¡¡

¡¡

The Role of Bilirubin in Diabetes, Metabolic Syndrome, and Cardiovascular Diseases

¡¡

¡¡

¡¡

Abstract

Bilirubin belongs to a phylogenetically old superfamily of tetrapyrrolic compounds, which have multiple biological functions. Although for decades bilirubin was believed to be only a waste product of the heme catabolic pathway at best, and a potentially toxic compound at worst; recent data has convincingly demonstrated that mildly elevated serum bilirubin levels are strongly associated with a lower prevalence of oxidative stress-mediated diseases. Indeed, serum bilirubin has been consistently shown to be negatively correlated to cardiovascular diseases (CVD), as well as to CVD-related diseases and risk factors such as arterial hypertension, diabetes mellitus, metabolic syndrome, and obesity. In addition, the clinical data are strongly supported by evidence arising from both in vitro and in vivo experimental studies. This data not only shows the protective effects of bilirubin per se; but additionally, of other products of the heme catabolic pathway such as biliverdin and carbon monoxide, as well as its key enzymes (heme oxygenase and biliverdin reductase); thus, further underlining the biological impacts of this pathway. In this review, detailed information on the experimental and clinical evidence between the heme catabolic pathway and CVD, and those related diseases such as diabetes, metabolic syndrome, and obesity is provided. All of these pathological conditions represent an important threat to human civilization, being the major killers in developed countries, with a steadily increasing prevalence. Thus, it is extremely important to search for novel markers of these diseases, as well as for novel therapeutic modalities to reverse this unfavorable situation. The heme catabolic pathway seems to fulfill the criteria for both diagnostic purposes as well as for potential therapeutical interventions.

 

Keywords: bilirubin, biliverdin, biliverdin reductase, cardiovascular diseases, diabetes, heme oxygenase, metabolic syndrome, UGT1A1

¡¡

Introduction

Bilirubin belongs to the superfamily of tetrapyrrolic compounds, which is one of the most highly conserved groups of molecules in nature. Tetrapyrroles have evolved to have pluripotent functions, including light harvesting in plants; as well as other roles in plants as well as in humans: chronobiology, energy generation, transport, and homeostasis of oxygen by hemoglobins and myoglobin ¨C to mention only a few of them. Although for decades bilirubin was believed to be a potentially toxic metabolite of heme catabolism, in particular for central nervous system, data from the last few decades clearly demonstrates that this tetrapyrrolic compound might also have numerous other beneficial effects for the human body, including the scavenging of overproduced reactive oxygen species, anti-inflammatory actions, or direct effects upon cell signaling.

 

Bilirubin is the end product of heme catabolism in the systemic circulation. It is formed by the action of heme oxygenase (HMOX), an enzyme that splits cyclic tetrapyrrole heme into biliverdin, carbon monoxide, and ferrous iron. Biliverdin is subsequently reduced to bilirubin by biliverdin reductase (BLVRA). HMOX exists in two isoforms: HMOX1, a highly inducible isoform responsible for reactions of acute phase immediate response against oxidative stress, and a constitutive isoenzyme HMOX2, playing important roles in the brain and testes. The third member of the HMOX family (HMOX3) has also been described, but it is generally believed that HMOX3 is only represented by a pseudogene, with no coding function (Hayashi et al., 2004). Nevertheless, in light of the recent discoveries of the possible regulating functions of pseudogenes (Pink et al., 2011), it is possible that HMOX3 might also have biological effects by contributing to gene regulation.

 

Bilirubin, as a non-polar molecule, is solubilized in the vascular bed by binding to albumin. When reaching the liver, it is actively transported by the basolateral OATP transporter, solubilized in the cytoplasm by binding to specific binding proteins, and subsequently conjugated by the action of bilirubin UDP-glucuronosyltransferase (UGT1A1) with two molecules of glucuronic acid. Bisglucuronosyl bilirubin is then actively secreted into bile and proceeds into the intestinal lumen, where it undergoes further metabolic changes. From the metabolic point of view, there are several crucial enzymatic steps (in particular those catalyzed by HMOX1, BLVRA, and UGT1A1 enzymes) which play an important role in bilirubin homeostasis with subsequent impacts on the risk of metabolic diseases, including cardiovascular diseases (CVD), diabetes, metabolic syndrome, arterial hypertension, and obesity.

 

Serum bilirubin concentrations are affected by many factors including cigarette smoking, gender, fasting, intake of numerous drugs and/or plant products, altitude, race, and age (for review see V¨ªtek and Schwertner, 2007b). All these factors are likely to influence biological impact of bilirubin on human body.

 

¡¡

Biological Properties of Bilirubin

Bilirubin has been recognized as a substance with potent antioxidant properties. The first report on the antioxidant effects of bilirubin was published as early as 1954 (Bernard et al., 1954), however, it took several more decades until the antioxidant properties of bilirubin attracted major scientific attention (Stocker et al., 1987). Indeed, bilirubin has been shown to be more effective at protecting lipids from oxidation than the water-soluble antioxidants such as glutathione, which primarily protect proteins from oxidation (Sedlak et al., 2009). However, bilirubin has also been demonstrated to be almost 30 times more potent toward the prevention of LDL oxidation compared to a vitamin E analog, Trolox, which represents a lipid-soluble antioxidant substance (Wu et al., 1994). Even more importantly, serum bilirubin has been demonstrated to be a major contributor to the total antioxidant capacity in blood plasma (Frei et al., 1988). Additionally, bilirubin has been proven to have anti-inflammatory properties (for review see V¨ªtek and Schwertner, 2007b). Bilirubin inhibited tumor necrosis factor ¦Á-induced up-regulation of E-selectin, vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule (ICAM-1) in vitro (Mazzone et al., 2009). The negative association between serum bilirubin levels and soluble forms of CD40 ligand and P-selectin which have also been demonstrated in in vivo study on subjects with Gilbert syndrome (Tapan et al., 2009). Bilirubin has also been shown to significantly interfere with the complement system, with all of the possible protective consequences (reviewed in Basiglio et al., 2010). Consistent with this data, several studies have demonstrated a negative relationship between serum bilirubin and C-reactive protein levels (V¨ªtek et al., 2007a; Hwang et al., 2011; Yoshino et al., 2011). In addition, the modulatory effects of bilirubin on T regulatory cell differentiation were recently reported (Rocuts et al., 2010), further underlining the protective role of bilirubin in the pathogenesis of chronic inflammatory as well as in autoimmune conditions.

 

¡¡

Negative Association between Bilirubin and Cardiovascular Diseases

The first report of a negative relationship between serum bilirubin levels and coronary artery disease was published as early as 1994 (Schwertner et al., 1994). Since then, numerous studies have been published, which have consistently demonstrated that subjects with lower bilirubin levels are at increased risk of both coronary and peripheral atherosclerotic disease (for reviews see recent papers; V¨ªtek and Schwertner, 2007b; Schwertner and V¨ªtek, 2008; V¨ªtek and Ostrow, 2009; Lin et al., 2010). Serum bilirubin concentrations were also found to be negatively related to coronary artery calcification (Tanaka et al., 2009), and also to ischemic stroke (Kimm et al., 2009); all consistent with our own data which demonstrated a marked postponement in the progression of the intimo-medial thickness of carotid arteries in hyperbilirubinemic subjects with Gilbert syndrome, when compared to normobilirubinemic individuals (V¨ªtek et al., 2006). In addition, our meta-analysis of studies focused on the association between CVD and bilirubin, involving almost 15,000 men, showed that each 1.0¦Ìmol/L increase in serum bilirubin was associated with a 6.5% decrease in CVD risk (Novotný and V¨ªtek, 2003). The same association was also reported in a recent Taiwanese prospective study on patients with cardiac X syndrome followed for 5years, in which patients with the lowest serum bilirubin levels had a higher incidence of non-fatal myocardial infarction, ischemic stroke, rehospitalization for unstable angina, and coronary revascularization procedures (Huang et al., 2010).

 

Logically, the opposite relationship is also true, i.e., subjects with moderately elevated levels of serum bilirubin, such as those with Gilbert syndrome, are at decreased risk of CVD (V¨ªtek et al., 2002).

 

Although the association of serum bilirubin levels and CVD has been known for almost two decades, the relationship between bilirubin and other players of the heme catabolic pathway, as well as pathologic conditions such as metabolic syndrome and/or diabetes has only been investigated in the last few years.

 

¡¡

Heme Oxygenase and Diabetes ¨C Experimental Evidence

Diabetes mellitus, the hallmark of which consists of elevated plasma glucose, is consistently associated with increased oxidative stress, as well as enhanced formations of advanced glycation end products (AGEs; Yamagishi et al., 2011). Overproduction of oxidizing molecules results in the progressive loss of pancreatic ¦Â-cells, depleting insulin levels. However, also numerous other mechanisms contributing to the pathogenesis of diabetes play an important pathogenic role, including reduction of the adiponectin levels (Soares et al., 2005), interference with insulin signaling, endothelial dysfunction, or direct damages of the endothelial wall (Unoki and Yamagishi, 2008; Potenza et al., 2009; Giacco and Brownlee, 2010). Thus, it is not surprising that HMOX1, as a key antioxidant enzyme, has consistently been shown to protect from the development of diabetes (for review see Ndisang, 2010). The mechanisms by which HMOX1 mediated these effects are pluripotent, and apart from the antioxidant action, HMOX1 also directly affects glucose metabolism, which might be due to the presence of a binding site for a glucocorticoid-responsive element in the HMOX1 gene promoter (Lavrovsky et al., 1996). In fact, HMOX1 was shown to stimulate insulin production in experimental animal models (Ndisang, 2010), at least partially, via the release of carbon monoxide (Henningsson et al., 1999), which is an immediate product of HMOX. Indeed, HMOX1 induction by hemin has been demonstrated to improve insulin signaling and glucose metabolism, and to have lowered insulin resistance in various animal models, including those of streptozotocin-induced diabetes (Ndisang and Jadhav, 2009a), insulin-resistant type 2 diabetes (Ndisang and Jadhav, 2009b), primary hyperaldosteronism (Ndisang and Jadhav, 2010a), as well as essential hypertension (Ndisang et al., 2010b). Apart from an improvement of insulin sensitivity, HMOX1 induction was also reported to reduce visceral and subcutaneous obesity in diabetic and obese mice (Li et al., 2008; Nicolai et al., 2009; Burgess et al., 2010) through mechanisms involving the attenuation of the inflammatory processes, as well as modulation of PPAR¦Ã signaling (Ndisang, 2010). Furthermore, chronic HMOX1 induction was shown to increase metabolic turnover, heat production, and physical activity in an experimental model of obesity (Csongradi et al., 2012), suggesting the complexity of the beneficial effects of HMOX1 induction upon these metabolic disorders.

 

¡¡

Heme Oxygenase and Diabetes ¨C Clinical Evidence

Experimental data on the role of HMOX in the pathogenesis of diabetes, metabolic syndrome, and obesity are also supported by the clinical evidence. Plasma concentrations of HMOX1 have been demonstrated to strongly correlate with type 2 diabetes mellitus (Bao et al., 2010b). In fact, subjects in the highest quartile of HMOX1 plasma levels had more than an 8X higher risk for development of type 2 diabetes, compared to the lowest quartile (Bao et al., 2010b). In this study, the HMOX1 plasma level might reflect increased oxidative stress preceding the expression of diabetes. In addition, it is known that high glucose exposure leads to HMOX1 induction (Jonas et al., 2003; Won et al., 2006), which also might account for the observed phenomenon. On the other hand, the same authors recently published a meta-analysis of six studies focused on the role of HMOX1 promoter gene variants and the risk of diabetes, covering almost 2,000 diabetic patients and 3,500 controls. These results revealed that subjects carrying longer (GT)n repeats in the HMOX1 gene promoter (associated, nevertheless, with decreased HMOX1 activity), had an increased risk of type 2 diabetes (Bao et al., 2010a). In addition to the effect on diabetes risk, diabetic subjects with the L/L genotype had an almost threefold increase in CVD risk, even after controlling for conventional risk factors in one of the studies involved in the reported meta-analysis (Chen et al., 2008). Further detailed clinical studies are needed to elucidate all of the mechanisms related to the roles of HMOX in the risks of development of diabetes.

 h

¡¡

Biliverdin Reductase and Diabetes ¨C Experimental Evidence and Possible Clinical Implications

Biliverdin reductase, similarly as HMOX, is an evolutionarily conserved enzyme, responsible for the hydrogenation of biliverdin on bilirubin. Besides that, BLVRA has multiple other functions affecting cell signaling, modulating immune system response, and also exerting substantial metabolic effects (Maines, 2005; Kapitulnik and Maines, 2009; Wegiel et al., 2011). BLVRA also serves as a unique serine/threonine/tyrosine kinase, significantly modulating (among other molecules) the phosphorylation of the insulin receptor substrate-1, as well as other insulin/IGF-1 signaling pathway targets (Kapitulnik and Maines, 2009). As a result, BLVRA diminishes insulin signaling, and acts as a negative regulator of glucose uptake (Lerner-Marmarosh et al., 2005). BLVRA also binds to the p85 regulatory subunit of PI3K/Akt (Pachori et al., 2007), a mechanism which might also lead to improved insulin sensitivity (Terauchi et al., 1999). Based upon this data, small BLVRA-based peptides which activate the insulin receptor signaling axis have been suggested as possible therapeutics to combat insulin resistance in diabetic patients (Lerner-Marmarosh et al., 2008).

 

Not only BLVRA itself, but also the products of its basic enzymatic activity, biliverdin (and consequently bilirubin), might significantly contribute to protection from diabetic complications. Oral administration of biliverdin to db/db mice partially prevented a worsening of hyperglycemia and glucose intolerance (Ikeda et al., 2011), as well as diabetic nephropathy in these animals, principally via inhibition of oxidative stress-induced damage, mediated by the interference with the NADPH oxidase pathway (Fujii et al., 2010). Biliverdin-treated animals had less albuminuria, and complete protection against the progression of mesangial expansion, accompanied by normalization of transforming growth factor-¦Â1 and the expression of fibronectin (Fujii et al., 2010).

 

There are no clinical data on the role of BLVRA/biliverdin in diabetes, metabolic syndrome, or obesity. Only recently, a paper describing the effect of a BLVRA gene variant (rs699512) on the risk of essential hypertension has just been reported (Lin et al., 2011). Detailed studies are certainly needed to fully elucidate the clinical relevance of the experimental findings discussed.

 

¡¡

Bilirubin and Diabetes ¨C Experimental Evidence

It is not surprising that bilirubin, a subsequent reduction product of biliverdin, also has salutary effects in terms of the prevention of diabetes mellitus and its complications. In addition, bilirubin was also reported to provide protection against metabolic syndrome, and to be negatively associated with overweight and obesity.

 

In fact, in a manner similar to biliverdin substantially improving renal pathology in db/db mice, specifically via inhibition of NADPH oxidase (as discussed above), a practically identical effect was observed in hyperbilirubinemic Gunn rats exposed to streptozocin (Fujii et al., 2010). Compared to their diabetic normobilirubinemic heterozygous siblings, diabetic Gunn rats developed less albuminuria, exhibited practically normal urinary levels of 8-hydroxy-2¡ä-deoxyguanosine (8-OHdG), as well as 8-epi-prostaglandin F2¦Á levels; both markers of systemic oxidative stress. Additionally, expression of NADPH oxidase was markedly attenuated in the kidneys of diabetic Gunn rats, who also exhibited complete inhibition of mesangial expansion when compared to normobilirubinemic diabetic controls (Fujii et al., 2010). Consistent with this data, Korean researchers reported substantial resistance of hyperbilirubinemic Gunn rats to developing diabetes after intraperitoneal exposure to streptozocin, compared to their normobilirubinemic littermates (Fu et al., 2010). Markers of diabetes, such as fasting blood glucose and HbA1c levels were much less pronounced in Gunn rats; on the other hand, insulin secretion by the pancreatic islets was preserved in hyperbilirubinemic animals. In addition, concentrations as low as 1.7¦Ìmol/L bilirubin prevented streptozocin-induced apoptosis in rat insulinoma cell culture; significantly supporting the in vivo data (Fu et al., 2010).

 

¡¡

Bilirubin, Diabetes, Metabolic Syndrome, and Obesity ¨C Clinical Evidence

Negative associations between serum bilirubin concentrations and abnormal glucose tolerance tests were reported as early as 1996 (Ko et al., 1996). Since then, numerous clinical studies evaluating the possible associations between serum/plasma bilirubin levels and the risk of diabetes (and its complications) have been published. Inoguchi et al. (2007) first indicated that diabetic patients who have concomitant Gilbert syndrome have a lower prevalence rate of vascular complications, compared to normobilirubinemic diabetics. Although not specifically mentioned in their study, the prevalence of Gilbert syndrome among more than 5,000 consecutive diabetics was at least three times lower than would be expected in the general population. This data is in line with our observation of low prevalence of diabetes in subjects with (TA)7 promoter variation in UGT1A1 responsible for systemic bilirubin homeostasis (Jir¨¢skov¨¢ et al., 2011), and with low levels of AGEs in Gilbert syndrome subjects, compared to the normobilirubinemic population (Kalousov¨¢ et al., 2005). Furthermore, low serum bilirubin levels were identified as an important predictor of CVD in Type 2 diabetes patients receiving haemodialysis (Fukui et al., 2011). Just recently, another group of Japanese investigators reported in their cross-sectional study on more than 3,000 participants, that compared to subjects within the lowest bilirubin quartile, those with the highest bilirubin levels had a four times lower prevalence of diabetic retinopathy (Yasuda et al., 2011). In a large Korean cross-sectional study on almost 94,000 subjects, high serum bilirubin was also found to be associated with the reduced risk of diabetes mellitus and diabetic nephropathy (Han et al., 2010). Higher serum bilirubin levels were also shown to protect from diabetes mellitus in the US National Health and Nutrition Examination Survey (NHANES) on almost 16,000 subjects. In this study, subjects with a bilirubin level above 10¦Ìmol/L had 20% less risk of developing diabetes, compared to those with bilirubin levels below this cut-off value, even after multiple adjustments (Cheriyath et al., 2010).

 

Similarly, additional studies have supported the protective effects of bilirubin on both clinical and laboratory outcomes of diabetic subjects. Serum bilirubin concentrations were demonstrated to be negatively associated with albuminuria in patients with type 2 diabetes (Fukui et al., 2008), HbA1c levels (being an independent risk factor of CVD) in Japanese populations (Oda, 2010; Oda and Kawai, 2010; Ohnaka et al., 2010); additionally, with both insulin resistance and prevalence rate of the metabolic syndrome among children and adolescents (Lin et al., 2009a), as well as in adult populations (Kwon et al., 2011; Wu et al., 2011). Consistent with this data, low prevalence of metabolic syndrome in subjects with phenotypic Gilbert syndrome was described in a recent large Korean study on more than 12,000 participants (Choi et al., 2011).

 

In line with these negative relationships between serum bilirubin levels and metabolic syndrome, a negative association between bilirubin levels and abdominal obesity per se has been shown in several recent studies (Bhuiyan et al., 2008; Lin et al., 2009a; Choi et al., 2011; Kwon et al., 2011; Wu et al., 2011). Since weight reduction is known to reduce several cardiovascular risk factors, it is important to note that each one percent decrease in weight loss was associated with a linear increase in serum bilirubin concentration (Andersson et al., 2009). The close relationship between serum bilirubin levels and the UGT1A1 gene promoter variants, responsible for manifestation of Gilbert syndrome in Asians, and the risk of non-alcoholic fatty liver disease (a condition commonly associated with obesity and metabolic syndrome) was recently demonstrated in Taiwanese children (Lin et al., 2009b); further underlining the importance of the heme catabolic pathway in the pathogenesis of obesity, metabolic syndrome, diabetes mellitus and its complications.

 

¡¡

Pharmacologic and/or Nutraceutic Interventions

Interventions which induce HMOX1 and/or BLVRA, or those that inhibit UGT1A1 activities might be effective in increasing serum and tissue bilirubin concentrations, as well as in modulating signaling pathways involved in energy homeostasis.

 

Heme oxygenase 1, as a highly inducible enzyme, which can be upregulated by many common drugs routinely used in clinical medicine (Bach, 2005), including non-steroidal anti-inflammatory drugs, hypolipidemics (Muchov¨¢ et al., 2007; Wu et al., 2012), PPAR¦Á agonists, certain immunosuppressive drugs, to mention just a few. It is likely that at least part of their therapeutic action might be mediated via induction of HMOX1. The therapeutic potential of HMOX1 induction is also supported by recent experimental data demonstrating that chronic HMOX1 induction lowers body weight, corrects hyperglycemia and hyperinsulinemia, as well as increases oxygen consumption, heat production, and activity in obese mice (Csongradi et al., 2012).

 

Based on recent data, it also becomes evident that BLVRA might have great therapeutic implications (Maines, 2010). In fact, small residue peptides designed according to the primary structure of BLVRA were shown to block important pathways implicated in the pathogenesis of diabetes, including the inhibition of the MAPK (Lerner-Marmarosh et al., 2008) and TNF-¦Á/PKC ¦Æ/NF-¦ÊB pathways (Lerner-Marmarosh et al., 2007), as well as the inhibition of activation and membrane translocation of PKC-¦ÂII (Maines et al., 2007). There are also pharmaceuticals capable of a partial inhibition of UGT1A1, which in turn result in a mild elevation of systemic bilirubin levels. Among the substances having UGT1A1 inhibiting activity is a uricosuric drug probenecid (McCarty, 2007), and also the antiviral drug atazanavir (Dekker et al., 2011). In fact, atazanavir has been demonstrated to improve endothelial function in patients with type 2 diabetes mellitus, most likely due to its bilirubin-increasing activities (Dekker et al., 2011).

 

However, the heme catabolic pathway might also be potentiated through non-pharmacologic means, and its salutary effects can, for instance, be mimicked by tetrapyrollic compounds widely occurring in plants and algae. There are numerous natural HMOX1 inducers originating from plants, including polyphenols, but also compounds such as curcumin, or silymarin (Bonifaz et al., 2009). Indeed, curcumin was reported to increase insulin secretion from rat-isolated pancreatic islets via HMOX1 induction (Abdel Aziz et al., 2010), as well as to improve clinical and laboratory markers of experimental diabetes (Gutierres et al., 2011; Soetikno et al., 2011); the same effect was also demonstrated for silymarin-treated diabetic rats (Vessal et al., 2010). Although the action of both compounds is certainly multifactorial, HMOX1 induction seems to play an important role in their anti-diabetic effects.

 

It is also tempting to speculate that oral supplementation with nutraceuticals containing plant tetrapyrolles, such as phycocyanobilins (Lee et al., 2008; Moura et al., 2011), or their increased consumption in the form of natural foods (Tonstad et al., 2009) might be used as a novel approach of the chemoprevention of obesity, metabolic syndrome, and diabetes.

 

¡¡

Concluding Remarks

In this review, the experimental as well as clinical studies on the associations between CVD, diabetes mellitus, metabolic syndrome, and obesity with the heme catabolic pathway have been discussed. These studies unanimously show that low serum bilirubin concentrations are associated with an increased risk of these pathologic conditions; whereas, mildly elevated serum bilirubin levels provide protection. Moreover, it is clear that apart from the direct beneficial effects of bile pigments, the enzymes HMOX1 and BLVRA also play an important role in the development of these metabolic diseases. Based on recent data, it seems that there is a therapeutical potential to modulate HMOX1, BLVRA, and UGT1A1 activities, and/or mildly increase bilirubin levels (or its analogs) in the systemic circulation to suppress pathways leading to the development of diabetes and its complications. Obviously, detailed studies to explore this potential further appear warranted, and will definitely provide much needed evidence that the whole heme catabolic pathway is also likely beneficial for this group of metabolic diseases.

¡¡

¡¡

Front Pharmacol. 2012; 3: 55.

Published online 2012 Apr 3. doi:  [10.3389/fphar.2012.00055]

PMCID: PMC3318228

PMID: 22493581

¡¡

Libor V¨ªtek1,*

Author information Article notes Copyright and License information Disclaimer

This article has been cited by other articles in PMC.

¡¡

¡¡

Conflict of Interest Statement

The Role of Bilirubin in Diabetes, Metabolic Syndrome, and Cardiovascular Diseases  https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3318228/

¡¡

¡¡

¡¡

Role of bilirubin in the prevention of cardiovascular diseases and cancer

¡¡

Format: AbstractSend to

Cas Lek Cesk. 2016;155(2):10-4.

¡¡

[Article in Czech]

V¨ªtek L.

Abstract

Oxidative stress contributes importantly to pathogenesis of numerous civilization diseases, including cardiovascular diseases, cancer, as well as autoimmune and neurodegenerative conditions. Bilirubin is the major product of the heme catabolic pathway in the intravascular compartment. For long time, bilirubin was considered to be only a waste product, however, recent data from the last two decades have proved its important protective properties, which contributes to defense against development of civilization diseases. Numerous experimental as well as clinical studies have demonstrated association between low bilirubin concentrations and cardiovascular diseases, as well as certain cancers. On the other hand, subjects with mildly elevated blood bilirubin levels, typical for Gilbert syndrome, have decreased risk of these diseases.

 

KEYWORDS:

Gilbert syndrome; antioxidant; bilirubin; cancer; cardiovascular diseases; oxidative stress.

PMID: 27088786

[Role of bilirubin in the prevention of cardiovascular diseases and cancer]. - PubMed - NCBI  https://www.ncbi.nlm.nih.gov/pubmed/27088786

¡¡

¡¡

¡¡

Trends Mol Med. 2016 Sep;22(9):758-768. doi: 10.1016/j.molmed.2016.07.004. Epub 2016 Aug 8.

A Novel Perspective on the Biology of Bilirubin in Health and Disease.

Gazzin S1, Vitek L2, Watchko J3, Shapiro SM4, Tiribelli C5.

Author information

Abstract

Unconjugated bilirubin (UCB) is known to be one of the most potent endogenous antioxidant substances. While hyperbilirubinemia has long been recognized as an ominous sign of liver dysfunction, recent data strongly indicate that mildly elevated bilirubin (BLB) levels can be protective against an array of diseases associated with increased oxidative stress. These clinical observations are supported by new discoveries relating to the role of BLB in immunosuppression and inhibition of protein phosphorylation, resulting in the modulation of intracellular signaling pathways in vascular biology and cancer, among others. Collectively, the evidence suggests that targeting BLB metabolism could be considered a potential therapeutic approach to ameliorate a variety of conditions.

 

KEYWORDS:

bilirubin; bilirubinomics; human health; intracellular events; redox state

PMID: 27515064 DOI: 10.1016/j.molmed.2016.07.004

[Indexed for MEDLINE]

A Novel Perspective on the Biology of Bilirubin in Health and Disease. - PubMed - NCBI  https://www.ncbi.nlm.nih.gov/pubmed/27515064

¡¡

¡¡

¡¡

Biochimie. 2012 Aug;94(8):1821-7. doi: 10.1016/j.biochi.2012.04.026. Epub 2012 May 10.

Intracellular accumulation of bilirubin as a defense mechanism against increased oxidative stress.

Zelenka J1, Muchova L, Zelenkova M, Vanova K, Vreman HJ, Wong RJ, Vitek L.

Author information

1

Institute of Physiology, Academy of Sciences, Dept. 75, Videnska 1083, Prague 142 20, Czech Republic. jar.zelenka@gmail.com

Abstract

Antioxidant, anti-inflammatory and anti-atherogenic effects have been associated with elevations of unconjugated bilirubin (UCB) in serum and with the induction of heme oxygenase-1 (HO-1), the rate-limiting enzyme in UCB synthesis. The aim of this study was to investigate the intracellular metabolism and antioxidant properties of UCB in human hepatoblastoma HepG2 cells and tissues of Wistar rats exposed to oxidative stressors and lipopolysaccharide (LPS), respectively. Intracellular UCB concentrations in HepG2 cells correlated with its levels in culture media (p < 0.001) and diminished lipid peroxidation in a dose-dependent manner (p < 0.001). Moreover, induction of HO-1 with sodium arsenite led to 2.4-fold (p = 0.01) accumulation of intracellular UCB over basal level while sodium azide-derived oxidative stress resulted in a 60% drop (p < 0.001). This decrease was ameliorated by UCB elevation in media or by simultaneous induction of HO-1. In addition, hyperbilirubinemia and liver HO-1 induction in LPS-treated rats resulted in a 2-fold accumulation of tissue UCB (p = 0.01) associated with enhanced protection against lipid peroxidation (p = 0.02). In conclusion, hyperbilirubinemia and HO-1 induction associated with inflammation and oxidative stress increase intracellular concentrations of UCB, thus enhancing the protection of cellular lipids against peroxidation. Therefore, the previously reported protective effects of hyperbilirubinemia and HO-1 induction are at least in part due to intracellular accumulation of UCB.

 

PMID: 22580386 DOI: 10.1016/j.biochi.2012.04.026

Intracellular accumulation of bilirubin as a defense mechanism against increased oxidative stress. - PubMed - NCBI  https://www.ncbi.nlm.nih.gov/pubmed/22580386

¡¡

¡¡

¡¡

Heme Oxygenase-1 May Affect Cell Signalling via Modulation of Ganglioside Composition.

¡¡

¡¡

Oxid Med Cell Longev. 2018 Sep 19;2018:3845027. doi: 10.1155/2018/3845027. eCollection 2018.

¡¡

Abstract

Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Gangliosides, sialic acid-containing glycosphingolipids expressed in all cells, are involved in cell recognition, signalling, and membrane stabilization. Their expression is often altered under many pathological and physiological conditions including cell death, proliferation, and differentiation. The aim of this study was to assess the possible role of Hmox1 in ganglioside metabolism in relation to oxidative stress. The content of liver and brain gangliosides, their cellular distribution, and mRNA as well as protein expression of key glycosyltransferases were determined in Hmox1 knockout mice as well as their wild-type littermates. To elucidate the possible underlying mechanisms between Hmox1 and ganglioside metabolism, hepatoblastoma HepG2 and neuroblastoma SH-SY5Y cell lines were used for in vitro experiments. Mice lacking Hmox1 exhibited a significant increase in concentrations of liver and brain gangliosides and in mRNA expression of the key enzymes of ganglioside metabolism. A marked shift of GM1 ganglioside from the subsinusoidal part of the intracellular compartment into sinusoidal membranes of hepatocytes was shown in Hmox1 knockout mice. Induction of oxidative stress by chenodeoxycholic acid in vitro resulted in a significant increase in GM3, GM2, and GD1a gangliosides in SH-SY5Y cells and GM3 and GM2 in the HepG2 cell line. These changes were abolished with administration of bilirubin, a potent antioxidant agent. These observations were closely related to oxidative stress-mediated changes in sialyltransferase expression regulated at least partially through the protein kinase C pathway. We conclude that oxidative stress is an important factor modulating synthesis and distribution of gangliosides in vivo and in vitro which might affect ganglioside signalling in higher organisms.

¡¡

Šm¨ªd V1,2, Šuk J1, Kachamakova-Trojanowska N3,4, Jašprov¨¢ J1, Val¨¢škov¨¢ P1, J¨®zkowicz A3, Dulak J3, Šm¨ªd F1, V¨ªtek L1,2, Muchov¨¢ L1.

Author information

1

Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine and General University Hospital in Prague, Charles University, Katerinska 32, 12108 Prague, Czech Republic.

2

4th Department of Internal Medicine, 1st Faculty of Medicine and General University Hospital in Prague, Charles University, U Nemocnice 499/2, 12801 Prague, Czech Republic.

3

Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Krakow, Poland.

4

Malopolska Centre for Biotechnology, Jagiellonian University, Gronostajowa str 7a, 30-387 Krakow, Poland.

¡¡

¡¡

PMID: 30327713 PMCID: PMC6169227 DOI: 10.1155/2018/3845027

Heme Oxygenase-1 May Affect Cell Signalling via Modulation of Ganglioside Composition. - PubMed - NCBI  https://www.ncbi.nlm.nih.gov/pubmed/30327713

¡¡

¡¡

Dev Neurosci. 2002;24(2-3):161-8.

Heme oxygenase-1 induction and dependent increase in ferritin. A protective antioxidant stratagem in hemin-treated rat brain.

Gonzales S1, Erario MA, Tomaro ML.

Author information

Abstract

The in vivo effect of hemin on both brain oxidative stress and heme oxygenase-1 (HO-1) induction was studied. A marked increase in lipid peroxidation was observed 1 h after hemin administration and antioxidant enzymes significantly decreased 3 h after hemin injection. HO-1 activity appeared 6 h after treatment, peaking 9 h after hemin administration. Such induction was preceded by a decrease in GSH pool and an increase in hydrogen peroxide concentration. Iron ferritin levels and ferritin content began to increase 6 h after HO-1 induction, and these increases remained high for at least 24 h after hemin injection. Administration of bilirubin entirely prevented HO-1 induction as well as the generation of oxidative stress parameters. These results indicate that the induction of heme oxygenase by hemin may be a general response to oxidant stress, by increasing bilirubin and ferritin levels and could therefore provide a major cellular defense mechanism against oxidative damage.

 

PMID: 12401954 DOI: 10.1159/000065686

Heme oxygenase-1 induction and dependent increase in ferritin. A protective antioxidant stratagem in hemin-treated rat brain. - PubMed - NCBI  https://www.ncbi.nlm.nih.gov/pubmed/12401954

¡¡

¡¡

Format: AbstractSend to

Int J Biochem Cell Biol. 1998 Feb;30(2):285-92.

Heme oxygenase induction by UVA radiation. A response to oxidative stress in rat liver.

Ossola JO1, Tomaro ML.

Author information

Abstract

Heme oxygenase is a key enzyme for heme catabolism and catalyzes the oxidative degradation of heme to form biliverdin IX alpha, an immediate precursor of bilirubin. In order to shed light on the mechanism by which UVA radiation causes oxidative damage, the relationship between heme oxygenase induction and oxidative stress was studied. HO-1 activity, lipid peroxidation and generation of active oxygen species (H2O2) were measured in rat liver exposed to UVA radiation. Besides, soluble and enzymatic antioxidant defenses (GSH, SOD, CAT and GSH-Px) were determined, while bilirubin antioxidant capacity was also evaluated. UVA radiation markedly increased both lipid peroxidation (180% +/- 7; S.E.M., n = 9 over control value of 0.1 +/- 0.01 nmol MDA/min per mg prot.) and steady state concentration of hydrogen peroxide (4 +/- 0.03 microM; S.E.M., n = 9) 3 h after treatment. At the same time, GSH content decreased to 3.6 +/- 0.2 mumol/g liver (S.E.M., n = 9) increasing thereafter. Antioxidant enzymes reached minimum values 6 h after UVA treatment (SOD: 7.2 +/- 0.2 U/mg protein, CAT: 7.8 +/- 0.2 pmol/mg protein, GSH-Px: 0.088 +/- 0.004 U/mg protein; S.E.M., n = 9), starting to increase 12 h after irradiation. HO-1 induction was observed 6 h after UVA irradiation, reaching a maximum value of 2.5 +/- 0.03 U/mg protein (S.E.M., n = 9) 12 h after treatment, and then declined until it reached control levels 24 h after exposure. Administration of bilirubin 2 h before UVA irradiation, entirely prevented HO-1 induction, the increase in MDA content and the decrease in GSH levels. This study shows that UVA irradiation leads to oxidative stress as evidenced by increased MDA content and H2O2 steady state levels, and depletion of GSH, SOD, CAT and GSH-Px. All these changes produced HO-1 induction. It is concluded that the induction of this enzyme could be a response to oxidative stress, since bilirubin can act as a physiological antioxidant.

Heme oxygenase induction by UVA radiation. A response to oxidative stress in rat liver. - PubMed - NCBI  https://www.ncbi.nlm.nih.gov/pubmed/9608682

¡¡

¡¡