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.
The liver has a strong regenerative capacity; however, chronic injury can lead to extracellular matrix accumulation, fibrosis and eventually liver failure, representing a major global health burden (1). In 2022, liver diseases such as cirrhosis, viral hepatitis and hepatocellular carcinoma (HCC) accounted for 2 million deaths worldwide (2). In China, the increasing prevalence of liver diseases highlights the requirement for more effective therapies (3). Conventional treatments offer limited efficacy in conditions such as non-alcoholic steatohepatitis (NASH), alcoholic liver disease (ALD) and HCC (4–7). Therefore, it is important to identify shared molecular mechanisms to improve the diagnosis of liver diseases and identify more effective treatment strategies.
Poly(ADP-ribose) polymerase-1 (PARP-1), a key enzyme in the DNA damage response (DDR), also regulates cellular metabolism by modulating oxidized nicotinamide adenine dinucleotide (NAD+) levels and sirtuin 1 (SIRT1) activity (8–10). PARP-1 expression is upregulated in both primary and recurrent HCC and has been implicated in chemoresistance (11). The involvement of PARP-1 in the progression of various liver diseases, including non-alcoholic fatty liver disease (NAFLD), ALD and HCC, suggests its potential as a therapeutic target (12–14).
Unlike previous studies focusing on single disease types, the present review comprehensively analyzes the role of PARP-1 in multiple liver pathologies, including NASH, ALD, fibrosis and HCC, revealing shared molecular mechanisms. The dual role of PARP-1 in liver diseases is systematically summarized, and its functions in liver regeneration and pathological fibrosis are integrated to provide a comprehensive understanding of its regulatory mechanisms. In addition to summarizing established evidence, novel mechanistic insights into the regulation of tumor metabolism, transcriptional programming and immune microenvironment remodeling by PARP-1 in HCC are discussed, and the therapeutic synergy between PARP inhibitors and immune checkpoint blockade is highlighted. Furthermore, the translational potential of PARP-1 inhibitors for autoimmune liver diseases (AILDs) is proposed based on preclinical evidence, addressing a critical research gap. Overall, the present review synthesizes current knowledge on the expression, regulatory networks and mechanisms of PARP-1 in various liver diseases, ultimately evaluating its clinical diagnostic and therapeutic value.
In 2022, liver disease resulted in 2 million fatalities worldwide, or 4% of total deaths globally, ranking it as the 11th largest cause of death (2). The main causes of mortality are complications arising from cirrhosis and HCC, while acute hepatitis accounts for a lower proportion of fatalities. The primary global causes of cirrhosis include viral hepatitis, alcohol use and NAFLD, which lead to a progressive deterioration of hepatic function. Other factors also contribute to liver disease, including age, sex, body composition, diet, physical activity, microbiome, and history of alcohol and tobacco use (15).
The liver performs essential biological tasks such as protein synthesis, glucose and lipid metabolism, detoxification and bile production. It exhibits marked immunological activity and functions as a systemic barrier against intestinal infections and toxins by maintaining a localized immune-regulating milieu and promoting tolerance to prevalent antigens (16). Optimal liver function is essential for the body to effectively combat bacteria and viruses; therefore, individuals with liver dysfunction are particularly susceptible to infections (17). The immune response generated by the liver relies upon its distinctive architecture, the presence of essential immune cells such as Kupffer cells (KCs), continuous immunological surveillance and rapid recruitment of immune cells (16,18). Chronic alcohol use increases the permeability of the gut to bacterial lipopolysaccharide (LPS) (19), elevates endotoxin levels in the hepatic portal vein, and activates KCs via interactions with Toll-like receptor-4 (14).
The liver has strong regenerative capacity and can recover considerably after resection. However, conditions such as ALD and chronic hepatitis B (CHB) may overly activate this regenerative response, resulting in the excessive accumulation of extracellular matrix and collagen, leading to liver fibrosis. Furthermore, decompensated liver fibrosis leads to the formation of hepatic scar tissue, which is known as cirrhosis. This significantly reduces liver function, and may progress to liver failure and death (1).
PARP-1, a DNA repair enzyme, plays a key role in hepatocyte apoptosis by promoting cell death and increasing the synthesis of pro-inflammatory mediators when overactivated by ROS and reactive nitrogen species, thereby influencing the self-healing capacity of the liver (10). NAFLD is a major contributor to chronic liver disease (CLD), with a spectrum ranging from moderate steatosis to advanced NASH. NASH is characterized by inflammation and progressive hepatocyte injury, which can lead to cirrhosis (20) and subsequently increase the risk of HCC.
PARPs are ADP-ribosyltransferases that catalyze ADP-ribosylation, a post-translational protein modification. ADP-ribosylation involves the cleavage of NAD+ to generate single ADP-ribose units, oligomers or polymers that are covalently linked to serine, glutamate, aspartate, arginine and lysine residues in target proteins (21). The PARP family comprises 17 proteins, with PARP-1 accounting for 85–90% of total PARP activity (22). PARP-1 contains several conserved and functionally distinct domains, including an auto-modification domain, a 55-kDa catalytic domain, and two zinc finger domains responsible for DNA binding (23).
PARP-1 is present in all human tissues, with the highest concentrations in the lymph nodes, appendix, brain, placenta, prostate, spleen and testes. It is an essential element in the DNA base excision repair mechanism (24,25) and participates in the repair of single-strand and double-strand breaks (DSBs) (26). Human PARPs, namely PARP-1, −2 and −3, are categorized as DDR proteins because of their involvement in DNA repair and reliance on DNA damage for activation (22). Among these, PARP-1 is the predominant enzyme, characterized by its abundance and strong enzymatic activity. It serves as the principal signaling factor in DDR, where its rapid activation is the most critical stage in the DDR process (8). PARP-1 serves as a primary sensor of DNA damage, capable of detecting DNA breaks within 3 sec of their occurrence, and triggers the recruitment of repair complexes via the ADP-ribosylation of proteins near the damage site. Its activity is further influenced by co-factors, such as histone PARylation factor 1 (HPF1), which enhances the catalytic activity of PARP-1 and modifies substrate selectivity. HPF1 interacts with the PARP-1 catalytic domain and colocalizes with it at sites of DNA damage (8).
A study evaluating the hepatotoxicity of columbin (CLB) demonstrated that CLB induces DNA damage both in vitro and in vivo, which is associated with the upregulation of PARP-1 (27). PARP-1 also interacts with SIRTs. SIRT1-7 are a family of NAD+-dependent protein deacetylases with extensive roles in metabolism and aging. In the absence of PARP-1, NAD+ levels rise, potentially increasing SIRT1 activity (22,28). In addition, PARP-1 and AMP-activated protein kinase (AMPK) mutually reinforce each other: The activation of PARP-1 stimulates AMPK, which in turn phosphorylates and activates PARP-1. Within the reactive oxygen species (ROS)-PARP-1-AMPK pathway, the interaction between PARP-1 and AMPK promotes the nuclear export of AMPK, which induces autophagic flux and cellular apoptosis (22,29).
PARP-1 plays an essential role in DNA repair and cellular stress responses in the liver. During oxidative stress, such as exposure to hydrogen peroxide, PARP-1 activation preserves hepatocyte survival, while PARP-1 inhibition or genetic ablation in primary hepatocytes diminishes oxidation-induced necrotic cell death, suggesting a preventive mechanism against oxidative injury (10).
A pilot study revealed that PARP-1 knockout mice exhibit impaired liver regeneration and reduced hepatocyte proliferation. It also showed that PARP-1 is required for both the early liver response and late tissue repair during liver regeneration, and that PARP-1 inhibitors can reduce hepatocyte proliferation (30). These results indicate that PARP-1 and its catalytic activity are essential for hepatocyte proliferation. This effect appears to be associated with Yes-associated protein (YAP), a key driver of hepatocyte proliferation (31). In PARP-1 knockout mice, YAP activity and the expression of cell cycle-related proteins in liver tissue are suppressed, resulting in reduced hepatocyte proliferation and impaired liver repair during liver regeneration (30). However, another study showed that chronic or excessive activation, such as that observed in severe or long-standing liver disease, depletes NAD+ and promotes fibrosis, ultimately impairing liver regeneration (32). In view of this duality, the inhibition of pathological activation while retaining the beneficial functions of PARP-1 is a therapeutic challenge.
Chromatin relaxation is essential for efficient DNA repair, and PARP-1 functions as a crucial mediator of this process. The inhibition of PARP-1 activity directly affects chromatin structure. Through poly(ADP-ribosyl)ation, PARP-1 binds to and modifies chromatin remodelers, thereby altering chromatin structure and facilitating the ensuing DDR (33).
PARP-1 also plays a pivotal role in inflammation. It activates nuclear factor κB (NF-κB), nuclear factor of activated T cells and activator protein 1, leading to the production of inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin (IL)-1β, and effector T cell cytokines, including IL-4 and IL-5. Excessive activation of PARP-1 promotes cell death and tissue damage, thereby exacerbating the inflammatory response (34). These functions are particularly relevant to chronic inflammatory liver conditions, including viral hepatitis, ALD and drug-induced liver injury (DILI).
PARP-1 is essential for sustaining proper immune function, and its dysregulation contributes to immune-mediated disorders. Under physiological conditions, PARP-1 expression is typically low but becomes significantly upregulated during early liver injury, chronic liver fibrosis and HCC (10). Consequently, aberrant PARP-1 activity is associated with the progression of several liver disorders.
Viral infection is a major cause of acute hepatitis, which leads to elevated liver transaminase levels and impaired liver function (35). China is among the top 20 nations with the highest viral hepatitis burdens. Between 2016 and 2022, the proportion of people living with viral hepatitis confirmed hepatitis B cases in China increased from 19 to 24%, while that of hepatitis C cases increased from 22 to 33% (36). CHB infection is the major cause of HCC. Transforming viral proteins such as hepatitis B virus (HBV) regulatory protein X (HBx) are key oncogenic factors (37). HBx induces the degradation of the structural maintenance of chromosome 5/6 complex, a process implicated in HBV-related HCC. This impairs homologous recombination (HR)-mediated DNA repair, thereby contributing to carcinogenesis. The accumulation of double-stranded DNA (dsDNA) breaks in cancer cells with HR deficit (HRD) can be lethal. PARP-1 inhibition prevents the repair of single-strand DNA breaks, which subsequently exacerbates the accumulation of dsDNA breaks (38,39). In addition to altering protein function, HBV infection regulates the expression of certain long non-coding RNAs (lncRNAs) in infected cells. For example, HBx inhibits p53 activity, leading to the upregulation of lncRNA-HUR1 expression, which promotes cell proliferation and cancer progression (40). The overexpression of lncRNA-HUR1 reduces caspase-3/7 activity and PARP-1 cleavage, while lncRNA-HUR1 knockdown increases caspase-3/7 activity and promotes PARP-1 cleavage (41). Together, these findings underscore the crucial role of PARP-1 in HBV-induced HCC.
In addition to HBV, other viral hepatitis subtypes may also interact with DNA damage and repair pathways. Although PARP-1 has not been extensively studied in these contexts, existing data suggest a potential involvement that is worthy of further exploration. Hepatitis C virus (HCV) is a leading cause of CLD and HCC that induces persistent oxidative stress and DNA damage in infected hepatocytes. Viral proteins, including core protein and the non-structural (NS) proteins NS3 and NS5A disrupt host DNA repair pathways by targeting key components, such as the ataxia telangiectasia-mutated (ATM)-nibrin-meiotic recombination 11 complex, thereby promoting chromosomal instability and malignant transformation (42,43). Although direct studies of PARP-1 in HCV-infected liver tissue are limited, the well-established roles of this enzyme in DDR and inflammation suggest that PARP-1 may be activated under conditions of HCV-induced genomic stress, potentially contributing to fibrosis and cancer progression. Hepatitis D virus (HDV), which often co-occurs with HBV, is associated with more severe disease and heightened oxidative stress (44), factors that may also induce PARP-1 activation. However, direct evidence associating PARP-1 with HDV-induced liver injury is lacking.
Collectively, these observations highlight the importance of investigating the role of PARP-1 in diverse forms of viral hepatitis, as its regulatory role in DNA repair, cell death and the inflammatory response may have major implications for the diagnosis and treatment of various liver infections.
Liver damage, inflammation, fibrosis, cirrhosis and cancer resulting from prolonged or excessive alcohol consumption are hallmarks of ALD. With 3.3 million associated fatalities, or 6% of all deaths globally, alcohol-related damage is one of the most prevalent preventable causes of mortality. Liver damage occurs in 20–30% of individuals who misuse alcohol (45). A key pathogenic characteristic of ALD is hepatocyte apoptosis mediated by PARP-1. Under normal physiological conditions, alcohol dehydrogenase is the primary enzyme that metabolizes ethanol in the liver. However, chronic alcohol intake stimulates the expression of cytochrome P450 2E1, which metabolizes ethanol to acetaldehyde and produces considerable amounts of ROS as byproducts (46). Acetaldehyde is a toxic metabolite that forms adducts with cellular macromolecules and further amplifies oxidative stress. ROS can induce oxidative DNA damage, which is sensed by PARP-1 and activates the TGF-β1-Smad pathway. In the early stages of DNA damage, PARP-1 binds to sites of DNA damage and uses NAD+ as a substrate to synthesize PARPs including PARP-1 (47). However, persistent or severe DNA damage leads to the overactivation of PARP-1, which consumes large amounts of NAD+ for poly(ADP-ribosyl)ation, leading to NAD+ and ATP depletion along with impaired mitochondrial function and energy metabolism, ultimately triggering hepatocyte necrosis or monocytic cell death (48,49).
Preventing and treating ALD requires the reversal of this imbalance caused by PARP-1 overactivation that leads to hepatocyte necrosis or monocyte death (50). In ALD, hepatocytes and liver macrophages exhibit upregulated C-C chemokine ligand type 2 (CCL2) levels. A pharmacological study of mice revealed that alcohol-induced liver damage can be prevented or reversed by blocking the activation of C-C chemokine receptor type 2 and 5 (51). Alcohol-induced increases in PARP-1 activity have also been shown to mediate hepatocyte apoptosis (14,52). In Raw264.7 macrophages, PARP-1 activation promotes the nuclear translocation of LPS-induced NF-kB, and increased KC activation exacerbates alcohol-induced liver steatosis by increasing hepatic M1 macrophage marker gene expression. Consistent with this, treatment of mice with the PARP-1 inhibitor PJ34 downregulates hepatic M1 marker gene expression (14). This suggests that limiting KC activation helps to restore hepatic lipid balance, an important protective mechanism of PARP-1 inhibition in alcohol-induced hepatic steatosis. Certain medications, such as cenicriviroc, have been demonstrated to reduce hepatocyte apoptosis and alleviate CCL2-induced hepatic steatosis (52).
Based on histological features, NAFLD can be classified as NASH, which comprises <20% of cases, and non-alcoholic fatty liver (NAFL), which comprises the remaining >80% (53,54). NAFL is defined as hepatic steatosis without substantial hepatocyte injury, or minor lobular inflammation is present, while NASH is characterized by hepatocyte damage in addition to steatosis (54). With an estimated global incidence of 25%, NAFLD is a growing public health concern. In the United States, severe NASH is now the second most frequent indication for liver transplantation in men and the most common indication in women (55). Nearly 75% of individuals in the United States with risk factors such as diabetes and obesity develop NAFLD (56).
In early NAFLD, the gut vascular barrier is disrupted, which allows intestinal bacteria to proliferate, leading to increased translocation of LPS to the liver, thereby triggering pyroptosis. LPS stimulation also activates PARP-1, which increases signaling via NF-κB, a crucial transcription factor that regulates the production of pro-inflammatory cytokines. Specifically, PARP-1 interacts with transcription factors and co-activators, including p300, to increase the transcriptional activity of NF-κB. This then influences the expression of cytokines and other pro-inflammatory mediators, including inducible nitric oxide synthase and cyclooxygenase-2, thereby contributing to the inflammatory response in hepatic tissue (34). In high-fat diet (HFD)-induced mouse models, DNA damage activates PARP-1, which stimulates the activation of NLR family pyrin domain containing 3 (NLRP3) and its downstream signaling proteins (57). This is associated with increased levels of cleaved PARP-1 together with elevated blood levels of IL-1β and C-reactive protein (58). Beyond inflammation, the development of NAFLD is strongly influenced by the SIRT1/AMPK pathway and NAD+ homeostasis. PARP-1 contributes to this dysregulation by depleting NAD+ and interfering with SIRT1 activity, which is essential for AMPK activation (59). SIRT1 modulates hepatic lipid metabolism by regulating the activity of peroxisome proliferator-activated receptors (PPARs) (60). PARP-1 activity also disrupts hepatic lipid homeostasis by modulating the SIRT1/PPARα axis. It depletes intracellular NAD+, directly poly(ADP-ribosyl)ates PPARα, and impairs both SIRT1 activity and the PPARα-mediated transcription of β-oxidation genes, leading to suppressed mitochondrial function and fatty acid oxidation (61,62). This promotes lipid accumulation and the progression of steatosis. By contrast, the activation of SIRT1 enhances hepatic lipid catabolism and mitochondrial efficiency, serving as a critical counterbalance in the maintenance of metabolic adaptation during NAFLD progression (63). Collectively, these insights highlight the pivotal regulatory function of the PARP-1/SIRT1/PPARα axis in the pathophysiology of NAFLD.
Unanticipated liver damage caused by drugs or other xenobiotics, known as DILI, is a major concern in clinical trials and drug development (64). A wide range of agents, including medications, natural products and supplements, can cause DILI (65). DILI is challenging to diagnose, requires the rigorous elimination of other causes, and is underreported to pharmacovigilance authorities; therefore, its prevalence is challenging to determine (66,67). While traditional and nutritional supplements are the primary causes of DILI in Asia, drug responses to conventional pharmaceuticals are most prevalent in the United States and Europe (67). With an estimated 23.8 cases per 100,000 individuals, the frequency of DILI is high in China (68). However, the actual incidence could be much higher than this.
The mechanisms underlying liver injury vary depending on the causative agents. Experimental models hased on DILI principles have been developed to assess the efficacy of liver protective and anti-fibrosis interventions. For example, carbon tetrachloride can induce liver fibrosis and injury, processes that are often accompanied by DNA damage and repair processes, with PARP-1 playing a key role in the DDR (49). The overactivation of PARP-1 promotes the binding of transcription factor activator protein 1 to the TGF-β1 gene, leading to chronic chemical damage in human and animal liver cells (69). Another example of a liver injury-inducing agent is CLB, which increases ROS production and depletes glutathione. The resulting oxidative stress promotes DNA breaks and upregulates PARP-1 expression (27).
PARP-1 is activated during acetaminophen overdose, resulting in poly(ADP-ribosyl)ation of the pregnane X receptor. This post-translational modification augments the transcription of the cytochrome P450, family 3, subfamily A, polypeptide 11 enzyme, thereby increasing the production of toxic acetaminophen metabolites (70). Furthermore, anti-tuberculosis medications, including isoniazid and rifampicin, can induce marked hepatic damage by inducing an inflammatory response and oxidative stress, subsequently activating the NLRP3 inflammasome. This induces PARP-1 activation, which contributes to hepatic damage (71).
There are three principal types of AILD, a chronic immune-mediated illness that affects hepatic and bile duct cells, which are autoimmune hepatitis (AIH), primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) (72,73). A clinical phenotype known as overlap syndrome occurs when symptoms of multiple categories co-exist within the spectrum of AILD; however, the majority of patients fit the criteria of one specific type (74). AILD has a male-to-female ratio of 1:5, and an annual yearly incidence worldwide of 1.37 cases per 100,000 individuals. PBC mostly affects women, and has a prevalence of 39.2 per 100,000 in the United States and 4.75 per 100,000 in Korea (75).
Studies have shown that the pathophysiology of autoimmune disorders is associated with increased PARP-1 activation (76,77). Trichloroethene exposure induces autoimmune reactions, leading to increased apoptosis, immune activation and the upregulation of PARP-1 expression following the development of anti-single-stranded DNA antibodies (78). Concanavalin A (ConA) causes liver damage via a mechanism comparable to the clinical alterations and immunological responses observed in patients with AIH (79). In AIH model mice, the liver exhibits widespread necrosis, marked macrophage infiltration, and elevated levels of the pro-inflammatory cytokine TNF-α (80). Following ConA induction, PARP-1 levels and TNF-α production increase, which contributes to PARP-1 activation (80).
Although PARP-1 has been implicated in the regulation of inflammation and T cell-mediated immune responses, direct evidence for the use of PARP inhibitors in animal models of AILD, including AIH, PBC and PSC, is currently lacking. The ConA-induced hepatitis model has been widely employed to investigate the immunopathogenesis of AIH, owing to its ability to simulate CD4+ T cell-mediated hepatic injury (81). Notably, genetic ablation of PARP-2, but not PARP-1, confers significant protection against ConA-induced liver damage, suggesting isoform-specific roles of PARP enzymes in immune liver injury (82). However, the role of PARP-1 remains incompletely understood in this context. Given the established involvement of PARP-1 in the amplification of NF-κB-mediated proinflammatory cytokine release and the modulation of CD4+ T cell polarization (83), it is plausible that PARP-1 inhibition may mitigate immune-driven hepatic inflammation in AIH (84). Accordingly, future studies utilizing pharmacological PARP-1 inhibitors such as PJ34 or olaparib, or gene-silencing approaches in AIH models, such as the ConA-induced hepatitis model, are warranted to determine whether PARP-1 is a viable therapeutic target in AILD.
Persistent liver injury, such as hepatitis, can lead to liver fibrosis. This condition is characterized by the buildup of extracellular matrix proteins, primarily cross-linked type I and III collagen, which replace damaged normal tissue following cholestatic and hepatotoxic injury. The resulting fibrous scarring impairs the structural integrity of the liver tissue and disrupts the homeostatic function of hepatocytes (85). Liver fibrosis may eventually develop into cirrhosis, which is associated with high morbidity and mortality, and increases the risk of HCC and chronic liver failure (86).
Hepatic stellate cells (HSCs) are non-parenchymal cells located in the subendothelial region of the liver and are the major mediators of liver fibrosis. A promising strategy to mitigate the development of liver fibrosis is the induction of senescence in activated HSCs (87). HSC apoptosis can be accurately predicted based on changes in PARP-1 levels (88). Studies have shown that autophagy exacerbates liver fibrosis by breaking down lipid droplets in HSCs, thereby activating the HSCs. Conversely, the suppression of autophagy promotes HSC death and reduces proliferation. Elevated levels of cleaved PARP are associated with HSC apoptosis, which can be induced by pharmacological agents such as berberine (BBR) and carvedilol (89,90). Specifically, BBR blocks autophagy in HSCs and downregulates autophagy protein 5 expression, which leads to HSC death (90). In addition, PARP-1 activation is associated with increased TGF-β expression. In activated KCs, upregulated PARP-1 expression promotes TGF-β production, further stimulating HSC activation (10).
Liver cancer ranks fourth in cancer-related mortality and is the sixth most prevalent cancer worldwide, with HCC accounting for >80% of cases (91,92). With a yearly incidence rate of 1–6%, cirrhosis of any etiology is the biggest risk factor for HCC. Indeed, HCC is the primary cause of mortality in individuals with cirrhosis (93). Compared with nearby normal tissues, HCC exhibits numerous chromosomal aberrations and elevated PARP expression, which accumulates during DNA replication (94,95). PARP-1 inhibition has been suggested to be an effective treatment for HCC, particularly in BRCA-deficient tumors (94).
Emerging evidence indicates that PARP-1 enhances glycolysis and metabolic reprogramming in tumor cells by physically interacting with hypoxia inducible factor-1α, stabilizing it, and acting as a transcriptional coactivator (96). This promotes the expression of hexokinase 2 and other glycolytic enzymes, thereby reinforcing the Warburg effect in HCC and facilitating tumor proliferation (97). Beyond its role in DNA repair, PARP-1 also functions as a transcriptional coregulator, interacting with NF-κB and STAT5 to facilitate the expression of pro-inflammatory and tumorigenic genes (98), which promotes chronic inflammation, cell proliferation and angiogenesis in HCC. PARP-1 also modulates the tumor immune microenvironment through multiple mechanisms. PARP inhibition leads to the accumulation of cytosolic DNA, activation of the cyclic GMP-AMP synthase-stimulator of IFN genes-type I IFN pathway and upregulation of programmed death ligand-1 (PD-L1), thereby reshaping immune cell infiltration and enhancing antitumor immunity (99). A preclinical study of HCC demonstrated that olaparib upregulates PD-L1 via the suppression of microRNA-513, and its combination with anti-programmed cell death protein-1 (PD-1) therapy significantly enhances CD8+T-cell infiltration and antitumor efficacy (100). Broader oncology studies indicate that PARP inhibitor plus immune checkpoint inhibitor combinations are well-tolerated and exhibit synergistic antitumor effects, which justifies their translational exploration in HCC (101–103).
Caspase-3 is an effector protein in cancer cell apoptosis, which promotes apoptosis by cleaving PARP-1 (104). Notably, HCC progression is associated with decreased levels of both PARP-1 and cleaved caspase-3. DNA damage repair fails when PARP-1 is cleaved, as the cleaved form of PARP-1 can no longer repair DNA (104–106). Peptides derived from Laminaria japonica promote apoptosis by increasing cleaved caspase-9 and caspase-3 levels and activating PARP, potentially through upstream apoptotic signal-regulating kinase 1 (ASK1) phosphorylation and subsequent p38 MAPK activation (107). Factors upstream of PARP-1, such as JNK1/2 and ERK1/2, may also regulate cell death following PARP-1 inhibition (108). This mechanism can kill cancer cells and provides a potential therapeutic avenue for HCC. However, the upregulation of MET expression in HCC presents a challenge: Oxidative DNA damage activates MET, which interacts with and phosphorylates PARP-1 at tyrosine 907, thereby limiting the effectiveness of PARP-1 inhibitors in HCC (109). Future studies on the use of PARP inhibitors for the treatment HCC should account for this mechanism.
A summary of the targets and associated molecules or complexes of PARP-1 in liver diseases is presented in Table I.
Table I.Targets, associated molecules and complexes of poly(ADP-ribose) polymerase-1 in liver diseases. |
The onset and progression of liver diseases involve intricate molecular pathways that are poorly understood and associated with a poor prognosis. PARP-1 activity is both directly or indirectly regulated by a variety of signals, rendering it a potentially useful biomarker for detecting the onset of liver disorders and monitoring their course. This section summarizes the key cellular signaling pathways in which PARP-1 participates and their contributions to the pathogenesis of various liver disorders.
The regulation of tumor cell growth and death is strongly influenced by lncRNAs. Numerous lncRNAs that promote the proliferation of tumor cells also decrease their apoptosis (110). For example, Chen et al (41) reported that lncRNA-HUR1 increases Bcl-2 expression and reduces Bax expression by suppressing p53 activity. This inhibits caspase-3/7 activation via the intrinsic apoptosis pathway, thereby preventing PARP-1 cleavage and cell death. Conversely, the study demonstrated that in cell lines with stable lncRNA-HUR1 knockdown, Bcl-2 expression is reduced, Bax expression is increased and caspase-3/7 activation is promoted, which in turn accelerates PARP-1 cleavage and cell death (41). Although few lncRNAs have been thoroughly investigated in the context HCC, lncRNAs in general have been demonstrated to be essential for the development and progression of HCC (111), indicating their potential as both diagnostic and therapeutic targets (Fig. 1).
PARP-1 can sense DNA damage and trigger the TGF-β1-Smad signaling pathway. TGF-β1 is produced by endothelial, hematopoietic and connective tissue cells, and has been associated with heart failure in several studies (49,112,113). A main cause of hepatic fibrosis (HF) is the activation of HSCs. When DNA damage occurs, PARP-1 is recruited and activates TGF-β1-Smad pathway by upregulating TNF-α in HSCs, which increases ASK1-JNK phosphorylation. This exacerbates HF by promoting epithelial-to-mesenchymal transition and the deposition of α-smooth muscle actin (α-SMA) and collagen I/III. Dihydrokaempferol has been shown to decrease TNF-α transcription and inhibit the PARP-1-regulated TGF-β1 pathway, thereby reducing the levels of phosphorylated (p-)Smad 2/3 and p-ERK 1/2 (MAPK1). This is associated with suppression of the ability of HSCs to produce α-SMA and collagen I/III, contributing to the mitigation of HF, potentially by inhibition of the downstream NF-κB and ASK1-JNK signaling pathways (49) (Fig. 2).
SIRT1 is an NAD+-dependent deacetylase that plays a key role in the regulation of PPARα activity. Its expression and activity are regulated by NAD+, which serves as its substrate (114,115). Consequently, intracellular NAD+ levels can directly influence PPARα signaling. In the liver, PPARα signaling is markedly inhibited in the absence of SIRT1. The inhibition of PARP-1 can raise intracellular NAD+ levels, thereby increasing SIRT1 activity, and upregulating PPARα signaling and mitochondrial oxidation. The PARP-1-mediated poly(ADP-ribosyl)ation of PPARα suppresses its transcriptional activity by preventing PPARα from binding to SIRT1 and from recruiting the PPARα/SIRT1/PPARg coactivator-1α complex to target promoters (115). Notably, the liver PARP activity of patients with severe NAFLD is much greater than that of patients with simple steatosis, suggesting that aberrant PARP-1 activation may contribute to the downregulation of liver PPARα expression in these individuals (115). These findings suggest that inhibition of PARP-1 may have therapeutic potential in the prevention and treatment of NAFLD (Fig. 3).
The accumulation of β-catenin has been observed in a mouse model of liver cancer (116). Valanejad et al (117) identified a subset of 250-bp human chromosomal domains, known as aggressive liver cancer domains (ALCDs), that are located close to genes associated with various cancer pathways and are regulated by PARP-1. Hepatoblastoma (HBL) is a type of pediatric liver cancer that primarily affects children <3 years old (118). Notably, the PARP-1-ALCD axis modulates β-catenin, a potent promoter of aggressive liver cancer, which is markedly increased in aggressive HBL (117). PARP-1 activation can contribute to the development of HBL by activating the Wnt/β-catenin pathway and inducing post-translational changes in tumor suppressor genes. Conversely, PARP-1 inhibition can decrease cell proliferation, block Wnt/β-catenin signaling, and restore the expression of tumor suppressor genes (119). These findings suggest that targeting proteins associated with the Wnt/β-catenin pathway may represent a potential therapeutic approach for liver cancer.
The PI3K/Akt/mTOR signaling pathway inhibits apoptosis while enhancing cell survival and proliferation, and is associated with angiogenesis, carcinogenesis, invasion, and metastasis in several cancers, including colorectal cancer and non-small cell lung cancer (120,121). Numerous studies have examined the effects of natural substances on HCC, some of which target this pathway. For example, in one study, the treatment of HepG2 cells with 13-acetoxysarcocrassolide reduced p-PI3K, p-AKT and p-mTOR levels, indicating suppression of the PI3K/Akt/mTOR signaling pathway. This inhibition decreased the mTOR-mediated phosphorylation of p70S6K and its downstream effectors S6 and eukaryotic translation initiation factor 4B, and also increased cleaved PARP-1 levels, indicating that the induced apoptosis was facilitated by mitochondrial dysfunction. These findings suggest that PARP-1 activity may be associated with the phosphorylation of AKT, which influences downstream mTOR signaling and promotes tumor cell proliferation and apoptotic resistance (122).
Mechanistically, PARP-1 further promotes HCC growth by forming a positive feedback loop with the PI3K/AKT/mTOR signaling axis. PARP-1 activation increases the phosphorylation of AKT at serine 473, which activates mTOR complex 1 (mTORC1) targets such as ribosomal protein S6 kinase 1 and eukaroytic translation initiator factor 4E binding protein 1, thereby driving protein synthesis, cell proliferation and survival while inhibiting apoptosis (123–125). Conversely, PARP-1 inhibition downregulates AKT activity via upregulation of PH domain leucine-rich repeat protein phosphatase 1, highlighting the direct role of PARP-1 in the modulation of AKT/mTOR signaling (126). The activation of AKT/mTOR signaling has been demonstrated to support the stability and activity of PARP-1, with PARP-1 activation modulating mTORC1 through AMPK-dependent mechanisms, and AKT activation enhancing DDR components, including PARP-1 itself (124,127). This mutual reinforcement establishes a negative feedback loop of pro-survival signaling and genomic maintenance that promotes HCC progression and therapy resistance. Preclinical studies indicate that disrupting either PARP-1 or the PI3K/AKT/mTOR pathway can disrupt this feedback loop, reduce tumor viability, and sensitize cells to treatment (125,126,128).
These findings indicate that PARP-1 regulates several cellular signaling pathways and is crucial for the onset, progression and clinical signs of several liver diseases. This suggests the potential of PARP-1 as a prognostic indicator and possible target for liver disease therapy.
In emerging nations, particularly in the Asia-Pacific region, liver diseases are becoming widely acknowledged as major health concerns. While viral hepatitis remains the primary cause of morbidity and death, ALD and NAFLD are contributing to the liver disease burden at an accelerating rate (129).
PARP-1 expression in upregulated in numerous cancers, and prompt evaluation of its expression is important when devising a treatment plan and monitoring therapeutic effectiveness. Positron emission tomography (PET) imaging is an efficient, noninvasive, real-time technique that can be used to assess PARP-1 activity throughout the body. In 2016, researchers conducted preclinical evaluations and the first-in-human study of PARP activity in cancer. Notably, the study observed increased uptake of 18F fluorthanatrace (18F-FTT) in a patient with HCC (130). However, investigations of PARP-targeted imaging in liver diseases, including NASH, ALD and HCC, remain at the preclinical stage (131), including a study of PARP-targeted PET imaging in NASH animal models (132). Clinical trials of PARP-1 PET probes such as 18F-FTT have so far been limited to other tumor types, including breast, ovarian and prostate cancer (133), with no direct applications in liver pathology. This highlights a critical gap and opportunity for the future clinical translation of PARP-1 in liver diseases.
A number of tracers targeting PARP-1 have been developed, including 18F-FTT and the olaparib analog 18F-PARPi. Patients with elevated PARP-1 expression can be identified by PET imaging using 18F-FTT (134). A recent study designed a high-affinity PARP-1 molecule, 18F-BIBD-300, by chemical modification of the main pharmacophore of 18F-FTT. Since 18F-FTT has been used to diagnose liver cancer,18F-BIBD-300 has been suggested to have potential for this purpose (135). 18F-PARPi and 18F-FTT both undergo hepatobiliary clearance, which may limit their effectiveness in the imaging of abdominal lesions, such as liver metastases (136). 18F-FPyPARP, a 6-fluoronicotinoyl analog of 18F-PARPi, has been found to be an excellent radioactive tracer for PARP expression that is simpler to manufacture and less lipophilic than 18F-PARPi (136).
PARP inhibitors have emerged as leading cancer treatments for BRCA-deficient patients with ovarian and breast cancers (137). Notably, although all PARP-1 drugs are classified as PARP inhibitors, not all PARP inhibitors are selective for PARP-1. Thus, PARP-1 inhibitors constitute a distinct subgroup within the broader category of PARP inhibitors. The clinical utility of PARP inhibitors may be attributed to their ability to efficiently kill cells with HRD in BRCA1/2-deficient cancers by driving the accumulation of unrepaired DNA damage and inducing synthetic lethality (138).
Common adverse reactions reported in cancer trials of various PARP inhibitors, including olaparib and niraparib, include anemia, leukopenia, thrombocytopenia, nausea, vomiting and fatigue (139,140). While these adverse reactions have been observed in patients with cancer, patients with liver disease often have cytopenias, such as thrombocytopenia due to cirrhosis (141); therefore, PARP inhibitors may exacerbate hematological complications that are already present. Nausea and vomiting are also common symptoms of PARP inhibitors, occurring in >20% of patients, which may exacerbate malnutrition in patients with advanced liver disease (142). In addition, niraparib has been associated with nephrotoxicity; impaired hepatic albumin synthesis, such as that occurring in cirrhosis, may increase free drug concentrations, thereby exacerbating nephrotoxicity (143,144).
Evidence for the efficacy of PARP-1 inhibitors in liver diseases, including NAFLD/NASH, ALD, liver fibrosis/cirrhosis and HCC, comes primarily from cell and animal models. For example, one study demonstrated that PARP-1 inhibition contributed to the antifibrotic effect of puerarin in mice (145), suggesting that targeting PARP-1 has therapeutic potential for liver fibrosis. Another study used mouse models of NASH to verify that PARP inhibition and PARP-1 deletion reduce hepatic triglyceride accumulation, metabolic disorders, inflammation and/or fibrosis (146), indicating that PARP inhibition may be a promising treatment strategy for fatty hepatitis. Limited data from patients are also available: A pilot study using liver samples from patients with NAFLD confirmed that PARP-1 is activated in human fatty liver, as well as in a HFD-induced mouse model (61). In addition, a study at the University of Minnesota analyzed liver tissue samples from patients with alcoholic and HBV-related cirrhosis (stage 3–4 fibrosis). The results of the study demonstrated the key pathogenic role of PARP-1 in liver fibrosis and the potential of PARP-1 inhibitors to restore liver function after fibrosis (10). Collectively, these studies suggest that repurposing PARP-1 inhibitors to treat liver diseases associated with injury, inflammation and fibrosis may have potential clinical applicability.
PARP-1 inhibitor monotherapy appears to be largely ineffective in HCC, likely due to the BRCA nature of this cancer type. However, one study investigated non-BRCA1/2 gene abnormalities, and found that they confer sensitivities to PARP inhibition comparable to those observed with BRCA1/2 mutations (147). In addition, marked suppression of HCC cell growth was observed both in vitro and in vivo following treatment with a combination of wild-type p53-induced protein phosphatase 1 and PARP inhibitors (138). Synergistic strategies comprising PARP inhibitors and radiotherapy have also been shown to improve HCC management, with olaparib increasing the radiosensitivity of HCC cells by promoting extensive DSBs (148). In addition, dual inhibition of PARP-1/2 and tankyrase 1/2 exhibited potent anticancer activity with strong synergy, suggesting an expanded therapeutic scope for PARP-1 inhibitors (149).
Likewise, both ALD and NAFLD may be prevented by blocking PARP-1. Since NAD+ plays a crucial role in acute alcoholic liver injury, preserving NAD+ levels by blocking PARP may help to maintain liver homeostasis (12). In addition, the pharmacological inhibition or genetic deletion of PARP-1 has been shown to protect against ethanol-induced hepatocyte damage and increased fatty acid oxidation, thereby preventing fatty liver development in a rat model of NAFLD (19).
Despite the encouraging results obtained for PARP-1 inhibitors in preclinical studies, several obstacles remain in the translation of these discoveries into successful clinical applications. A major issue is the intrinsic disparity between animal or in vitro systems and the intricate pathophysiology of human disease. Preclinical models cannot accurately replicate the tumor microenvironment, genetic diversity or immune responses observed in patients. In addition, drug metabolism and pharmacokinetics differ significantly across species, resulting in variations in efficacy and toxicity profiles. Another major barrier is the development of resistance mechanisms in patients, which are not always predictable from preclinical data (150). Collectively, these factors contribute to the frequent failure of preclinically promising drugs in clinical trials and highlight the requirement for improved prediction models and biomarkers. Nevertheless, PARP-1 inhibition remains a promising treatment strategy for various liver diseases, if these translational challenges can be addressed.
As research has progressed, understanding of the role of PARP-1 in liver disorders has advanced. Studies have shown that conditions such as ALD (52), liver cirrhosis (88) and HCC (94,95) are associated with elevated PARP-1 levels, and that reducing PARP-1 expression and/or activity in hepatocytes can alleviate these disorders. This section of the review examines future directions for PARP-1 research by integrating the latest findings from both medical and and pharmaceutical perspectives.
PARP-1 inhibitors are commonly used to treat malignancies, such as breast and ovarian cancers, with BRCA1/2 mutations (151). However, these inhibitors have limited effectiveness in individuals who develop resistance to PARP-1 inhibitors and in other malignancies, such as HCC (152). Therefore, clinical development has focused on overcoming PARP-1 inhibitor resistance.
First, PARP-1 inhibitors can be combined with other DNA damage repair inhibitors. Resistance to PARP inhibitors is often not due to target protein mutations, but arises from the ability of tumor cells to change their DNA damage repair pathways (153). In addition to PARP-1, several other proteins participate in DNA damage repair. For example, DSBs recruit ATM proteins, which mediate checkpoint signaling and DNA repair (154). Replication stress activates ATM and Rad3-related (ATR), which stabilize and restart replication forks, with checkpoint kinases 1 and 2 acting downstream of ATR and ATM (155). The effectiveness of DDR inhibitors as monotherapies depends on their specific biological activity, and combining inhibitors may be a beneficial therapeutic approach when complementary pathways are targeted (156).
Secondly, PARP-1 inhibitors can be combined with immune checkpoint inhibitors. PD-1, PD-L1 and cytotoxic T-lymphocyte-associated antigen-4 are the most studied immunological checkpoints. Immune checkpoint inhibitors targeting these molecules exert antitumor effects by stimulating the tumor immune response (157). Treatment with a combination of PARP and immune checkpoint inhibitors has been shown to exhibit higher antitumor efficacy than monotherapy. Immune checkpoint inhibitors are able to sensitize tumor cells to PARP inhibitors, which represents a promising therapeutic approach (158).
A recent study identified numerous novel liver cancer susceptibility genes and characterized the landscape of rare genetic variants of liver cancer in a Chinese population. These variants were found to be strongly associated with the risk of liver cancer. Notably, the nuclear RNAi-defective 2 (NRDE2) gene was shown to enhance the assembly and activity of the casein kinase 2 complex, thereby promoting the phosphorylation of mediator of DNA damage checkpoint protein 1, initiating the HR repair pathway for DNA DSB repair, and ultimately inhibiting the development of liver cancer. NRDE2 may be a potential synthetic lethal target as rare genetic mutations that impair its HR repair function markedly increase the susceptibility of HCC to PARP-1 inhibitors. Overall, the study demonstrated that NRDE2 is a regulatory component that suppresses HCC and facilitates DNA damage repair, and that its deficiency in HCC cells results in greater susceptibility to PARP-1 inhibitors (159). This study identified NRDE2 as a potential biomarker for the treatment of HCC with PARP-1 inhibitors. However, further studies are necessary to determine whether NRDE2 deficiency could be used as a biomarker for PARP-1 inhibitor response in other cancer types or if the loss of other genes may have an impact comparable to that of NRDE2 (158).
PARP-1 plays a crucial role in liver disease by managing responses to DNA damage, inflammation, apoptosis and metabolic balance. When PARP-1 is dysregulated, it significantly contributes to the progression of conditions such as NAFLD, ALD, viral hepatitis, autoimmune liver diseases, hepatic fibrosis and the serious development of HCC. Research has shown that inhibiting PARP-1 can help reduce liver cell injury, decrease inflammation and fibrosis, and enhance antitumor immunity, highlighting its potential as a therapeutic target. Additionally, combining PARP-1 inhibitors with immune checkpoint blockers or agents that target DNA repair may improve treatment effectiveness. Future research should focus on the specific roles of different PARP-1 isoforms, the mechanisms behind treatment resistance and how to apply these findings in clinical settings. Overall, PARP-1 represents a promising target for both treatment and diagnosis, with the potential to improve outcomes for various liver diseases.
Not applicable.
This study was supported by the Natural Science Research Project of Anhui Educational Committee (grant no. 2024AH050702) and the Anhui Traditional Chinese Medicine Inheritance and Innovation Research Project (grant no. 2024CCCX288).
Not applicable.
KH, HT and SC wrote the manuscript. YL and RW created the figures and table. YJ and BC supervised the research, revised the manuscript, obtained financial support, conceptualized the review and performed the literature search. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Neshat SY, Quiroz VM, Wang Y, Tamayo S and Doloff JC: Liver Disease: induction, progression, immunological mechanisms, and therapeutic interventions. Int J Mol Sci. 22:67772021. 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 | |
|
Xiao J, Wang F, Wong NK, He J, Zhang R, Sun R, Xu Y, Liu Y, Li W, Koike K, et al: Global liver disease burdens and research trends: Analysis from a Chinese perspective. J Hepatol. 71:212–221. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Younossi ZM, Loomba R, Rinella ME, Bugianesi E, Marchesini G, Neuschwander-Tetri BA, Serfaty L, Negro F, Caldwell SH, Ratziu V, et al: Current and future therapeutic regimens for nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Hepatology. 68:361–371. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Alvarado-Tapias E, Pose E, Gratacós-Ginès J, Clemente-Sánchez A, López-Pelayo H and Bataller R: Alcohol-associated liver disease: Natural history, management and novel targeted therapies. Clin Mol Hepatol. 31 (Suppl 1):S112–S133. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Llovet JM, De Baere T, Kulik L, Haber PK, Greten TF, Meyer T and Lencioni R: Locoregional therapies in the era of molecular and immune treatments for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 18:293–313. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ishikawa T: Efficacy and features of balloon-occluded transarterial chemoembolization for hepatocellular carcinoma: A narrative review. Transl Gastroenterol Hepatol. 9:482024. View Article : Google Scholar : PubMed/NCBI | |
|
Pandey N and Black BE: Rapid detection and signaling of DNA damage by PARP-1. Trends Biochem Sci. 46:744–757. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Galindo-Campos MA, Bedora-Faure M, Farrés J, Lescale C, Moreno-Lama L, Martínez C, Martín-Caballero J, Ampurdanés C, Aparicio P, Dantzer F, et al: Coordinated signals from the DNA repair enzymes PARP-1 and PARP-2 promotes B-cell development and function. Cell Death Differ. 26:2667–2681. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Mukhopadhyay P, Rajesh M, Cao Z, Horváth B, Park O, Wang H, Erdelyi K, Holovac E, Wang Y, Liaudet L, et al: Poly (ADP-ribose) polymerase-1 is a key mediator of liver inflammation and fibrosis. Hepatology. 59:1998–2009. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Xu XL, Xing BC, Han HB, Zhao W, Hu MH, Xu ZL, Li JY, Xie Y, Gu J, Wang Y and Zhang ZQ: The properties of tumor-initiating cells from a hepatocellular carcinoma patient's primary and recurrent tumor. Carcinogenesis. 31:167–174. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Gariani K, Ryu D, Menzies KJ, Yi HS, Stein S, Zhang H, Perino A, Lemos V, Katsyuba E, Jha P, et al: Inhibiting poly ADP-ribosylation increases fatty acid oxidation and protects against fatty liver disease. J Hepatol. 66:132–141. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Guillot C, Hall J, Herceg Z, Merle P and Chemin I: Update on hepatocellular carcinoma breakthroughs: Poly(ADP-ribose) polymerase inhibitors as a promising therapeutic strategy. Clin Res Hepatol Gastroenterol. 38:137–142. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Wang C, Tian Y, Zhang F, Xu W, Li X, Shu Z, Wang Y, Huang K and Huang D: Inhibition of poly(ADP-ribose) polymerase-1 protects chronic alcoholic liver injury. Am J Pathol. 186:3117–3130. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Hardy T and Mann DA: Epigenetics in liver disease: From biology to therapeutics. Gut. 65:1895–1905. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng ML, Nakib D, Perciani CT and MacParland SA: The immune niche of the liver. Clin Sci (Lond). 135:2445–2466. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Piano S, Bunchorntavakul C, Marciano S and Rajender Reddy K: Infections in cirrhosis. Lancet Gastroenterol Hepatol. 9:745–757. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kubes P and Jenne C: Immune responses in the liver. Annu Rev Immunol. 36:247–277. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Mukhopadhyay P, Horváth B, Rajesh M, Varga ZV, Gariani K, Ryu D, Cao Z, Holovac E, Park O, Zhou Z, et al: PARP inhibition protects against alcoholic and non-alcoholic steatohepatitis. J Hepatol. 66:589–600. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
De Siervi S, Cannito S and Turato C: Chronic liver disease: latest research in pathogenesis, detection and treatment. Int J Mol Sci. 24:106332023. View Article : Google Scholar : PubMed/NCBI | |
|
Santinelli-Pestana DV, Aikawa E, Singh SA and Aikawa M: PARPs and ADP-ribosylation in chronic inflammation: A focus on macrophages. Pathogens. 12:9642023. View Article : Google Scholar : PubMed/NCBI | |
|
Vida A, Márton J, Mikó E and Bai P: Metabolic roles of poly(ADP-ribose) polymerases. Semin Cell Dev Biol. 63:135–143. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Li WH, Wang F, Song GY, Yu QH, Du RP and Xu P: PARP-1: A critical regulator in radioprotection and radiotherapy-mechanisms, challenges, and therapeutic opportunities. Front Pharmacol. 14:11989482023. View Article : Google Scholar : PubMed/NCBI | |
|
Szántó M, Gupte R, Kraus WL, Pacher P and Bai P: PARPs in lipid metabolism and related diseases. Prog Lipid Res. 84:1011172021. View Article : Google Scholar : PubMed/NCBI | |
|
Demin AA, Hirota K, Tsuda M, Adamowicz M, Hailstone R, Brazina J, Gittens W, Kalasova I, Shao Z, Zha S, et al: XRCC1 prevents toxic PARP1 trapping during DNA base excision repair. Mol Cell. 81:3018–3030.e5. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ogawa K, Masutani M, Kato K, Tang M, Kamada N, Suzuki H, Nakagama H, Sugimura T and Shirai T: Parp-1 deficiency does not enhance liver carcinogenesis induced by 2-amino-3-methylimidazo[4,5-f]quinoline in mice. Cancer Lett. 236:32–38. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Liao Y, Wang X, Ran G, Zhang S, Wu C, Tan R, Liu Y, He Y, Liu T, Wu Z, et al: DNA damage and up-regulation of PARP-1 induced by columbin in vitro and in vivo. Toxicol Lett. 379:20–34. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cantó C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer MH, Cen Y, Fernandez-Marcos PJ, Yamamoto H, Andreux PA, Cettour-Rose P, et al: The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 15:838–847. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Wu AY, Sekar P, Huang DY, Hsu SH, Chan CM and Lin WW: Spatiotemporal roles of AMPK in PARP-1- and autophagy-dependent retinal pigment epithelial cell death caused by UVA. J Biomed Sci. 30:912023. View Article : Google Scholar : PubMed/NCBI | |
|
Ju C, Liu C, Yan S, Wang Y, Mao X, Liang M and Huang K: Poly(ADP-ribose) Polymerase-1 is required for hepatocyte proliferation and liver regeneration in mice. Biochem Biophys Res Commun. 511:531–535. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yimlamai D, Christodoulou C, Galli GG, Yanger K, Pepe-Mooney B, Gurung B, Shrestha K, Cahan P, Stanger BZ and Camargo FD: Hippo pathway activity influences liver cell fate. Cell. 157:1324–1338. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Cover C, Fickert P, Knight TR, Fuchsbichler A, Farhood A, Trauner M and Jaeschke H: Pathophysiological role of poly(ADP-ribose) polymerase (PARP) activation during acetaminophen-induced liver cell necrosis in mice. Toxicol Sci. 84:201–208. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Andronikou C and Rottenberg S: Studying PAR-dependent chromatin remodeling to tackle PARPi resistance. Trends Mol Med. 27:630–642. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Pazzaglia S and Pioli C: Multifaceted role of PARP-1 in DNA repair and inflammation: Pathological and therapeutic implications in cancer and non-cancer diseases. Cells. 9:412019. View Article : Google Scholar : PubMed/NCBI | |
|
Llaneras J, Riveiro-Barciela M, Rando-Segura A, Marcos-Fosch C, Roade L, Velázquez F, Rodríguez-Frías F, Esteban R and Buti M: Etiologies and features of acute viral hepatitis in Spain. Clin Gastroenterol Hepatol. 19:1030–1037. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Cooke GS, Flower B, Cunningham E, Marshall AD, Lazarus JV, Palayew A, Jia J, Aggarwal R, Al-Mahtab M, Tanaka Y, et al: Progress towards elimination of viral hepatitis: A lancet gastroenterology and hepatology commission update. Lancet Gastroenterol Hepatol. 9:346–365. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Yeh SH, Li CL, Lin YY, Ho MC, Wang YC, Tseng ST and Chen PJ: Hepatitis B virus DNA integration drives carcinogenesis and provides a new biomarker for HBV-related HCC. Cell Mol Gastroenterol Hepatol. 15:921–929. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Funato K, Otsuka M, Sekiba K, Miyakawa Y, Seimiya T, Shibata C, Kishikawa T and Fujishiro M: Hepatitis B virus-associated hepatocellular carcinoma with Smc5/6 complex deficiency is susceptible to PARP inhibitors. Biochem Biophys Res Commun. 607:89–95. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou L, Liu CH, Lv D, Sample KM, Rojas Á, Zhang Y, Qiu H, He L, Zheng L, Chen L, et al: Halting hepatocellular carcinoma: Identifying intercellular crosstalk in HBV-driven disease. Cell Rep. 44:1154572025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu N, Liu Q, Yang X, Zhang F, Li X, Ma Y, Guan F, Zhao X, Li Z, Zhang L and Ye X: Hepatitis B virus-upregulated LNC-HUR1 promotes cell proliferation and tumorigenesis by blocking p53 activity. Hepatology. 68:2130–2144. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Wen J, Qi D, Tong X, Liu N and Ye X: HBV-upregulated Lnc-HUR1 inhibits the apoptosis of liver cancer cells. Sheng Wu Gong Cheng Xue Bao. 38:3501–3514. 2022.(In Chinese). PubMed/NCBI | |
|
Machida K, Tsukamoto H, Mkrtchyan H, Duan L, Dynnyk A, Liu HM, Asahina K, Govindarajan S, Ray R, Ou JH, et al: Toll-like receptor 4 mediates synergism between alcohol and HCV in hepatic oncogenesis involving stem cell marker Nanog. Proc Natl Acad Sci USA. 106:1548–1553. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Pal S, Polyak SJ, Bano N, Qiu WC, Carithers RL, Shuhart M, Gretch DR and Das A: Hepatitis C virus induces oxidative stress, DNA damage and modulates the DNA repair enzyme NEIL1. J Gastroenterol Hepatol. 25:627–634. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Smirnova OA, Ivanova ON, Mukhtarov F, Valuev-Elliston VT, Fedulov AP, Rubtsov PM, Zakirova NF, Kochetkov SN, Bartosch B and Ivanov AV: Hepatitis Delta virus antigens trigger oxidative stress, activate antioxidant Nrf2/ARE pathway, and induce unfolded protein response. Antioxidants (Basel). 12:9742023. View Article : Google Scholar : PubMed/NCBI | |
|
Bajaj JS: Alcohol, liver disease and the gut microbiota. Nat Rev Gastroenterol Hepatol. 16:235–246. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Contreras-Zentella ML, Villalobos-García D and Hernández-Muñoz R: Ethanol metabolism in the liver, the induction of oxidant stress, and the antioxidant defense system. Antioxidants (Basel). 11:12582022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Luo W and Wang Y: PARP-1 and its associated nucleases in DNA damage response. DNA Repair (Amst). 81:1026512019. View Article : Google Scholar : PubMed/NCBI | |
|
Langelier MF and Pascal JM: PARP-1 mechanism for coupling DNA damage detection to poly(ADP-ribose) synthesis. Curr Opin Struct Biol. 23:134–143. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Huang H, Wei S, Wu X, Zhang M, Zhou B, Huang D and Dong W: Dihydrokaempferol attenuates CCl4-induced hepatic fibrosis by inhibiting PARP-1 to affect multiple downstream pathways and cytokines. Toxicol Appl Pharmacol. 464:1164382023. View Article : Google Scholar : PubMed/NCBI | |
|
Ma XY, Zhang M, Fang G, Cheng CJ, Wang MK, Han YM, Hou XT, Hao EW, Hou YY and Bai G: Ursolic acid reduces hepatocellular apoptosis and alleviates alcohol-induced liver injury via irreversible inhibition of CASP3 in vivo. Acta Pharmacol Sin. 42:1101–1110. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yin F, Wu MM, Wei XL, Ren RX, Liu MH, Chen CQ, Yang L, Xie RQ, Jiang SY, Wang XF and Wang H: Hepatic NCoR1 deletion exacerbates alcohol-induced liver injury in mice by promoting CCL2-mediated monocyte-derived macrophage infiltration. Acta Pharmacol Sin. 43:2351–2361. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ambade A, Lowe P, Kodys K, Catalano D, Gyongyosi B, Cho Y, Iracheta-Vellve A, Adejumo A, Saha B, Calenda C, et al: Pharmacological inhibition of CCR2/5 signaling prevents and reverses alcohol-induced liver damage, steatosis, and inflammation in mice. Hepatology. 69:1105–1121. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang XJ and Malhi H: Nonalcoholic fatty liver disease. Ann Intern Med. 169:ITC65–ITC80. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Guo X, Yin X, Liu Z and Wang J: Non-alcoholic fatty liver disease (NAFLD) pathogenesis and natural products for prevention and treatment. Int J Mol Sci. 23:154892022. View Article : Google Scholar : PubMed/NCBI | |
|
Harrison SA, Allen AM, Dubourg J, Noureddin M and Alkhouri N: Challenges and opportunities in NASH drug development. Nat Med. 29:562–573. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Saiman Y, Duarte-Rojo A and Rinella ME: Fatty liver disease: Diagnosis and stratification. Annu Rev Med. 73:529–544. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ye H, Ma S, Qiu Z, Huang S, Deng G, Li Y, Xu S, Yang M, Shi H, Wu C, et al: Poria cocos polysaccharides rescue pyroptosis-driven gut vascular barrier disruption in order to alleviates non-alcoholic steatohepatitis. J Ethnopharmacol. 296:1154572022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang JH, Byeon EH, Kang D, Hong SG, Yang J, Kim DR, Yun SP, Park SW, Kim HJ, Huh JW, et al: Fermented soybean paste attenuates biogenic amine-induced liver damage in obese mice. Cells. 12:8222023. View Article : Google Scholar : PubMed/NCBI | |
|
Guo R, Li Y, Song Q, Huang R, Ge X, Nieto N, Jiang Y and Song Z: Increasing cellular NAD+ protects hepatocytes against palmitate-induced lipotoxicity by preventing PARP-1 inhibition and the mTORC1-p300 pathway activation. Am J Physiol Cell Physiol. 328:C776–C790. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Salomone F, Barbagallo I, Godos J, Lembo V, Currenti W, Cinà D, Avola R, D'Orazio N, Morisco F, Galvano F and Li Volti G: Silibinin restores NAD+ levels and induces the SIRT1/AMPK pathway in non-alcoholic fatty liver. Nutrients. 9:10862017. View Article : Google Scholar : PubMed/NCBI | |
|
Huang K, Du M, Tan X, Yang L, Li X, Jiang Y, Wang C, Zhang F, Zhu F, Cheng M, et al: PARP1-mediated PPARα poly(ADP-ribosyl)ation suppresses fatty acid oxidation in non-alcoholic fatty liver disease. J Hepatol. 66:962–977. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Majeed Y, Halabi N, Madani AY, Engelke R, Bhagwat AM, Abdesselem H, Agha MV, Vakayil M, Courjaret R, Goswami N, et al: SIRT1 promotes lipid metabolism and mitochondrial biogenesis in adipocytes and coordinates adipogenesis by targeting key enzymatic pathways. Sci Rep. 11:81772021. View Article : Google Scholar : PubMed/NCBI | |
|
Ding RB, Bao J and Deng CX: Emerging roles of SIRT1 in fatty liver diseases. Int J Biol Sci. 13:852–867. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Ray K: Developing a toolbox for drug-induced liver injury. Nat Rev Gastroenterol Hepatol. 17:7142020. View Article : Google Scholar : PubMed/NCBI | |
|
Kumachev A and Wu PE: Drug-induced liver injury. CMAJ. 193:E3102021. View Article : Google Scholar : PubMed/NCBI | |
|
Björnsson HK and Björnsson ES: Drug-induced liver injury: Pathogenesis, epidemiology, clinical features, and practical management. Eur J Intern Med. 97:26–31. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
European Association for the Study of the Liver, . EASL clinical practice guidelines: Drug-induced liver injury. J Hepatol. 70:1222–1261. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Tang J and Mao Y: Incidence and risk factors of drug-induced liver injury. Liver Int. 42:1999–2014. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Gallyas F Jr and Sumegi B: Mitochondrial protection by PARP inhibition. Int J Mol Sci. 21:27672020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Xu W, Zhang Y, Huang D and Huang K: Poly(ADP-ribosyl)ated PXR is a critical regulator of acetaminophen-induced hepatotoxicity. Cell Death Dis. 9:8192018. View Article : Google Scholar : PubMed/NCBI | |
|
Su Q, Kuang W, Hao W, Liang J, Wu L, Tang C, Wang Y and Liu T: Antituberculosis drugs (rifampicin and isoniazid) induce liver injury by regulating NLRP3 inflammasomes. Mediators Inflamm. 2021:80862532021. View Article : Google Scholar : PubMed/NCBI | |
|
Carbone M and Neuberger JM: Autoimmune liver disease, autoimmunity and liver transplantation. J Hepatol. 60:210–223. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Trivedi PJ, Hirschfield GM, Adams DH and Vierling JM: Immunopathogenesis of primary biliary cholangitis, primary sclerosing cholangitis and autoimmune hepatitis: Themes and concepts. Gastroenterology. 166:995–1019. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Yilmaz K, Haeberle S, Kim YO, Fritzler MJ, Weng SY, Goeppert B, Raker VK, Steinbrink K, Schuppan D, Enk A and Hadaschik EN: Regulatory T-cell deficiency leads to features of autoimmune liver disease overlap syndrome in scurfy mice. Front Immunol. 14:12536492023. View Article : Google Scholar : PubMed/NCBI | |
|
Heo NY and Kim H: Epidemiology and updated management for autoimmune liver disease. Clin Mol Hepatol. 29:194–196. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Pötter S, Chen CW, Liang R, Gelse K, Ludolph I, Horch RE, Distler O, Schett G, Distler JHW and Dees C: Poly(ADP-ribose) polymerase-1 regulates fibroblast activation in systemic sclerosis. Ann Rheum Dis. 77:744–751. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Su X, Ye L, Chen X, Zhang H, Zhou Y, Ding X, Chen D, Lin Q and Chen C: MiR-199-3p promotes ERK-mediated IL-10 production by targeting poly (ADP-ribose) polymerase-1 in patients with systemic lupus erythematosus. Chem Biol Interact. 306:110–116. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang G, Ma H, Wang J and Khan MF: Contribution of poly(ADP-ribose)polymerase-1 activation and apoptosis in trichloroethene-mediated autoimmunity. Toxicol Appl Pharmacol. 362:28–34. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hao J, Sun W and Xu H: Pathogenesis of concanavalin A induced autoimmune hepatitis in mice. Int Immunopharmacol. 102:1084112022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Q, Yang H, Kang X, Tian H, Kang Y, Li L, Yang X, Liu H, Ren P, Kuang X, et al: A synbiotic ameliorates con A-induced autoimmune hepatitis in mice through modulation of gut microbiota and immune imbalance. Mol Nutr Food Res. 67:e22004282023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Hao H and Hou T: Concanavalin A-induced autoimmune hepatitis model in mice: Mechanisms and future outlook. Open Life Sci. 17:91–101. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Filliol A, Piquet-Pellorce C, Dion S, Genet V, Lucas-Clerc C, Dantzer F and Samson M: PARP2 deficiency affects invariant-NKT-cell maturation and protects mice from concanavalin A-induced liver injury. Am J Physiol Gastrointest Liver Physiol. 313:G399–G409. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Virág L and Szabó C: The therapeutic potential of poly(ADP-ribose) polymerase inhibitors. Pharmacol Rev. 54:375–429. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Tanaka A: Emerging novel treatments for autoimmune liver diseases. Hepatol Res. 49:489–499. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kisseleva T and Brenner D: Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 18:151–166. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Horn P and Tacke F: Metabolic reprogramming in liver fibrosis. Cell Metab. 36:1439–1455. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gu L, Zhao C, Wang Y, Wang C, Yin X, Ye Q, Liu Y, Zou X, Wang L, Zhuge Y, et al: Senescence of hepatic stellate cells by specific delivery of manganese for limiting liver fibrosis. Nano Lett. 24:1062–1073. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Nithyananthan S and Thirunavukkarasu C: Arsenic trioxide, a cancer chemo drug hampers fibrotic liver regeneration by interrupting oxidative stress rekindling and stellate cell rejuvenation. J Cell Physiol. 235:1222–1234. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Meng D, Li Z, Wang G, Ling L, Wu Y and Zhang C: Carvedilol attenuates liver fibrosis by suppressing autophagy and promoting apoptosis in hepatic stellate cells. Biomed Pharmacother. 108:1617–1627. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Tan Y, Li C, Zhou J, Deng F and Liu Y: Berberine attenuates liver fibrosis by autophagy inhibition triggering apoptosis via the miR-30a-5p/ATG5 axis. Exp Cell Res. 427:1136002023. View Article : Google Scholar : PubMed/NCBI | |
|
Singal AG, Kanwal F and Llovet JM: Global trends in hepatocellular carcinoma epidemiology: Implications for screening, prevention and therapy. Nat Rev Clin Oncol. 20:864–884. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Pan J, Zhang M, Dong L, Ji S, Zhang J, Zhang S, Lin Y, Wang X, Ding Z, Qiu S, et al: Genome-Scale CRISPR screen identifies LAPTM5 driving lenvatinib resistance in hepatocellular carcinoma. Autophagy. 19:1184–1198. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
No authors listed. Hepatocellular carcinoma. Nat Rev Dis Primers. 7:72021. View Article : Google Scholar : PubMed/NCBI | |
|
Zai W, Chen W, Han Y, Wu Z, Fan J, Zhang X, Luan J, Tang S, Jin X, Fu X, et al: Targeting PARP and autophagy evoked synergistic lethality in hepatocellular carcinoma. Carcinogenesis. 41:345–357. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gerossier L, Dubois A, Paturel A, Fares N, Cohen D, Merle P, Lachuer J, Wierinckx A, Saintigny P, Bancel B, et al: PARP inhibitors and radiation potentiate liver cell death in vitro. Do hepatocellular carcinomas have an achilles' heel? Clin Res Hepatol Gastroenterol. 45:1015532021.PubMed/NCBI | |
|
Jeong KY and Park MH: The significance of targeting Poly (ADP-ribose) polymerase-1 in pancreatic cancer for providing a new therapeutic paradigm. Int J Mol Sci. 22:35092021. View Article : Google Scholar : PubMed/NCBI | |
|
Demény MA and Virág L: The PARP enzyme family and the hallmarks of cancer part 1. Cell intrinsic hallmarks. Cancers (Basel). 13:20422021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu T, Zheng JY, Huang LL, Wang YH, Yao DF and Dai HB: Human PARP1 substrates and regulators of its catalytic activity: An updated overview. Front Pharmacol. 14:11371512023. View Article : Google Scholar : PubMed/NCBI | |
|
Wong CW, Evangelou C, Sefton KN, Leshem R, Zhang W, Gopalan V, Chattrakarn S, Fernandez Carro ML, Uzuner E, Mole H, et al: PARP14 inhibition restores PD-1 immune checkpoint inhibitor response following IFNγ-driven acquired resistance in preclinical cancer models. Nat Commun. 14:59832023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun G, Miao G, Li Z, Zheng W, Zhou C, Sun G, Cao H, Li Z and Tang W: Inhibition of PARP potentiates immune checkpoint therapy through miR-513/PD-L1 pathway in hepatocellular carcinoma. J Oncol. 2022:69889232022. View Article : Google Scholar : PubMed/NCBI | |
|
Konstantinopoulos PA, Waggoner S, Vidal GA, Mita M, Moroney JW, Holloway R, Van Le L, Sachdev JC, Chapman-Davis E, Colon-Otero G, et al: Single-arm phases 1 and 2 trial of niraparib in combination with pembrolizumab in patients with recurrent platinum-resistant ovarian carcinoma. JAMA Oncol. 5:1141–1149. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yu EY, Piulats JM, Gravis G, Fong PCC, Todenhöfer T, Laguerre B, Arranz JA, Oudard S, Massard C, Heinzelbecker J, et al: Pembrolizumab plus olaparib in patients with metastatic castration-resistant prostate cancer: Long-term results from the phase 1b/2 KEYNOTE-365 cohort a study. Eur Urol. 83:15–26. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yap TA, Bardia A, Dvorkin M, Galsky MD, Beck JT, Wise DR, Karyakin O, Rubovszky G, Kislov N, Rohrberg K, et al: Avelumab plus talazoparib in patients with advanced solid tumors: The JAVELIN PARP medley nonrandomized controlled trial. JAMA Oncol. 9:40–50. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ishteyaque S, Singh G, Yadav KS, Verma S, Sharma RK, Sen S, Srivastava AK, Mitra K, Lahiri A, Bawankule DU, et al: Cooperative STAT3-NFkB signaling modulates mitochondrial dysfunction and metabolic profiling in hepatocellular carcinoma. Metabolism. 152:1557712024. View Article : Google Scholar : PubMed/NCBI | |
|
Zein N, Elewa YHA, Alruwaili MK, Dewaard M, Alorabi M, Albogami SM, Batiha GE and Zahran MH: Barhi date (Phoenix dactylifera) extract ameliorates hepatocellular carcinoma in male rats. Biomed Pharmacother. 156:1139762022. View Article : Google Scholar : PubMed/NCBI | |
|
Pasaol JC, Dejnaka E, Mucignat G, Bajzert J, Henklewska M, Obmińska-Mrukowicz B, Giantin M, Pauletto M, Zdyrski C, Dacasto M and Pawlak A: PARP inhibitor olaparib induces DNA damage and acts as a drug sensitizer in an in vitro model of canine hematopoietic cancer. BMC Vet Res. 21:4392025. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Li Y, Guo W, Liu J, Lao W, Hu P, Lin Y and Chen H: Laminaria japonica peptides suppress liver cancer by inducing apoptosis: Possible signaling pathways and mechanism. Mar Drugs. 20:7042022. View Article : Google Scholar : PubMed/NCBI | |
|
Luo T, Yuan Y, Yu Q, Liu G, Long M, Zhang K, Bian J, Gu J, Zou H, Wang Y, et al: PARP-1 overexpression contributes to Cadmium-induced death in rat proximal tubular cells via parthanatos and the MAPK signalling pathway. Sci Rep. 7:43312017. View Article : Google Scholar : PubMed/NCBI | |
|
Dong Q, Du Y, Li H, Liu C, Wei Y, Chen MK, Zhao X, Chu YY, Qiu Y, Qin L, et al: EGFR and c-MET cooperate to enhance resistance to PARP inhibitors in hepatocellular carcinoma. Cancer Res. 79:819–829. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Merdrignac A, Papoutsoglou P and Coulouarn C: Long noncoding RNAs in cholangiocarcinoma. Hepatology. 73:1213–1226. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Z, Zhou JK, Peng Y, He W and Huang C: The role of long noncoding RNAs in hepatocellular carcinoma. Mol Cancer. 19:772020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Chen J, Wang Y, Fang L, Guo C, Sang T, Peng H, Zhao Q, Chen S, Lin X and Wang X: Ganoderma lucidum polysaccharide inhibits HSC activation and liver fibrosis via targeting inflammation, apoptosis, cell cycle, and ECM-receptor interaction mediated by TGF-β/Smad signaling. Phytomedicine. 110:1546262023. View Article : Google Scholar : PubMed/NCBI | |
|
Ren Y, Chen Y, Tang EH, Hu Y, Niu B, Liang H, Xi C, Zhao F and Cao Z: Arbidol attenuates liver fibrosis and activation of hepatic stellate cells by blocking TGF-β1 signaling. Eur J Pharmacol. 967:1763672024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Zhang J, Zheng Y, Zhang Y, Zhang XJ, Wang H, Du Y, Guan J, Wang X and Fu J: NAD+ improves cognitive function and reduces neuroinflammation by ameliorating mitochondrial damage and decreasing ROS production in chronic cerebral hypoperfusion models through Sirt1/PGC-1α pathway. J Neuroinflammation. 18:2072021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang K, Du M, Tan X, Yang L, Li X, Jiang Y, Wang C, Zhang F, Zhu F, Cheng M, et al: PARP1-mediated PPARα poly(ADP-ribosyl)ation suppresses fatty acid oxidation in non-alcoholic fatty liver disease. J Hepatol. 66:962–977. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Nozaki T, Fujihara H, Watanabe M, Tsutsumi M, Nakamoto K, Kusuoka O, Kamada N, Suzuki H, Nakagama H, Sugimura T and Masutani M: Parp-1 deficiency implicated in colon and liver tumorigenesis induced by azoxymethane. Cancer Sci. 94:497–500. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Valanejad L, Cast A, Wright M, Bissig KD, Karns R, Weirauch MT and Timchenko N: PARP1 activation increases expression of modified tumor suppressors and pathways underlying development of aggressive hepatoblastoma. Commun Biol. 1:672018. View Article : Google Scholar : PubMed/NCBI | |
|
Pio L, O'Neill AF, Woodley H, Murphy AJ, Tiao G, Franchi-Abella S, Fresneau B, Watanabe K, Alaggio R, Lopez-Terrada D, et al: Hepatoblastoma. Nat Rev Dis Primers. 11:362025. View Article : Google Scholar : PubMed/NCBI | |
|
Sha YL, Liu S, Yan WW and Dong B: Wnt/β-catenin signaling as a useful therapeutic target in hepatoblastoma. Biosci Rep. 39:BSR201924662019. View Article : Google Scholar : PubMed/NCBI | |
|
Maharati A and Moghbeli M: PI3K/AKT signaling pathway as a critical regulator of epithelial-mesenchymal transition in colorectal tumor cells. Cell Commun Signal. 21:2012023. View Article : Google Scholar : PubMed/NCBI | |
|
Tan AC: Targeting the PI3K/Akt/mTOR pathway in non-small cell lung cancer (NSCLC). Thorac Cancer. 11:511–518. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Hsu CM, Lin JJ, Su JH and Liu CI: 13-Acetoxysarcocrassolide induces apoptosis in human hepatocellular carcinoma cells through mitochondrial dysfunction and suppression of the PI3K/AKT/mTOR/p70S6K signalling pathway. Pharm Biol. 60:2276–2285. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ethier C, Tardif M, Arul L and Poirier GG: PARP-1 modulation of mTOR signaling in response to a DNA alkylating agent. PLoS One. 7:e479782012. View Article : Google Scholar : PubMed/NCBI | |
|
Sun EJ, Wankell M, Palamuthusingam P, McFarlane C and Hebbard L: Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Biomedicines. 9:16392021. View Article : Google Scholar : PubMed/NCBI | |
|
Gallyas F Jr, Sumegi B and Szabo C: Role of Akt activation in PARP inhibitor resistance in cancer. Cancers (Basel). 12:5322020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Wang H, Davis BC, Liang J, Cui R, Chen SJ and Xu ZX: PARP1 inhibitors attenuate AKT phosphorylation via the upregulation of PHLPP1. Biochem Biophys Res Commun. 412:379–384. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Xu H, Ma Z, Mo X, Chen X, Xu F, Wu F, Chen H, Zhou G, Xia H and Zhang C: Inducing synergistic DNA damage by TRIP13 and PARP1 inhibitors provides a potential treatment for hepatocellular carcinoma. J Cancer. 13:2226–2237. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Bhamidipati D, Haro-Silerio JI, Yap TA and Ngoi N: PARP inhibitors: Enhancing efficacy through rational combinations. Br J Cancer. 129:904–916. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sarin SK, Kumar M, Eslam M, George J, Al Mahtab M, Akbar SMF, Jia J, Tian Q, Aggarwal R, Muljono DH, et al: Liver diseases in the Asia-Pacific region: A lancet gastroenterology & hepatology commission. Lancet Gastroenterol Hepatol. 5:167–228. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Michel LS, Dyroff S, Brooks FJ, Spayd KJ, Lim S, Engle JT, Phillips S, Tan B, Wang-Gillam A, Bognar C, et al: PET of poly (ADP-Ribose) polymerase activity in cancer: Preclinical assessment and first in-human studies. Radiology. 282:453–463. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Destro G, Rizzo R, Rua C, Azimi RR and Morbelli S: Exploring the radiochemistry of PARP inhibitors: A new era in therapy and imaging. EJNMMI Radiopharm Chem. 10:372025. View Article : Google Scholar : PubMed/NCBI | |
|
Jeppesen TE, Shao T, Chen J, Patel JS, Zhou X, Kjaer A and Liang SH: Poly(ADP-ribose) polymerase (PARP)-targeted PET imaging in non-oncology application: A pilot study in preclinical models of nonalcoholic steatohepatitis. Am J Nucl Med Mol Imaging. 14:41–47. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
McDonald ES, Pantel AR, Shah PD, Farwell MD, Clark AS, Doot RK, Pryma DA and Carlin SD: In vivo visualization of PARP inhibitor pharmacodynamics. JCI Insight. 6:e1465922021. View Article : Google Scholar : PubMed/NCBI | |
|
Lee HS, Schwarz SW, Schubert EK, Chen DL, Doot RK, Makvandi M, Lin LL, McDonald ES, Mankoff DA and Mach RH: The development of 18F fluorthanatrace: A PET radiotracer for imaging poly (ADP-ribose) polymerase-1. Radiol Imaging Cancer. 4:e2100702022. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng W, Huang Y, Xie Y, Yang T, Cheng X, Chen H, Li C, Jiang Z, Yu Z, Li Z, et al: Design, synthesis, and evaluation of [18F]BIBD-300 as a positron emission tomography tracer for poly(ADP-ribose) polymerase-1. Mol Pharm. 21:2606–2621. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Stotz S, Kinzler J, Nies AT, Schwab M and Maurer A: Two experts and a newbie: [18F]PARPi vs. [18F]FTT vs. [18F]FPyPARP-a comparison of PARP imaging agents. Eur J Nucl Med Mol Imaging. 49:834–846. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Liu ZY, Wu N, Chen YC, Cheng Q and Wang J: PARP inhibitor resistance: The underlying mechanisms and clinical implications. Mol Cancer. 19:1072020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen M, Wang W, Hu S, Tong Y, Li Y, Wei Q, Yu L, Zhu L, Zhu Y, Liu L, et al: Co-targeting WIP1 and PARP induces synthetic lethality in hepatocellular carcinoma. Cell Commun Signal. 20:392022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Y, Zhao S, Wu T and Zhang H: Comparison of adverse reactions caused by olaparib for different indications. Front Pharmacol. 13:9681632022. View Article : Google Scholar : PubMed/NCBI | |
|
LaFargue CJ, Dal Molin GZ, Sood AK and Coleman RL: Exploring and comparing adverse events between PARP inhibitors. Lancet Oncol. 20:e15–e28. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Peck-Radosavljevic M: Thrombocytopenia in chronic liver disease. Liver Int. 37:778–793. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Shin S, Jun DW, Saeed WK and Koh DH: A narrative review of malnutrition in chronic liver disease. Ann Transl Med. 9:1722021. View Article : Google Scholar : PubMed/NCBI | |
|
Ren X, Sun P and Wang Y: PARP inhibitor-related acute renal failure: A real-world study based on the FDA adverse event reporting system database. Expert Opin Drug Saf. 23:1463–1471. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lazareth H, Delanoy N, Cohen R, Boissier E, Ayari H, Combe P, Crespel C, Mercadier-Riaz E, Karras A, Courbebaisse M, et al: Nephrotoxicity associated with niraparib. Am J Kidney Dis. 76:898–900. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Shi XL, Feng M, Wang X, Zhang ZH, Zhao X, Han B, Ma HC, Dai B and Ding YT: Puerarin protects against CCl4-induced liver fibrosis in mice: Possible role of PARP-1 inhibition. Int Immunopharmacol. 38:238–245. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Mukhopadhyay P, Horváth B, Rajesh M, Varga ZV, Gariani K, Ryu D, Cao Z, Holovac E, Park O, Zhou Z, et al: PARP inhibition protects against alcoholic and non-alcoholic steatohepatitis. J Hepatol. 66:589–600. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Guo M and Wang SM: The BRCAness landscape of cancer. Cells. 11:38772022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen G, Zheng D, Zhou Y, Du S and Zeng Z: Olaparib enhances radiation-induced systemic anti-tumor effects via activating STING-chemokine signaling in hepatocellular carcinoma. Cancer Lett. 582:2165072024. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Y, Wu H, Huang L, Zhai B, Li X, Xu S, Wu X, Zhu Q and Xu Q: Rational design, synthesis and biological evaluation of dual PARP-1/2 and TNKS1/2 inhibitors for cancer therapy. Eur J Med Chem. 237:1144172022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang YQ, Wang PY, Wang YT, Yang GF, Zhang A and Miao ZH: An update on poly(ADP-ribose)polymerase-1 (PARP-1) inhibitors: Opportunities and challenges in cancer therapy. J Med Chem. 59:9575–9598. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Chan CY, Tan KV and Cornelissen B: PARP inhibitors in cancer diagnosis and therapy. Clin Cancer Res. 27:1585–1594. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kim DS, Camacho CV and Kraus WL: Alternate therapeutic pathways for PARP inhibitors and potential mechanisms of resistance. Exp Mol Med. 53:42–51. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mweempwa A and Wilson MK: Mechanisms of resistance to PARP inhibitors-an evolving challenge in oncology. Cancer Drug Resist. 2:608–617. 2019.PubMed/NCBI | |
|
Priya B, Ravi S and Kirubakaran S: Targeting ATM and ATR for cancer therapeutics: Inhibitors in clinic. Drug Discov Today. 28:1036622023. View Article : Google Scholar : PubMed/NCBI | |
|
Wilson Z, Odedra R, Wallez Y, Wijnhoven PWG, Hughes AM, Gerrard J, Jones GN, Bargh-Dawson H, Brown E, Young LA, et al: ATR inhibitor AZD6738 (ceralasertib) exerts antitumor activity as a monotherapy and in combination with chemotherapy and the PARP inhibitor olaparib. Cancer Res. 82:1140–1152. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Jaidee R, Pocasap P, Jusakul A, Senggunprai L, Prawan A, Hong JH, Heng HL, Kukongviriyapan V, Teh BT and Kongpetch S: Synergistic suppression of cholangiocarcinoma cells via DNA damage response and cell cycle arrest by dual targeting PARP and ATM in DNA damage repair pathway. Biomed Pharmacother. 189:1182732025. View Article : Google Scholar : PubMed/NCBI | |
|
Pandey P, Khan F, Qari HA, Upadhyay TK, Alkhateeb AF and Oves M: Revolutionization in cancer therapeutics via targeting major immune checkpoints PD-1, PD-L1 and CTLA-4. Pharmaceuticals (Basel). 15:3352022. View Article : Google Scholar : PubMed/NCBI | |
|
Stewart RA, Pilié PG and Yap TA: Development of PARP and immune-checkpoint inhibitor combinations. Cancer Res. 78:6717–6725. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Liu X, Zuo X, Wang C, Zhang Z, Zhang H, Zeng T, Chen S, Liu M, Chen H, et al: NRDE2 deficiency impairs homologous recombination repair and sensitizes hepatocellular carcinoma to PARP inhibitors. Cell Genom. 4:1005502024. View Article : Google Scholar : PubMed/NCBI |