Biliverdin reductase

Last updated
biliverdin reductase
1hdo.jpg
Identifiers
EC no. 1.3.1.24
CAS no. 9074-10-6
Databases
IntEnz IntEnz view
BRENDA BRENDA entry
ExPASy NiceZyme view
KEGG KEGG entry
MetaCyc metabolic pathway
PRIAM profile
PDB structures RCSB PDB PDBe PDBsum
Gene Ontology AmiGO / QuickGO
Search
PMC articles
PubMed articles
NCBI proteins
biliverdin reductase A
BLVRA 2H63.png
Crystallographic structure of human biliverdin reductase A based on the PDB: 2H63 coordinates. The enzyme is displayed as a rainbow colored cartoon (N-terminus = blue, C-terminus = red) while the NADP cofactor is displayed as space-filling model (carbon = white, oxygen = red, nitrogen = blue, phosphorus = orange).
Identifiers
Symbol BLVRA
Alt. symbolsBLVR
NCBI gene 644
HGNC 1062
OMIM 109750
RefSeq NM_000712
UniProt P53004
Other data
EC number 1.3.1.24
Locus Chr. 7 p14-cen
Search for
Structures Swiss-model
Domains InterPro
biliverdin reductase B
Identifiers
Symbol BLVRB
Alt. symbolsFLR
NCBI gene 645
HGNC 1063
OMIM 600941
RefSeq NM_000713
UniProt P30043
Other data
EC number 1.3.1.24
Locus Chr. 19 q13.1-13.2
Search for
Structures Swiss-model
Domains InterPro
Biliverdin reductase, catalytic
PDB 1lc3 EBI.jpg
crystal structure of a biliverdin reductase enzyme-cofactor complex
Identifiers
SymbolBiliv-reduc_cat
Pfam PF09166
InterPro IPR015249
SCOP2 1lc0 / SCOPe / SUPFAM
Available protein structures:
Pfam   structures / ECOD  
PDB RCSB PDB; PDBe; PDBj
PDBsum structure summary

Biliverdin reductase (BVR) is an enzyme (EC 1.3.1.24) found in all tissues under normal conditions, but especially in reticulo-macrophages of the liver and spleen. BVR facilitates the conversion of biliverdin to bilirubin via the reduction of a double bond between the second and third pyrrole ring into a single bond.

Contents

There are two isozymes in humans, each encoded by its own gene, biliverdin reductase A (BLVRA) and biliverdin reductase B (BLVRB).

Mechanism of catalysis

BVR acts on biliverdin by reducing its double-bond between the pyrrole rings into a single-bond. [1] It accomplishes this using NADPH + H+ as an electron donor, forming bilirubin and NADP+ as products.

BVR catalyzes this reaction through an overlapping binding site including Lys18, Lys22, Lys179, Arg183, and Arg185 as key residues. [2] This binding site attaches to biliverdin, and causes its dissociation from heme oxygenase (HO) (which catalyzes reaction of ferric heme --> biliverdin), causing the subsequent reduction to bilirubin. [3]

Reduction of biliverdin to bilirubin catalyzed by biliverdin reductase. BVR mechanism.png
Reduction of biliverdin to bilirubin catalyzed by biliverdin reductase.

Structure

BVR is composed of two closely packed domains, between 247 and 415 amino acids long and containing a Rossmann fold. [4] BVR has also been determined to be a zinc-binding protein with each enzyme protein having one strong-binding zinc atom. [5] [6]

The C-terminal half of BVR contains the catalytic domain, which adopts a structure containing a six-stranded beta-sheet that is flanked on one face by several alpha-helices. This domain contains the catalytic active site, which reduces the gamma-methene bridge of the open tetrapyrrole, biliverdin IX alpha, to bilirubin with the concomitant oxidation of a NADH or NADPH cofactor. [7]

Function

BVR works with the biliverdin/bilirubin redox cycle. It converts biliverdin to bilirubin (a strong antioxidant), which is then converted back into biliverdin through the actions of reactive oxygen species (ROS). This cycle allows for the neutralization of ROS, and the reuse of biliverdin products. Biliverdin also is replenished in the cycle with its formation from heme units through heme oxygenase (HO) localized from the endoplasmic reticulum. [8]

Bilirubin, being one of the last products of heme degradation in the liver, is further processed and excreted in bile after conjugation with glucuronic acid. [9] In this way, BVR is essential in many mammals for the disposal of heme catabolites – especially in the fetus where the placental membranes are bilirubin-permeable but not biliverdin-permeable – aiding in the removal of potentially toxic protein build-up. [10]

BVR has also more recently been recognized as a regulator of glucose metabolism and in cell growth and apoptosis control, due to its dual-specificity kinase character. [11] This control over glucose metabolism indicates that BVR may play a role in pathogenesis of multiple metabolic diseases – notably diabetes – by control of the upstream activator of insulin growth factor-1 (IGF-1) and mitogen-activated protein kinase (MAPK) signaling pathway. [12]

Disease relevance

BVR acts as a means to regenerate bilirubin in a repeating redox cycle without significantly modifying the concentration of available bilirubin. With these levels maintained, it appears that BVR represents a new strategy for the treatment of multiple sclerosis and other types of oxidative stress-mediated diseases. [13] The mechanism is due to the amplification of the potent antioxidant actions of bilirubin, as this can ameliorate free radical-mediated diseases. [14]

Studies have shown that the BVR redox cycle is essential in providing physiological cytoprotection. Genetic knock-outs and reduced BVR levels have demonstrated increased formation of ROS, and results in augmented cell death. Cells that experienced a 90% reduction in BVR experienced three times normal ROS levels. [15] Through this protective and amplifying cycle, BVR allows low concentrations of bilirubin to overcome 10,000-fold higher concentrations of ROS. [16]

Related Research Articles

<span class="mw-page-title-main">Bilirubin</span> Red pigment of the bile

Bilirubin (BR) is a red-orange compound that occurs in the normal catabolic pathway that breaks down heme in vertebrates. This catabolism is a necessary process in the body's clearance of waste products that arise from the destruction of aged or abnormal red blood cells. In the first step of bilirubin synthesis, the heme molecule is stripped from the hemoglobin molecule. Heme then passes through various processes of porphyrin catabolism, which varies according to the region of the body in which the breakdown occurs. For example, the molecules excreted in the urine differ from those in the feces. The production of biliverdin from heme is the first major step in the catabolic pathway, after which the enzyme biliverdin reductase performs the second step, producing bilirubin from biliverdin.

<span class="mw-page-title-main">Coenzyme A</span> Coenzyme, notable for its synthesis and oxidation role

Coenzyme A (CoA, SHCoA, CoASH) is a coenzyme, notable for its role in the synthesis and oxidation of fatty acids, and the oxidation of pyruvate in the citric acid cycle. All genomes sequenced to date encode enzymes that use coenzyme A as a substrate, and around 4% of cellular enzymes use it (or a thioester) as a substrate. In humans, CoA biosynthesis requires cysteine, pantothenate (vitamin B5), and adenosine triphosphate (ATP).

<span class="mw-page-title-main">Heme</span> Chemical coordination complex of an iron ion chelated to a porphyrin

Heme, or haem, is a ring-shaped iron-containing molecular component of hemoglobin, which is necessary to bind oxygen in the bloodstream. It is composed of four pyrrole rings with 2 vinyl and 2 propionic acid side chains. Heme is biosynthesized in both the bone marrow and the liver.

<span class="mw-page-title-main">Nitric oxide synthase</span> Enzyme catalysing the formation of the gasotransmitter NO(nitric oxide)

Nitric oxide synthases (NOSs) are a family of enzymes catalyzing the production of nitric oxide (NO) from L-arginine. NO is an important cellular signaling molecule. It helps modulate vascular tone, insulin secretion, airway tone, and peristalsis, and is involved in angiogenesis and neural development. It may function as a retrograde neurotransmitter. Nitric oxide is mediated in mammals by the calcium-calmodulin controlled isoenzymes eNOS and nNOS. The inducible isoform, iNOS, involved in immune response, binds calmodulin at physiologically relevant concentrations, and produces NO as an immune defense mechanism, as NO is a free radical with an unpaired electron. It is the proximate cause of septic shock and may function in autoimmune disease.

<span class="mw-page-title-main">Succinate dehydrogenase</span> Enzyme

Succinate dehydrogenase (SDH) or succinate-coenzyme Q reductase (SQR) or respiratory complex II is an enzyme complex, found in many bacterial cells and in the inner mitochondrial membrane of eukaryotes. It is the only enzyme that participates in both the citric acid cycle and the electron transport chain. Histochemical analysis showing high succinate dehydrogenase in muscle demonstrates high mitochondrial content and high oxidative potential.

<span class="mw-page-title-main">Thioredoxin</span> Class of reduction–oxidation proteins

Thioredoxin is a class of small redox proteins known to be present in all organisms. It plays a role in many important biological processes, including redox signaling. In humans, thioredoxins are encoded by TXN and TXN2 genes. Loss-of-function mutation of either of the two human thioredoxin genes is lethal at the four-cell stage of the developing embryo. Although not entirely understood, thioredoxin is linked to medicine through their response to reactive oxygen species (ROS). In plants, thioredoxins regulate a spectrum of critical functions, ranging from photosynthesis to growth, flowering and the development and germination of seeds. Thioredoxins play a role in cell-to-cell communication.

<span class="mw-page-title-main">HMOX1</span> Mammalian protein found in Homo sapiens

HMOX1 is a human gene that encodes for the enzyme heme oxygenase 1. Heme oxygenase mediates the first step of heme catabolism, it cleaves heme to form biliverdin.

<span class="mw-page-title-main">Heme oxygenase</span> Class of enzymes

Heme oxygenase, or haem oxygenase, is an enzyme that catalyzes the degradation of heme to produce biliverdin, ferrous ion, and carbon monoxide.

<span class="mw-page-title-main">Inositol oxygenase</span> Protein-coding gene in the species Homo sapiens

Inositol oxygenase, also commonly referred to as myo-inositol oxygenase (MIOX), is a non-heme di-iron enzyme that oxidizes myo-inositol to glucuronic acid. The enzyme employs a unique four-electron transfer at its Fe(II)/Fe(III) coordination sites and the reaction proceeds through the direct binding of myo-inositol followed by attack of the iron center by diatomic oxygen. This enzyme is part of the only known pathway for the catabolism of inositol in humans and is expressed primarily in the kidneys. Recent medical research regarding MIOX has focused on understanding its role in metabolic and kidney diseases such as diabetes, obesity and acute kidney injury. Industrially-focused engineering efforts are centered on improving MIOX activity in order to produce glucaric acid in heterologous hosts.

<span class="mw-page-title-main">Cystathionine beta synthase</span> Mammalian protein found in humans

Cystathionine-β-synthase, also known as CBS, is an enzyme (EC 4.2.1.22) that in humans is encoded by the CBS gene. It catalyzes the first step of the transsulfuration pathway, from homocysteine to cystathionine:

<span class="mw-page-title-main">NFE2L2</span> Human protein and coding gene

Nuclear factor erythroid 2-related factor 2 (NRF2), also known as nuclear factor erythroid-derived 2-like 2, is a transcription factor that in humans is encoded by the NFE2L2 gene. NRF2 is a basic leucine zipper (bZIP) protein that may regulate the expression of antioxidant proteins that protect against oxidative damage triggered by injury and inflammation, according to preliminary research. In vitro, NRF2 binds to antioxidant response elements (AREs) in the promoter regions of genes encoding cytoprotective proteins. NRF2 induces the expression of heme oxygenase 1 in vitro leading to an increase in phase II enzymes. NRF2 also inhibits the NLRP3 inflammasome.

<span class="mw-page-title-main">Thiosulfate dehydrogenase</span>

Thiosulfate dehydrogenase is an enzyme that catalyzes the chemical reaction:

<span class="mw-page-title-main">HMOX2</span> Protein-coding gene in the species Homo sapiens

Heme oxygenase 2 is an enzyme that in humans is encoded by the HMOX2 gene.

<span class="mw-page-title-main">GLRX2</span> Protein-coding gene in the species Homo sapiens

Glutaredoxin 2 (GLRX2) is an enzyme that in humans encoded by the GLRX2 gene. GLRX2, also known as GRX2, is a glutaredoxin family protein and a thiol-disulfide oxidoreductase that maintains cellular thiol homeostasis. This gene consists of four exons and three introns, spanned 10 kilobase pairs, and localized to chromosome 1q31.2–31.3.

<span class="mw-page-title-main">TXN2</span> Protein-coding gene in the species Homo sapiens

Thioredoxin, mitochondrial also known as thioredoxin-2 is a protein that in humans is encoded by the TXN2 gene on chromosome 22. This nuclear gene encodes a mitochondrial member of the thioredoxin family, a group of small multifunctional redox-active proteins. The encoded protein may play important roles in the regulation of the mitochondrial membrane potential and in protection against oxidant-induced apoptosis.

Plant seed peroxygenase (EC 1.11.2.3, plant peroxygenase, soybean peroxygenase) is an enzyme with systematic name substrate:hydroperoxide oxidoreductase (RH-hydroxylating or epoxidising). This enzyme catalyses the following chemical reaction

1,8-Cineole 2-endo-monooxygenase (EC 1.14.14.133, Formerly EC 1.14.13.156, P450cin, CYP176A, CYP176A1) is an enzyme with systematic name 1,8-cineole,NADPH:oxygen oxidoreductase (2-endo-hydroxylating). This enzyme catalyses the following chemical reaction

<span class="mw-page-title-main">Carbon monoxide-releasing molecules</span> Substances delivering CO within the body

Carbon monoxide-releasing molecules (CORMs) are chemical compounds designed to release controlled amounts of carbon monoxide (CO). CORMs are being developed as potential therapeutic agents to locally deliver CO to cells and tissues, thus overcoming limitations of CO gas inhalation protocols.

<span class="mw-page-title-main">Biliverdin reductase B</span> Protein-coding gene in the species Homo sapiens

Biliverdin reductase B is a protein that in humans is encoded by the BLVRB gene.

Morphinone reductase is an enzyme which catalyzes the NADH-dependent saturation of the carbon-carbon double bond of morphinone and codeinone, yielding hydromorphone and hydrocodone respectively. This saturation reaction is assisted by a FMN cofactor and the enzyme is a member of the α/β-barrel flavoprotein family. The sequence of the enzyme has been obtained from bacteria Pseudomonas putida M10 and has been successfully expressed in yeast and other bacterial species. The enzyme is reported to harbor high sequence and structural similarity to the Old Yellow Enzyme, a large group of flavin-dependent redox biocatalysts of yeast species, and an oestrogen-binding protein of Candida albicans. The enzyme has demonstrated value in biosynthesis of semi-opiate drugs in microorganisms, expanding the chemical diversity of BIA biosynthesis.

References

  1. Rigney E, Mantle TJ (Nov 1988). "The reaction mechanism of bovine kidney biliverdin reductase". Biochimica et Biophysica Acta (BBA) - Protein Structure and Molecular Enzymology. 957 (2): 237–42. doi:10.1016/0167-4838(88)90278-6. PMID   3191141.
  2. Wang J, de Montellano PR (May 2003). "The binding sites on human heme oxygenase-1 for cytochrome p450 reductase and biliverdin reductase". The Journal of Biological Chemistry. 278 (22): 20069–76. doi: 10.1074/jbc.M300989200 . PMID   12626517.
  3. Ahmad Z, Salim M, Maines MD (Mar 2002). "Human biliverdin reductase is a leucine zipper-like DNA-binding protein and functions in transcriptional activation of heme oxygenase-1 by oxidative stress". The Journal of Biological Chemistry. 277 (11): 9226–32. doi: 10.1074/jbc.M108239200 . PMID   11773068.
  4. Bellamacina CR (Sep 1996). "The nicotinamide dinucleotide binding motif: a comparison of nucleotide binding proteins". FASEB Journal. 10 (11): 1257–69. doi: 10.1096/fasebj.10.11.8836039 . PMID   8836039. S2CID   24189316.
  5. Maines MD, Polevoda BV, Huang TJ, McCoubrey WK (Jan 1996). "Human biliverdin IXalpha reductase is a zinc-metalloprotein. Characterization of purified and Escherichia coli expressed enzymes". European Journal of Biochemistry. 235 (1–2): 372–81. doi: 10.1111/j.1432-1033.1996.00372.x . PMID   8631357.
  6. PDB: 1GCU ; Kikuchi A, Park SY, Miyatake H, Sun D, Sato M, Yoshida T, Shiro Y (Mar 2001). "Crystal structure of rat biliverdin reductase". Nature Structural Biology. 8 (3): 221–5. doi:10.1038/84955. PMID   11224565. S2CID   42293456.
  7. Whitby FG, Phillips JD, Hill CP, McCoubrey W, Maines MD (Jun 2002). "Crystal structure of a biliverdin IXalpha reductase enzyme-cofactor complex". Journal of Molecular Biology. 319 (5): 1199–210. doi:10.1016/S0022-2836(02)00383-2. PMID   12079357.
  8. Kravets A, Hu Z, Miralem T, Torno MD, Maines MD (May 2004). "Biliverdin reductase, a novel regulator for induction of activating transcription factor-2 and heme oxygenase-1". The Journal of Biological Chemistry. 279 (19): 19916–23. doi: 10.1074/jbc.M314251200 . PMID   14988408.
  9. Bosma PJ, Seppen J, Goldhoorn B, Bakker C, Oude Elferink RP, Chowdhury JR, Chowdhury NR, Jansen PL (Jul 1994). "Bilirubin UDP-glucuronosyltransferase 1 is the only relevant bilirubin glucuronidating isoform in man". The Journal of Biological Chemistry. 269 (27): 17960–4. doi: 10.1016/S0021-9258(17)32403-1 . PMID   8027054.
  10. McDonagh AF, Palma LA, Schmid R (Jan 1981). "Reduction of biliverdin and placental transfer of bilirubin and biliverdin in the pregnant guinea pig". The Biochemical Journal. 194 (1): 273–82. doi:10.1042/bj1940273. PMC   1162741 . PMID   7305981.
  11. Florczyk UM, Jozkowicz A, Dulak J (January–February 2008). "Biliverdin reductase: new features of an old enzyme and its potential therapeutic significance". Pharmacological Reports. 60 (1): 38–48. PMC   5536200 . PMID   18276984.
  12. Kapitulnik J, Maines MD (Mar 2009). "Pleiotropic functions of biliverdin reductase: cellular signaling and generation of cytoprotective and cytotoxic bilirubin". Trends in Pharmacological Sciences. 30 (3): 129–37. doi:10.1016/j.tips.2008.12.003. PMID   19217170.
  13. Maghzal GJ, Leck MC, Collinson E, Li C, Stocker R (Oct 2009). "Limited role for the bilirubin-biliverdin redox amplification cycle in the cellular antioxidant protection by biliverdin reductase". The Journal of Biological Chemistry. 284 (43): 29251–9. doi: 10.1074/jbc.M109.037119 . PMC   2785555 . PMID   19690164.
  14. Liu Y, Li P, Lu J, Xiong W, Oger J, Tetzlaff W, Cynader M (Aug 2008). "Bilirubin possesses powerful immunomodulatory activity and suppresses experimental autoimmune encephalomyelitis". Journal of Immunology. 181 (3): 1887–97. doi: 10.4049/jimmunol.181.3.1887 . PMID   18641326.
  15. Baranano DE, Rao M, Ferris CD, Snyder SH (Dec 2002). "Biliverdin reductase: a major physiologic cytoprotectant". Proceedings of the National Academy of Sciences of the United States of America. 99 (25): 16093–8. Bibcode:2002PNAS...9916093B. doi: 10.1073/pnas.252626999 . PMC   138570 . PMID   12456881.
  16. Sedlak TW, Snyder SH (Jun 2004). "Bilirubin benefits: cellular protection by a biliverdin reductase antioxidant cycle". Pediatrics. 113 (6): 1776–82. doi:10.1542/peds.113.6.1776. PMID   15173506.
This article incorporates text from the public domain Pfam and InterPro: IPR015249