Abstract
The number of individuals aged 65 years and older is expected to grow significantly in the coming decades. As life expectancy improves, the likelihood of developing chronic diseases, such as liver diseases, rises sharply with age. Aging is characterized by 3 main categories of hallmarks: primary, antagonistic, and integrative hallmarks. These categories are also observed in the liver, which ages more slowly than other organs. In this review, we summarize the current understanding of the mechanisms of aging as they pertain to the liver. This includes aging-related structural and functional changes in the liver, the roles of liver parenchymal and nonparenchymal cells, oxidative stress, and the sirtuin 1 protein. We also discuss how aging may influence the development and therapeutic management of various common liver diseases, including ischemia-reperfusion injury, DILI, alcohol-associated liver disease, and metabolic dysfunction–associated liver diseases.
Keywords: alcohol, autophagy, DILI, ischemia-reperfusion, MASLD
INTRODUCTION
The average life expectancy during the 1800s and early 1900s was projected to be around 40–50 years. During these times, lack of effective medical treatment, untreated infectious diseases, improper sanitation practices, and war were the leading causes of shortened lifespan. Since then, average expected life expectancy has drastically improved as people can live longer due to the advancement in medical practices, sanitation, and overall healthier lifestyles.1,2 Population studies performed by both the US Census Bureau and the World Health Organization (WHO) show that the number of individuals aged 65 and older gradually increases yearly. In the United States alone, this age group is projected to comprise ~25% of the population by 2050. Though people are living longer, increasing age is associated with a decline in quality of life as basic survival-related functions are lost. 3 In its simplest terms, aging is defined as a natural biological process in which an organism experiences a decline in physiological and cellular function over time. 4 The loss of cellular homeostasis associated with aging results in an inability to respond to stress caused by both internal and external insults, such as increased oxidative stress or exposure to environmental toxins. 4
In aging, several processes occur and culminate in the aged phenotype. Collectively, these processes are termed the “hallmarks of aging” and divided into 3 subgroups. The first subgroup is categorized as primary hallmarks and includes telomere attrition, genomic instability, epigenetic alterations, loss of proteostasis, and disabled autophagy. Primary hallmarks are events that initiate cellular damage that disrupt repair and regenerative mechanisms at the molecular level, leading to the increased occurrence of the remaining hallmarks.5–7 The second subgroup of hallmarks encompasses cellular mechanisms that function as restorative processes initially, as these processes will attempt to alleviate the damage created by the primary hallmarks and are termed as antagonistic pathways. The antagonistic subgroup hallmarks are dysregulated nutrient sensing, mitochondrial dysfunction, and cellular senescence.4–6 Nutrient sensing involves a complex network of pathways that are responsible for detecting alterations in nutrient availability and energy imbalances. Aging can promote dysregulated nutrient sensing due to the constant upregulation of promoters of aging, such as mechanistic target of rapamycin, insulin and IGF-1 1 or the functional loss of proteins that effectively extend lifespan, such as sirtuin proteins and AMP kinase (AMP-activated protein kinase [AMPK]). 8 The removal of senescent cells has been of interest in targeted therapies to improve cellular and organismal longevity. 9 The third subgroup of hallmarks is termed the integrative hallmarks and includes altered intracellular communication, stem cell exhaustion, dysbiosis, and chronic inflammation. 5 Many of the hallmarks in all 3 subgroups will subsequently activate inflammatory signaling in cells, tissues, and organ systems overall. The consistent presence of elevated inflammatory signaling molecules resulting from the persistent stress associated with aging will generate a state of chronic inflammation, leading to a further decline in the organism’s health.4–6,10 Overall, these 3 layers of hallmarks create complex interactions that may help explain certain aspects of the aging process.
Like other highly metabolic organs such as the heart and brain, the liver experiences a slower rate of aging, which increases its susceptibility to chronic liver pathologies that culminate in the degeneration of liver cells and the overall loss of hepatic function. 11 This review will discuss the mechanisms of liver cell aging and the relationship between hepatic aging and the development of liver dysfunction and diseases.
MECHANISMS OF AGING IN THE LIVER
Age-induced changes in liver structure and function
The liver is a dynamic organ responsible for performing many homeostatic functions, such as metabolic maintenance, detoxification, and bile production for fat digestion. 12 The liver undergoes significant alterations in structure and function as a person ages. Research utilizing ultrasound technology has shown that liver volume can decline by 20%–40% with increasing age. 13 Furthermore, both the rate of blood flow and alterations in the hepatic vascular endothelium, including loss of porosity or fenestration, were found to correlate with a decrease in liver volume in aged livers.14–16
The liver possesses a unique ability to regenerate after tissue damage or injury. This regeneration facilitates the restoration of liver mass through the replication of liver cells, primarily hepatocytes, in response to a signaling network initiated by cytokine release, followed by cell cycle progression stimulated by growth factors.17,18 In aging, liver regeneration declines or becomes impaired due to disrupted signaling pathways stemming from insufficient growth ligand-to-receptor binding or an increase in cell cycle inhibitory complexes, such as the one formed by CCAAT/enhancer-binding protein alpha (CEBP/α), histone deacetylase 1, and Brm, a chromatin remodeling protein.18,19 In addition to a decline in regenerative capacity, studies on aged rat livers have shown significant reductions in drug metabolism related to Cytochrome P450 (CYP) enzymes.20,21 However, studies on CYP-mediated metabolism in humans have produced mixed results. While some research has indicated a substantial age-related decline in the protein levels of CYP isozymes, such as CYP3A and CYP2E1, other studies have reported no significant functional changes in other CYP isozymes.22–24 Furthermore, as people age, there is a decrease in the protein content of albumin, which is produced by the liver. This reduction in albumin affects the drug-protein binding interactions necessary for adequate drug clearance by the liver. 25
Changes in parenchymal and nonparenchymal hepatic cells during liver aging
The liver is a heterogeneous organ containing various cell populations, essential for the liver’s overall function.12,26 The liver’s most prominent cell type, or parenchymal cell, is the hepatocyte, which comprises ~60%–80% of liver volume and performs most hepatic functions. Hepatocytes play a vital role in regulating glucose metabolism by facilitating glycolysis and gluconeogenesis. They also synthesize essential plasma proteins, such as albumin and complement proteins, which circulate in the body. In addition, hepatocytes are involved in lipid metabolism by synthesizing and secreting lipids. Furthermore, they produce bile acids, crucial for the absorption of lipids from the intestine. Moreover, hepatocytes are the primary cells responsible for detoxifying xenobiotics in the body.12,26,27 The remaining 20%–40% of liver volume consists of nonparenchymal hepatic cells, including hepatic stellate cells (HSCs), cholangiocytes, kupffer cells (KCs), and liver sinusoidal endothelial cells (LSECs).12,26 In the liver, HSCs are usually quiescent cells that store vitamin A and lipids and help to maintain the extracellular matrix (ECM). Upon activation due to stress or injury, HSCs promote the accumulation of scar tissue in the liver, leading to fibrosis and cirrhosis.26,28 Cholangiocytes are cells that line the epithelium of the bile duct and are involved in the modification and secretion of bile from the liver. 29 As the name implies, LSECs are specialized cells that line the hepatic sinusoids and are fenestrated to allow for blood filtration.28,30 Lastly, the liver contains its unique population of immune cells, KCs, that regulate innate immunity and perform phagocytic activity 28
The 3 general layers of aging hallmarks that apply to other tissues and organs we discussed above also pertain to the liver. In the first layer, telomere shortening, a standard marker of aging, occurs in the liver and is linked to hepatocyte senescence. This shortening is associated with a reduced regenerative capacity and an increased risk of chronic liver diseases, such as cirrhosis and hepatocellular carcinoma (HCC). 31 Hepatocyte polyploidy, decreased telomere length, and hypertrophy were observed in hepatocytes from aged rodent livers and human hepatocytes.11,32 Moreover, aged hepatocytes exhibit changes in DNA methylation and histone acetylation, contributing to increased genomic instability in the aging liver.33,34 The liver’s cellular proteostasis and organelle quality control are primarily regulated by 2 major degradation pathways: the proteasome system and the autophagy-lysosomal system. Aging causes age-dependent decreases in the liver’s 26S and 20S proteasome activity. 35 The liver has 3 types of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy. These types differ in the routes used to deliver autophagic cargo to the lysosome. 36 Aging was connected to decreased autophagy-related proteins such as microtubule-associated protein 1B-light chain 3 (LC3), autophagosome structures, and increases in primary, or unfused, lysosomes. 37 In addition, LAMP2A levels, key receptors for chaperone-mediated autophagy, decreased in the aged liver.38–40 Intriguingly, elevated protein levels of cathepsin B were observed in the livers of aged rats, likely a compensatory response to increased expression of an endogenous cysteine peptidase inhibitor and impaired lysosomal functions. 41 Indeed, aged hepatocytes exhibit increased lysosomal leakage resulting in the progressive accumulation of lysosomal content, such as cathepsin D, in the cytosol. 42 One of the most common features of aging’s effects on autophagy and lysosomal function is the excessive accumulation of lipofuscin within lysosomes. 43 Lipofuscin is termed the “age-pigment” as it commonly accumulates in aged tissues. 44 Notably, hepatocyte lysosomes have increased lipofuscin accumulation in aged livers, suggesting impaired lysosomal degradation. 45 While changes in autophagy within the liver can impair its overall function, pharmacological and genetic strategies to enhance autophagy have been shown to improve liver function in aging. 46
In the second layer of aging liver hallmarks, mechanistic target of rapamycin complex 1, a key regulator of protein synthesis and cellular metabolism, is decreased in aged animal livers. 47 AMPK is a key sensor of cellular energy and an essential regulator of cellular metabolism. In aged mice, the basal activity of AMPK α1 increases, whereas AMPK α2 does not show this increase. In addition, both isoforms of AMPK in the livers of aged mice demonstrate a lack of response to hypoxia, suggesting an overall impairment of the AMPK pathway. 48 Aging caused increased cristae fragmentation and vacuole formation in the mitochondria of hepatocytes. 49 The maintenance and sustainability of ATP production is highly reliant on the mitochondrial membrane potential generated by the proton pumps in the electron transport chain located on the inner mitochondrial membrane. 50 A loss of mitochondrial membrane potential leads to mitochondrial damage and subsequent cell death. Mitochondrial membrane potential is significantly decreased in aged hepatocytes with elevated levels of oxidized mitochondrial proteins. 51 In addition, hepatocyte aging is associated with various markers of cellular senescence, such as increased senescence-associated-β-galactosidase activity, p16INK4a (p16), p21, and γ-H2AX. 52 Aged hepatocytes also exhibit enlarged vacuolated nuclei, which are associated with increased DNA damage and cellular senescence. 53
In the third layer of liver aging hallmarks, the blood-borne systemic factors with proaging or antiaging properties for cell-to-cell communication and changes in the liver ECM are closely associated with the nonparenchymal hepatic cells. Transplanting young blood into aged rats reverses age-related hepatic dysfunction and restores liver regeneration, although the specific antiaging factors in the young blood have not yet been identified. 54 Aged livers exhibit impaired ECM remodeling, altered composition (increased collagen and decreased growth factors), and increased stiffness, all contributing to fibrosis and liver dysfunction.55,56 Studies evaluating the correlation between HSCs, aging, and fibrosis found that HSCs in aged livers exhibited characteristics more typical of myofibroblasts, demonstrated by increased positive staining of alpha-smooth muscle actin. Evidence also indicated elevated expression levels of the profibrogenic genes such as TGF-β and platelet-derived growth factor receptor-beta in aged livers.32,57,58 This transition of HSCs from quiescent to activated can be partly attributable to the elevation of circulating inflammatory mediators associated with aging. 57 Notably, the loss of efficient hepatic blood flow in the aging liver is primarily due to the thickening of the endothelium of hepatic sinusoids. Furthermore, the expression of p16, along with the reduced expression of VEGF receptor-2 and CD32b, proteins essential for LSEC development and function, is present in the aging liver.15,16,58 In addition, the age-induced changes to LSEC impact drug clearance due to a loss of fenestration in aged livers.59,60 Though the effect of aging on the liver microenvironment has been investigated for many years, the breadth of research surrounding changes in KC composition and function in aged livers is scarce. The number and activities of KCs increased and are associated with increased IL-6 production in the aged liver.61,62 The phagocytic activity of KCs in aged livers has been controversial, as some studies have reported a substantial increase in KC endocytic activity, while others have noted a decline in this function of KCs in aged rat livers.63,64 Studies on age-related effects in cholangiocytes are also limited and primarily focus on the role of cellular senescence in the progression of severe cholestatic diseases, which are more prevalent in older adults. 15 Moreover, aged mice show increased intestinal barrier impairment and higher bacterial endotoxin levels, activating Toll-like receptor 4 signaling in the liver. These changes are associated with liver degeneration, inflammation, and fibrosis. 65 Over time and as the organism ages, the effectiveness of the immune response diminishes, leading to the occurrence of immunosenescence and a continuation of chronic inflammation, or inflammaging, where low-grade production of cytokines occurs.66,67 As one of the molecular drivers of inflammatory signaling in macrophages, the upregulated expression of forkhead box protein O1 was correlated to a significant increase in inflammaging-associated markers such as NF-κβ and circulating cytokines like IL-1β in aged mice as well as isolated macrophages from aged mouse livers. 68 Notably, declined liver function in aged individuals can be reversed using hepatic stem cells or liver progenitor cells, which can regenerate and repair the liver. In aged mouse livers, the activation of liver progenitor cells decreases, likely due to increased production of reactive oxygen species from infiltrating neutrophils. 69 Accordingly, a stem cell–based approach has been proposed as a promising therapeutic strategy for enhancing liver regeneration and combating liver aging and its associated diseases. 70 The current understanding of the 3 layers of hallmarks in liver aging is summarized in Figure 1.
FIGURE 1.
Three sets of aging hallmarks in the liver. The scheme illustrates the 3 subsets of proposed hallmarks in liver aging. Primary hallmarks include a decline of proteasome and autophagy, an increase of hepatocyte polyploidy, telomere attrition, and epigenetic and genomic instability. Antagonistic hallmarks include decreased mTORC1 and AMPK, increased senescence, mitochondrial dysfunction, and vacuolated hepatocyte nuclei. Integrative hallmarks include altered dysbiosis, cell-cell communication, decreased LPC/stem cells, increased inflammation, and ECM. Abbreviations: AMPK, AMP-activated protein kinase; ECM, extracellular matrix; LPC, liver progenitor cell; mTOR, mechanistic target of rapamycin.
The role of oxidative stress in the aged liver
Oxidative stress increases across various tissue types in the aged organism, contributing to the increased occurrence of many of the aging hallmarks.5–7 Oxidative stress occurs because of the imbalance between reactive oxygen and nitrogen species and the antioxidant defense systems responsible for reducing the harmful effects of reactive oxygen and nitrogen species on many intracellular biomolecules, including lipids, proteins, and DNA. Oxidative stress has been connected to the development and progression of aging as it stimulates the onset of cellular senescence through the induction of several intracellular mechanisms.71,72 These include the onset of cell cycle arrest through the induction of cell cycle arrest mediators like p16 and p21, induction of proinflammatory mediators like NF-κB, and a persistent activation of cellular matrix remodeling proteins.72,73 Furthermore, oxidative damage to cellular membranes induced by the elevated levels of reactive oxygen and nitrogen species can result in the release of “danger” signals from the cell, termed damage-associated molecular patterns, sparking an innate inflammatory response. 74 In rodent models, oxidative stress was confirmed to accumulate in the aged liver as evidenced by substantial rises in lipofuscin and lipid peroxidation content in conjunction with a diminished capacity for eliminating toxic fatty acid species.32,43,45,75 Oxidative stress and aging cooperatively impair the liver’s ability to regenerate after injury. 18
Sirtuin 1 in the aged liver: controversial topic
In aging research, several signaling pathways have been identified as playing a significant role in the progression of aging. One family of molecules extensively studied is the sirtuin family, a class of NAD+-dependent deacetylases. The sirtuin family comprises 7 members (1–7) that rely on NAD+ to function and regulate metabolism in response to cellular stress. Though all 7 sirtuins have an important biological function, mammalian Sirtuin 1 (SIRT1) is the most thoroughly studied.76,77 SIRT1 is found in both the cytoplasm and nucleus and functions as a deacetylase to remove acetyl groups from histones, regulating gene expression. In metabolic regulation, SIRT1 activates transcriptional activators and coactivators like peroxisome proliferator–activated receptor alpha (PPARα) and PPARgamma coactivator 1 alpha (PGC1α) to induce transcription of genes involved in mitochondrial ATP production, gluconeogenesis, and fatty acid metabolism. 78 In response to oxidative stress, SIRT1 deacetylates multiple proteins involved in DNA repair, antioxidant enzymes for mitigating reactive oxygen species in mitochondria, and genes involved in cell death versus cellular repair pathways.78,79 Decreased SIRT1 levels in aged livers have been correlated to higher levels of oxidative stress, increased lipid accumulation, and increased expression of fibrotic factors such as alpha-smooth muscle actin and TGF-β in the presence of various hepatotoxins, as well as in metabolic dysfunction–associated steatotic liver diseases (MASLD).80,81 Furthermore, the age-associated loss of hepatic SIRT1 or genetic ablation of SIRT1 resulted in upregulated inflammatory signaling as well as inflammasome induction, as evidenced by an increase in NLRP3 (NACHT, LRR, and PYD domains–containing protein 3) in aging rodent models. 80 Genetic or pharmacological activation of SIRT1 decreases lipogenesis, induces fatty acid β-oxidation, decreases the presence of senescence-associated genes such as p16, and improves glucose tolerance and utilization in aged rodent livers.81,82
Numerous studies have highlighted the roles of NAD and Sirtuins in extending lifespan, leading to a strong interest in finding small-molecule activators for SIRT1 to address aging.77,78,83 However, the role of SIRT1 in regulating aging and longevity remains controversial. The Sir2 gene was first identified in yeast, which functions in transcriptional silencing. Subsequent research revealed that Sir2 and its orthologs are critical for extending the lifespan of yeast, worms, and flies.84,85 Despite this, overexpressing Sir2 has not consistently resulted in lifespan extension in Caenorhabditis elegans and Drosophila. 39 Only mice with Sirt1 overexpressed in specific brain regions, rather than those with whole body Sirt1 overexpression, exhibit lifespan extension.82,86 This raises questions about the significance of Sir2 orthologs in aging and longevity. Furthermore, while the supplementation of NAD+ or its precursors has demonstrated beneficial effects on improving insulin resistance, glucose metabolism, heart health, hepatic steatosis, and chronic inflammation contributing to a “healthspan,” such supplementation does not extend lifespan in mice. 87 It is also unclear if these benefits are primarily due to the activation of SIRT1. 76 Regardless of its impact on lifespan, the role of SIRT1 in maintaining liver health during aging can be explored using liver-specific gain-of-function approaches (such as transgenic models or viral vector-mediated overexpression) and loss-of-function strategies (like liver-specific knockout) in various liver disease models in the future. Below, we discuss the role and mechanisms of aging in various liver diseases, including hepatic ischemia-reperfusion injury (IRI), DILI, alcohol-associated liver disease (ALD), and MASLD.
AGING IN LIVER DISEASES
Aging in hepatic IRI
Liver transplantation remains the only effective clinical method for treating end-stage liver disease. However, the shortage of donor livers has resulted in a growing number of patients on the waiting list for transplantation. There is an increasing need to expand the donor pool to include steatotic livers, older donors, and donations after cardiac death. In addition, the rising prevalence of severe liver diseases such as ALD, MASLD, and HCC in the elderly population heightens the demand for liver transplantation, which contributes to the need for more donors. 88 Given the shortage of donor grafts, it is essential to investigate whether older patients might have poorer posttransplant survival rates and consume excessive resources postoperatively, thus affecting the procedure’s economic viability. Below, we discuss the potential impacts of aging patients receiving liver transplants and the effects of aged liver donors on transplantation outcomes.
The prospect of performing liver transplantations in elderly adults raises significant concerns. Many age-related factors exacerbate hepatic IRI and negatively impact the success and survival rates of liver transplantation. 89 Animal studies have shown that older age worsens hepatic IRI due to changes in the hepatic microvasculature, in which LSECs respond less to signals for vasodilation, along with significant increases in capillarization. 90 One early study indicated that patient survival rates were significantly lower among recipients of liver transplant over 60. 40 However, this excess mortality was attributed not to liver conditions but to nonhepatic age-related issues such as infections, cardiac problems, and neurological diseases occurring within 6 months after transplant. 40 More recent studies have indicated that the operative course, length of hospitalization, and incidence of perioperative complications for patients older than 60 years are comparable to those of their younger counterparts. As a result, advanced age is no longer deemed a contraindication to transplantation.91,92 However, it is vital to screen for cardiopulmonary comorbidities, asymptomatic malignancies, nutritional status, and frailty to ensure optimal outcomes and prevent futile transplantation at most centers.
In experimental animal models, aged liver grafts have shown lower ATP production and increased generation of free radicals, both of which exacerbate the inflammatory response. Inflammation, driven by activated KCs and neutrophils, is increased in aged livers due to elevated NF-κB signaling, which worsens hepatocyte injury.89,93 In addition, age-related loss of autophagy and changes in mitochondrial dynamics have been linked to increased injury in hepatic IRI. Specifically, the age-associated decline of autophagy-related protein 4b (Atg4b), impaired mitophagy, and a significant reduction in ATP production have all contributed to increased signs of IRI in aged rodent livers. 94 Overexpression of either Atg4b or Beclin-1 enhanced autophagy, blocked the onset of mitochondrial permeability transition, and suppressed cell death following ischemia-reperfusion in old hepatocytes. 94 Moreover, aging also exacerbates hepatic IRI by impairing age-dependent mitophagy due to insufficient PARKIN expression in mice. 95 Consequently, strategies targeting the mechanisms involved in energy metabolism, the inflammatory response, and autophagy may be particularly effective in preventing the increased risk of IRI in aged livers following major hepatic surgery.
What about younger recipients receiving aged liver grafts? A retrospective cohort study indicated that transplanting livers from donors aged 40–59 and 60 years and older was associated with poorer graft survival in recipients younger than 40. 96 However, due to advancements in patient care, surgical techniques, and improved donor-to-recipient matching, more recent studies have shown improved posttransplant mortality, reduced all-cause graft loss, and utilization of comparable resources in liver transplant recipients ≥ 60 years of age versus younger recipients. 97 While there is an effort to prioritize younger organs for younger recipients, expanding the donor pool by using liver grafts from older donors may be a reasonable approach in the future.
Aging in DILI
The liver is essential for drug metabolism and clearance, which are mediated by phase I and phase II drug metabolism. Phase I is mediated by CYPs, which regulate reactions involving modifications such as oxidation, reduction, and hydrolysis. In contrast, phase II metabolism involves conjugation reactions with endogenous molecules like glucuronic acid, sulfate, or glutathione, increasing their water solubility for easier excretion. 98 Reductions in certain CYPs occur due to advanced age, as well as decreases in hepatic blood flow and volume, all of which can alter drug metabolism in the elderly.99,100 In this age group, hepatic drug clearance for specific medications can be reduced by up to 30%, while phase II metabolism is relatively preserved. 100 These notable changes in drug metabolizing enzymes, decreased drug absorption, and clearance can lead to developing more severe adverse drug reactions and increase DILI risks in aged individuals.25,60,101
Increased oxidative stress, the loss of function of antioxidant regulators such as superoxide dismutase and glutathione, and decreased liver regeneration all contribute to the exacerbation of acetaminophen hepatotoxicity in aged mice. 102 Clinical studies have shown that the severity of DILI is linked to a higher prevalence of severe adverse drug reactions in older patients. This is often due to the presence of comorbidities such as chronic liver disease, cardiovascular disease, and impaired renal function, along with the use of multiple prescribed medications, known as polypharmacy.101,103,104 Age also affects the phenotypes of DILI. For instance, hepatocellular-type DILI is more common among pediatric patients, while cholestatic-type DILI and chronic DILI are more prevalent in the elderly. 105 In addition, susceptibility to DILI in older adults may be influenced by certain medications and patterns of exposure (such as a higher prevalence of medication use, polypharmacy, and drug interactions). The cognitive impairments in the elderly also affect adherence, or age-related changes in pharmacokinetics and pharmacodynamics. 106 While there are currently no specific therapies for DILI, understanding the risk factors associated with DILI in the aging population may help enhance diagnostic and prognostic capabilities to protect these vulnerable populations.
Aging in ALD
ALD is one of the leading causes of liver disease and alcohol-associated deaths, with reported mortality rates as high as 44% in the United States alone. 107 ALD begins with lipid accumulation in hepatocytes and progresses to increased cell death. It can further develop into more severe conditions such as steatohepatitis, fibrosis, cirrhosis, and ultimately HCC.108,109 The pathogenesis of alcohol-induced liver injury involves disrupting numerous physiological processes in the liver. In brief, alcohol is primarily metabolized in the cytoplasm by alcohol dehydrogenases 1A (ADH1A), ADH1B, ADH1C, and ADH4. 110 This process is followed by the breakdown of alcohol through enzymes such as mitochondrial acetaldehyde dehydrogenase 2 (ALDH2), CYP2E1 in microsomes, and catalases in peroxisomes, resulting in the production of acetaldehyde and acetate.110,111 The metabolism of ethanol by the ADH and ALDH2 enzymes requires NAD+, resulting in an initial decline of hepatic NAD+ and a concomitant increase in the production of the reduced form of nicotinamide adenine dinucleotide (NADH). Decreased levels of hepatic NAD+ and an increased NADH/NAD+ ratio lead to impaired alcohol metabolism, mitochondrial dysfunction, megamitochondria accumulation, oxidative stress, and hepatocyte degeneration in the liver.112,113
In older adults and the elderly, alcohol use has reportedly increased in recent years. In a drug use and health survey conducted by the Substance Abuse and Mental Health Services Administration (SAMHSA), ~15% of the total participants aged 65 years and older reported that they engaged in binge drinking as well as heavy alcohol use within a 1-month timeframe in 2023. In 2019, alcohol-associated cirrhosis accounted for 21.8% of cases among the elderly, totaling 3.23 million. Furthermore, liver cancer from alcohol in this group made up 51.27% of all related cases. 114 Many mechanisms may explain the increased sensitivity to alcohol-induced liver injury in the elderly. First, alcohol concentrations are elevated in the blood due to diminished activity of alcohol-metabolizing enzymes such as ADH, ALDH2, and CYP2E1 in aging livers, along with a concurrent decrease in liver volume in these older livers.13,115 Second, decreased hepatic cellular proteostasis and organelle quality control pathways involve both the proteasome and autophagy-lysosomal pathways in the aged liver.35,45 Third, increased cellular senescence, exacerbated by alcohol consumption, may contribute to the overall decline of hepatocytes and the progression of ALD. 116 However, more direct studies are needed to clarify the role of senescence in alcohol-induced liver issues in aged individuals. Fourth, an elevated basal inflammatory environment in aged livers further promotes alcohol-induced inflammation and fibrosis.117,118 Fifth, lower levels of SIRT1 in hepatocytes and HSC in aged mouse livers worsen alcohol-induced liver fibrosis. This was demonstrated by HSC-specific SIRT1 knockout mice, which were more susceptible to short-term-plus-binge ethanol-induced liver fibrosis, exhibiting upregulation of platelet-derived growth factor receptor-α expression. Restoration of SIRT1 expression in the mouse liver and HSCs improved short-term-plus-binge ethanol-induced liver injury and fibrosis in middle-aged mice. 118 Moreover, excessive alcohol intake can not only exacerbate the harmful effects on the aging liver but also worsen commonly occurring age-related comorbidities such as hypertension, cardiovascular disease, and diabetes mellitus, which further impair the liver’s ability to manage injury in older adults.119,120 Therefore, alcohol treatment programs specifically designed for older adults may be more effective in meeting their unique needs and challenges by considering age-related physiological changes, comorbidities, and cognitive impairment.
Aging in MASLD
MASLD, previously known as NAFLD, affects more than 30% of the global population. MASLD is highlighted by >5% steatosis in conjunction with at least one of the cardiometabolic risk factors.121,122 According to the consensus on the diagnostic criteria in adults, the cardiometabolic risk factors associated with MASLD include hypertriglyceridemia, hyperglycemia, hypertension, obesity, and severely decreased HDL levels.122,123 The molecular pathogenesis driving MASLD is similar to that of ALD; however, the key difference between the 2 diseases is excessive alcohol intake.124,125
The incidence of MASLD in the elderly population continues to grow globally. An Australian study found that the estimated rate of MASLD in patients over 70 was 33%. 126 Elderly patients exhibited worsening conditions of fibrosis and steatohepatitis. 127 Cellular senescence associated with aging correlates with increased hepatic fat accumulation in MASLD. 128 Moreover, the aging liver exhibits increased lipogenesis and decreased peroxisomal fatty acid oxidation,129,130 enhancing hepatic lipid accumulation in older individuals. In aged animals, the mitochondrial network is often more heterogeneous, fragmented, and made up of large, swollen mitochondria, which are more challenging to remove through mitophagy.131,132 This leads to the accumulation of megamitochondria. While the formation of megamitochondria is initially an adaptive response, it may become maladaptive and contribute to increased oxidative stress and susceptibility to the pathogenesis of MASLD and ALD in the aging liver.112,113,132,133 Moreover, general hepatic autophagy may also decline during aging, contributing to the dysregulation of lipid metabolism, upregulated inflammatory signaling, and the development of fibrosis in advanced MASLD.46,134 Animal studies conducted by You-Jin Choi and colleagues investigated the role of dysfunctional chaperone-mediated autophagy in isolated aged mouse hepatocytes with a knocked-down Lamp2a gene. They found that fatty acid oxidation decreased due to a significant increase in nuclear receptor corepressor 1 (NCoR1), a negative regulator of PPARα. The elevated levels of NCoR1 are likely the result of decreased autophagy in the livers of aged mice, as autophagy plays a role in selectively degrading NCoR1. 135 This change in fatty acid metabolism was linked to increased lipid accumulation and the subsequent development of fatty liver in aged mice. Older mice experience more hepatocyte ferroptosis and liver degeneration than younger mice when fed diets that induce MASLD. By inhibiting ferroptosis, older mice’s liver gene expression profile shifts to resemble that of younger mice, effectively reversing the liver damage caused by aging. 136 In addition to aging, sex is a significant determinant of liver gene expression and function, especially in response to a high-fat diet. Aging leads to a decline in metabolic flexibility, which affects the liver’s ability to adjust its gene expression programs. High-fat diet feeding causes higher expression levels of genes related to inflammation, fibrosis, and the ECM in aged male mice, but not in aged female mice, suggesting a sexual dimorphism in how the aged liver responds to a high-fat diet. 137 Necroptosis is a form of nonapoptotic cell death regulated by the receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage domain kinase-like (MLKL) pathways. 138 Increased levels of phosphorylated RIPK1, RIPK3, and MLKL were observed in the livers of aged mice, which may exacerbate diet-induced MASLD in older individuals. 139
To improve and treat MASLD in older patients, lifestyle modifications such as weight loss through diet and exercise, as well as the management of related conditions like diabetes, high cholesterol, and high blood pressure, can be beneficial. Medications like GLP-1 receptor agonists and resmetirom (brand name Rezdiffra), the first FDA-approved treatment for MASH, may also aid in managing this condition in elderly patients. Research has indicated that in liver biopsies from patients with MASH, the expression of thyroid hormone receptor beta is inversely correlated with the MASH score and tends to decline with age. 140 Therefore, it is crucial to consider these age-related changes when designing management and treatment plans for older patients with MASLD.
CONCLUSIONS AND FUTURE PERSPECTIVES
As the global population grows, the number of older adults is increasing rapidly. While the liver can maintain some of its homeostatic functions throughout aging, it becomes highly susceptible to liver injury and chronic liver diseases. This susceptibility is mainly due to decreased hepatic volume, reduced blood flow, altered microvasculature, defective metabolizing enzymes, impaired proteostasis, mitochondrial dysfunction, and reduced expression of hormone receptors. Furthermore, chronic exposure to external factors such as polypharmacy, excessive alcohol consumption, and nutritional imbalances may exacerbate harmful inflammatory signaling, cellular senescence, and elevated oxidative stress. These changes ultimately hinder the aged liver’s ability to manage cell death and disease progression effectively. These age-related anatomical and molecular changes in the aged liver exacerbate IRI, DILI, ALD, and MASLD (Figure 2). The aging-related changes also significantly influence how liver diseases are managed and treated in elderly patients. In addition, there is limited information regarding sexual differences in aging liver disease, which warrants further studies using aged animal models to explore how sex hormones and menopause affect liver diseases in older females. Given the rapid advancements in artificial intelligence within biological research, it will be exciting to see how artificial intelligence can predict liver disease progression in aging populations in future studies.
FIGURE 2.
The proposed role of aging in liver diseases. (A) IRI: Aging livers experience reduced levels of ATG4B and PARKIN, leading to decreased autophagy/mitophagy and increased ROS. Enhanced NF-κB activation in macrophages causes greater inflammation, worsening IRI. (B) DILI: Older adults have decreased liver drug metabolism due to reduced activity of CYP enzymes and lower clearance, along with diminished GSH and SOD levels. These factors, combined with polypharmacy, raise susceptibility to DILI. (C) ALD: Aging diminishes alcohol-metabolizing enzymes and proteostasis mechanisms like autophagy/proteasome. Decreased SIRT1 levels and increased inflammation further exacerbate ALD. (D) MASLD: The aging liver shows elevated necroptosis and ferroptosis proteins, increased lipogenesis, and inflammation, while fatty acid beta-oxidation and autophagy decrease, leading to more severe MASLD. Abbreviations: ADH, alcohol dehydrogenase; ALD, alcohol-associated liver disease; ALDH2, acetaldehyde dehydrogenase 2; CYP, cytochrome; GSH, glutathione; IRI, ischemia-reperfusion injury; MASLD, metabolic dysfunction–associated steatotic liver disease; ROS, reactive oxygen species; SIRT1, sirtuin 1; SOD, superoxide dismutase.
AUTHOR CONTRIBUTIONS
Wen-Xing Ding conceived and supervised the project. Sha Neisha Williams wrote the draft. Wen-Xing Ding revised the manuscript and made the figures.
FUNDING INFORMATION
This study was partly supported by the National Institute of Health (NIH) funds R37 AA020518, R21 AA030617, and R01AA031230 (Wen-Xing Ding) as well as William Warner Abercrombie endowment from The Cancer Center of KUMC (Wen-Xing Ding). Sha Neisha Williams was supported by 1F31AA031623-01.
CONFLICTS OF INTEREST
The authors have no conflicts to report.
Footnotes
Abbreviations: ADH1A, alcohol dehydrogenase 1A; ADH1B, alcohol dehydrogenase 1B; ADH1C, alcohol dehydrogenase 1C; ADH4, alcohol dehydrogenase 4; AMPK, AMP-activated protein kinase; ALD, alcohol-associated liver disease; ALDH2, acetaldehyde dehydrogenase 2; Atg4b, autophagy-related protein 4b;CEBP/α, CCAAT/enhancer-binding protein alpha, C. elegans, Caenorhabditis elegans; CYP, cytochrome -P450; ECM, extracellular matrix; FDA, Food and Drug Administration; GLP-1, glucagon-like peptide-1; IRI, ischemia-reperfusion injury; KCs, Kϋpffer cells; LAMP2A, lysosomal-associated membrane protein 2A; LC-3, microtubule-associated protein 1B-light chain 3; MASH, metabolic dysfunction-associated steato-hepatitis; MASLD, metabolic dysfunction–associated steatotic liver disease; MLKL, mixed lineage domain kinase-like; p16, p16INK4a; p21, cyclin-dependent kinase inhibitor 1; PGC1α, PPARgamma coactivator 1 alpha; PPARα, peroxisome proliferator-activated receptor alpha; RIPK1, receptor-interacting protein kinase 1; RIPK3, receptor-interacting protein kinase 3; SAMHSA, Substance Abuse and Mental Health Services Administration; Sir2, sirtuin 2; SIRT1, mammalian sirtuin-1; TGF-β, transforming growth factor-beta; TLR4, toll-like receptor 4; WHO, World Health Organization.
Contributor Information
Sha Neisha Williams, Email: swilliams30@kumc.edu.
Wen-Xing Ding, Email: wxding@kumc.edu.
REFERENCES
- 1. Crimmins EM. Lifespan and healthspan: Past, present, and promise. Gerontologist. 2015;55:901–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Oeppen J, Vaupel JW. Demography. Broken limits to life expectancy. Science. 2002;296:1029–1031. [DOI] [PubMed] [Google Scholar]
- 3. Brown GC. Living too long: The current focus of medical research on increasing the quantity, rather than the quality, of life is damaging our health and harming the economy. EMBO Rep. 2015;16:137–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Tenchov R, Sasso JM, Wang X, Zhou QA. Aging hallmarks and progression and age-related diseases: A landscape view of research advancement. ACS Chem Neurosci. 2024;15:1–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: An expanding universe. Cell. 2023;186:243–278. [DOI] [PubMed] [Google Scholar]
- 6. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, et al. Aging and aging-related diseases: From molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 2022;7:391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Pan H, Finkel T. Key proteins and pathways that regulate lifespan. J Biol Chem. 2017;292:6452–6460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018;24:1246–1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Tartiere AG, Freije JMP, Lopez-Otin C. The hallmarks of aging as a conceptual framework for health and longevity research. Front Aging. 2024;5:1334261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Chatterjee N, Sharma R, Kale PR, Trehanpati N, Ramakrishna G. Is the liver resilient to the process of ageing? Ann Hepatol. 2024;30:101580. [DOI] [PubMed] [Google Scholar]
- 12. Trefts E, Gannon M, Wasserman DH. The liver. Curr Biol. 2017;27:R1147–R1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Wang YXJ. Gender-specific liver aging and magnetic resonance imaging. Quant Imaging Med Surg. 2021;11:2893–2904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Wakabayashi H, Nishiyama Y, Ushiyama T, Maeba T, Maeta H. Evaluation of the effect of age on functioning hepatocyte mass and liver blood flow using liver scintigraphy in preoperative estimations for surgical patients: Comparison with CT volumetry. J Surg Res. 2002;106:246–253. [DOI] [PubMed] [Google Scholar]
- 15. Baiocchi L, Glaser S, Francis H, Kennedy L, Felli E, Alpini G, et al. Impact of aging on liver cells and liver disease: Focus on the biliary and vascular compartments. Hepatol Commun. 2021;5:1125–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. McLean AJ, Cogger VC, Chong GC, Warren A, Markus AM, Dahlstrom JE, et al. Age-related pseudocapillarization of the human liver. J Pathol. 2003;200:112–117. [DOI] [PubMed] [Google Scholar]
- 17. Fausto N, Campbell JS, Riehle KJ. Liver regeneration. Hepatology. 2006;43:S45–S53. [DOI] [PubMed] [Google Scholar]
- 18. Michalopoulos GK, Bhushan B. Liver regeneration: Biological and pathological mechanisms and implications. Nat Rev Gastroenterol Hepatol. 2021;18:40–55. [DOI] [PubMed] [Google Scholar]
- 19. Timchenko NA. Aging and liver regeneration. Trends Endocrinol Metab. 2009;20:171–176. [DOI] [PubMed] [Google Scholar]
- 20. Schmucker DL. Age-related changes in liver structure and function: Implications for disease? Exp Gerontol. 2005;40:650–659. [DOI] [PubMed] [Google Scholar]
- 21. Schmucker DL. Aging and the liver: An update. J Gerontol A Biol Sci Med Sci. 1998;53:B315–B320. [DOI] [PubMed] [Google Scholar]
- 22. Tanaka E. In vivo age-related changes in hepatic drug-oxidizing capacity in humans. J Clin Pharm Ther. 1998;23:247–255. [DOI] [PubMed] [Google Scholar]
- 23. Konstandi M, Johnson EO. Age-related modifications in CYP-dependent drug metabolism: Role of stress. Front Endocrinol (Lausanne). 2023;14:1143835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. George J, Byth K, Farrell GC. Age but not gender selectively affects expression of individual cytochrome P450 proteins in human liver. Biochem Pharmacol. 1995;50:727–730. [DOI] [PubMed] [Google Scholar]
- 25. Le Couteur DG, McLean AJ. The aging liver. Drug clearance and an oxygen diffusion barrier hypothesis. Clin Pharmacokinet. 1998;34:359–373. [DOI] [PubMed] [Google Scholar]
- 26. Kalra A, Yetiskul E, Wehrle CJ, Tuma F. Physiology, liver. In: StatPearls, StatPearls Publishing LLC; 2025. [PubMed] [Google Scholar]
- 27. Kmiec Z. Cooperation of liver cells in health and disease. Adv Anat Embryol Cell Biol. 2001;161:1–151; III–XIII. [DOI] [PubMed] [Google Scholar]
- 28. Ishibashi H, Nakamura M, Komori A, Migita K, Shimoda S. Liver architecture, cell function, and disease. Semin Immunopathol. 2009;31:399–409. [DOI] [PubMed] [Google Scholar]
- 29. Cai X, Tacke F, Guillot A, Liu H. Cholangiokines: Undervalued modulators in the hepatic microenvironment. Front Immunol. 2023;14:1192840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Shetty S, Lalor PF, Adams DH. Liver sinusoidal endothelial cells—Gatekeepers of hepatic immunity. Nat Rev Gastroenterol Hepatol. 2018;15:555–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Wiemann SU, Satyanarayana A, Tsahuridu M, Tillmann HL, Zender L, Klempnauer J, et al. Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis. FASEB J. 2002;16:935–942. [DOI] [PubMed] [Google Scholar]
- 32. Hunt NJ, Kang SWS, Lockwood GP, Le Couteur DG, Cogger VC. Hallmarks of aging in the liver. Comput Struct Biotechnol J. 2019;17:1151–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Bacalini MG, Franceschi C, Gentilini D, Ravaioli F, Zhou X, Remondini D, et al. Molecular aging of human liver: An epigenetic/transcriptomic signature. J Gerontol A Biol Sci Med Sci. 2019;74:1–8. [DOI] [PubMed] [Google Scholar]
- 34. Price AJ, Manjegowda MC, Kain J, Anandh S, Bochkis IM. Hdac3, Setdb1, and Kap1 mark H3K9me3/H3K14ac bivalent regions in young and aged liver. Aging Cell. 2020;19:e13092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Dasuri K, Zhang L, Ebenezer P, Liu Y, Fernandez-Kim SO, Keller JN. Aging and dietary restriction alter proteasome biogenesis and composition in the brain and liver. Mech Ageing Dev. 2009;130:777–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Qian H, Chao X, Williams J, Fulte S, Li T, Yang L, et al. Autophagy in liver diseases: A review. Mol Aspects Med. 2021;82:100973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Uddin MN, Nishio N, Ito S, Suzuki H, Isobe K. Autophagic activity in thymus and liver during aging. Age. 2012;34:75–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Kaushik S, Cuervo AM. The coming of age of chaperone-mediated autophagy. Nat Rev Mol Cell Biol. 2018;19:365–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Burnett C, Valentini S, Cabreiro F, Goss M, Somogyvari M, Piper MD, et al. Absence of effects of Sir2 overexpression on lifespan in C. elegans and Drosophila. Nature. 2011;477:482–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Zetterman RK, Belle SH, Hoofnagle JH, Lawlor S, Wei Y, Everhart J, et al. Age and liver transplantation: A report of the Liver Transplantation Database. Transplantation. 1998;66:500–506. [DOI] [PubMed] [Google Scholar]
- 41. Keppler D, Walter R, Perez C, Sierra F. Increased expression of mature cathepsin B in aging rat liver. Cell Tissue Res. 2000;302:181–188. [DOI] [PubMed] [Google Scholar]
- 42. Dutta RK, Lee JN, Maharjan Y, Park C, Choe SK, Ho YS, et al. Catalase-deficient mice induce aging faster through lysosomal dysfunction. Cell Commun Signal. 2022;20:192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Gomez-Sintes R, Ledesma MD, Boya P. Lysosomal cell death mechanisms in aging. Ageing Res Rev. 2016;32:150–168. [DOI] [PubMed] [Google Scholar]
- 44. Gray DA, Woulfe J. Lipofuscin and aging: A matter of toxic waste. Sci Aging Knowledge Environ. 2005;2005:re1. [DOI] [PubMed] [Google Scholar]
- 45. Schmucker DL, Sachs H. Quantifying dense bodies and lipofuscin during aging: A morphologist’s perspective. Arch Gerontol Geriatr. 2002;34:249–261. [DOI] [PubMed] [Google Scholar]
- 46. Xu F, Hua C, Tautenhahn HM, Dirsch O, Dahmen U. The role of autophagy for the regeneration of the aging liver. Int J Mol Sci. 2020;21:3606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Dominick G, Bowman J, Li X, Miller RA, Garcia GG. mTOR regulates the expression of DNA damage response enzymes in long-lived Snell dwarf, GHRKO, and PAPPA-KO mice. Aging Cell. 2017;16:52–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Mulligan JD, Gonzalez AA, Kumar R, Davis AJ, Saupe KW. Aging elevates basal adenosine monophosphate-activated protein kinase (AMPK) activity and eliminates hypoxic activation of AMPK in mouse liver. J Gerontol A Biol Sci Med Sci. 2005;60:21–27. [DOI] [PubMed] [Google Scholar]
- 49. Łysek-Gładysińska M, Wieczorek A, Jóźwik A, Walaszczyk A, Jelonek K, Szczukiewicz-Markowska G, et al. Aging-related changes in the ultrastructure of hepatocytes and cardiomyocytes of elderly mice are enhanced in ApoE-deficient animals. Cells. 2021;10:502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Zorova LD, Popkov VA, Plotnikov EY, Silachev DN, Pevzner IB, Jankauskas SS, et al. Mitochondrial membrane potential. Anal Biochem. 2018;552:50–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Sastre J, Pallardo FV, Pla R, Pellin A, Juan G, O’Connor JE, et al. Aging of the liver: Age-associated mitochondrial damage in intact hepatocytes. Hepatology. 1996;24:1199–1205. [DOI] [PubMed] [Google Scholar]
- 52. Irvine KM, Skoien R, Bokil NJ, Melino M, Thomas GP, Loo D, et al. Senescent human hepatocytes express a unique secretory phenotype and promote macrophage migration. World J Gastroenterol. 2014;20:17851–17862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Aravinthan A, Verma S, Coleman N, Davies S, Allison M, Alexander G. Vacuolation in hepatocyte nuclei is a marker of senescence. J Clin Pathol. 2012;65:557–560. [DOI] [PubMed] [Google Scholar]
- 54. Liu A, Guo E, Yang J, Yang Y, Liu S, Jiang X, et al. Young plasma reverses age-dependent alterations in hepatic function through the restoration of autophagy. Aging Cell. 2018;17:e12708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Delire B, Lebrun V, Selvais C, Henriet P, Bertrand A, Horsmans Y, et al. Aging enhances liver fibrotic response in mice through hampering extracellular matrix remodeling. Aging (Albany NY). 2016;9:98–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Kim IH, Kisseleva T, Brenner DA. Aging and liver disease. Curr Opin Gastroenterol. 2015;31:184–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Wahid RM, Hassan NH, Samy W, Abdelhadi AA, Saadawy SF, Elsayed SF, et al. Unraveling the hepatic stellate cells mediated mechanisms in aging’s influence on liver fibrosis. Sci Rep. 2024;14:13473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Maeso-Diaz R, Ortega-Ribera M, Fernandez-Iglesias A, Hide D, Munoz L, Hessheimer AJ, et al. Effects of aging on liver microcirculatory function and sinusoidal phenotype. Aging Cell. 2018;17:e12829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Cogger VC, Svistounov D, Warren A, Zykova S, Melvin RG, Solon-Biet SM, et al. Liver aging and pseudocapillarization in a Werner syndrome mouse model. J Gerontol A Biol Sci Med Sci. 2014;69:1076–1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. McLachlan AJ, Pont LG. Drug metabolism in older people—A key consideration in achieving optimal outcomes with medicines. J Gerontol A Biol Sci Med Sci. 2012;67:175–180. [DOI] [PubMed] [Google Scholar]
- 61. Hilmer SN, Cogger VC, Le Couteur DG. Basal activity of Kupffer cells increases with old age. J Gerontol A Biol Sci Med Sci. 2007;62:973–978. [DOI] [PubMed] [Google Scholar]
- 62. Baiocchi L, Glaser S, Francis H, Kennedy L, Felli E, Alpini G, et al. Impact of aging on liver cells and liver disease: Focus on the biliary and vascular compartments. Hepatol Commun. 2021;5:1125–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Brouwer A, Barelds RJ, Knook DL. Age-related changes in the endocytic capacity of rat liver Kupffer and endothelial cells. Hepatology. 1985;5:362–366. [DOI] [PubMed] [Google Scholar]
- 64. Sun WB, Han BL, Peng ZM, Li K, Ji Q, Chen J, et al. Effect of aging on cytoskeleton system of Kupffer cell and its phagocytic capacity. World J Gastroenterol. 1998;4:77–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Baumann A, Hernandez-Arriaga A, Brandt A, Sanchez V, Nier A, Jung F, et al. Microbiota profiling in aging-associated inflammation and liver degeneration. Int J Med Microbiol. 2021;311:151500. [DOI] [PubMed] [Google Scholar]
- 66. Fulop T, Larbi A, Dupuis G, Le Page A, Frost EH, Cohen AA, et al. Immunosenescence and inflamm-aging as two sides of the same coin: Friends or foes? Front Immunol. 2017;8:1960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Li X, Li C, Zhang W, Wang Y, Qian P, Huang H. Inflammation and aging: Signaling pathways and intervention therapies. Signal Transduct Target Ther. 2023;8:239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Yang W, Kim DM, Jiang W, Ai W, Pan Q, Rahman S, et al. Suppression of FOXO1 attenuates inflamm-aging and improves liver function during aging. Aging Cell. 2023;22:e13968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Cheng Y, Wang X, Wang B, Zhou H, Dang S, Shi Y, et al. Aging-associated oxidative stress inhibits liver progenitor cell activation in mice. Aging (Albany NY). 2017;9:1359–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Messina A, Luce E, Hussein M, Dubart-Kupperschmitt A. Pluripotent-stem-cell-derived hepatic cells: Hepatocytes and organoids for liver therapy and regeneration. Cells. 2020;9:420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Warraich UE, Hussain F, Kayani HUR. Aging—Oxidative stress, antioxidants and computational modeling. Heliyon. 2020;6:e04107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Chandrasekaran A, Idelchik M, Melendez JA. Redox control of senescence and age-related disease. Redox Biol. 2017;11:91–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018;13:757–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Land WG. The role of damage-associated molecular patterns in human diseases: Part I—Promoting inflammation and immunity. Sultan Qaboos Univ Med J. 2015;15:e9–e21. [PMC free article] [PubMed] [Google Scholar]
- 75. Giorgi C, Marchi S, Simoes ICM, Ren Z, Morciano G, Perrone M, et al. Mitochondria and reactive oxygen species in aging and age-related diseases. Int Rev Cell Mol Biol. 2018;340:209–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Brenner C. Sirtuins are not conserved longevity genes. Life Metab. 2022;1:122–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Zhao L, Cao J, Hu K, He X, Yun D, Tong T, et al. Sirtuins and their biological relevance in aging and age-related diseases. Aging Dis. 2020;11:927–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Chen C, Zhou M, Ge Y, Wang X. SIRT1 and aging related signaling pathways. Mech Ageing Dev. 2020;187:111215. [DOI] [PubMed] [Google Scholar]
- 79. Yuan Y, Cruzat VF, Newsholme P, Cheng J, Chen Y, Lu Y. Regulation of SIRT1 in aging: Roles in mitochondrial function and biogenesis. Mech Ageing Dev. 2016;155:10–21. [DOI] [PubMed] [Google Scholar]
- 80. Adjei-Mosi J, Sun Q, Smithson SB, Shealy GL, Amerineni KD, Liang Z, et al. Age-dependent loss of hepatic SIRT1 enhances NLRP3 inflammasome signaling and impairs capacity for liver fibrosis resolution. Aging Cell. 2023;22:e13811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Zhang Y, Gong C, Tao L, Zhai J, Huang F, Zhang S. Involvement of SIRT1-mediated aging in liver diseases. Front Cell Dev Biol. 2025;13:1548015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Herranz D, Munoz-Martin M, Canamero M, Mulero F, Martinez-Pastor B, Fernandez-Capetillo O, et al. Sirt1 improves healthy ageing and protects from metabolic syndrome-associated cancer. Nat Commun. 2010;1:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Imai S, Guarente L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014;24:464–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Tissenbaum HA, Guarente L. Increased dosage of a gene extends lifespan in. Nature. 2001;410:227–230. [DOI] [PubMed] [Google Scholar]
- 85. Rogina B, Helfand SL. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc Natl Acad Sci U S A. 2004;101:15998–16003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Satoh A, Brace CS, Rensing N, Cliften P, Wozniak DF, Herzog ED, et al. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 2013;18:416–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Mitchell SJ, Bernier M, Aon MA, Cortassa S, Kim EY, Fang EF, et al. Nicotinamide improves aspects of healthspan, but not lifespan, in mice. Cell Metab. 2018;27:667–676 e664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Younossi ZM, Stepanova M, Al Shabeeb R, Eberly KE, Shah D, Nguyen V, et al. The changing epidemiology of adult liver transplantation in the United States in 2013-2022: The dominance of metabolic dysfunction-associated steatotic liver disease and alcohol-associated liver disease. Hepatol Commun. 2024;8:e0352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Kan C, Ungelenk L, Lupp A, Dirsch O, Dahmen U. Ischemia-reperfusion injury in aged livers—The energy metabolism, inflammatory response, and autophagy. Transplantation. 2018;102:368–377. [DOI] [PubMed] [Google Scholar]
- 90. Hide D, Warren A, Fernandez-Iglesias A, Maeso-Diaz R, Peralta C, Le Couteur DG, et al. Ischemia/reperfusion injury in the aged liver: The importance of the sinusoidal endothelium in developing therapeutic strategies for the elderly. J Gerontol A Biol Sci Med Sci. 2020;75:268–277. [DOI] [PubMed] [Google Scholar]
- 91. Adani GL, Baccarani U, Lorenzin D, Rossetto A, Nicolini D, Vecchi A, et al. Elderly versus young liver transplant recipients: Patient and graft survival. Transplant Proc. 2009;41:1293–1294. [DOI] [PubMed] [Google Scholar]
- 92. Dolnikov S, Adam R, Cherqui D, Allard MA. Liver transplantation in elderly patients: What do we know at the beginning of 2020? Surg Today. 2020;50:533–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Okaya T, Blanchard J, Schuster R, Kuboki S, Husted T, Caldwell CC, et al. Age-dependent responses to hepatic ischemia/reperfusion injury. Shock. 2005;24:421–427. [DOI] [PubMed] [Google Scholar]
- 94. Wang JH, Ahn IS, Fischer TD, Byeon JI, Dunn WA, Jr, Behrns KE, et al. Autophagy suppresses age-dependent ischemia and reperfusion injury in livers of mice. Gastroenterology. 2011;141:2188–2199 e2186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Li Y, Ruan DY, Jia CC, Zheng J, Wang GY, Zhao H, et al. Aging aggravates hepatic ischemia-reperfusion injury in mice by impairing mitophagy with the involvement of the EIF2alpha-parkin pathway. Aging (Albany NY). 2018;10:1902–1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Bittermann T, Goldberg DS. Quantifying the effect of transplanting older donor livers into younger recipients: The need for donor-recipient age matching. Transplantation. 2018;102:2033–2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Shankar N, AlBasheer M, Marotta P, Wall W, McAlister V, Chandok N. Do older patients utilize excess health care resources after liver transplantation? Ann Hepatol. 2011;10:477–481. [PubMed] [Google Scholar]
- 98. Almazroo OA, Miah MK, Venkataramanan R. Drug metabolism in the liver. Clin Liver Dis. 2017;21:1–20. [DOI] [PubMed] [Google Scholar]
- 99. Kinirons MT, O’Mahony MS. Drug metabolism and ageing. Br J Clin Pharmacol. 2004;57:540–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Klotz U. Pharmacokinetics and drug metabolism in the elderly. Drug Metab Rev. 2009;41:67–76. [DOI] [PubMed] [Google Scholar]
- 101. Cohen EB, Patwardhan M, Raheja R, Alpers DH, Andrade RJ, Avigan MI, et al. Drug-induced liver injury in the elderly: Consensus statements and recommendations from the IQ-DILI Initiative. Drug Saf. 2024;47:301–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Tanimizu N, Ichinohe N, Suzuki H, Mitaka T. Prolonged oxidative stress and delayed tissue repair exacerbate acetaminophen-induced liver injury in aged mice. Aging (Albany NY). 2020;12:18907–18927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Onji M, Fujioka S, Takeuchi Y, Takaki T, Osawa T, Yamamoto K, et al. Clinical characteristics of drug-induced liver injury in the elderly. Hepatol Res. 2009;39:546–552. [DOI] [PubMed] [Google Scholar]
- 104. Mitchell SJ, Hilmer SN. Drug-induced liver injury in older adults. Ther Adv Drug Saf. 2010;1:65–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Yu S, Li J, He T, Zheng H, Wang S, Sun Y, et al. Age-related differences in drug-induced liver injury: A retrospective single-center study from a large liver disease specialty hospital in China, 2002-2022. Hepatol Int. 2024;18:1202–1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Lucena MI, Sanabria J, Garcia-Cortes M, Stephens C, Andrade RJ. Drug-induced liver injury in older people. Lancet Gastroenterol Hepatol. 2020;5:862–874. [DOI] [PubMed] [Google Scholar]
- 107. Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol. 2019;70:151–171. [DOI] [PubMed] [Google Scholar]
- 108. Mackowiak B, Fu Y, Maccioni L, Gao B. Alcohol-associated liver disease. J Clin Invest. 2024;134:e176345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Williams JA, Manley S, Ding WX. New advances in molecular mechanisms and emerging therapeutic targets in alcoholic liver diseases. World J Gastroenterol. 2014;20:12908–12933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Edenberg HJ. The genetics of alcohol metabolism: Role of alcohol dehydrogenase and aldehyde dehydrogenase variants. Alcohol Res Health. 2007;30:5–13. [PMC free article] [PubMed] [Google Scholar]
- 111. Lehner T, Gao B, Mackowiak B. Alcohol metabolism in alcohol use disorder: A potential therapeutic target. Alcohol Alcohol. 2024;59:agad077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Ma X, Ni HM, Ding WX. Perspectives of mitochondria-lysosome-related organelle in hepatocyte dedifferentiation and implications in chronic liver disease. eGastroenterology. 2024;2:e100046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Ma X, Chen A, Melo L, Clemente-Sanchez A, Chao X, Ahmadi AR, et al. Loss of hepatic DRP1 exacerbates alcoholic hepatitis by inducing megamitochondria and mitochondrial maladaptation. Hepatology. 2023;77:159–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Danpanichkul P, Suparan K, Ng CH, Dejvajara D, Kongarin S, Panpradist N, et al. Global and regional burden of alcohol-associated liver disease and alcohol use disorder in the elderly. JHEP Rep. 2024;6:101020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Meier P, Seitz HK. Age, alcohol metabolism and liver disease. Curr Opin Clin Nutr Metab Care. 2008;11:21–26. [DOI] [PubMed] [Google Scholar]
- 116. Lu C, Ge T, Shao Y, Cui W, Li Z, Xu W, et al. ZNF281 drives hepatocyte senescence in alcoholic liver disease by reducing HK2-stabilized PINK1/Parkin-mediated mitophagy. Cell Prolif. 2023;56:e13378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Kim IH, Xu J, Liu X, Koyama Y, Ma HY, Diggle K, et al. Aging increases the susceptibility of hepatic inflammation, liver fibrosis and aging in response to high-fat diet in mice. Age (Dordr). 2016;38:291–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Ramirez T, Li YM, Yin S, Xu MJ, Feng D, Zhou Z, et al. Aging aggravates alcoholic liver injury and fibrosis in mice by downregulating sirtuin 1 expression. J Hepatol. 2017;66:601–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Seitz HK, Stickel F. Alcoholic liver disease in the elderly. Clin Geriatr Med. 2007;23:905–921. [DOI] [PubMed] [Google Scholar]
- 120. Dombestein Elde K, Jepsen P, Von Wowern N, Winther-Jensen M, Eliasen MH, Christensen AI, et al. Influence of comorbidities on health-related quality of life in alcohol-related liver disease: A population-based survey. Clin Gastroenterol Hepatol. 2025;23:1174–1182. [DOI] [PubMed] [Google Scholar]
- 121. Younossi ZM, Kalligeros M, Henry L. Epidemiology of metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol. 2025;31:S32–S50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Rinella ME, Sookoian S. From NAFLD to MASLD: Updated naming and diagnosis criteria for fatty liver disease. J Lipid Res. 2024;65:100485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Rinella ME, Lazarus JV, Ratziu V, Francque SM, Sanyal AJ, Kanwal F, et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. J Hepatol. 2023;79:1542–1556. [DOI] [PubMed] [Google Scholar]
- 124. Fukushima M, Miyaaki H, Nakao Y, Sasaki R, Haraguchi M, Takahashi K, et al. Characterizing alcohol-related and metabolic dysfunction-associated steatotic liver disease cirrhosis via fibrotic pattern analysis. Sci Rep. 2024;14:23679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Mejía-Guzmán JE, Belmont-Hernández RA, Chávez-Tapia NC, Uribe M, Nuño-Lámbarri N. Metabolic-dysfunction-associated steatotic liver disease: Molecular mechanisms, clinical implications, and emerging therapeutic strategies. Int J Mol Sci. 2025;26:2959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Clayton-Chubb D, Kemp WW, Majeed A, Lubel JS, Woods RL, Tran C, et al. Metabolic dysfunction-associated steatotic liver disease in older adults is associated with frailty and social disadvantage. Liver Int. 2024;44:39–51. [DOI] [PubMed] [Google Scholar]
- 127. Bertolotti M, Lonardo A, Mussi C, Baldelli E, Pellegrini E, Ballestri S, et al. Nonalcoholic fatty liver disease and aging: Epidemiology to management. World J Gastroenterol. 2014;20:14185–14204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 2017;8:15691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Kuhla A, Blei T, Jaster R, Vollmar B. Aging is associated with a shift of fatty metabolism toward lipogenesis. J Gerontol A Biol Sci Med Sci. 2011;66:1192–1200. [DOI] [PubMed] [Google Scholar]
- 130. Perichon R, Bourre JM. Aging-related decrease in liver peroxisomal fatty acid oxidation in control and clofibrate-treated mice. A biochemical study and mechanistic approach. Mech Ageing Dev. 1996;87:115–126. [DOI] [PubMed] [Google Scholar]
- 131. Sharma A, Smith HJ, Yao P, Mair WB. Causal roles of mitochondrial dynamics in longevity and healthy aging. EMBO Rep. 2019;20:e48395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Ma X, Niu M, Ni HM, Ding WX. Mitochondrial dynamics, quality control, and mtDNA in alcohol-associated liver disease and liver cancer. Hepatol. 2024. doi: 10.1097/HEP.0000000000000910. Online ahead of print. [DOI] [PubMed] [Google Scholar]
- 133. Yamada T, Murata D, Kleiner DE, Anders R, Rosenberg AZ, Kaplan J, et al. Prevention and regression of megamitochondria and steatosis by blocking mitochondrial fusion in the liver. iScience. 2022;25:103996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Schneider JL, Suh Y, Cuervo AM. Deficient chaperone-mediated autophagy in liver leads to metabolic dysregulation. Cell Metab. 2014;20:417–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Saito T, Kuma A, Sugiura Y, Ichimura Y, Obata M, Kitamura H, et al. Autophagy regulates lipid metabolism through selective turnover of NCoR1. Nat Commun. 2019;10:1567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Du K, Wang L, Jun JH, Dutta RK, Maeso-Diaz R, Oh SH, et al. Aging promotes metabolic dysfunction-associated steatotic liver disease by inducing ferroptotic stress. Nat Aging. 2024;4:949–968. [DOI] [PubMed] [Google Scholar]
- 137. Yadav AK, MacNeill JJ, Krylov A, Ashrafi N, Mimi RA, Saxena R, et al. Sex- and age-associated factors drive the pathophysiology of MASLD. Hepatol Commun. 2024;8:e0523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Cao P, Jaeschke H, Ni HM, Ding WX. The ways to die: Cell death in liver pathophysiology. Semin Liver Dis. 2025. doi: 10.1055/a-2576-4332. Online ahead of print. [DOI] [PubMed] [Google Scholar]
- 139. Mohammed S, Thadathil N, Selvarani R, Nicklas EH, Wang D, Miller BF, et al. Necroptosis contributes to chronic inflammation and fibrosis in aging liver. Aging Cell. 2021;20:e13512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Krause C, Grohs M, El Gammal AT, Wolter S, Lehnert H, Mann O, et al. Reduced expression of thyroid hormone receptor beta in human nonalcoholic steatohepatitis. Endocr Connect. 2018;7:1448–1456. [DOI] [PMC free article] [PubMed] [Google Scholar]