v
Search
Advanced Search

Publications > Journals > Journal of Clinical and Translational Hepatology > Article Full Text

  • OPEN ACCESS

The Multifaceted Role of Bilirubin in Liver Disease: A Literature Review

  • Mariana M. Ramírez-Mejía1,2 ,
  • Stephany M. Castillo-Castañeda2,3 ,
  • Shreya C. Pal2,4 ,
  • Xingshun Qi5  and
  • Nahum Méndez-Sánchez2,4,*
 Author information
Journal of Clinical and Translational Hepatology   2024

doi: 10.14218/JCTH.2024.00156

Abstract

Bilirubin, the primary breakdown product of hemoproteins, particularly hemoglobin, plays a key role in the diagnosis, prognosis, and monitoring of liver diseases. In acute liver diseases, such as acute liver failure, drug-induced liver injury, and viral hepatitis, bilirubin serves as a biomarker reflecting the extent of hepatocyte loss and liver damage. Chronic liver diseases, including alcohol-related liver disease, chronic hepatitis C virus infection, metabolic dysfunction-associated fatty liver disease, and autoimmune liver diseases, are marked by persistent liver injury and inflammation. Bilirubin levels in chronic liver diseases provide insight into liver function, disease severity, and prognosis. As a versatile biomarker, bilirubin offers valuable information on the pathophysiology of liver diseases and aids in guiding clinical decision-making regarding the treatment of liver diseases and their complications. This review aimed to explore the multifunctional role of bilirubin in liver diseases by analyzing its biological functions beyond its role as a biomarker of liver damage.

Graphical Abstract

Keywords

Bilirubin, Liver failure, Chronic liver failure, Metabolic dysfunction-associated fatty liver disease, Hepatitis, Acute-on-chronic liver failure

Introduction

Bilirubin, the primary bile pigment, is a degradation product of hemoproteins, primarily hemoglobin. Hemoglobin accounts for approximately 80% of daily bilirubin production, which ranges from 250 to 400 mg.1,2 This assessment is particularly important for the early detection, prevention, and prognosis of liver diseases .3 Under normal physiological conditions, total bilirubin (TB) levels in the bloodstream are typically less than 1.2 mg/dL (20.5 µmol/L), with a narrow physiologic range between 0.1 to 1 mg/dL (2–17.1 µmol/L). Circulating TB exists in either unconjugated (UCB) or conjugated (CB) forms.1,2 UCB, which is lipid-soluble, is initially produced in the spleen and bone marrow during the breakdown of hemoglobin from senescent red blood cells. It is then transported to the liver bound to albumin, where it is enzymatically converted to CB by the action of uridine diphosphate-glucuronosyltransferase (UGT1A1).3

The liver is the primary organ involved in bilirubin metabolism, and any alteration in liver function can significantly affect serum bilirubin levels. Consequently, elevated levels of bilirubin in the bloodstream, known as hyperbilirubinemia, can be caused by disruptions in production (e.g., increased red blood cell turnover), metabolism (e.g., altered conjugation), or transport (e.g., impaired uptake or excretion) of either CB or UCB.4,5 Unconjugated hyperbilirubinemia usually occurs in conditions such as hemolytic anemia or ineffective erythropoiesis, where excessive bilirubin production overwhelms the liver’s ability to conjugate bilirubin.6 Additionally, genetic disorders such as Gilbert’s syndrome or Crigler-Najjar syndrome, characterized by deficiencies in the enzyme UGT1A1, further impair bilirubin conjugation, leading to elevated UCB levels.7 Conversely, conjugated hyperbilirubinemia usually reflects a defect in the hepatic excretion of bilirubin. This can be due to intrahepatic causes, such as hepatocellular injury, cholestasis, or cirrhosis, where liver cells are unable to properly excrete CB into bile.8 Alternatively, extrahepatic causes, such as biliary obstruction due to gallstones, tumors, or strictures, may impede bile flow, leading to an accumulation of CB in the blood.9

Chronic liver disease can arise from various causes and, over time, can lead to progressive liver dysfunction that eventually results in cirrhosis.10 The severity of cirrhosis is assessed by a combination of clinical and biochemical parameters, including the presence of ascites or encephalopathy, and levels of serum albumin, prothrombin, and bilirubin.11 Although cirrhosis represents the end stage of chronic liver disease, bilirubin continues to serve as a critical marker in the progression of various liver conditions (Table 1).12–23

Table 1

Scores for determining prognosis and mortality of liver diseases using bilirubin as a parameter

Liver diseaseScore nameYearParametersCut-off pointsReference
Alcoholic HepatitisMaddrey Discriminant Function (MDF)1978Bilirubin concentrations, prothrombin timeMDF ≥ 3210
Glasgow Alcoholic Hepatitis Score (GAHS)1995Bilirubin concentrations, creatinine, PT, ageGAHS ≥ 911
Lille Model2007Bilirubin concentrations, creatinine, age, albumin, MELD, Improvement of bilirubinLille score >0.45 after 7 days of corticosteroid therapy12
ABIC Score (Age, Bilirubin, INR, Creatinine)2006Bilirubin concentrations, INR, Creatinine, AgeABIC > 6.7113
Drug-Induced Liver Injury (DILI)Roussel Uclaf Causality Assessment Method (RUCAM)1993Total bilirubin, ALT/ALP ratio, Previous hepatotoxicity, time to onset<0: Excluded; 1–2: Unlikely; 3–5: Possible; 6–8: Probable; >8: Highly probable14
Acute Liver Failure (ALF)Kings College Criteria for Non-Acetaminophen Related Acute Liver Failure1989INR, age, bilirubin, etiologyNot applicable15
Chronic Liver Disease (CLD)Model for End-Stage Liver Disease (MELD Score)2000Bilirubin, INR, creatinineHigher scores indicating more severe liver dysfunction and a higher risk of mortality.16
MELD-Na Score2016Bilirubin, INR, creatinine, sodiumHigher scores indicating more severe liver dysfunction and a higher risk of mortality.17
MELD 3.02021Bilirubin, INR, creatinine, sodium, age, albuminHigher scores indicating more severe liver dysfunction and a higher risk of mortality.18
Child-Turcotte-Pugh Score1973Presence of ascites, bilirubin, albumin, INR, hepatic encephalopathyClass A: 5–6 points, Class B:7–9 points, Class C: 10–15 points19
Acute-On-ChronicChronic Liver Failure-Sequential Organ Failure (CLIF-SOFA)2013Bilirubin, creatinine, hepatic encephalopathy, mean arterial pressure, INRGrade I: single kidney failure or single failure of the liver, coagulation, circulation, or respiration with creatinine level ranging from 1.5 to 1.9 mg/dL; Grade II: two organ failures; Grade III: three organ failures or more20

Beyond its role as a biomarker, bilirubin possesses significant antioxidant and immunomodulatory properties.24 As an antioxidant, bilirubin neutralizes reactive oxygen species (ROS) and reduces oxidative stress, protecting cells from damage.25 These antioxidant properties are particularly beneficial in conditions characterized by high oxidative stress.26,27 Additionally, the immunomodulatory effects of bilirubin include the inhibition of proinflammatory cytokines and suppression of immune cell activation, which helps mitigate inflammation-induced liver damage.4,28 These protective functions underscore the dual role of bilirubin in liver disease, highlighting not only its diagnostic utility but also its potential therapeutic implications.29

This review aimed to provide a comprehensive analysis of the current literature on bilirubin, exploring its role as both a biomarker and a multifunctional molecule in liver diseases. Furthermore, it evaluated the significance of bilirubin in clinical assessments and its potential in managing complications associated with liver conditions.

Bilirubin metabolism

The first phase of bilirubin metabolism occurs in reticuloendothelial cells, where the enzyme heme oxygenase (HO) catalyzes the breakdown of heme into biliverdin, carbon monoxide, and free iron.30 HO is the rate-limiting enzyme in the first phase of bilirubin metabolism and exists in two main isoforms: HO-1, which is inducible and typically increases in response to stress, and HO-2, which is constitutively expressed.31 HO-1 is highly inducible and responds to a wide range of stressors, including oxidative stress, inflammatory cytokines, growth factors, hormones, and physical stressors (e.g., ischemia/reperfusion injury and hypoxia/hyperoxia).32,33 It is especially sensitive to prooxidant stimuli, such as ultraviolet radiation, iron-containing molecules, and heavy metals, which trigger its upregulation.34

Following heme breakdown by HO, biliverdin is reduced to UCB by the enzyme biliverdin reductase, completing the initial phase of bilirubin metabolism (Fig. 1).1,2,27 UCB, which is hydrophobic and insoluble, is transported through the bloodstream bound to soluble complexes, primarily plasma proteins like albumin (90%) and apolipoprotein D of HDL (10%).1,5,12,27 Once free-circulating UCB and the albumin-binding complex reach the liver, UCB dissociates from albumin. Subsequently, UCB is transported across the basolateral membrane of hepatocytes by two mechanisms: passive diffusion and receptor-mediated facilitated diffusion via the organic anion transporter polypeptide OATP1B1 (alternatively known as liver-specific transporter 1, OATPC, SLC21A6, or OATP2) and OATP1B3.1,2,4 Out of the more than 300 known members of the OATP transporter superfamily, only 11 are found in humans.35 Among these, OATP1B1 and OATP1B3 are liver-specific transporters located in the basolateral membrane of hepatocytes. Notably, OATP1B3 exhibits higher expression around the central vein compared to the periportal zone.6,35

Bilirubin synthesis. After the half-life of erythrocytes, the heme group of hemoglobin is degraded by the enzyme heme oxygenase into carbon monoxide, iron, and biliverdin.
Fig. 1  Bilirubin synthesis. After the half-life of erythrocytes, the heme group of hemoglobin is degraded by the enzyme heme oxygenase into carbon monoxide, iron, and biliverdin.

Subsequently, biliverdin is reduced to unconjugated bilirubin (UCB) in its natural isomeric form, IXα, by the action of biliverdin reductase. NADP, nicotinamide adenine dinucleotide phosphate.

Once UCB reaches the cytosol of the hepatocyte, it undergoes a crucial biochemical process known as glucuronidation, facilitated by the enzyme uridine-diphosphate glucuronosyltransferase (Fig. 2). Although several isoforms of uridine-diphosphate glucuronosyltransferase exist, the primary one responsible for bilirubin glucuronidation is UGT1A1.4 This enzyme is predominantly located in the microsomes (smooth endoplasmic reticulum, rough endoplasmic reticulum), Golgi membranes, and the nuclear envelope.36,37 UGT1A1 catalyzes the esterification of two glucuronic acid molecules to the propionic acid side chains of bilirubin. Under normal conditions, bilirubin diglucuronoside (BDG) is the predominant conjugated form produced. However, when the conjugation system is overwhelmed, a significant portion of bilirubin can be conjugated as bilirubin monoglucuronoside (BMG).38 The conversion of bilirubin to glucuronosides is essential for its effective elimination through the biliary system.27,39 The expression of UGT1A1 is regulated by various multifunctional nuclear receptors, including the constitutive androstane receptor, pregnane X receptor, glucocorticoid receptor, aryl hydrocarbon receptor, and hepatocyte nuclear factor 1α. This regulation occurs through the phenobarbital-responsive enhancer module.40–42

Bilirubin metabolism.
Fig. 2  Bilirubin metabolism.

After about 100–120 days, (1) erythrocytes are removed from circulation to the spleen or liver, where they are broken down into globulins, heme, and iron. (2) In the liver, heme is catalyzed by heme oxygenase (HMOX) and biliverdin reductase (BVR) to form biliverdin and then unconjugated bilirubin (UCB), which primarily circulates bound to albumin. (3) The UCB passes through the sinusoids without albumin and (4) enters the hepatocyte by binding to a transporter protein (OATP1B1) or via passive diffusion, crossing the cell membrane. Inside the cell, it binds to Y and Z proteins and then to ligandin (not shown) for transport to the smooth endoplasmic reticulum (SER). (5) In the SER, bilirubin is conjugated to glucuronic acid by UDP-glucuronosyltransferase 1 (UGT1A1), producing monoglucuronides (BMG) and diglucuronides (BDG) of bilirubin. Finally, (6) conjugated bilirubin (CB) is secreted into the canaliculus by the adenosine triphosphate–binding cassette transporter protein (MRP2).

BDG and BMG are water-soluble conjugates that facilitate bilirubin excretion from hepatocytes. Once formed, they are actively transported across the canalicular membrane into bile via the multidrug resistance-associated protein 2 (MRP2).1,43 MRP2 is an ATP-binding cassette transporter localized in the canalicular membrane of hepatocytes, the apical membrane of enterocytes in the duodenum and jejunum, and the renal proximal tubule epithelial cells.44 MRP2 is the primary mechanism for the bile-independent efflux of BMG, BDG, and other organic anions into the bile canaliculus.45 When the formation and/or uptake of BMG and BDG exceeds the transport capacity of MRP2 in the canalicular membrane, the sinusoidal efflux via MRP3 provides a pathway for compensation.43,46 MRP3, located on the basolateral membrane of hepatocytes, redirects BMG and BDG into the bloodstream, from where they are reabsorbed into hepatocytes by OATP1B1 and OATP1B3. OATP1B1 facilitates unidirectional, high-affinity transport of BMG and BDG across the sinusoidal membrane of hepatocytes, while OATP1B3 specifically transports BMG.47,48

In bile, bilirubin conjugates mix with bile acids, phospholipids, and cholesterol to form mixed micelles, which are transported through the bile ducts into the intestine.49 CB passes through the gastrointestinal tract without being absorbed by the intestinal mucosa and reaches the duodenum.50 In the gut lumen, CB is metabolized by the gut microbiota through two main processes: 1) deconjugation of BMG and BDG molecules and 2) reduction of UCB to urobilinogen.51 Gut bacteria hydrolyze bilirubin glucuronosides, releasing UCB, which is then reduced to urobilinogen by bacteria containing β-glucuronidases, such as Clostridioides difficile, Clostridium ramosum, Clostridium perfringens, and Bacteroides fragilis.52 Urobilinogen can be reduced to stercobilinogen, and both urobilinogen and stercobilinogen can be oxidized to form urobilin and stercobilin, respectively. These metabolites are responsible for the characteristic colors of urine and feces.27 Additionally, urobilinogen can be passively absorbed by the intestine into the portal system.53

Molecular mechanisms of bilirubin in liver diseases

Bilirubin plays a crucial role in the pathophysiology of liver diseases, serving as both a diagnostic marker and a contributor to disease processes. Elevated levels of bilirubin, whether UCB or CB, indicate various forms of liver dysfunction and provide insights into the severity and progression of liver diseases.54 Liver dysfunction can result from various causes, such as viral hepatitis, alcoholic liver disease, metabolic dysfunction-associated steatotic liver disease (MASLD), and autoimmune liver diseases. The liver performs many essential functions, such as metabolizing nutrients, detoxifying harmful substances, and synthesizing vital proteins such as albumin and clotting factors. When the liver is damaged, these functions are impaired, leading to a cascade of physiological disorders.2,55 The main cellular component of the liver is the hepatocyte. Injury to these cells, either through direct toxic effects (such as drug-induced liver injury) or immune-mediated mechanisms (such as viral hepatitis), results in the release of intracellular contents, including bilirubin, into the bloodstream, leading to elevated serum bilirubin levels that reflect the extent of hepatocyte damage.6,56

Altered metabolism

In liver diseases, normal bilirubin metabolism is often disturbed. This can occur at multiple stages: from excessive bilirubin production to defects in its conjugation by UGT1A1 and problems with the secretion of CB into the bile.57 In certain liver conditions, such as cirrhosis and chronic liver disease, increased bilirubin production may occur due to accelerated red blood cell breakdown (hemolysis) or ineffective erythropoiesis.58 This excessive bilirubin production can overwhelm the hepatocytes’ ability to absorb and conjugate UCB, leading to its accumulation in the bloodstream and causing jaundice.59

On the other side, defects in the conjugation of UCB by the UGT1A1 enzyme may be due to genetic variations, as seen in Gilbert’s syndrome and Crigler-Najjar syndrome (CN), or acquired causes.60 In CN type 1 (CN1), there is a complete absence of UGT1A1 activity due to genetic lesions in the UGT1A1 gene, causing a severe form of unconjugated hyperbilirubinemia and a high risk of developing kernicterus, a potentially fatal neurological condition.61 CN1 is typically caused by genetic changes that result in premature truncation or substitution of critical amino acid residues, which completely eliminate the enzyme’s function.62 In CN type 2 (CN2), UGT1A1 activity is partially deficient rather than completely absent, resulting in milder elevations of UCB compared to CN1. CN2 is due to the substitution of individual amino acid residues that markedly reduce, but do not abolish, the catalytic activity of the enzyme. Both types of the syndrome follow an autosomal recessive inheritance pattern.62 In contrast, Gilbert’s syndrome is a much milder condition caused by a partial deficiency of UGT1A1 activity, linked to a specific genetic variation known as the UGT1A1*28 polymorphism (A(TA)7TAA promoter sequence, rs8175347).63 This is the most common inherited cause of unconjugated hyperbilirubinemia and follows an autosomal recessive inheritance pattern, similar to CN1 and CN2. However, in Gilbert’s syndrome, UGT1A1 activity is only slightly reduced, resulting in intermittent, mild elevations of UCB, usually triggered by factors such as fasting, stress, illness, or strenuous exercise.64 In addition to genetic disorders, conditions such as advanced hepatitis or cirrhosis may cause a slight reduction in bilirubin conjugation capacity. However, in these cases, conjugation is usually better preserved than other aspects of bilirubin metabolism.3 Moreover, several drugs, such as pregnanediol, novobiocin, chloramphenicol, gentamicin, and atazanavir, can induce unconjugated hyperbilirubinemia by inhibiting UGT1A1 activity.65

Finally, CB secretion problems in bile can lead to conjugated hyperbilirubinemia. As previously discussed, this step in bilirubin metabolism involves the active transport of CB from hepatocytes into bile canaliculi via specific transporters.1,43 In cholestatic diseases, such as primary biliary cholangitis and primary sclerosing cholangitis, inflammation or scarring of the bile ducts obstructs bile flow, resulting in the retention of CB and bile acids in the liver. This not only causes jaundice but also risks hepatocyte damage due to the toxic accumulation of bile acids.66,67 Genetic disorders, such as Dubin-Johnson syndrome, also illustrate how defects in CB secretion can occur at the transporter level. In this disease, genetic changes in the ABCC2 gene (encoding the MRP2 transporter) prevent the efficient excretion of CB into the bile, leading to the accumulation of CB in hepatocytes and its subsequent release into the bloodstream via MRP3.68 Rotor syndrome is another genetic disorder affecting CB secretion, although it differs from Dubin-Johnson syndrome in its underlying mechanism. In Rotor syndrome, both CB and UCB accumulate in the bloodstream due to defects in hepatic uptake and storage of bilirubin, resulting from genetic variations affecting the OATP1B1 and OATP1B3 transporters.7,69 Beyond genetic conditions, it has been observed that in patients with advanced liver disease, especially of cholestatic origin, there is an upregulation of MRP3 and downregulation of MRP2. This could represent an adaptation to protect hepatocytes from the accumulation of toxic biliary components, including bilirubin.70 Furthermore, the upregulation of MRP3 expression may be related to the inflammatory response triggered in chronic liver diseases.71 Nevertheless, the mechanisms behind this association are not yet fully understood.

Molecular associated pathways

Studies have highlighted bilirubin’s antioxidant, anti-inflammatory, and cytoprotective properties.72–75 The antioxidant properties of bilirubin are closely associated with the biliverdin–bilirubin antioxidant cycle. This cycle, driven by HO-1 and biliverdin reductase, neutralizes ROS and maintains cellular redox balance.76 Bilirubin contains an extensive system of conjugated double bonds and a pair of reactive hydrogen atoms, which are key to its antioxidant properties. It has been proposed that these hydrogen atoms participate in antioxidant activity by donating to free radicals, thereby neutralizing them and preventing oxidative damage.77 In contrast, biliverdin possesses only the conjugated double bond system, and its antioxidant capacity probably comes from the formation of resonance-stabilized structures that can absorb and disperse oxidative energy.77 This regenerative antioxidant cycling between biliverdin and bilirubin provides a unique advantage: unlike many antioxidants that are consumed in the process of neutralizing ROS, bilirubin, through its cycling with biliverdin, can continue to exert protective effects.78 Furthermore, bilirubin inhibits nicotinamide adenine dinucleotide phosphate oxidase activity and acts synergistically with other antioxidants to decrease lipid peroxidation.25,79,80

Additionally, bilirubin influences lipid metabolism by reducing lipogenesis and promoting fatty acid oxidation through peroxisome proliferator-activated receptors.81,82 In glucose metabolism, bilirubin inhibits gluconeogenic enzymes, increases insulin sensitivity by enhancing insulin signaling, and reduces inflammatory mediators.83,84 Moreover, the anti-inflammatory effects of bilirubin are significant in conditions where chronic inflammation is prevalent. It reduces proinflammatory cytokines, suppresses immune cell activation, and inhibits the NF-κB pathway, thereby mitigating inflammation-induced liver damage.28 Finally, bilirubin modulates the activity of nuclear receptors such as the aryl hydrocarbon receptor, constitutive androstane receptor, and pregnane X receptor. These receptors are involved in detoxification and metabolic regulation, influencing the expression of genes associated with lipid and glucose metabolism, and overall energy homeostasis.85

In line with these beneficial effects that may prevent the development of MASLD, it has been observed that individuals with MASLD tend to have low bilirubin concentrations.86 This observation suggests a possible inverse relationship between bilirubin concentrations and the incidence of MASLD, indicating that higher bilirubin concentrations may be associated with a lower risk of developing MASLD.87 It has been proposed that bilirubin metabolism is dysregulated in MASLD patients, likely due to increased oxidative stress.86

Neurotoxicity of bilirubin

Bilirubin neurotoxicity is a significant concern in the pathophysiology of liver diseases, especially in conditions associated with severe hyperbilirubinemia. UCB, in particular, can cross the blood-brain barrier and accumulate in neural tissues, leading to bilirubin encephalopathy.88 This condition, characterized by neurological impairment, occurs when bilirubin levels overwhelm the brain’s capacity to detoxify and eliminate this pigment.89 Studies have shown that bilirubin-induced neurotoxicity results from its ability to interfere with various cellular functions, including neurotransmitter synthesis, release, and uptake, as well as mitochondrial function and energy metabolism.90,91 Bilirubin can alter mitochondrial function, reducing ATP production and increasing oxidative stress in neurons. Bilirubin-induced oxidative stress is a major contributor to neurotoxicity, as UCB accumulation in neuronal tissues generates ROS, which damages cellular components such as lipids, proteins, and DNA. This oxidative damage can trigger cell death pathways, including apoptosis and necrosis.92,93 Additionally, bilirubin can induce an inflammatory response in the brain. Activation of microglia, the resident immune cells of the central nervous system, leads to the production of proinflammatory cytokines, further aggravating neuronal damage. This inflammation can amplify the neurotoxic effects of bilirubin.94,95 Moreover, bilirubin inhibits glutamate uptake by astrocytes, leading to excessive glutamate accumulation in the synaptic cleft. This phenomenon, known as excitotoxicity, leads to overactivation of glutamate receptors in neurons, causing calcium overload and subsequent neuronal injury and death.91,96

Hepatic encephalopathy is a severe neurotoxic complication of liver disease, arising from the liver’s inability to detoxify harmful substances that subsequently accumulate in the bloodstream and impair central nervous system function.97,98 Elevated bilirubin levels have been shown to exacerbate oxidative stress and contribute to neuronal dysfunction, as evidenced by bilirubin’s inhibition of glutamate uptake in astrocytes, which can lead to excitotoxicity and neuronal cell death.96 Nevertheless, the exact role of bilirubin in the pathogenesis of hepatic encephalopathy remains unclear and warrants further investigation.

Gut-liver axis

It has been demonstrated that gut microbiota composition can affect bilirubin metabolism, highlighting the importance of a balanced microbiota for normal bilirubin catabolism.99 A study by Walker et al. showed that a high-fat diet alters gut microbiota and increases urobilinoid levels in obese mice, suggesting a role for bilirubin in lipid metabolism and obesity.100 Further investigations have revealed that specific bacterial populations in the gut are responsible for the deconjugation and reduction of bilirubin. For instance, bacteria such as Clostridium ramosum, Clostridium perfringens, Clostridium difficile, and Bacteroides fragilis play critical roles in transforming bilirubin into urobilinogen and other metabolites.101 Recent research has advanced our understanding of this process by identifying BilR, an intestinal microbial enzyme responsible for the reduction of bilirubin to urobilinogen, highlighting the critical role of microbial metabolism in maintaining bilirubin homeostasis within the gut-liver axis. This enzyme is predominantly encoded by Firmicutes species in the gut microbiome and is considered vital in preventing the accumulation of UCB, which might otherwise be reabsorbed into the bloodstream. Dysregulation of this microbial reduction process can result in elevated serum bilirubin levels.102 While the protective roles of bilirubin and its metabolites are recognized, many mechanisms and functions within the gut-liver axis remain unknown, highlighting the need for further research.

Bilirubin is an integral part of the pathophysiology of liver diseases, serving as both a diagnostic marker and an active participant in pathological processes (Fig. 3). Its role in pathways associated with metabolism, anti-inflammatory responses, regulation of apoptosis and autophagy, and interaction with nuclear receptors highlights its multifaceted nature. Despite its beneficial effects, dysregulation of bilirubin metabolism can lead to neurotoxicity, particularly in conditions of severe hyperbilirubinemia. Understanding the complex molecular mechanisms of bilirubin in liver diseases provides crucial information on its diagnostic and therapeutic potential, underscoring the need for further research to fully exploit its clinical benefits while mitigating the associated risks.4

The dual roles of bilirubin in liver disease.
Fig. 3  The dual roles of bilirubin in liver disease.

In cases of severe hyperbilirubinemia, elevated bilirubin levels cross the blood-brain barrier (BBB) and accumulate in neural tissues. This accumulation leads to the generation of reactive oxygen species (ROS), activation of microglia, alteration of mitochondrial function, and inhibition of glutamate uptake by astrocytes. These processes result in neurotoxic effects such as neuronal damage and excitotoxicity. Conversely, in mild hyperbilirubinemia, bilirubin exerts protective effects. It neutralizes ROS, decreases lipid peroxidation, increases insulin sensitivity, reduces the production of pro-inflammatory cytokines, suppresses immune cell activation, and promotes fatty acid oxidation through the activation of peroxisome proliferator-activated receptors (PPARs). These actions contribute to its antioxidant, anti-inflammatory, and metabolic regulatory roles. The transition from mild to severe hyperbilirubinemia underscores the importance of maintaining bilirubin within a physiological range to harness its protective effects while avoiding neurotoxicity.

Clinical implications

Hyperbilirubinemia, particularly seen in genetic conditions such as Gilbert’s syndrome, CN syndrome, Dubin-Johnson syndrome, and Rotor syndrome, holds significant clinical implications.103 In CN, clinical attention is focused on preventing neurological damage caused by extremely high levels of UCB. Early and aggressive treatment, such as phototherapy or liver transplantation, is essential to prevent complications such as kernicterus.104 In contrast, Dubin-Johnson syndrome and Rotor syndrome, characterized by conjugated hyperbilirubinemia, do not usually pose serious health risks beyond jaundice and, in some cases, fatigue and abdominal pain. In most cases, hyperbilirubinemia is an incidental finding.105 On the other hand, the mild hyperbilirubinemia observed in Gilbert’s syndrome has been associated with cardiovascular and metabolic benefits.84 Studies have shown that individuals with Gilbert’s syndrome may have a reduced risk of cardiovascular disease due to bilirubin’s role as a potent antioxidant, protecting against oxidative stress and low-density lipoprotein oxidation, which is a key factor in the development of atherosclerosis.106,107 Additionally, the protective role of bilirubin extends to metabolic disorders. Elevated levels of UCB have been linked to a reduced risk of developing metabolic syndrome, MASLD, and diabetes.108,109 Interestingly, studies have demonstrated that both acute and chronic oral administration of zinc sulfate significantly reduces serum UCB levels in individuals with Gilbert’s syndrome, likely by inhibiting the normal enterohepatic circulation of UCB.110,111 This suggests potential therapeutic strategies for managing UCB levels in cases where reductions are clinically desired.

Understanding the role of bilirubin in acute and chronic liver diseases has important clinical implications. In acute liver diseases, such as acute liver failure, rapid elevation of bilirubin levels reflects severe hepatocellular injury and decreased liver function.112 Monitoring bilirubin concentrations is crucial for diagnosing acute liver failure, assessing its severity, and guiding therapeutic decisions. Elevated bilirubin levels are integrated into prognostic scoring systems such as the Model for End-Stage Liver Disease (MELD) score and the Maddrey Discriminant Function (Maddrey score), underscoring their importance in predicting patient prognosis and guiding therapeutic decisions.10 The MELD score is widely used to assess the severity of chronic liver disease and prioritize patients for liver transplantation. It incorporates serum bilirubin levels, along with creatinine and the international normalized ratio of prothrombin time, to calculate a score that predicts the three-month mortality risk.21 The Maddrey score is used specifically to assess the prognosis of patients with alcoholic hepatitis, calculated using the patient’s prothrombin time and serum bilirubin levels.13 Both the MELD and Maddrey scores illustrate the enduring importance of bilirubin as a biomarker in liver disease, highlighting its reliability and relevance in assessing liver function, guiding therapeutic decisions, and predicting patient outcomes.113 In chronic liver diseases, such as cirrhosis, hepatitis, and alcohol-related liver diseases, bilirubin concentrations are essential indicators of liver function and disease progression. Elevated bilirubin levels indicate advanced liver dysfunction and are associated with complications such as hepatic encephalopathy.114,115

Bilirubin is also a critical biomarker in the management of drug-induced liver injury, as higher bilirubin concentrations at the onset and peak of drug-induced liver injury are associated with more severe liver injury and worse outcomes.116 In conditions such as metabolic dysfunction-associated fatty liver disease and autoimmune liver diseases, bilirubin provides valuable information on liver function, disease severity, and progression. Interestingly, slightly elevated bilirubin concentrations may have a protective effect against metabolic syndrome and metabolic dysfunction-associated fatty liver disease, suggesting that higher bilirubin levels, particularly indirect bilirubin, may reduce the risk of developing these diseases.86,87,117 Overall, bilirubin is a critical biomarker in several liver diseases, guiding clinical decision-making, assessing treatment efficacy, and predicting patient outcomes. Our recent study on molecular species of bilirubin in acute-on-chronic liver failure (ACLF) has further elucidated the clinical significance of bilirubin. We observed that individuals with ACLF exhibit significantly higher levels of bilirubin diglucuronide and bilirubin monoglucuronide compared to healthy subjects and those with compensated cirrhosis. In addition, we identified distinct bilirubin species profiles that correlate with the severity and prognosis of ACLF.118

Furthermore, hyperbilirubinemia in genetic disorders underscores the importance of understanding bilirubin metabolism for accurate diagnosis and management.

How to assess hyperbilirubinemia?

Direct hyperbilirubinemia is defined as a CB concentration above 1.0 mg/dL (conjugated fraction >50% of TB). Indirect hyperbilirubinemia is considered when UCB comprises >85% of TB.119,120 While unconjugated hyperbilirubinemia can be clinically ambiguous, the presence of conjugated hyperbilirubinemia always suggests liver disease.55,121 Although we have mentioned the importance of bilirubin concentrations in various scales for stratifying disease severity, it is important to note that bilirubin concentrations vary widely across different populations. Ethnicity, age, race, and other potentially confounding variables can make the currently used physiological ranges inaccurate, as noted by Vitek et al.122 Nevertheless, analyzing bilirubin levels in different clinical entities might still provide an indicator of expected ranges, which could be used as diagnostic support after adjusting for confounding factors. Furthermore, the variations in bilirubin concentrations in disease states are greater than what can be attributed to these confounding factors (in contrast to concentrations in healthy individuals). Even if used as a reference interval rather than as a decision value, bilirubin remains a vital biomarker. Understanding its variability in different diseases is fundamental as an initial step.122

Future directions

Looking ahead, several promising avenues of research and clinical application could significantly improve our understanding and management of the dual role of bilirubin in liver disease.123 One promising field is the development of targeted therapies that can selectively enhance the beneficial effects of bilirubin, such as its antioxidant and anti-inflammatory properties, while mitigating its neurotoxic risks. This could include the use of bilirubin analogues or compounds that promote endogenous bilirubin production and stability.29,124 In addition, integrating a holistic approach that combines dietary modifications, lifestyle changes, and pharmacological treatments could improve the treatment of liver diseases.125 For example, dietary interventions that support a healthy gut microbiota could improve bilirubin metabolism and its protective functions.123 Interdisciplinary research efforts that bring together experts in hepatology, neurology, pharmacology, and microbiology will be essential to fully understand the complex mechanisms through which bilirubin operates and to develop comprehensive treatment strategies.

Conclusions

Although bilirubin has traditionally been defined as a waste product, its role in liver metabolism is undisputed. The measurement of bilirubin and its fractions in serum is crucial for diagnosis, prognosis, and treatment evaluation, as its concentration is directly related to liver function. Therefore, liver damage can be detected quickly. As shown in various studies presented in this review, bilirubin concentrations can vary from normal to very high, leading to serious consequences such as organ failure and death. Based on this review, it is important to study specific bilirubin molecular species in different diseases, given that total bilirubin concentrations (without understanding the ratio of its species) do not show clear differences among pathological entities. Continuing to study bilirubin concentrations in various diseases is essential to identify critical cutpoints, develop better treatments, and identify patients who may benefit from early liver transplantation or a life-support system.

Declarations

Funding

None to declare.

Conflict of interest

XQ has been an Editorial Board Member of Journal of Clinical and Translational Hepatology since 2023, NMS has been an Associate Editor of Journal of Clinical and Translational Hepatology since 2021. The other authors have no conflict of interests related to this publication.

Authors’ contributions

Concept and design (NMS, SMCC), drafting of the manuscript (SMCC, SXP, MMRM, XQ), critical revision of the manuscript for important intellectual content (NMS, SMCC, MMRM), and supervision (NMS). All authors have made significant contributions to this study and have approved the final manuscript. All authors have approved the final version and publication of the manuscript.

References

  1. Bhagavan NV, Ha C-E. Essentials of Medical Biochemistry (Second Edition). San Diego: Academic Press; 2015, 511-529
  2. Wang X, Chowdhury JR, Chowdhury NR. Bilirubin metabolism: Applied physiology. Current Paediatrics 2006;16(1):70-74 View Article
  3. Sticova E, Jirsa M. New insights in bilirubin metabolism and their clinical implications. World J Gastroenterol 2013;19(38):6398-6407 View Article PubMed/NCBI
  4. Vitek L, Hinds TD, Stec DE, Tiribelli C. The physiology of bilirubin: health and disease equilibrium. Trends Mol Med 2023;29(4):315-328 View Article PubMed/NCBI
  5. Chowdhury NR, Li Y, Chowdhury JR. The Liver. Wiley; 2020, 229-244
  6. Fevery J. Bilirubin in clinical practice: a review. Liver Int 2008;28(5):592-605 View Article PubMed/NCBI
  7. Strassburg CP. Hyperbilirubinemia syndromes (Gilbert-Meulengracht, Crigler-Najjar, Dubin-Johnson, and Rotor syndrome). Best Pract Res Clin Gastroenterol 2010;24(5):555-571 View Article PubMed/NCBI
  8. Gazzin S, Masutti F, Vitek L, Tiribelli C. The molecular basis of jaundice: An old symptom revisited. Liver Int 2017;37(8):1094-1102 View Article PubMed/NCBI
  9. Liu JJ, Sun YM, Xu Y, Mei HW, Guo W, Li ZL. Pathophysiological consequences and treatment strategy of obstructive jaundice. World J Gastrointest Surg 2023;15(7):1262-1276 View Article PubMed/NCBI
  10. Tsochatzis EA, Bosch J, Burroughs AK. Liver cirrhosis. Lancet 2014;383(9930):1749-1761 View Article PubMed/NCBI
  11. Ginès P, Krag A, Abraldes JG, Solà E, Fabrellas N, Kamath PS. Liver cirrhosis. Lancet 2021;398(10308):1359-1376 View Article PubMed/NCBI
  12. Méndez-Sánchez N, Vítek L, Aguilar-Olivos NE, Uribe M. Biomarkers in Liver Disease. Dordrecht: Springer Netherlands; 2016, 1-25
  13. Maddrey WC, Boitnott JK, Bedine MS, Weber FL, Mezey E, White RI. Corticosteroid therapy of alcoholic hepatitis. Gastroenterology 1978;75(2):193-199 PubMed/NCBI
  14. Forrest EH, Morris AJ, Stewart S, Phillips M, Oo YH, Fisher NC, et al. The Glasgow alcoholic hepatitis score identifies patients who may benefit from corticosteroids. Gut 2007;56(12):1743-1746 View Article PubMed/NCBI
  15. Louvet A, Naveau S, Abdelnour M, Ramond MJ, Diaz E, Fartoux L, et al. The Lille model: a new tool for therapeutic strategy in patients with severe alcoholic hepatitis treated with steroids. Hepatology 2007;45(6):1348-1354 View Article PubMed/NCBI
  16. Dominguez M, Rincón D, Abraldes JG, Miquel R, Colmenero J, Bellot P, et al. A new scoring system for prognostic stratification of patients with alcoholic hepatitis. Am J Gastroenterol 2008;103(11):2747-2756 View Article PubMed/NCBI
  17. Danan G, Benichou C. Causality assessment of adverse reactions to drugs—I. A novel method based on the conclusions of international consensus meetings: application to drug-induced liver injuries. J Clin Epidemiol 1993;46(11):1323-1330 View Article PubMed/NCBI
  18. O’Grady JG, Alexander GJ, Hayllar KM, Williams R. Early indicators of prognosis in fulminant hepatic failure. Gastroenterology 1989;97(2):439-445 View Article PubMed/NCBI
  19. Malinchoc M, Kamath PS, Gordon FD, Peine CJ, Rank J, ter Borg PC. A model to predict poor survival in patients undergoing transjugular intrahepatic portosystemic shunts. Hepatology 2000;31(4):864-871 View Article PubMed/NCBI
  20. Kalra A, Wedd JP, Biggins SW. Changing prioritization for transplantation: MELD-Na, hepatocellular carcinoma exceptions, and more. Curr Opin Organ Transplant 2016;21(2):120-126 View Article PubMed/NCBI
  21. Kim WR, Mannalithara A, Heimbach JK, Kamath PS, Asrani SK, Biggins SW, et al. MELD 3.0: The Model for End-Stage Liver Disease Updated for the Modern Era. Gastroenterology 2021;161(6):1887-1895.e1884 View Article PubMed/NCBI
  22. Pugh RN, Murray-Lyon IM, Dawson JL, Pietroni MC, Williams R. Transection of the oesophagus for bleeding oesophageal varices. Br J Surg 1973;60(8):646-649 View Article PubMed/NCBI
  23. Moreau R, Jalan R, Gines P, Pavesi M, Angeli P, Cordoba J, et al. Acute-on-chronic liver failure is a distinct syndrome that develops in patients with acute decompensation of cirrhosis. Gastroenterology 2013;144(7):1426-1437.e1-19 View Article PubMed/NCBI
  24. Peng Y-F, Goyal H, Xu G-D. Serum bilirubin has an important role in multiple clinical applications. J Lab Precis Med 2017;2:82 View Article
  25. Punzo A, Silla A, Fogacci F, Perillo M, Cicero AFG, Caliceti C. Bile Acids and Bilirubin Role in Oxidative Stress and Inflammation in Cardiovascular Diseases. Diseases 2024;12(5):103 View Article PubMed/NCBI
  26. Vítek L. Bilirubin as a signaling molecule. Med Res Rev 2020;40(4):1335-1351 View Article PubMed/NCBI
  27. Vítek L, Ostrow JD. Bilirubin chemistry and metabolism; harmful and protective aspects. Curr Pharm Des 2009;15(25):2869-2883 View Article PubMed/NCBI
  28. Jangi S, Otterbein L, Robson S. The molecular basis for the immunomodulatory activities of unconjugated bilirubin. Int J Biochem Cell Biol 2013;45(12):2843-2851 View Article PubMed/NCBI
  29. Adin CA. Bilirubin as a Therapeutic Molecule: Challenges and Opportunities. Antioxidants (Basel) 2021;10(10):1536 View Article PubMed/NCBI
  30. Soares MP, Bach FH. Heme oxygenase-1: from biology to therapeutic potential. Trends Mol Med 2009;15(2):50-58 View Article PubMed/NCBI
  31. Bucolo C, Drago F. Focus on molecules: heme oxygenase-1. Exp Eye Res 2009;89(6):822-823 View Article PubMed/NCBI
  32. Chiang SK, Chen SE, Chang LC. The Role of HO-1 and Its Crosstalk with Oxidative Stress in Cancer Cell Survival. Cells 2021;10(9):2401 View Article PubMed/NCBI
  33. Tift MS, Alves de Souza RW, Weber J, Heinrich EC, Villafuerte FC, Malhotra A, et al. Adaptive Potential of the Heme Oxygenase/Carbon Monoxide Pathway During Hypoxia. Front Physiol 2020;11:886 View Article PubMed/NCBI
  34. Vanella L, Barbagallo I, Tibullo D, Forte S, Zappalà A, Li Volti G. The non-canonical functions of the heme oxygenases. Oncotarget 2016;7(42):69075-69086 View Article PubMed/NCBI
  35. Hagenbuch B, Meier PJ. The superfamily of organic anion transporting polypeptides. Biochim Biophys Acta 2003;1609(1):1-18 View Article PubMed/NCBI
  36. Xu B, Gao S, Wu B, Yin T, Hu M. Absolute quantification of UGT1A1 in various tissues and cell lines using isotope label-free UPLC-MS/MS method determines its turnover number and correlates with its glucuronidation activities. J Pharm Biomed Anal 2014;88:180-190 View Article PubMed/NCBI
  37. Rowland A, Miners JO, Mackenzie PI. The UDP-glucuronosyltransferases: their role in drug metabolism and detoxification. Int J Biochem Cell Biol 2013;45(6):1121-1132 View Article PubMed/NCBI
  38. Chowdhury NR, Chowdhury JR, Avsar Y. Emery and Rimoin’s Principles and Practice of Medical Genetics (Sixth Edition). Oxford: Academic Press; 2013, 1-34
  39. Stieger B, Hagenbuch B. Organic anion-transporting polypeptides. Curr Top Membr 2014;73:205-232 View Article PubMed/NCBI
  40. Sugatani J, Sueyoshi T, Negishi M, Miwa M. Regulation of the human UGT1A1 gene by nuclear receptors constitutive active/androstane receptor, pregnane X receptor, and glucocorticoid receptor. Methods Enzymol 2005;400:92-104 View Article PubMed/NCBI
  41. Sugatani J, Kojima H, Ueda A, Kakizaki S, Yoshinari K, Gong QH, et al. The phenobarbital response enhancer module in the human bilirubin UDP-glucuronosyltransferase UGT1A1 gene and regulation by the nuclear receptor CAR. Hepatology 2001;33(5):1232-1238 View Article PubMed/NCBI
  42. Sugatani J, Uchida T, Kurosawa M, Yamaguchi M, Yamazaki Y, Ikari A, et al. Regulation of pregnane X receptor (PXR) function and UGT1A1 gene expression by posttranslational modification of PXR protein. Drug Metab Dispos 2012;40(10):2031-2040 View Article PubMed/NCBI
  43. Keppler D. The roles of MRP2, MRP3, OATP1B1, and OATP1B3 in conjugated hyperbilirubinemia. Drug Metab Dispos 2014;42(4):561-565 View Article PubMed/NCBI
  44. Karpen HE, Karpen SJ. Fetal and Neonatal Physiology (Fourth Edition). Philadelphia: W.B. Saunders; 2011, 1280-1291
  45. Čvorović J, Passamonti S. Membrane Transporters for Bilirubin and Its Conjugates: A Systematic Review. Front Pharmacol 2017;8:887 View Article PubMed/NCBI
  46. Keppler D, Kamisako T, Leier I, Cui Y, Nies AT, Tsujii H, et al. Localization, substrate specificity, and drug resistance conferred by conjugate export pumps of the MRP family. Adv Enzyme Regul 2000;40:339-349 View Article PubMed/NCBI
  47. Cui Y, König J, Leier I, Buchholz U, Keppler D. Hepatic uptake of bilirubin and its conjugates by the human organic anion transporter SLC21A6. J Biol Chem 2001;276(13):9626-9630 View Article PubMed/NCBI
  48. König J, Cui Y, Nies AT, Keppler D. A novel human organic anion transporting polypeptide localized to the basolateral hepatocyte membrane. Am J Physiol Gastrointest Liver Physiol 2000;278(1):G156-164 View Article PubMed/NCBI
  49. Chiang JY. Bile acid metabolism and signaling. Compr Physiol 2013;3(3):1191-1212 View Article PubMed/NCBI
  50. Pratt DS. Sleisenger and Fordtran’s Gastrointestinal and Liver Disease (Ninth Edition). Philadelphia: W.B. Saunders; 2010, 1227-1237.e1222
  51. Vítek L, Tiribelli C. Bilirubin: The yellow hormone?. J Hepatol 2021;75(6):1485-1490 View Article PubMed/NCBI
  52. Hall B, Levy S, Dufault-Thompson K, Ndjite GM, Weiss A, Braccia D, et al. Discovery of the gut microbial enzyme responsible for bilirubin reduction to urobilinogen. bioRxiv 2023 View Article PubMed/NCBI
  53. Vítek L, Majer F, Muchová L, Zelenka J, Jirásková A, Branný P, et al. Identification of bilirubin reduction products formed by Clostridium perfringens isolated from human neonatal fecal flora. J Chromatogr B Analyt Technol Biomed Life Sci 2006;833(2):149-157 View Article PubMed/NCBI
  54. Guerra Ruiz AR, Crespo J, López Martínez RM, Iruzubieta P, Casals Mercadal G, Lalana Garcés M, et al. Measurement and clinical usefulness of bilirubin in liver disease. Adv Lab Med 2021;2(3):352-372 View Article PubMed/NCBI
  55. Agrawal S, Dhiman RK, Limdi JK. Evaluation of abnormal liver function tests. Postgrad Med J 2016;92(1086):223-234 View Article PubMed/NCBI
  56. Rutherford A, Chung RT. Acute liver failure: mechanisms of hepatocyte injury and regeneration. Semin Liver Dis 2008;28(2):167-174 View Article PubMed/NCBI
  57. Memon N, Weinberger BI, Hegyi T, Aleksunes LM. Inherited disorders of bilirubin clearance. Pediatr Res 2016;79(3):378-386 View Article PubMed/NCBI
  58. Gonzalez-Casas R, Jones EA, Moreno-Otero R. Spectrum of anemia associated with chronic liver disease. World J Gastroenterol 2009;15(37):4653-4658 View Article PubMed/NCBI
  59. Fargo MV, Grogan SP, Saguil A. Evaluation of Jaundice in Adults. Am Fam Physician 2017;95(3):164-168 PubMed/NCBI
  60. Bosma PJ. Inherited disorders of bilirubin metabolism. J Hepatol 2003;38(1):107-117 View Article PubMed/NCBI
  61. Strauss KA, Ahlfors CE, Soltys K, Mazareigos GV, Young M, Bowser LE, et al. Crigler-Najjar Syndrome Type 1: Pathophysiology, Natural History, and Therapeutic Frontier. Hepatology 2020;71(6):1923-1939 View Article PubMed/NCBI
  62. Servedio V, d’Apolito M, Maiorano N, Minuti B, Torricelli F, Ronchi F, et al. Spectrum of UGT1A1 mutations in Crigler-Najjar (CN) syndrome patients: identification of twelve novel alleles and genotype-phenotype correlation. Hum Mutat 2005;25(3):325 View Article PubMed/NCBI
  63. Kadakol A, Ghosh SS, Sappal BS, Sharma G, Chowdhury JR, Chowdhury NR. Genetic lesions of bilirubin uridine-diphosphoglucuronate glucuronosyltransferase (UGT1A1) causing Crigler-Najjar and Gilbert syndromes: correlation of genotype to phenotype. Hum Mutat 2000;16(4):297-306 View Article PubMed/NCBI
  64. Vítek L, Tiribelli C. Gilbert’s syndrome revisited. J Hepatol 2023;79(4):1049-1055 View Article PubMed/NCBI
  65. Marques SC, Ikediobi ON. The clinical application of UGT1A1 pharmacogenetic testing: gene-environment interactions. Hum Genomics 2010;4(4):238-249 View Article PubMed/NCBI
  66. Park JW, Kim JH, Kim SE, Jung JH, Jang MK, Park SH, et al. Primary Biliary Cholangitis and Primary Sclerosing Cholangitis: Current Knowledge of Pathogenesis and Therapeutics. Biomedicines 2022;10(6):1288 View Article PubMed/NCBI
  67. Chiang JYL, Ferrell JM. Bile Acid Biology, Pathophysiology, and Therapeutics. Clin Liver Dis (Hoboken) 2020;15(3):91-94 View Article PubMed/NCBI
  68. Kartenbeck J, Leuschner U, Mayer R, Keppler D. Absence of the canalicular isoform of the MRP gene-encoded conjugate export pump from the hepatocytes in Dubin-Johnson syndrome. Hepatology 1996;23(5):1061-1066 View Article PubMed/NCBI
  69. Saxena R. Practical Hepatic Pathology: a Diagnostic Approach (Second Edition). Philadelphia: Elsevier; 2018, 445-464
  70. Ros JE, Libbrecht L, Geuken M, Jansen PL, Roskams TA. High expression of MDR1, MRP1, and MRP3 in the hepatic progenitor cell compartment and hepatocytes in severe human liver disease. J Pathol 2003;200(5):553-560 View Article PubMed/NCBI
  71. Chai J, He Y, Cai SY, Jiang Z, Wang H, Li Q, et al. Elevated hepatic multidrug resistance-associated protein 3/ATP-binding cassette subfamily C 3 expression in human obstructive cholestasis is mediated through tumor necrosis factor alpha and c-Jun NH2-terminal kinase/stress-activated protein kinase-signaling pathway. Hepatology 2012;55(5):1485-1494 View Article PubMed/NCBI
  72. Datla SR, Dusting GJ, Mori TA, Taylor CJ, Croft KD, Jiang F. Induction of heme oxygenase-1 in vivo suppresses NADPH oxidase derived oxidative stress. Hypertension 2007;50(4):636-642 View Article PubMed/NCBI
  73. Ziberna L, Martelanc M, Franko M, Passamonti S. Bilirubin is an Endogenous Antioxidant in Human Vascular Endothelial Cells. Sci Rep 2016;6:29240 View Article PubMed/NCBI
  74. Inoguchi T, Sasaki S, Kobayashi K, Takayanagi R, Yamada T. Relationship between Gilbert syndrome and prevalence of vascular complications in patients with diabetes. Jama 2007;298(12):1398-1400 View Article PubMed/NCBI
  75. Quan S, Kaminski PM, Yang L, Morita T, Inaba M, Ikehara S, et al. Heme oxygenase-1 prevents superoxide anion-associated endothelial cell sloughing in diabetic rats. Biochem Biophys Res Commun 2004;315(2):509-516 View Article PubMed/NCBI
  76. Sedlak TW, Snyder SH. Bilirubin benefits: cellular protection by a biliverdin reductase antioxidant cycle. Pediatrics 2004;113(6):1776-1782 View Article PubMed/NCBI
  77. Stocker R. Antioxidant activities of bile pigments. Antioxid Redox Signal 2004;6(5):841-849 View Article PubMed/NCBI
  78. McDonagh AF. The biliverdin-bilirubin antioxidant cycle of cellular protection: Missing a wheel?. Free Radic Biol Med 2010;49(5):814-820 View Article PubMed/NCBI
  79. Nitti M, Furfaro AL, Mann GE. Heme Oxygenase Dependent Bilirubin Generation in Vascular Cells: A Role in Preventing Endothelial Dysfunction in Local Tissue Microenvironment?. Front Physiol 2020;11:23 View Article PubMed/NCBI
  80. Basuroy S, Bhattacharya S, Leffler CW, Parfenova H. Nox4 NADPH oxidase mediates oxidative stress and apoptosis caused by TNF-alpha in cerebral vascular endothelial cells. Am J Physiol Cell Physiol 2009;296(3):C422-432 View Article PubMed/NCBI
  81. Hinds TD, Adeosun SO, Alamodi AA, Stec DE. Does bilirubin prevent hepatic steatosis through activation of the PPARα nuclear receptor?. Med Hypotheses 2016;95:54-57 View Article PubMed/NCBI
  82. Stec DE, John K, Trabbic CJ, Luniwal A, Hankins MW, Baum J, et al. Bilirubin Binding to PPARα Inhibits Lipid Accumulation. PLoS One 2016;11(4):e0153427 View Article PubMed/NCBI
  83. Zhang F, Guan W, Fu Z, Zhou L, Guo W, Ma Y, et al. Relationship between Serum Indirect Bilirubin Level and Insulin Sensitivity: Results from Two Independent Cohorts of Obese Patients with Impaired Glucose Regulation and Type 2 Diabetes Mellitus in China. Int J Endocrinol 2020;2020:5681296 View Article PubMed/NCBI
  84. Vítek L. The role of bilirubin in diabetes, metabolic syndrome, and cardiovascular diseases. Front Pharmacol 2012;3:55 View Article PubMed/NCBI
  85. Wagner M, Halilbasic E, Marschall HU, Zollner G, Fickert P, Langner C, et al. CAR and PXR agonists stimulate hepatic bile acid and bilirubin detoxification and elimination pathways in mice. Hepatology 2005;42(2):420-430 View Article PubMed/NCBI
  86. Petrtýl J, Dvořák K, Stříteský J, Leníček M, Jirásková A, Šmíd V, et al. Association of Serum Bilirubin and Functional Variants of Heme Oxygenase 1 and Bilirubin UDP-Glucuronosyl Transferase Genes in Czech Adult Patients with Non-Alcoholic Fatty Liver Disease. Antioxidants (Basel) 2021;10(12):2000 View Article PubMed/NCBI
  87. Chang Y, Ryu S, Zhang Y, Son HJ, Kim JY, Cho J, et al. A cohort study of serum bilirubin levels and incident non-alcoholic fatty liver disease in middle aged Korean workers. PLoS One 2012;7(5):e37241 View Article PubMed/NCBI
  88. Pranty AI, Shumka S, Adjaye J. Bilirubin-Induced Neurological Damage: Current and Emerging iPSC-Derived Brain Organoid Models. Cells 2022;11(17):2647 View Article PubMed/NCBI
  89. Rose CF, Amodio P, Bajaj JS, Dhiman RK, Montagnese S, Taylor-Robinson SD, et al. Hepatic encephalopathy: Novel insights into classification, pathophysiology and therapy. J Hepatol 2020;73(6):1526-1547 View Article PubMed/NCBI
  90. Ostrow JD, Pascolo L, Brites D, Tiribelli C. Molecular basis of bilirubin-induced neurotoxicity. Trends Mol Med 2004;10(2):65-70 View Article PubMed/NCBI
  91. Wu C, Jin Y, Cui Y, Zhu Y, Yin S, Li C. Effects of bilirubin on the development and electrical activity of neural circuits. Front Cell Neurosci 2023;17:1136250 View Article PubMed/NCBI
  92. Brito MA, Lima S, Fernandes A, Falcão AS, Silva RF, Butterfield DA, et al. Bilirubin injury to neurons: contribution of oxidative stress and rescue by glycoursodeoxycholic acid. Neurotoxicology 2008;29(2):259-269 View Article PubMed/NCBI
  93. Rawat V, Bortolussi G, Gazzin S, Tiribelli C, Muro AF. Bilirubin-Induced Oxidative Stress Leads to DNA Damage in the Cerebellum of Hyperbilirubinemic Neonatal Mice and Activates DNA Double-Strand Break Repair Pathways in Human Cells. Oxid Med Cell Longev 2018;2018:1801243 View Article PubMed/NCBI
  94. Silva SL, Vaz AR, Barateiro A, Falcão AS, Fernandes A, Brito MA, et al. Features of bilirubin-induced reactive microglia: from phagocytosis to inflammation. Neurobiol Dis 2010;40(3):663-675 View Article PubMed/NCBI
  95. Vaz AR, Falcão AS, Scarpa E, Semproni C, Brites D. Microglia Susceptibility to Free Bilirubin Is Age-Dependent. Front Pharmacol 2020;11:1012 View Article PubMed/NCBI
  96. Silva R, Mata LR, Gulbenkian S, Brito MA, Tiribelli C, Brites D. Inhibition of glutamate uptake by unconjugated bilirubin in cultured cortical rat astrocytes: role of concentration and pH. Biochem Biophys Res Commun 1999;265(1):67-72 View Article PubMed/NCBI
  97. Sharma K, Akre S, Chakole S, Wanjari MB. Hepatic Encephalopathy and Treatment Modalities: A Review Article. Cureus 2022;14(8):e28016 View Article PubMed/NCBI
  98. Häussinger D, Dhiman RK, Felipo V, Görg B, Jalan R, Kircheis G, et al. Hepatic encephalopathy. Nat Rev Dis Primers 2022;8(1):43 View Article PubMed/NCBI
  99. Gustafsson BE, Lanke LS. Bilirubin and urobilins in germfree, ex-germfree, and conventional rats. J Exp Med 1960;112(6):975-981 View Article PubMed/NCBI
  100. Walker A, Pfitzner B, Neschen S, Kahle M, Harir M, Lucio M, et al. Distinct signatures of host-microbial meta-metabolome and gut microbiome in two C57BL/6 strains under high-fat diet. Isme j 2014;8(12):2380-2396 View Article PubMed/NCBI
  101. Fahmy K, Gray CH, Nicholson DC. The reduction of bile pigments by faecal and intestinal bacteria. Biochim Biophys Acta 1972;264(1):85-97 View Article PubMed/NCBI
  102. Hall B, Levy S, Dufault-Thompson K, Arp G, Zhong A, Ndjite GM, et al. BilR is a gut microbial enzyme that reduces bilirubin to urobilinogen. Nat Microbiol 2024;9(1):173-184 View Article PubMed/NCBI
  103. Rossi F, Francese M, Iodice RM, Falcone E, Vetrella S, Punzo F, et al. [Inherited disorders of bilirubin metabolism]. Minerva Pediatr 2005;57(2):53-63 PubMed/NCBI
  104. Tcaciuc E, Podurean M, Tcaciuc A. Management of Crigler-Najjar syndrome. Med Pharm Rep 2021;94(Suppl No 1):S64-s67 View Article PubMed/NCBI
  105. Hay JWW, Levin MJ, Deterding RR, Abzug MJ. Dubin-Johnson Syndrome & Rotor Syndrome. Quick Medical Diagnosis & Treatment Pediatrics. New York, NY: McGraw-Hill Education; 2017
  106. Novotný L, Vítek L. Inverse relationship between serum bilirubin and atherosclerosis in men: a meta-analysis of published studies. Exp Biol Med (Maywood) 2003;228(5):568-571 View Article PubMed/NCBI
  107. Bulmer AC, Blanchfield JT, Toth I, Fassett RG, Coombes JS. Improved resistance to serum oxidation in Gilbert’s syndrome: a mechanism for cardiovascular protection. Atherosclerosis 2008;199(2):390-396 View Article PubMed/NCBI
  108. Nikouei M, Cheraghi M, Ghaempanah F, Kohneposhi P, Saniee N, Hemmatpour S, et al. The association between bilirubin levels, and the incidence of metabolic syndrome and diabetes mellitus: a systematic review and meta-analysis of cohort studies. Clin Diabetes Endocrinol 2024;10(1):1 View Article PubMed/NCBI
  109. Jang BK. Elevated serum bilirubin levels are inversely associated with nonalcoholic fatty liver disease. Clin Mol Hepatol 2012;18(4):357-359 View Article PubMed/NCBI
  110. Méndez-Sánchez N, Martínez M, González V, Roldán-Valadez E, Flores MA, Uribe M. Zinc sulfate inhibits the enterohepatic cycling of unconjugated bilirubin in subjects with Gilbert’s syndrome. Ann Hepatol 2002;1(1):40-43 PubMed/NCBI
  111. Méndez-Sánchez N, Roldán-Valadez E, Flores MA, Cárdenas-Vázquez R, Uribe M. Zinc salts precipitate unconjugated bilirubin in vitro and inhibit enterohepatic cycling of bilirubin in hamsters. Eur J Clin Invest 2001;31(9):773-780 View Article PubMed/NCBI
  112. Bernal W, Auzinger G, Dhawan A, Wendon J. Acute liver failure. Lancet 2010;376(9736):190-201 View Article PubMed/NCBI
  113. Morales-Arráez D, Ventura-Cots M, Altamirano J, Abraldes JG, Cruz-Lemini M, Thursz MR, et al. The MELD Score Is Superior to the Maddrey Discriminant Function Score to Predict Short-Term Mortality in Alcohol-Associated Hepatitis: A Global Study. Am J Gastroenterol 2022;117(2):301-310 View Article PubMed/NCBI
  114. Moon AM, Singal AG, Tapper EB. Contemporary Epidemiology of Chronic Liver Disease and Cirrhosis. Clin Gastroenterol Hepatol 2020;18(12):2650-2666 View Article PubMed/NCBI
  115. Seitz HK, Bataller R, Cortez-Pinto H, Gao B, Gual A, Lackner C, et al. Alcoholic liver disease. Nat Rev Dis Primers 2018;4(1):16 View Article PubMed/NCBI
  116. Fontana RJ, Hayashi PH, Gu J, Reddy KR, Barnhart H, Watkins PB, et al. Idiosyncratic drug-induced liver injury is associated with substantial morbidity and mortality within 6 months from onset. Gastroenterology 2014;147(1):96-108.e104 View Article PubMed/NCBI
  117. Liang C, Yu Z, Bai L, Hou W, Tang S, Zhang W, et al. Association of Serum Bilirubin With Metabolic Syndrome and Non-Alcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2022;13:869579 View Article PubMed/NCBI
  118. Ramírez-Mejía MM, Méndez-Sánchez N. What Is in a Name: from NAFLD to MAFLD and MASLD—Unraveling the Complexities and Implications. Curr Hepatology Rep 2023;22:221-227 View Article
  119. Tafesh ZH, Salcedo RO, Pyrsopoulos NT. Classification and Epidemiologic Aspects of Acute-on-Chronic Liver Failure. Clin Liver Dis 2023;27(3):553-562 View Article PubMed/NCBI
  120. Pievsky D, Rustgi N, Pyrsopoulos NT. Classification and Epidemiologic Aspects of Acute Liver Failure. Clin Liver Dis 2018;22(2):229-241 View Article PubMed/NCBI
  121. Dong V, Nanchal R, Karvellas CJ. Pathophysiology of Acute Liver Failure. Nutr Clin Pract 2020;35(1):24-29 View Article PubMed/NCBI
  122. Vítek L. Bilirubin as a predictor of diseases of civilization. Is it time to establish decision limits for serum bilirubin concentrations?. Arch Biochem Biophys 2019;672:108062 View Article PubMed/NCBI
  123. Hamoud AR, Weaver L, Stec DE, Hinds TD. Bilirubin in the Liver-Gut Signaling Axis. Trends Endocrinol Metab 2018;29(3):140-150 View Article PubMed/NCBI
  124. Luckring EJ, Parker PD, Hani H, Grace MH, Lila MA, Pierce JG, et al. In Vitro Evaluation of a Novel Synthetic Bilirubin Analog as an Antioxidant and Cytoprotective Agent for Pancreatic Islet Transplantation. Cell Transplant 2020;29:963689720906417 View Article PubMed/NCBI
  125. Nobili V, Carter-Kent C, Feldstein AE. The role of lifestyle changes in the management of chronic liver disease. BMC Med 2011;9:70 View Article PubMed/NCBI
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

The Multifaceted Role of Bilirubin in Liver Disease: A Literature Review

Mariana M. Ramírez-Mejía, Stephany M. Castillo-Castañeda, Shreya C. Pal, Xingshun Qi, Nahum Méndez-Sánchez
  • Reset Zoom
  • Download TIFF