Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 389
Filter
Add more filters

Publication year range
1.
J Am Chem Soc ; 146(23): 15955-15964, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38814055

ABSTRACT

Artificial syntheses of biologically active molecules have been fruitful in many bioinspired catalysis applications. Specifically, verdoheme and biliverdin, bearing polypyrrole frameworks, have inspired catalyst designs to address energy and environmental challenges. Despite remarkable progress in benchtop synthesis of verdoheme and biliverdin derivatives, all reported syntheses, starting from metalloporphyrins or inaccessible biliverdin precursors, require multiple steps to achieve the final desired products. Additionally, such synthetic procedures use multiple reactants/redox agents and involve multistep purification/extraction processes that often lower the yield. However, in a single step using atmospheric oxygen, heme oxygenases selectively generate verdoheme or biliverdin from heme. Motivated by such enzymatic pathways, we report a single-step electrosynthesis of verdoheme or biliverdin derivatives from their corresponding meso-aryl-substituted metalloporphyrin precursors. Our electrosynthetic methods have produced a copper-coordinating verdoheme analog in >80% yield at an applied potential of 0.65 V vs ferrocene/ferrocenium in air-exposed acetonitrile solution with a suitable electrolyte. These electrosynthetic routes reached a maximum product yield within 8 h of electrolysis at room temperature. The major products of verdoheme and biliverdin derivatives were isolated, purified, and characterized using electrospray mass spectrometry, absorption spectroscopy, cyclic voltammetry, and nuclear magnetic resonance spectroscopy techniques. Furthermore, X-ray crystallographic data were collected for select cobalt (Co)- and Cu-chelating verdoheme and metal-free biliverdin products. Electrosynthesis routes for the selective modification at the macrocycle ring in a single step are not known yet, and therefore, we believe that this report would advance the scopes of electrosynthesis strategies.


Subject(s)
Biliverdine , Biliverdine/chemistry , Biliverdine/metabolism , Biliverdine/analogs & derivatives , Heme/chemistry , Heme/analogs & derivatives , Electrochemical Techniques , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/chemistry , Porphyrins/chemistry , Molecular Structure
2.
Chembiochem ; 25(11): e202400068, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38623786

ABSTRACT

Far-red and near-infrared fluorescent proteins have regions of maximum transmission in most tissues and can be widely used as fluorescent biomarkers. We report that fluorescent phycobiliproteins originating from the phycobilisome core subunit ApcF2 can covalently bind biliverdin, named BDFPs. To further improve BDFPs, we conducted a series of studies. Firstly, we mutated K53Q and T144A of BDFPs to increase their effective brightness up to 190 % in vivo. Secondly, by homochromatic tandem fusion of high-brightness BDFPs to achieve monomerization, which increases the effective brightness by up to 180 % in vivo, and can effectively improve the labeling effect. By combining the above two approaches, the brightness of the tandem BDFPs was much improved compared with that of the previously reported fluorescent proteins in a similar spectral range. The tandem BDFPs were expressed stably while maintaining fluorescence in mammalian cells and Caenorhabditis elegans. They were also photostable and resistant to high temperature, low pH, and chemical denaturation. The tandem BDFPs advantages were proved in applications as biomarkers for imaging in super-resolution microscopy.


Subject(s)
Caenorhabditis elegans , Luminescent Proteins , Animals , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Caenorhabditis elegans/metabolism , Humans , Phycobiliproteins/chemistry , Phycobiliproteins/metabolism , Biliverdine/chemistry , Biliverdine/metabolism , Fluorescent Dyes/chemistry , HEK293 Cells
3.
Plant Physiol ; 193(1): 246-258, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37311159

ABSTRACT

Phytochromes are biliprotein photoreceptors present in plants, algae, certain bacteria, and fungi. Land plant phytochromes use phytochromobilin (PΦB) as the bilin chromophore. Phytochromes of streptophyte algae, the clade within which land plants evolved, employ phycocyanobilin (PCB), leading to a more blue-shifted absorption spectrum. Both chromophores are synthesized by ferredoxin-dependent bilin reductases (FDBRs) starting from biliverdin IXα (BV). In cyanobacteria and chlorophyta, BV is reduced to PCB by the FDBR phycocyanobilin:ferredoxin oxidoreductase (PcyA), whereas, in land plants, BV is reduced to PФB by phytochromobilin synthase (HY2). However, phylogenetic studies suggested the absence of any ortholog of PcyA in streptophyte algae and the presence of only PФB biosynthesis-related genes (HY2). The HY2 of the streptophyte alga Klebsormidium nitens (formerly Klebsormidium flaccidum) has already indirectly been indicated to participate in PCB biosynthesis. Here, we overexpressed and purified a His6-tagged variant of K. nitens HY2 (KflaHY2) in Escherichia coli. Employing anaerobic bilin reductase activity assays and coupled phytochrome assembly assays, we confirmed the product and identified intermediates of the reaction. Site-directed mutagenesis revealed 2 aspartate residues critical for catalysis. While it was not possible to convert KflaHY2 into a PΦB-producing enzyme by simply exchanging the catalytic pair, the biochemical investigation of 2 additional members of the HY2 lineage enabled us to define 2 distinct clades, the PCB-HY2 and the PΦB-HY2 clade. Overall, our study gives insight into the evolution of the HY2 lineage of FDBRs.


Subject(s)
Cyanobacteria , Phytochrome , Phylogeny , Ferredoxins/genetics , Plants/metabolism , Bile Pigments/metabolism , Biliverdine/chemistry , Biliverdine/genetics , Biliverdine/metabolism , Phytochrome/genetics , Phytochrome/metabolism , Cyanobacteria/genetics , Cyanobacteria/metabolism
4.
Nature ; 563(7732): 555-558, 2018 11.
Article in English | MEDLINE | ID: mdl-30464264

ABSTRACT

Birds are the only living amniotes with coloured eggs1-4, which have long been considered to be an avian innovation1,3. A recent study has demonstrated the presence of both red-brown protoporphyrin IX and blue-green biliverdin5-the pigments responsible for all the variation in avian egg colour-in fossilized eggshell of a nonavian dinosaur6. This raises the fundamental question of whether modern birds inherited egg colour from their nonavian dinosaur ancestors, or whether egg colour evolved independently multiple times. Here we present a phylogenetic assessment of egg colour in nonavian dinosaurs. We applied high-resolution Raman microspectroscopy to eggshells that represent all of the major clades of dinosaurs, and found that egg colour pigments were preserved in all eumaniraptorans: egg colour had a single evolutionary origin in nonavian theropod dinosaurs. The absence of colour in ornithischian and sauropod eggs represents a true signal rather than a taphonomic artefact. Pigment surface maps revealed that nonavian eumaniraptoran eggs were spotted and speckled, and colour pattern diversity in these eggs approaches that in extant birds, which indicates that reproductive behaviours in nonavian dinosaurs were far more complex than previously known3. Depth profiles demonstrated identical mechanisms of pigment deposition in nonavian and avian dinosaur eggs. Birds were not the first amniotes to produce coloured eggs: as with many other characteristics7,8 this is an attribute that evolved deep within the dinosaur tree and long before the spectacular radiation of modern birds.


Subject(s)
Biological Evolution , Dinosaurs , Egg Shell/anatomy & histology , Pigmentation/physiology , Animals , Biliverdine/metabolism , Color , Dinosaurs/classification , Fossils , Phylogeny , Principal Component Analysis , Protoporphyrins/metabolism
5.
Physiol Rev ; 96(4): 1449-508, 2016 10.
Article in English | MEDLINE | ID: mdl-27604527

ABSTRACT

Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. While initially considered to be waste products, CO and biliverdin/bilirubin have been shown over the last 20 years to modulate key cellular processes, such as inflammation, cell proliferation, and apoptosis, as well as antioxidant defense. This shift in paradigm has led to the importance of heme oxygenases and their products in cell physiology now being well accepted. The identification of the two human cases thus far of heme oxygenase deficiency and the generation of mice deficient in Hmox1 or Hmox2 have reiterated a role for these enzymes in both normal cell function and disease pathogenesis, especially in the context of cardiovascular disease. This review covers the current knowledge on the function of both Hmox1 and Hmox2 at both a cellular and tissue level in the cardiovascular system. Initially, the roles of heme oxygenases in vascular health and the regulation of processes central to vascular diseases are outlined, followed by an evaluation of the role(s) of Hmox1 and Hmox2 in various diseases such as atherosclerosis, intimal hyperplasia, myocardial infarction, and angiogenesis. Finally, the therapeutic potential of heme oxygenases and their products are examined in a cardiovascular disease context, with a focus on how the knowledge we have gained on these enzymes may be capitalized in future clinical studies.


Subject(s)
Cardiovascular Diseases/enzymology , Cardiovascular System/enzymology , Heme Oxygenase (Decyclizing)/metabolism , Heme/metabolism , Animals , Biliverdine/metabolism , Carbon Monoxide/metabolism , Humans , Iron/metabolism
6.
New Phytol ; 239(5): 1989-2006, 2023 09.
Article in English | MEDLINE | ID: mdl-37329247

ABSTRACT

Legume nodules produce large quantities of heme required for the synthesis of leghemoglobin (Lb) and other hemoproteins. Despite the crucial function of Lb in nitrogen fixation and the toxicity of free heme, the mechanisms of heme homeostasis remain elusive. Biochemical, cellular, and genetic approaches were used to study the role of heme oxygenases (HOs) in heme degradation in the model legume Lotus japonicus. Heme and biliverdin were quantified and localized, HOs were characterized, and knockout LORE1 and CRISPR/Cas9 mutants for LjHO1 were generated and phenotyped. We show that LjHO1, but not the LjHO2 isoform, is responsible for heme catabolism in nodules and identify biliverdin as the in vivo product of the enzyme in senescing green nodules. Spatiotemporal expression analysis revealed that LjHO1 expression and biliverdin production are restricted to the plastids of uninfected interstitial cells. The nodules of ho1 mutants showed decreased nitrogen fixation, and the development of brown, rather than green, nodules during senescence. Increased superoxide production was observed in ho1 nodules, underscoring the importance of LjHO1 in antioxidant defense. We conclude that LjHO1 plays an essential role in degradation of Lb heme, uncovering a novel function of nodule plastids and uninfected interstitial cells in nitrogen fixation.


Subject(s)
Lotus , Nitrogen Fixation , Nitrogen Fixation/genetics , Lotus/metabolism , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Biliverdine/metabolism , Leghemoglobin/genetics , Symbiosis/genetics , Root Nodules, Plant/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Gene Expression Regulation, Plant
7.
Proc Natl Acad Sci U S A ; 117(31): 18574-18581, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32661155

ABSTRACT

Many vertebrates have distinctive blue-green bones and other tissues due to unusually high biliverdin concentrations-a phenomenon called chlorosis. Despite its prevalence, the biochemical basis, biology, and evolution of chlorosis are poorly understood. In this study, we show that the occurrence of high biliverdin in anurans (frogs and toads) has evolved multiple times during their evolutionary history, and relies on the same mechanism-the presence of a class of serpin family proteins that bind biliverdin. Using a diverse combination of techniques, we purified these serpins from several species of nonmodel treefrogs and developed a pipeline that allowed us to assemble their complete amino acid and nucleotide sequences. The described proteins, hereafter named biliverdin-binding serpins (BBS), have absorption spectra that mimic those of phytochromes and bacteriophytochromes. Our models showed that physiological concentration of BBSs fine-tune the color of the animals, providing the physiological basis for crypsis in green foliage even under near-infrared light. Additionally, we found that these BBSs are most similar to human glycoprotein alpha-1-antitrypsin, but with a remarkable functional diversification. Our results present molecular and functional evidence of recurrent evolution of chlorosis, describe a biliverdin-binding protein in vertebrates, and introduce a function for a member of the serpin superfamily, the largest and most ubiquitous group of protease inhibitors.


Subject(s)
Anura/physiology , Biliverdine/metabolism , Serpins/metabolism , Skin Pigmentation/physiology , Animals , Anura/classification , Anura/genetics , Biliverdine/chemistry , Biological Mimicry/physiology , Serpins/chemistry , Serpins/genetics , Skin Pigmentation/genetics
8.
J Biol Chem ; 296: 100666, 2021.
Article in English | MEDLINE | ID: mdl-33862082

ABSTRACT

Heme oxygenases (HOs) play a critical role in recouping iron from the labile heme pool. The acquisition and liberation of heme iron are especially important for the survival of pathogenic bacteria. All characterized HOs, including those belonging to the HugZ superfamily, preferentially cleave free b-type heme. Another common form of heme found in nature is c-type heme, which is covalently linked to proteinaceous cysteine residues. However, mechanisms for direct iron acquisition from the c-type heme pool are unknown. Here we identify a HugZ homolog from the oligopeptide permease (opp) gene cluster of Paracoccus denitrificans that lacks any observable reactivity with heme b and show that it instead rapidly degrades c-type hemopeptides. This c-type heme oxygenase catalyzes the oxidative cleavage of the model substrate microperoxidase-11 at the ß- and/or δ-meso position(s), yielding the corresponding peptide-linked biliverdin, CO, and free iron. X-ray crystallographic analysis suggests that the switch in substrate specificity from b-to c-type heme involves loss of the N-terminal α/ß domain and C-terminal loop containing the coordinating histidine residue characteristic of HugZ homologs, thereby accommodating a larger substrate that provides its own iron ligand. These structural features are also absent in certain heme utilization/storage proteins from human pathogens that exhibit low or no HO activity with free heme. This study thus expands the scope of known iron acquisition strategies to include direct oxidative cleavage of heme-containing proteolytic fragments of c-type cytochromes and helps to explain why certain oligopeptide permeases show specificity for the import of heme in addition to peptides.


Subject(s)
Bacterial Proteins/metabolism , Biliverdine/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Heme/analogs & derivatives , Heme/metabolism , Iron/metabolism , Membrane Transport Proteins/metabolism , Paracoccus denitrificans/enzymology , Catalysis , Crystallography, X-Ray , Heme Oxygenase (Decyclizing)/chemistry , Substrate Specificity
9.
J Biol Chem ; 296: 100217, 2021.
Article in English | MEDLINE | ID: mdl-33839679

ABSTRACT

Heme oxygenase (HO) converts heme to carbon monoxide, biliverdin, and free iron, products that are essential in cellular redox signaling and iron recycling. In higher plants, HO is also involved in the biosynthesis of photoreceptor pigment precursors. Despite many common enzymatic reactions, the amino acid sequence identity between plant-type and other HOs is exceptionally low (∼19.5%), and amino acids that are catalytically important in mammalian HO are not conserved in plant-type HOs. Structural characterization of plant-type HO is limited to spectroscopic characterization by electron spin resonance, and it remains unclear how the structure of plant-type HO differs from that of other HOs. Here, we have solved the crystal structure of Glycine max (soybean) HO-1 (GmHO-1) at a resolution of 1.06 Å and carried out the isothermal titration calorimetry measurements and NMR spectroscopic studies of its interaction with ferredoxin, the plant-specific electron donor. The high-resolution X-ray structure of GmHO-1 reveals several novel structural components: an additional irregularly structured region, a new water tunnel from the active site to the surface, and a hydrogen-bonding network unique to plant-type HOs. Structurally important features in other HOs, such as His ligation to the bound heme, are conserved in GmHO-1. Based on combined data from X-ray crystallography, isothermal titration calorimetry, and NMR measurements, we propose the evolutionary fine-tuning of plant-type HOs for ferredoxin dependency in order to allow adaptation to dynamic pH changes on the stroma side of the thylakoid membrane in chloroplast without losing enzymatic activity under conditions of fluctuating light.


Subject(s)
Ferredoxins/chemistry , Glycine max/chemistry , Heme Oxygenase-1/chemistry , Heme/chemistry , Iron/chemistry , Plant Proteins/chemistry , Amino Acid Sequence , Biliverdine/chemistry , Biliverdine/metabolism , Carbon Monoxide/chemistry , Carbon Monoxide/metabolism , Catalytic Domain , Chloroplasts/chemistry , Chloroplasts/enzymology , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Ferredoxins/genetics , Ferredoxins/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Heme/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hydrogen Bonding , Iron/metabolism , Molecular Docking Simulation , Nuclear Magnetic Resonance, Biomolecular , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Glycine max/enzymology , Glycine max/genetics , Thylakoids/chemistry , Thylakoids/enzymology
10.
Environ Microbiol ; 24(4): 2047-2058, 2022 04.
Article in English | MEDLINE | ID: mdl-35172392

ABSTRACT

Light is the crucial environmental signal for desiccation-tolerant cyanobacteria to activate photosynthesis and prepare for desiccation at dawn. However, the photobiological characteristics of desert cyanobacteria adaptation to one of the harshest habitats on Earth remain unresolved. In this study, we surveyed the genome of a subaerial desert cyanobacterium Nostoc flagelliforme and identified two phytochromes and seven cyanobacteriochromes (CBCRs) with one or more bilin-binding GAF (cGMP phosphodiesterase/adenylyl cyclase/FhlA) domains. Biochemical and spectroscopic analyses of 69 purified GAF-containing proteins from recombinant phycocyanobilin (PCB), biliverdin or phycoerythrobilin-producing Escherichia coli indicated that nine of these proteins bind chromophores. Further investigation revealed that 11 GAFs form covalent adducts responsive to near-UV and visible light: eight GAFs contained PCB chromophores, three GAFs contained biliverdin chromophores and one contained the PCB isomer, phycoviolobilin. Interestingly, COO91_03972 is the first-ever reported GAF-only CBCR capable of sensing five wavelengths of light. Bioinformatics and biochemical analyses revealed that residue P132 of COO91_03972 is essential for chromophore binding to dual-cysteine CBCRs. Furthermore, the complement of N. flagelliforme CBCRs is enriched in red light sensors. We hypothesize that these sensors are critical for the acclimatization of N. flagelliforme to weak light environments at dawn.


Subject(s)
Bile Pigments , Nostoc , Bacterial Proteins/metabolism , Bile Pigments/metabolism , Biliverdine/metabolism , Light , Nostoc/genetics , Nostoc/metabolism
11.
J Biochem Mol Toxicol ; 36(10): e23168, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35838105

ABSTRACT

Acute liver failure, associated with oxidative stress and sustained inflammation is the major clinical manifestation of liver diseases with a high mortality rate due to limited therapeutic options. Purpurin is a bioactive compound of Rubia cordifolia that has been used in textile staining, as a food additive, and as a treatment of multiple chronic and metabolic diseases associated with inflammation and oxidative stress. The present work aimed to investigate the protective efficacy of purpurin against hepatorenal damage. Thirty-six female albino rats were equally assigned into six groups. Purpurin was administered orally once a day for 6 days at doses of 05, 10, and 20 mg/kg, respectively. Intraperitoneal injection of lipopolysaccharide (50 µg/kg) was administered to the animals on 6th day evening, 1 h after d-galactosamine (300 mg/kg) administration to induce hepatorenal injury. The results revealed that purpurin alleviated alterations in serological and hematological parameters as well as restored histoarchitectural and cellular integrity of the liver and kidney. Purpurin restored superoxide dismutase, catalase, glutathione reductase, glutathione peroxidase, and glutathione content in hepatorenal tissues. Accompanied by the diminution of increased bilirubin and biliverdin, purpurin also diminished total cholesterol, triglyceride, and lipid peroxidation in hepatorenal tissues. Purpurin markedly attenuated the elevation of CYP2E1, restored glutathione-S-transferase, and prevented DNA damage in hepatorenal tissues. Purpurin reduced iron overload by reducing heme depletion and recycling of ferritin and hemosiderin. It also reinforced biliverdin reductase, heme oxygenase-1 to employ hepatorenal protection by regulating antioxidant enzymes and other pathways that produced NADPH. Thus, it may be concluded that purpurin has protective potential against acute hepatorenal injury.


Subject(s)
Galactosamine , Heme Oxygenase-1 , Animals , Female , Rats , Anthraquinones , Antioxidants/metabolism , Antioxidants/pharmacology , Biliverdine/metabolism , Catalase/metabolism , Cholesterol/metabolism , Cytochrome P-450 CYP2E1/metabolism , Ferritins , Food Additives , Galactosamine/toxicity , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Heme , Heme Oxygenase-1/metabolism , Hemosiderin/metabolism , Inflammation/metabolism , Lipopolysaccharides/toxicity , Liver/metabolism , NADP/metabolism , Superoxide Dismutase/metabolism , Transferases/metabolism , Triglycerides , Up-Regulation
13.
Proc Natl Acad Sci U S A ; 116(17): 8301-8309, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30948637

ABSTRACT

Because cyanobacteriochrome photoreceptors need only a single compact domain for chromophore incorporation and for absorption of visible spectra including the long-wavelength far-red region, these molecules have been paid much attention for application to bioimaging and optogenetics. Most cyanobacteriochromes, however, have a drawback to incorporate phycocyanobilin that is not available in the mammalian cells. In this study, we focused on biliverdin (BV) that is a mammalian intrinsic chromophore and absorbs the far-red region and revealed that replacement of only four residues was enough for conversion from BV-rejective cyanobacteriochromes into BV-acceptable molecules. We succeeded in determining the crystal structure of one of such engineered molecules, AnPixJg2_BV4, at 1.6 Å resolution. This structure identified unusual covalent bond linkage, which resulted in deep BV insertion into the protein pocket. The four mutated residues contributed to reducing steric hindrances derived from the deeper insertion. We introduced these residues into other domains, and one of them, NpF2164g5_BV4, produced bright near-infrared fluorescence from mammalian liver in vivo. Collectively, this study provides not only molecular basis to incorporate BV by the cyanobacteriochromes but also rational strategy to open the door for application of cyanobacteriochromes to visualization and regulation of deep mammalian tissues.


Subject(s)
Biliverdine , Photoreceptors, Microbial , Protein Engineering/methods , Animals , Biliverdine/chemistry , Biliverdine/metabolism , COS Cells , Chlorocebus aethiops , Cyanobacteria/genetics , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Liver/chemistry , Liver/diagnostic imaging , Liver/metabolism , Mice , Models, Molecular , Optical Imaging , Photoreceptors, Microbial/chemistry , Photoreceptors, Microbial/genetics , Photoreceptors, Microbial/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
14.
Bioprocess Biosyst Eng ; 45(3): 563-571, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35066674

ABSTRACT

Bilirubin, a natural intermediate in heme degradation, is a valuable Chinese medicine used in more than 50 traditional Chinese medicine (TCM) preparations. At present, bilirubin is mainly produced by extraction from pig bile, but a shortage of the raw material has increased the price, to about US$10,000/kg in the Chinese market. Biliverdin, the precursor of bilirubin, is more abundant and less expensive than bilirubin, but it is not used in TCM. Thus, the biotransformation of biliverdin by biliverdin reductase (BvdR) may be a practical way to produce bilirubin. In this study, the codon-optimized gene of biliverdin reductase (mbvdR) from the cyanobacterium Synechocystis was cloned into Escherichia coli BL21(DE3), and the conditions for BL21-mBvdR expressing BvdR were optimized. Resting BL21-mBvdR cells were employed as biocatalysts to biotransform biliverdin to bilirubin. At a concentration of biliverdin substrate of 450 mg/L in the reaction mixture, the bilirubin content in dry cells reached 20.8 ± 0.8 mg/g, with a conversion yield of 72.3%. Therefore, recombinant E. coli expressing BvdR can be applied to biotransform biliverdin to bilirubin, providing a potential alternative process for bilirubin production.


Subject(s)
Biliverdine , Cyanobacteria , Animals , Bilirubin/metabolism , Biliverdine/genetics , Biliverdine/metabolism , Biotransformation , Cyanobacteria/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Swine
15.
Int J Mol Sci ; 23(13)2022 Jun 24.
Article in English | MEDLINE | ID: mdl-35806040

ABSTRACT

Heme oxygenase (HO) has both beneficial and detrimental effects via its metabolites, including carbon monoxide (CO), biliverdin or bilirubin, and ferrous iron. HO-1 is an inducible form of HO that is upregulated by oxidative stress, nitric oxide, CO, and hypoxia, whereas HO-2 is a constitutive form that regulates vascular tone and homeostasis. In brains injured by trauma, ischemia-reperfusion, or Alzheimer's disease (AD), the long-term expression of HO-1 can be detected, which can lead to cytotoxic ferroptosis via iron accumulation. In contrast, the transient induction of HO-1 in the peri-injured region may have regenerative potential (e.g., angiogenesis, neurogenesis, and mitochondrial biogenesis) and neurovascular protective effects through the CO-mediated signaling pathway, the antioxidant properties of bilirubin, and the iron-mediated ferritin synthesis. In this review, we discuss the dual roles of HO-1 and its metabolites in various neurovascular diseases, including age-related macular degeneration, ischemia-reperfusion injury, traumatic brain injury, Gilbert's syndrome, and AD.


Subject(s)
Heme Oxygenase (Decyclizing) , Heme Oxygenase-1 , Bilirubin/metabolism , Biliverdine/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Iron/metabolism
16.
Int J Mol Sci ; 23(19)2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36233341

ABSTRACT

A diabetic foot ulcer (DFU) is one of the major complications of diabetes. Wound healing under diabetic conditions is often impaired. This is in part due to the excessive oxidative stress, prolonged inflammation, immune cell dysfunction, delayed re-epithelialization, and decreased angiogenesis present at the wound site. As a result of these multifactorial impaired healing pathways, it has been difficult to develop effective therapeutic strategies for DFU. Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme degradation generating carbon monoxide (CO), biliverdin (BV) which is converted into bilirubin (BR), and iron. HO-1 is a potent antioxidant. It can act as an anti-inflammatory, proliferative, angiogenic and cytoprotective enzyme. Due to its biological functions, HO-1 plays a very important role in wound healing, in part mediated through the biologically active end products generated by its enzymatic activity, particularly CO, BV, and BR. Therapeutic strategies involving the activation of HO-1, or the topical application of its biologically active end products are important in diabetic wound healing. Therefore, HO-1 is an attractive therapeutic target for DFU treatment. This review will provide an overview and discussion of the importance of HO-1 as a therapeutic target for diabetic wound healing.


Subject(s)
Diabetes Mellitus , Diabetic Foot , Anti-Inflammatory Agents , Antioxidants , Biliverdine/metabolism , Biliverdine/therapeutic use , Carbon Monoxide/metabolism , Diabetic Foot/drug therapy , Heme/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Humans , Iron/metabolism
17.
Br Poult Sci ; 63(3): 421-425, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34585996

ABSTRACT

1. The goal of this study was to investigate the colour diversity of egg shells and expression of related genes in the uterus of chickens that produce eggs of different colours.2. Four colour types of Changshun blue-shell chickens, producing dark or light blue, greenish-brown and brown shelled eggs, were selected. The eggshell pigment concentration and colour values in each group were examined. The relative gene expression of solute carrier organic anion transporter family member 1C1 (SLCO1C1), ferrochelatase (FECH), haem oxygenase 1 (HO-1), ovotransferrin (OF) and biliverdin reductase A (BLVRA) in eggshell gland were measured.3. The Δb, ΔE and protoporphyrin in brown and greenish-brown groups were significantly higher in the blue egg group (P < 0.01), whereas ΔL was significantly lower than that in the blue eggs (P < 0.01). There was no significant difference in biliverdin concentration between the brown and blue groups.4. The Δa values, in descending order, were 8.27 ± 2.76 in the brown, -3.79 ± 2.39 in the greenish-brown and -7.29 ± 2.27 in the blue groups, respectively. The relative expression of HO-1 in the greenish-brown and light blue groups was significantly higher than in the dark blue and brown groups. The relative expression of FECH in the light blue group was significantly lower than that in the dark blue, greenish-brown or brown group (P < 0.01). The relative expression of HO-1 and BLVRA genes in the dark blue group was significantly higher than that in the light blue, greenish-brown and the brown group (P < 0.01).5. The Δa might provide a better index than protoporphyrin and biliverdin contents for eggshell colour breeding. Overall, HO-1 as well as BLVRA were important candidate genes for the selection of dark blue eggs.


Subject(s)
Chickens , Egg Shell , Animals , Biliverdine/genetics , Biliverdine/metabolism , Chickens/genetics , Chickens/metabolism , Female , Gene Expression , Ovum , Pigmentation/genetics , Protoporphyrins/genetics , Protoporphyrins/metabolism , Uterus/metabolism
18.
Phys Chem Chem Phys ; 23(37): 20867-20874, 2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34374395

ABSTRACT

Cyanobacteriochromes (CBCRs) are bi-stable photoreceptor proteins with high potential for biotechnological applications. Most of these proteins utilize phycocyanobilin (PCB) as a light-sensing co-factor, which is unique to cyanobacteria, but some variants also incorporate biliverdin (BV). The latter are of particular interest for biotechnology due to the natural abundance and red-shifted absorption of BV. Here, AmI-g2 was investigated, a CBCR capable of binding both PCB and BV. The assembly kinetics and primary photochemistry of AmI-g2 with both chromophores were studied in vitro. The assembly reaction with PCB is roughly 10× faster than BV, and the formation of a non-covalent intermediate was identified as the rate-limiting step in the case of BV. This step is fast for PCB, where the formation of the covalent thioether bond between AmI-g2 and PCB becomes rate-limiting. The photochemical quantum yields of the forward and backward reactions of AmI-g2 were estimated and discussed in the context of homologous CBCRs.


Subject(s)
Biliverdine/chemistry , Cyanobacteria/metabolism , Photoreceptors, Microbial/chemistry , Phycobilins/chemistry , Phycocyanin/chemistry , Biliverdine/metabolism , Kinetics , Photoreceptors, Microbial/genetics , Photoreceptors, Microbial/metabolism , Phycobilins/metabolism , Phycocyanin/metabolism , Protein Binding , Quantum Theory , Spectrophotometry
19.
J Chem Phys ; 154(6): 065101, 2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33588533

ABSTRACT

An enhanced interest in the phytochrome-based fluorescent proteins is explained by their ability to absorb and emit light in the far-red and infra-red regions particularly suitable for bioimaging. The fluorescent protein IFP1.4 was engineered from the chromophore-binding domain of a bacteriophytochrome in attempts to increase the fluorescence quantum yield. We report the results of simulations of structures in the ground S0 and excited S1 electronic states of IFP1.4 using the methods of quantum chemistry and quantum mechanics/molecular mechanics. We construct different protonation states of the biliverdin (BV) chromophore in the red-absorbing form of the protein by moving protons from the BV pyrrole rings to a suitable acceptor within the system and show that these structures are close in energy but differ by absorption bands. For the first time, we report structures of the minimum energy conical intersection points S1/S0 on the energy surfaces of BV in the protein environment and describe their connection to the local minima in the excited S1 state. These simulations allow us to characterize the deactivation routes in IFP1.4.


Subject(s)
Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Molecular Dynamics Simulation , Phytochrome/metabolism , Bacterial Proteins , Biliverdine/metabolism , Hydrogen Bonding , Protein Conformation , Protein Domains , Quantum Theory
20.
Biochem J ; 477(3): 601-614, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31913441

ABSTRACT

The pro-oxidant effect of free heme (Fe2+-protoporphyrin IX) is neutralized by phylogenetically-conserved heme oxygenases (HMOX) that generate carbon monoxide, free ferrous iron, and biliverdin (BV) tetrapyrrole(s), with downstream BV reduction by non-redundant NADPH-dependent BV reductases (BLVRA and BLVRB) that retain isomer-restricted functional activity for bilirubin (BR) generation. Regioselectivity for the heme α-meso carbon resulting in predominant BV IXα generation is a defining characteristic of canonical HMOXs, thereby limiting generation and availability of BVs IXß, IXδ, and IXγ as BLVRB substrates. We have now exploited the unique capacity of the Pseudomonas aeruginosa (P. aeruginosa) hemO/pigA gene for focused generation of isomeric BVs (IXß and IXδ). A scalable system followed by isomeric separation yielded highly pure samples with predicted hydrogen-bonded structure(s) as documented by 1H NMR spectroscopy. Detailed kinetic studies established near-identical activity of BV IXß and BV IXδ as BLVRB-selective substrates, with confirmation of an ordered sequential mechanism of BR/NADP+ dissociation. Halogenated xanthene-based compounds previously identified as BLVRB-targeted flavin reductase inhibitors displayed comparable inhibition parameters using BV IXß as substrate, documenting common structural features of the cofactor/substrate-binding pocket. These data provide further insights into structure/activity mechanisms of isomeric BVs as BLVRB substrates, with potential applicability to further dissect redox-regulated functions in cytoprotection and hematopoiesis.


Subject(s)
Biliverdine , Heme Oxygenase (Decyclizing) , Heme/metabolism , Pseudomonas aeruginosa/metabolism , Biliverdine/chemistry , Biliverdine/metabolism , Genes, Bacterial/physiology , Heme Oxygenase (Decyclizing)/chemistry , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Kinetics , Oxidation-Reduction , Oxidoreductases/metabolism , Pseudomonas aeruginosa/genetics
SELECTION OF CITATIONS
SEARCH DETAIL