio R, Cassata G, Gulino A, Belmonte B, Gramignoli R, et al: Targeting HSP90 with the small molecule inhibitor AUY922 (luminespib) as a treatment strategy against hepatocellular carcinoma. Int J Cancer. 144:2613–2624. 2019. View Article : Google Scholar | |
|
Zhang W, Xue H, Zhou C, Zheng Z, Xing M, Chu H, Li P, Zhang N, Dang Y and Xu X: 7-Aminocephalosporanic acid, a novel HSP90β inhibitor, attenuates HFD-induced hepatic steatosis. Biochem Biophys Res Commun. 622:184–191. 2022. View Article : Google Scholar | |
|
Di Micco R, Krizhanovsky V, Baker D and d'Adda di Fagagna F: Cellular senescence in ageing: From mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol. 22:75–95. 2021. View Article : Google Scholar : | |
|
Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ and Robbins PD: Targeting cellular senescence with senotherapeutics: Senolytics and senomorphics. FEBS J. 290:1362–1383. 2023. View Article : Google Scholar | |
|
Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, et al: Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 460:392–395. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Selvarani R, Mohammed S and Richardson A: Effect of rapamycin on aging and age-related diseases-past and future. Geroscience. 43:1135–1158. 2021. View Article : Google Scholar | |
|
Laberge RM, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, Curran SC, Davalos AR, Wilson-Edell KA, Liu S, et al: MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat Cell Biol. 17:1049–1061. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Lee HS, Kim JY, Ro SW, Kim MS, Kim H and Joo DJ: Antitumor effect of low-dose of rapamycin in a transgenic mouse model of liver cancer. Yonsei Med J. 63:1007–1015. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kang HG, Park H, Myong GE, Kim WJ, Mun CE, Kim CR, You CY, Kim SK, Park MS and Park SI: Beneficial effect of rapamycin on liver fibrosis in a mouse model (C57bl/6 mouse). Transplant Proc. 56:701–704. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chao X, Williams SN and Ding WX: Role of mechanistic target of rapamycin in autophagy and alcohol-associated liver disease. Am J Physiol Cell Physiol. 323:C1100–C1111. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ge C, Ma C, Cui J, Dong X, Sun L, Li Y and Yu A: Rapamycin suppresses inflammation and increases the interaction between p65 and IκBα in rapamycin-induced fatty livers. PLoS One. 18:e02818882023. View Article : Google Scholar | |
|
Umemura A, Park EJ, Taniguchi K, Lee JH, Shalapour S, Valasek MA, Aghajan M, Nakagawa H, Seki E, Hall MN and Karin M: Liver damage, inflammation, and enhanced tumorigenesis after persistent mTORC1 inhibition. Cell Metab. 20:133–144. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Pallet N and Legendre C: Adverse events associated with mTOR inhibitors. Expert Opin Drug Saf. 12:177–186. 2013. View Article : Google Scholar | |
|
Moiseeva O, Deschênes-Simard X, St-Germain E, Igelmann S, Huot G, Cadar AE, Bourdeau V, Pollak MN and Ferbeyre G: Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell. 12:489–498. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Xu L, Wang X, Chen Y, Soong L, Chen Y, Cai J, Liang Y and Sun J: Metformin modulates T cell function and alleviates liver injury through bioenergetic regulation in viral hepatitis. Front Immunol. 12:6385752021. View Article : Google Scholar : PubMed/NCBI | |
|
Gkiourtzis N, Michou P, Moutafi M, Glava A, Cheirakis K, Christakopoulos A, Vouksinou E and Fotoulaki M: The benefit of metformin in the treatment of pediatric non-alcoholic fatty liver disease: A systematic review and meta-analysis of randomized controlled trials. Eur J Pediatr. 182:4795–4806. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Hunt NJ, Lockwood GP, Kang SWS, Pulpitel T, Clark X, Mao H, McCourt PAG, Cooney GJ, Wali JA, Le Couteur FH, et al: The effects of metformin on age-related changes in the liver sinusoidal endothelial cell. J Gerontol A Biol Sci Med Sci. 75:278–285. 2020. | |
|
Vacante F, Senesi P, Montesano A, Paini S, Luzi L and Terruzzi I: Metformin counteracts HCC progression and metastasis enhancing KLF6/p21 expression and downregulating the IGF axis. Int J Endocrinol. 2019:75701462019. View Article : Google Scholar : PubMed/NCBI | |
|
Antwi SO, Li Z, Mody K, Roberts LR and Patel T: Independent and joint use of statins and metformin by elderly patients with diabetes and overall survival following HCC diagnosis. J Clin Gastroenterol. 54:468–476. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Rauf A, Imran M, Butt MS, Nadeem M, Peters DG and Mubarak MS: Resveratrol as an anti-cancer agent: A review. Crit Rev Food Sci Nutr. 58:1428–1447. 2018. View Article : Google Scholar | |
|
Chen Q, Zhang H, Yang Y, Zhang S, Wang J, Zhang D and Yu H: Metformin attenuates UVA-induced skin photoaging by suppressing mitophagy and the PI3K/AKT/mTOR pathway. Int J Mol Sci. 23:69602022. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Han B, Li J, Lv Z, Jiang H, Liu Y, Yang X, Lu J and Zhang Z: Resveratrol alleviates liver fibrosis induced by long-term inorganic mercury exposure through activating the Sirt 1/PGC-1α signaling pathway. J Agric Food Chem. 72:15985–15997. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chai R, Fu H, Zheng Z, Liu T, Ji S and Li G: Resveratrol inhibits proliferation and migration through SIRT1 mediated post-translational modification of PI3K/AKT signaling in hepatocellular carcinoma cells. Mol Med Rep. 16:8037–8044. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang B, Yin X and Sui S: Resveratrol inhibited the progression of human hepatocellular carcinoma by inducing autophagy via regulating p53 and the phosphoinositide 3-kinase/protein kinase B pathway. Oncol Re. 40:2758–2765. 2018. | |
|
Shaito A, Posadino AM, Younes N, Hasan H, Halabi S, Alhababi D, Al-Mohannadi A, Abdel-Rahman WM, Eid AH, Nasrallah GK and Pintus G: Potential adverse effects of resveratrol: A literature review. Int J Mol Sci. 21:20842020. View Article : Google Scholar : PubMed/NCBI | |
|
Pallauf K, Rimbach G, Rupp PM, Chin D and Wolf IM: Resveratrol and lifespan in model organisms. Curr Med Chem. 23:4639–4680. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Fu J, Deng W, Ge J, Fu S, Li P, Wu H, Wang J, Gao Y, Gao H and Wu T: Sirtuin 1 alleviates alcoholic liver disease by inhibiting HMGB1 acetylation and translocation. PeerJ. 11:e164802023. View Article : Google Scholar : PubMed/NCBI | |
|
Yamazaki Y, Usui I, Kanatani Y, Matsuya Y, Tsuneyama K, Fujisaka S, Bukhari A, Suzuki H, Senda S, Imanishi S, et al: Treatment with SRT1720, a SIRT1 activator, ameliorates fatty liver with reduced expression of lipogenic enzymes in MSG mice. Am J Physiol Endocrinol Metab. 297:E1179–E1186. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
He Y, Su Y, Duan C, Wang S, He W, Zhang Y, An X and He M: Emerging role of aging in the progression of NAFLD to HCC. Ageing Res Rev. 84:1018332023. View Article : Google Scholar | |
|
Rastegar M, Marjani HA, Yazdani Y, Shahbazi M, Golalipour M and Farazmandfar T: Investigating effect of rapamycin and metformin on angiogenesis in hepatocellular carcinoma cell line. Adv Pharm Bull. 8:63–68. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Smith FC, Stocker SL, Danta M, Carland JE, Kumar SS, Liu Z, Greenfield JR, Braithwaite HE, Cheng TS, Graham GG, et al: The safety and pharmacokinetics of metformin in patients with chronic liver disease. Aliment Pharmacol Ther. 51:565–575. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Sato Y, Qiu J, Hirose T, Miura T, Sato Y, Kohzuki M and Ito O: Metformin slows liver cyst formation and fibrosis in experimental model of polycystic liver disease. Am J Physiol Gastrointest Liver Physiol. 320:G464–G473. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
de Oliveira S, Houseright RA, Graves AL, Golenberg N, Korte BG, Miskolci V and Huttenlocher A: Metformin modulates innate immune-mediated inflammation and early progression of NAFLD-associated hepatocellular carcinoma in zebrafish. J Hepatol. 70:710–721. 2019. View Article : Google Scholar | |
|
He Y, Wang H, Lin S, Chen T, Chang D, Sun Y, Wang C, Liu Y, Lu Y, Song J, et al: Advanced effect of curcumin and resveratrol on mitigating hepatic steatosis in metabolic associated fatty liver disease via the PI3K/AKT/mTOR and HIF-1/VEGF cascade. Biomed Pharmacother. 165:1152792023. View Article : Google Scholar : PubMed/NCBI | |
|
Elmorsy EA, Elsisi HA, Alkhamiss AS, Alsoqih NS, Khodeir MM, Alsalloom AA, Almeman AA, Elghandour SR, Nadwa EH, Khalifa AK, et al: Activation of SIRT1 by SRT1720 alleviates dyslipidemia, improves insulin sensitivity and exhibits liver-protective effects in diabetic rats on a high-fat diet: New insights into the SIRT1/Nrf2/NFκB signaling pathway. Eur J Pharm Sci. 206:1070022025. View Article : Google Scholar | |
|
Al-Gayyar MMH, Bagalagel A, Noor AO, Almasri DM and Diri R: The therapeutic effects of nicotinamide in hepatocellular carcinoma through blocking IGF-1 and effecting the balance between Nrf2 and PKB. Biomed Pharmacother. 112:1086532019. View Article : Google Scholar : PubMed/NCBI | |
|
Evangelou K, Vasileiou PVS, Papaspyropoulos A, Hazapis O, Petty R, Demaria M and Gorgoulis VG: Cellular senescence and cardiovascular diseases: Moving to the 'heart' of the problem. Physiol Rev. 103:609–647. 2023. View Article : Google Scholar | |
|
Li X, Li C, Zhang W, Wang Y, Qian P and Huang H: Inflammation and aging: Signaling pathways and intervention therapies. Signal Transduct Target Ther. 8:2392023. View Article : Google Scholar : PubMed/NCBI | |
|
Baumann A, Hernández-Arriaga A, Brandt A, Sánchez V, Nier A, Jung F, Kehm R, Höhn A, Grune T, Frahm C, et al: Microbiota profiling in aging-associated inflammation and liver degeneration. Int J Med Microbiol. 311:1515002021. View Article : Google Scholar : PubMed/NCBI |