1. Introduction
Liver diseases pose a significant global
health challenge, affecting millions of individuals and resulting in
substantial morbidity and mortality. Conditions such as hepatitis, cirrhosis,
fatty liver disease, and hepatocellular carcinoma (HCC) are among the most
common liver conditions, often leading to serious complications like liver
failure, portal hypertension, and increased susceptibility to infections.
According to the World Health Organization (WHO), liver diseases contribute to
approximately 2 million deaths annually worldwide, highlighting the urgent need
for improved diagnostic tools and therapeutic strategies [
1].
The complex nature of liver diseases is
evident in their intricate pathogenesis, involving critical signaling pathways
that regulate cellular responses to injury, inflammation, and fibrosis. These
pathways are influenced by factors such as genetic predisposition,
environmental exposures (e.g., alcohol consumption and viral infections),
metabolic disturbances (such as obesity and diabetes), and immune system
dysregulation. The interplay of these factors can lead to a series of events
ultimately resulting in hepatic injury and disease progression [
2].
Recent advancements in molecular biology
have unveiled the intricate networks of signaling cascades pivotal in the
development and progression of liver diseases. Key pathways like the
mitogen-activated protein kinase (MAPK) pathway, nuclear factor kappa B (NF-κB)
signaling, transforming growth factor-beta (TGF-β) signaling, and various
apoptotic pathways have been identified as essential players in mediating
hepatic inflammation, fibrosis, and tumorigenesis [
3].
Understanding these signaling pathways not only elucidates the underlying
mechanisms driving liver pathology but also presents opportunities for novel
therapeutic interventions to mitigate disease progression.
In addition to traditional pharmacological
approaches, emerging therapies targeting specific signaling molecules or
pathways show promise for more effective treatment options. For example, the
inhibitors of TGF-β signaling are under investigation for their potential to
reduce fibrosis in chronic liver diseases (CLDs). Recent research indicates
that the circadian clock plays a crucial role in regulating TGF-β signaling and
liver fibrosis, with disruptions in circadian clock regulation leading to
increased hepatic fibrosis [
4]. Similarly,
modulating immune responses through checkpoint inhibitors has shown efficacy in
certain cases of HCC. These innovative strategies underscore the importance of
ongoing research into the molecular foundations of liver disease [
5].
In this Topic titled “Signaling Pathways in
Liver Disease”, 110 authors from eight countries (USA, UK, Germany, France,
Italy, Spain, Greece, and China) provide a comprehensive overview of the recent
research findings emphasizing the crucial role of signaling mechanisms in liver
pathology (
Figure 1).
This Topic presents a comprehensive
collection of research that delves into the mechanisms of liver disorders. It
features four insightful review articles that synthesize the current knowledge
on various signaling pathways involved in liver disease, highlighting their
roles in hepatocyte function, inflammation, and fibrosis. Each article explores
various aspects of these complex signaling networks, from basic research
elucidating fundamental biological processes to establishing novel clinically
relevant targets. Additionally, this Topic aims to encourage further research
efforts that will enhance our understanding of liver diseases and ultimately
contribute to the development of more effective management strategies for
affected individuals. My co-editors, Ruchi Bansal (University of Twente, The
Netherlands), Gabriele Grassi (University of Trieste, Italy), and Leo A. van
Grunsven (Vrije Universiteit Brussel, Belgium), and I hope that this
compilation will serve as a valuable resource for researchers and clinicians
dedicated to unraveling the complexities surrounding liver disease
pathogenesis.
2. An Overview of Published Articles
The review by Zhong et al. (contribution 1)
titled “From Inflammation to Fibrosis: Novel Insights into the Roles of High
Mobility Group Protein Box 1 in
Schistosome-Induced Liver Damage” discusses the significant role of High
Mobility Group Box 1 (HMGB1) in
schistosomiasis, a chronic parasitic disease that can lead to severe liver
damage. The authors highlight that schistosome eggs trapped in the liver
trigger granuloma formation, chronic inflammation, and eventual fibrosis,
contributing to high morbidity and mortality rates. While praziquantel is
effective against mature worms, there are limited options for reversing liver
damage. HMGB1 is identified as a multifunctional cytokine involved in liver
injury and immune responses by interacting with various receptors. Elevated
levels of HMGB1 have been observed in patients with schistosomiasis,
correlating with hepatic fibrosis development. The review emphasizes HMGB1’s
potential as a therapeutic target due to its involvement in inflammatory
pathways and fibrogenesis mediated by activated hepatic stellate cells (HSCs).
The authors summarize the existing literature on HMGB1’s structure, functions,
and mechanistic roles in both inflammation and fibrosis within the context of
schistosomiasis. They suggest that inhibiting HMGB1 could provide protective
effects against liver damage caused by schistosomes. Overall, the review calls
for further research into HMGB1 as a potential treatment avenue for managing
schistosome-induced liver diseases.
The article titled “Modulation of the Bile
Acid Enterohepatic Cycle by Intestinal Microbiota Alleviates Alcohol Liver
Disease” by Ciocan and colleagues (contribution 2) investigates the effects of
pectin, a soluble fiber, on alcohol-induced liver disease (ALD) in mice. The
study demonstrates that pectin supplementation improves liver health by
reshaping the intestinal microbiota and enhancing bile acid (BA) metabolism.
Pectin treatment led to decreased BA levels in plasma and liver while
increasing their levels in the cecum, indicating enhanced BA excretion. This
shift was associated with a change in BA composition towards less toxic
hydrophilic forms. The beneficial effects of pectin were linked to an increase
in intestinal bacteria with bile acid-metabolizing enzymes, which facilitated
these metabolic changes. Furthermore, pectin influenced signaling pathways
related to bile acids by inhibiting farnesoid X receptor (FXR) signaling in the
ileum and impacting the expression of various BA transporters. Despite
increased BA synthesis due to altered signaling, pectin promoted their
excretion into feces. Overall, the findings suggest that dietary fiber like
pectin could serve as a therapeutic strategy for managing ALD by modifying the
gut microbiota and enhancing bile acid clearance. The authors advocate for
further clinical trials to explore the potential of dietary interventions in
ALD management among patients who often consume low-fiber diets.
In the article titled “Exercise Affects the
Formation and Recovery of Alcoholic Liver Disease through the IL-6-p47phox
Oxidative-Stress Axis”, the team of Cui et al. (contribution 3) explores the
impact of exercise on alcoholic liver disease (ALD) and its association with
the interleukin-6 (IL-6)–p47phox oxidative stress pathway. The study
involved two experiments using male C57BL/6J mice, where ALD was induced
through a high-fat alcoholic diet. In the first experiment, exercise
intervention over six weeks following ALD model establishment showed that
exercise significantly reduced serum triglycerides, improved liver function,
and decreased inflammation in liver tissue. The combination of exercise with a
NADPH oxidase inhibitor (apocynin) further enhanced these effects by reducing
oxidative stress markers. In the second experiment, simultaneous alcohol
consumption during exercise exacerbated dyslipidemia and oxidative stress,
leading to increased liver injury. The findings highlighted that while exercise
generally ameliorates ALD symptoms via the IL-6–p47phox axis,
concurrent alcohol intake during exercise negatively impacts lipid metabolism
and increases oxidative stress levels. Overall, the review concludes that
regular aerobic exercise can help mitigate hepatocyte damage and dyslipidemia
in ALD through the modulation of inflammatory pathways, but drinking alcohol
while exercising can counteract these benefits.
In the review article titled “Pituitary
Tumor-Transforming Gene 1/Delta like Non-Canonical Notch Ligand 1 Signaling in
Chronic Liver Diseases”, Perramón and Jiménez discuss the potential of
targeting the pituitary tumor transforming gene 1/delta like non-canonical
Notch ligand (PTTG1/DLK1) signaling axis as a therapeutic strategy for CLDs,
including non-alcoholic fatty liver disease (NAFLD), liver fibrosis, and HCC
(contribution 4). PTTG1 is identified as a proto-oncogene associated with
cellular proliferation, inflammation, and fibrogenesis. DLK1, a target of
PTTG1, has been shown to contribute to hepatic fibrosis by promoting the
activation of HSCs. The authors highlight that both proteins are involved in
regulating key processes such as metabolism, cell differentiation, and response
to injury within the liver. The review discusses how the dysregulation of the
PTTG1/DLK1 pathway is implicated in the progression of CLDs and suggests that
inhibiting this signaling could mitigate the tissue remodeling and fibrosis
associated with these conditions. The authors conclude that further research
into this axis may provide new insights for developing effective treatments for
CLDs.
The article “Caveolin-1 Alleviates
Acetaminophen-Induced Hepatotoxicity in Alcoholic Fatty Liver Disease by
Regulating the Ang II/EGFR/ERK Axis” explores the protective role of Caveolin-1
(CAV1) against liver injury caused by acetaminophen (APAP) in a model of
alcoholic fatty liver disease (AFLD) (contribution 5). In their study, Xin and
colleagues demonstrate that APAP exacerbates lipid accumulation and oxidative
stress in AFLD, leading to increased levels of angiotensin II (Ang II) and
decreased expression of CAV1 and ACE2. Through both in vivo and in vitro
experiments, the researchers found that the overexpression of CAV1 alleviated
APAP-induced hepatotoxicity by reducing Ang II levels and inhibiting the
activation of the epidermal growth factor receptor (EGFR) and its downstream
extracellular signal-regulated kinase (ERK) signaling pathway. Additionally,
CAV1 was shown to restore autophagic flux, which is crucial for mitigating
lipid accumulation. The findings suggest that targeting the CAV1-mediated regulation
of the Ang II/EGFR/ERK axis could provide new therapeutic strategies for
managing APAP-induced liver injury in patients with AFLD. Overall, this study
highlights the potential of CAV1 as a protective factor against hepatic damage
associated with alcohol consumption and APAP overdose.
The article titled “Role of Hepatocyte
Senescence in the Activation of Hepatic Stellate Cells and Liver Fibrosis
Progression” explores the relationship between hepatocyte senescence and liver
fibrosis, with a particular focus on how senescent hepatocytes influence HSC
activation (contribution 6). Wijaysiri and colleagues conducted an analysis of
liver biopsy specimens from patients with NAFLD and found a significant
correlation between the presence of senescent hepatocytes (marked by p16
expression) and HSC activation (indicated by αSMA expression) as well as the
fibrosis stage. Using in vitro models, the researchers discovered that
conditioned media from senescent HepG2 cells significantly upregulated
inflammatory and fibrogenic gene expression in cultured HSCs, suggesting that
the factors secreted by senescent hepatocytes activate these cells. Notably,
the platelet-derived growth factor (PDGF) levels were higher in media from the
senescent cells compared to controls. The findings support a causal link between
hepatocyte senescence and liver fibrosis progression through the secretion of
senescence-associated secretory phenotype (SASP) factors. The authors propose
that targeting this pathway could offer new therapeutic strategies for managing
CLDs, emphasizing the importance of understanding cellular mechanisms in liver
pathology.
The article by Charbonnier titled “ATP7B-Deficient
Hepatocytes Reveal the Importance of Protein Misfolding Induced at Low Copper
Concentration” examines the role of ATP7B, a copper transporter, in
hepatocyte function and its implications for Wilson disease (contribution 7).
The study employs CRISPR/Cas9 technology to generate ATP7B-deficient
HepG2/C3a cell lines to explore how these cells respond to copper exposure
compared to their wild-type counterparts. Key findings indicate that ATP7B
deficiency leads to increased sensitivity to copper-induced stress, resulting
in significant protein misfolding and the enhanced expression of heat shock
proteins like HSPA6. The research highlights that low concentrations of copper
trigger oxidative stress responses and activate critical signaling pathways
involving Ang II, EGFR, and ERK1/2, which are linked to liver injury and
fibrosis progression. Furthermore, the study shows that CAV1 can mitigate
APAP-induced lipid accumulation in AFLD by regulating these signaling pathways.
Overall, the findings suggest that targeting the mechanisms related to ATP7B
may provide new therapeutic strategies for managing copper-related liver
diseases such as Wilson’s disease.
In the article “FYB2 Is a Potential
Prognostic Biomarker for Hepatocellular Carcinoma”, authored by Qu and
colleagues, the role of FYN-binding protein 2 (FYB2, C1orf168) as a prognostic
biomarker in HCC is analyzed (contribution 8). The study reveals that FYB2
expression is significantly downregulated in HCC tissues compared to normal
liver tissues. Lower levels of FYB2 correlate with poorer survival outcomes,
advanced tumor grades, and higher pathological stages. Using bioinformatics
analyses from public databases such as TCGA and GEO, the researchers found that
FYB2 can serve as an independent prognostic factor alongside AJCC-M staging.
Gene Set Enrichment Analysis (GSEA) indicated that FYB2 is associated with
cellular metabolism-related pathways and cancer regulation. Single-cell
transcriptome analysis showed that FYB2-positive cells are primarily located in
hepatocytes. Spatial transcriptomics revealed higher FYB2 expression in
adjacent non-tumor areas compared to tumor regions. The findings suggest that targeting
the mechanisms involving FYB2 could provide new therapeutic strategies for HCC.
However, further validation in larger clinical cohorts and the exploration of
the biological mechanisms behind FYB2’s role in tumorigenesis are necessary.
Overall, this study highlights the potential of FYB2 as a valuable biomarker
for prognosis and treatment decision making in patients with HCC.
The contribution “Oxygen Gradient Induced
in Microfluidic Chips Can Be Used as a Model for Liver Zonation” by Ghafoory et
al. presents a novel microfluidic system designed to create oxygen gradients
that mimic the conditions found in liver acini (contribution 9). The study aims
to explore how these gradients affect hepatocyte function, specifically
focusing on the expression of hypoxia-inducible factor 1-alpha (Hif1α) and
albumin. Utilizing interconnected microfluidic chips, the authors established a
controlled flow of media that allowed for the measurement of oxygen levels over
time. They observed a significant reduction in oxygen concentration from inlet
to outlet, which correlated with increased Hif1α expression and decreased
albumin production in HepG2 cells, indicating that even slight changes in
oxygen levels can trigger metabolic responses relevant to liver zonation. The
study also demonstrated that conditioned media from senescent HepG2 cells could
activate HSCs, suggesting a link between hepatocyte senescence and liver
fibrosis progression. Overall, this research provides insights into the
cellular mechanisms underlying liver zonation and offers a promising platform
for studying liver metabolism and pathology in vitro, paving the way for future
investigations into therapeutic strategies for liver diseases.
The article “The Clostridium Metabolite
P-Cresol Sulfate Relieves Inflammation of Primary Biliary Cholangitis by
Regulating Kupffer Cells” by Fu et al. evaluates the role of p-Cresol sulfate
(PCS), a metabolite produced by Clostridium, in alleviating inflammation
associated with primary biliary cholangitis (PBC) (contribution 10). The study
highlights that PCS levels are significantly reduced in PBC patients and animal
models, particularly at advanced stages of the disease. Using both in vivo and
in vitro experiments, the researchers demonstrated that dietary supplementation
with tyrosine, which increases PCS levels, led to decreased liver inflammation
and improved inflammatory cytokine profiles in PBC mice. The findings indicate
that PCS modulates the polarization of hepatic macrophages (Kupffer cells),
shifting them from a pro-inflammatory M1 phenotype to an anti-inflammatory M2
phenotype. The study suggests that PCS could serve as a potential therapeutic
agent for managing PBC by regulating immune responses within the liver.
Additionally, decreased levels of PCS might serve as an early diagnostic marker
for PBC onset. Overall, this research underscores the importance of microbial
metabolites like PCS in liver health and disease management.
The article titled “PNPLA3(I148M) Inhibits
Lipolysis by Perilipin-5-Dependent Competition with ATGL” by Witzel et al.
investigates the role of the Patatin-Like Phospholipase Domain Containing
Protein 3 (PNPLA3) I148M polymorphism in lipid metabolism and its
implications for liver diseases, particularly steatohepatitis (contribution
11). The study reveals that the I148M variant of PNPLA3 negatively
affects lipolysis by competing with adipose triglyceride lipase (ATGL) for
binding to perilipin-5, a protein crucial for lipid droplet (LD) metabolism.
Using a combination of human liver biopsies, mouse models, and cell culture
experiments, the researchers demonstrated that hepatocytes carrying the I148M
variant exhibited increased lipid accumulation and impaired lipolytic activity.
Immunohistochemical analyses showed that PNPLA3 localized to LDs in patients
with steatosis and inflammation, correlating with disease severity. The
findings suggest that the interaction between PNPLA3, perilipin-5, and ATGL is
critical in regulating lipid metabolism in the liver. The authors propose that
targeting this pathway may provide therapeutic opportunities for managing fatty
liver diseases associated with the PNPLA3 polymorphism. Overall, this
study enhances our understanding of how genetic variations impact lipid
metabolism and the progression of liver disease.
In their article titled “Cellular
Senescence in Hepatocellular Carcinoma: The Passenger or the Driver?”, the Cai
research team explores the complex role of cellular senescence in the
progression of HCC (contribution 12). The authors discuss how senescence,
characterized by stable cell cycle arrest, can have dual effects on liver
health, acting as a protective mechanism against tumorigenesis while also
contributing to inflammation and fibrosis that promote cancer development. The
review highlights that senescent hepatocytes secrete a variety of factors known
as SASP, which can influence neighboring cells and drive fibrogenesis. It
emphasizes the importance of understanding how senescent cells interact with
immune cells and other components of the liver microenvironment during HCC
progression. Additionally, the article discusses potential therapeutic
strategies targeting cellular senescence, including inducing or eliminating
senescent cells and modulating SASP factors. The authors argue for further
investigation into specific biomarkers for senescence and its role in drug
resistance to improve treatment outcomes for HCC patients. Overall, this review
underscores that cellular senescence is not merely a consequence but may
actively drive liver carcinogenesis, suggesting new avenues for diagnosis and
therapy in HCC management.
The article titled “Analysis of the Role of
Stellate Cell VCAM-1 in NASH Models in Mice” by Chung et al. analyzes the role
of vascular cell adhesion molecule-1 (VCAM-1) expressed in HSCs during the
development and progression of non-alcoholic steatohepatitis (NASH)
(contribution 13). The researchers utilized two different mouse models to
explore whether VCAM-1 influences liver inflammation, fibrosis, and steatosis.
The study found that while VCAM-1 expression was upregulated in HSCs during
NASH, HSC-specific deletion of VCAM-1 did not significantly affect steatosis,
inflammation, or fibrosis in either model used. This suggests that VCAM-1 on
HSCs is dispensable for NASH development. However, the authors noted that other
adhesion molecules might compensate for the absence of VCAM-1. Overall, the
findings indicate that although VCAM-1 is associated with activated HSCs during
NASH, it does not play a critical role in mediating liver damage or
fibrogenesis in the context of the two experimental models studied. The
research highlights the need for further exploration into other potential
mechanisms and factors involved in NASH pathology.
Finally, the article titled “Sex
Differences Affect the NRF2 Signaling Pathway in the Early Phase of Liver
Steatosis: A High-Fat-Diet-Fed Rat Model Supplemented with Liquid Fructose” by
Di Veroli et al. evaluates how sex differences influence the Kelch-like
ECH-associated protein 1/Nuclear factor erythroid 2-related factor 2
(KEAP1/NRF2) signaling pathway in liver steatosis induced by a high-fat diet
and liquid fructose (contribution 14). The study utilized male and female
Sprague Dawley rats, feeding them either a control diet or a
high-fat–high-fructose diet for three months. Key findings indicate that female
rats exhibited significant hepatic steatosis characterized by increased lipid
levels, while males showed a more resilient metabolic phenotype despite similar
dietary intake. The researchers found that NRF2 expression was upregulated in
males but downregulated in females under dietary conditions, suggesting that
males have a better capacity to cope with oxidative stress through enhanced
autophagy and antioxidant defenses. Furthermore, the study demonstrated that
while both sexes activated autophagic processes, only males displayed proper
autophagic flux. In contrast, females showed impaired responses to endoplasmic
reticulum stress markers and reduced activity of antioxidant proteins such as
NQO1 and HO-1. Overall, this research highlights the importance of considering
sex differences when studying metabolic disorders like metabolic
dysfunction-associated steatotic liver disease (MASLD). The findings suggest
potential pathways for developing targeted therapies that take into account
these differences to improve treatment outcomes for MASLD patients.
3. Concluding Remarks
The collection of articles reviewed
presents a comprehensive exploration of various mechanisms and factors involved
in liver diseases, with a particular focus on conditions such as NAFLD/MASLD,
ALD, HCC, and PBC. Key themes emerge across the studies. Several articles delve
into specific molecular pathways, including the KEAP1/NRF2 axis, which plays a
crucial role in antioxidant defense and cellular responses to oxidative stress.
The dysregulation of this pathway is linked to increased susceptibility to liver
damage and fibrosis. The role of cellular senescence is highlighted as both a
protective mechanism against tumorigenesis and a contributor to chronic
inflammation and fibrosis in the liver. The SASP can influence neighboring
cells, promoting either tissue repair or exacerbating disease progression.
Genetic polymorphisms, such as PNPLA3 (I148M), are shown to significantly
impact lipid metabolism and the progression of fatty liver diseases. These
genetic variations can lead to altered interactions between key proteins
involved in lipolysis, contributing to steatosis and inflammation.
Additionally, several studies emphasize the importance of sex differences in
metabolic responses and disease progression, indicating that males and females
may exhibit distinct biochemical pathways when exposed to similar dietary
challenges or stressors. The potential for targeting specific pathways, such as
inhibiting pro-inflammatory cytokines or modulating autophagy, emerges as a
promising strategy for developing effective treatments for CLDs. Furthermore,
dietary interventions aimed at regulating metabolite levels show potential for
therapeutic benefits.
In conclusion, these articles collectively
underscore the complexity of liver pathophysiology where multiple factors,
including genetics, cellular behavior, environmental influences, and
sex-specific responses, interact to drive disease progression. A deeper
understanding of these mechanisms not only enhances our knowledge of liver
diseases but also paves the way for innovative therapeutic strategies tailored
to individual patient profiles based on their unique biological contexts.
Future research should continue exploring these intricate relationships with an
aim toward personalized medicine approaches in managing liver-related
disorders.
It should be noted that this Topic
attracted a significant number of submissions, indicating that liver fibrosis
remains a prominent subject in Hepatology research. As a result, the publisher
has tasked me with editing a second Topic focused on this field. The new Topic
titled “Signaling Pathways in Liver Disease 2nd Edition” is now accepting
submissions. I warmly invite research articles, reviews, and concise
perspective pieces from experts covering all facets related to this topic.