Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Inflamm Res ; 67(5): 407-422, 2018 May.
Article in English | MEDLINE | ID: mdl-29362850

ABSTRACT

OBJECTIVE AND DESIGN: To investigate the role of heme oxygenase-1 (HO-1), carbon monoxide (CO), and biliverdin (BVD) in the zymosan-induced TMJ arthritis in rats. MATERIALS AND METHODS: Mechanical threshold was assessed before and 4 h after TMJ arthritis induction in rats. Cell influx, myeloperoxidase activity, and histological changes were measured in the TMJ lavages and tissues. Trigeminal ganglion and periarticular tissues were used for HO-1, TNF-α, and IL-1ß mRNA time course expression and immunohistochemical analyses. Hemin (0.1, 0.3, or 1 mg kg-1), DMDC (0.025, 0.25, or 2.5 µmol kg-1), biliverdin (1, 3, or 10 mg kg-1), or ZnPP-IX (1, 3 or 9 mg kg-1) were injected (s.c.) 60 min before zymosan. ODQ (12.5 µmol kg-1; s.c.) or glibenclamide (10 mg kg-1; i.p.) was administered 1 h and 30 min prior to DMDC (2.5 µmol kg-1; s.c), respectively. RESULTS: Hemin (1 mg kg-1), DMDC (2.5 µmol kg-1), and BVD (10 mg kg-1) reduced hypernociception and leukocyte migration, which ZnPP (3 mg kg-1) enhanced. The effects of DMDC were counteracted by ODQ and glibenclamide. The HO-1, TNF-α, and IL-1ß mRNA expression and immunolabelling increased. CONCLUSIONS: HO-1/BVD/CO pathway activation provides anti-nociceptive and anti-inflammatory effects on the zymosan-induced TMJ hypernociception in rats.


Subject(s)
Biliverdine/physiology , Carbon Monoxide/physiology , Cyclic GMP , Heme Oxygenase-1/physiology , KATP Channels , Nociception/drug effects , Signal Transduction/drug effects , Animals , Arthritis/chemically induced , Biliverdine/genetics , Cytokines/metabolism , Down-Regulation/drug effects , Heme Oxygenase-1/genetics , Male , Pain Threshold , Peroxidase/metabolism , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Temporomandibular Joint Disorders/chemically induced , Temporomandibular Joint Disorders/pathology , Trigeminal Ganglion/drug effects , Zymosan
2.
Free Radic Biol Med ; 97: 466-477, 2016 08.
Article in English | MEDLINE | ID: mdl-27387768

ABSTRACT

The accumulation of bile acids affects mitochondria causing oxidative stress. Antioxidant defense is accepted to include biotransformation of biliverdin (BV) into bilirubin (BR) through BV reductase α (BVRα). The mutation (c.214C>A) in BLVRA results in a non-functional enzyme (mutBVRα). Consequently, homozygous carriers suffering from cholestasis develop green jaundice. Whether BVRα deficiency reduces BV-dependent protection against bile acids is a relevant question because a screening of the mut-BLVRA allele (a) in 311 individuals in Greenland revealed that this SNP was relatively frequent in the Inuit population studied (1% a/a and 4.5% A/a). In three human liver cell lines an inverse correlation between BVRα expression (HepG2>Alexander>HuH-7) and basal reactive oxygen species (ROS) levels was found, however the ability of BV to reduce oxidative stress and cell death induced by deoxycholic acid (DCA) or potassium dichromate (PDC) was similar in these cells. The transduction of BVRα or mutBVRα in human placenta JAr cells with negligible BVRα expression or the silencing of endogenous BVRα expression in liver cells had no effect on DCA-induced oxidative stress and cell death or BV-mediated cytoprotection. DCA stimulated both superoxide anion and hydrogen peroxide production, whereas BV only inhibited the latter. DCA and other dihydroxy-bile acids, but not PDC, induced up-regulation of both BVRα and heme oxygenase-1 (HO-1) in liver cells through a FXR independent and BV insensitive mechanism. In conclusion, BV exerts direct and BVRα-independent antioxidant and cytoprotective effects, whereas bile acid accumulation in cholestasis stimulates the expression of enzymes favoring the heme biotransformation into BV and BR.


Subject(s)
Biliverdine/physiology , Deoxycholic Acid/physiology , Oxidative Stress , Animals , Biliverdine/pharmacology , Cholestasis/metabolism , Deoxycholic Acid/pharmacology , Free Radical Scavengers/pharmacology , Gene Expression , HEK293 Cells , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Potassium Dichromate/pharmacology , Protective Factors , Reactive Oxygen Species/metabolism
3.
Antioxid Redox Signal ; 20(11): 1789-809, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24131232

ABSTRACT

SIGNIFICANCE: Heme oxygenases (HO-1 and HO-2) catalyze the degradation of the pro-oxidant heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. In the vasculature, particular interest has focused on antioxidant and anti-inflammatory properties of the inducible HO-1 isoform in the vascular endothelium. This review will present evidence that illustrates the potential therapeutic significance of HO-1 and its products, with special emphasis placed on their beneficial effects on the endothelium in vascular diseases. RECENT ADVANCES: The understanding of the molecular basis for the regulation and functions of HO-1 has led to the identification of a variety of drugs that increase HO-1 activity in the vascular endothelium. Moreover, therapeutic delivery of HO-1 products CO, biliverdin, and bilirubin has been shown to have favorable effects, notably on endothelial cells and in animal models of vascular disease. CRITICAL ISSUES: To date, mechanistic data identifying the downstream target genes utilized by HO-1 and its products to exert their actions remain relatively sparse. Likewise, studies in man to investigate the efficacy of therapeutics known to induce HO-1 or the consequences of the tissue-specific delivery of CO or biliverdin/bilirubin are rarely performed. FUTURE DIRECTIONS: Based on the promising in vivo data from animal models, clinical trials to explore the safety and efficacy of the therapeutic induction of HO-1 and the delivery of its products should now be pursued further, targeting, for example, patients with severe atherosclerotic disease, ischemic limbs, restenosis injury, or at high risk of organ rejection.


Subject(s)
Endothelium, Vascular/enzymology , Heme Oxygenase-1/physiology , Animals , Atherosclerosis/enzymology , Atherosclerosis/therapy , Bilirubin/physiology , Biliverdine/physiology , Carbon Monoxide/physiology , Endothelial Cells/enzymology , Endothelium, Vascular/pathology , Enzyme Induction , Humans , Neovascularization, Physiologic
4.
Annu Rev Pharmacol Toxicol ; 50: 323-54, 2010.
Article in English | MEDLINE | ID: mdl-20055707

ABSTRACT

Heme oxygenases (HO) catabolize free heme, that is, iron (Fe) protoporphyrin (IX), into equimolar amounts of Fe(2+), carbon monoxide (CO), and biliverdin. The stress-responsive HO-1 isoenzyme affords protection against programmed cell death. The mechanism underlying this cytoprotective effect relies on the ability of HO-1 to catabolize free heme and prevent it from sensitizing cells to undergo programmed cell death. This cytoprotective effect inhibits the pathogenesis of a variety of immune-mediated inflammatory diseases.


Subject(s)
Cytoprotection , Heme Oxygenase-1/physiology , Animals , Apoptosis/drug effects , Biliverdine/physiology , Carbon Monoxide/physiology , Gene Expression Regulation, Enzymologic , Heme/metabolism , Heme/toxicity , Heme Oxygenase-1/genetics , Humans , Inflammation/prevention & control , Tumor Necrosis Factor-alpha/pharmacology
5.
Nephron Exp Nephrol ; 104(4): e135-9, 2006.
Article in English | MEDLINE | ID: mdl-16902317

ABSTRACT

Heme oxygenase (HO) isoforms catalyze the conversion of heme to carbon monoxide (CO) and biliverdin/bilirubin with a concurrent release of iron. There is strong evidence that HO activity and products play a major role in renoprotection, however the exact molecular mechanisms underlying the beneficial effects exerted by this pathway are not fully understood. This review is aimed at illustrating the possible mechanism/s by which HO is renoprotective in the context of ischemia/reperfusion. We will first analyze the effects of exogenous administration of bilirubin/biliverdin and CO and then describe their biological activities once generated endogenously following stimulation of the HO pathway by either pharmacological means or gene targeting-mediated approaches.


Subject(s)
Biliverdine/physiology , Carbon Monoxide/physiology , Heme Oxygenase (Decyclizing)/metabolism , Kidney/blood supply , Reperfusion Injury/physiopathology , Animals , Bilirubin/physiology , Humans
6.
Inflamm Bowel Dis ; 11(4): 350-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15803024

ABSTRACT

BACKGROUND: Heme oxygenase-1 (HO-1) seems to have an important protective role in acute and chronic inflammation. The products of heme catalysis, biliverdin/bilirubin, carbon monoxide (CO), and iron (that induces apoferritin) mediate the beneficial effects of HO-1. Blockade of HO-1 activity results in exacerbation of experimental colitis. We tested whether HO-1 has protective effects in the development of colitis and determined that specific enzymatic products of HO-1 are responsible for these effects. METHODS: Colitis was induced by oral administration of dextran sodium sulfate (5%) to C57BL/6 mice for 7 days. HO-1 was up-regulated by cobalt-protoporphyrin (5 mg/kg, intraperitoneally). Biliverdin, exogenous CO, or the iron chelator desferrioxamine was administered to other groups. RESULTS: Cobalt-protoporphyrin treatment resulted in significant up-regulation of HO-1 protein in mucosal and submucosal cells. Induction of HO-1 was associated with significantly less loss of body weight in mice with induced colitis (-12% versus -22% in the control animals, P < 0.001). Development of diarrhea and gastrointestinal hemorrhage was substantially delayed in animals in which HO-1 was induced, and mucosal injury was significantly attenuated. Administration of CO or desferrioxamine alone had no significant effects, whereas enhanced protection with lesser evidence of bowel inflammation was observed with systemic biliverdin administration (50 micromol/kg, 3 times per day, intraperitoneally). CONCLUSIONS: We conclude that heightened HO-1 expression or administration of biliverdin ameliorates dextran sodium sulfate-induced experimental colitis. Novel therapeutic strategies based on HO-1 and/or biliverdin administration may have use in inflammatory bowel disease.


Subject(s)
Biliverdine/physiology , Colitis/enzymology , Colitis/prevention & control , Heme Oxygenase (Decyclizing)/physiology , Acute Disease , Animals , Colitis/pathology , Disease Models, Animal , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/drug effects , Heme Oxygenase-1 , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Protoporphyrins/pharmacology
7.
Eur J Neurosci ; 17(11): 2249-55, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12814358

ABSTRACT

Previously we found that some cyclopenteone prostaglandin derivatives (PGs), referred to as neurite outgrowth-promoting PGs (NEPPs), have dual biological activities of promoting neurite outgrowth and preventing neuronal death [Satoh et al. (2000) J. Neurochem., 75, 1092-1102; Satoh et al. (2001) J. Neurochem., 77, 50-62; Satoh et al. (2002) In Kikuchi, II. (ed.), Strategenic Medical Science Against Brain Attack. Springer-Verlag, Tokyo, pp. 78-93]. To investigate possible cellular mechanisms of the neuroprotective effects, we performed oligo hybridization-based DNA array analysis with mRNA isolated from HT22, a cell line that originated from a mouse hippocampal neuron. Several transcripts up-regulated by NEPP11 were identified. Because heme oxygenase 1 (HO-1) mRNA was the most prominently induced and was earlier reported to protect neuronal and non-neuronal cells against oxidative stress, we focused on it as a possible candidate responsible for the neuroprotective effects. We found NEPP11 to induce HO-1 protein (32 kDa) in HT22 cells in both the presence and the absence of glutamate, whereas non-neuroprotective prostaglandins (PGs) Delta12-PGJ2 or PGA2 did not. Overexpression of HO-1-green fluorescence protein (GFP) fusion protein significantly protected HT22 cells against oxidative glutamate toxicity, whereas that of GFP alone did not. Furthermore, biliverdin and bilirubin, products of HO-1 enzymatic activity on heme, protected HT22 cells from oxidative glutamate toxicity. These results, together with our previous results, suggest that NEPP11 activates the expression of HO-1 and that HO-1 produces biliverdin and bilirubin, which result in the inhibition of neuronal death induced by oxidative stress. NEPP11 is the first molecular probe reported to have a neuroprotective action through induction of HO-1 in neuronal cells.


Subject(s)
Gene Expression Regulation/drug effects , Heme Oxygenase (Decyclizing)/physiology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/physiology , Prostaglandin D2/analogs & derivatives , Prostaglandins/pharmacology , Animals , Antineoplastic Agents/pharmacology , Bilirubin/physiology , Biliverdine/physiology , Blotting, Western/methods , Cell Line , Cell Survival/drug effects , Cyclopentanes/metabolism , Excitatory Amino Acids/pharmacology , Fluorescent Antibody Technique/methods , Glutamic Acid/pharmacology , Green Fluorescent Proteins , Heme Oxygenase-1 , Luminescent Proteins/metabolism , Membrane Proteins , Mice , Neuroprotective Agents/metabolism , Oligonucleotide Array Sequence Analysis/methods , Oxidative Stress/genetics , Prostaglandin D2/pharmacology , Prostaglandins/genetics , Prostaglandins/metabolism , Prostaglandins/physiology , Prostaglandins A/agonists , Prostaglandins A/pharmacology , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Transfection/methods
9.
Transpl Immunol ; 9(2-4): 227-33, 2002 May.
Article in English | MEDLINE | ID: mdl-12180835

ABSTRACT

Much interest has recently been focused on the physiological/pathological role of the heme oxygenase (HO) system, the rate-limiting step in the conversion of heme, in inflammatory events. The HO system may be instrumental in mediating a number of cytoprotective effects, because of its end products, biliverdin, carbon monoxide (CO) and ferrous free iron (Fe2+). As each of the byproducts acts dependently and/or co-operatively with each other, their in vivo effects are complex. In general, the HO system is thought to exert three major functions in ischemia/reperfusion injury: (1) anti-oxidant effects; (2) maintenance of microcirculation; and (3) modulatory effects upon the cell cycle. The anti-oxidant functions depend on heme degradation, oxygen consumption and the production of biliverdin/ferritin via iron accumulation. On the other hand, the production of CO, which has vasodilatory and anti-platelet aggregative properties, can maintain tissue microcirculation. Strikingly, CO may also be instrumental in anti-apoptotic and cell arrest mechanisms. The HO system prevents early injury in the re-perfused organ, and inhibits the function of immune reactive cells, such as neutrophils, macrophages and lymphocytes. The role of the HO system as a novel strategy to mitigate an antigen-independent ischemia/reperfusion injury has been documented in a number of transplantation models.


Subject(s)
Cytoprotection/physiology , Heme Oxygenase (Decyclizing)/physiology , Ischemia/prevention & control , Reperfusion Injury/prevention & control , Animals , Biliverdine/physiology , Carbon Monoxide/physiology , Enzyme Induction , Free Radicals , Heme/metabolism , Heme Oxygenase-1 , Humans , Iron/metabolism , Membrane Proteins , Oxygen/metabolism
10.
Biochemistry ; 41(31): 10115-22, 2002 Aug 06.
Article in English | MEDLINE | ID: mdl-12146976

ABSTRACT

Biliverdin is required for Xenopus laevis embryo dorsal axis formation. When the tetrapyrrole is inactivated by phototransforming it with ultraviolet light prior to the first division, the embryo fails to synthesize dorsal mRNAs, such as goosecoid or chordin, yet forms increased amounts of ventral transcripts, such as Vent 1, and, consequently, develops ventralized morphology. Here we describe the metabolism of biliverdin during oogenesis and early embryogenesis. Estrogen induces frog hepatocytes to synthesize biliverdin and vitelogenin. The two molecules form a complex that is secreted into and transported in the plasma to be taken up by the oocyte as it matures through its six stages of oogenesis. In the oocyte, the biliverdin-vitellogenin complex is processed and stored in the yolk platelets. In these organelles, biliverdin is associated entirely with the lipovitellin domain of the processed vitellogenin. Once the egg is fertilized, its biliverdin content decreases over a 5-6 h period to participate in the chemical machinery required for dorsal axis formation. This participation must be initiated during the period encompassing the first embryonic mitosis. The results describe the pathway that generates, transports, and stores biliverdin as part of oogenesis, define the time course for its utilization after fertilization, and link biliverdin to the metabolism of the phosphoglycolipometalloprotein, vitellogenin.


Subject(s)
Biliverdine/physiology , Embryonic Development , Oogenesis/physiology , Animals , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Spectrophotometry, Ultraviolet , Xenopus laevis
11.
Proc Natl Acad Sci U S A ; 99(1): 251-6, 2002 Jan 08.
Article in English | MEDLINE | ID: mdl-11782548

ABSTRACT

The determinants of Xenopus laevis embryos that act before their first cell division are mandatory for the formation of mRNas required to establish the dorsal axis. Although their chemical identities are unknown, a number of their properties have long been recognized. One of the determinants is present in the cytoplasm and is sensitive to UV light. Thus, exposing stage 1 embryos to either standard 254-nm or, as shown here, to 366-nm UV light during the 0.3-0.4 time fraction of their first cycle inactivates the cytoplasmic determinant. As a consequence, both types of irradiated embryos fail to express dorsal markers, e.g., goosecoid and chordin, without affecting formation of ventral markers, e.g., Vent-1. The developmental outcome is dorsal axis-deficient morphology. We report here that biliverdin IXalpha, a normal constituent of cytoplasmic yolk platelets, is photo-transformed by irradiation with either 254- or 366-nm UV light and that the transformation triggers the dorsal axis deficiency. When the 254- or 366-nm UV-irradiated embryos, fated to dorsal axis deficiency, are incubated solely with microM amounts of biliverdin, they recover and form the axis. In contrast, incubation with either in vitro photo-transformed biliverdin or biliverdin IXalpha dimethyl ester does not induce recovery. The results define an approach to produce dorsal axis-deficient embryos by photo-transforming its biliverdin by irradiation with 366-nm UV light and identify an unsuspected role for biliverdin IXalpha in X. laevis embryogenesis.


Subject(s)
Biliverdine/physiology , Embryo, Nonmammalian/metabolism , Animals , Chromatography, High Pressure Liquid , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Mitosis , Models, Chemical , Time Factors , Ultraviolet Rays , Xenopus laevis
12.
Nihon Yakurigaku Zasshi ; 114(5): 255-64, 1999 Nov.
Article in Japanese | MEDLINE | ID: mdl-10621938

ABSTRACT

One of the most remarkable revolutions during the history of animal kingdom is the adaptation to oxygen, a highly toxic gas produced by plants. Based on the high affinity of heme toward oxygen, it has been believed that heme itself and/or hemoprotein should play significant roles in the processes of adaptation. Recently, two novel functions of heme and its metabolites were identified; namely, hemoprotein is an oxygen sensor and biliverdin and bilirubin, catabolites of heme, are antioxidants. Thus, heme is a key molecule in the responses to environmental stress, including oxygen. Although activation of hypoxia inducible factor-1, which induces expression of genes encoding erythropoietin and heme oxygenase (HO-1), is generally accepted to be controlled by oxygen sensor, the precise signaling pathway has not yet been well elucidated. Various stresses such as hypoxia are known to activate the HO-1 gene, the key enzyme of heme degradation, resulting in the marked conversion of pro-oxidant (heme) into its antioxidative catabolites. The induction is, therefore, supposed to have protective effects. Recent reports on HO-1 deficiency also support this hypothesis that the activation of the HO-1 gene is one of the most important defense mechanisms against environmental stress.


Subject(s)
Heme/physiology , Oxidative Stress , Animals , Antioxidants , Bilirubin/physiology , Biliverdine/physiology , Humans , Hypoxia/metabolism
13.
Biochim Biophys Acta ; 1158(2): 189-93, 1993 Oct 03.
Article in English | MEDLINE | ID: mdl-8399320

ABSTRACT

Bile pigments possess an anti-complement property and could be involved in tissue protection. In this study, we examined the physiological actions of bile pigments, which had been generally regarded as waste catabolites. Biliverdin inhibited complement cascade reactions in vitro, especially at the C1 step in the classical pathway at low micromolar concentrations. Further, Forssman anaphylaxis in guinea pigs, being closely associated with complement reactions, was inhibited by oral or intravenous administration of biliverdin. Conjugated bilirubin also showed an inhibitory effect on complement-dependent reactions in vitro. From these observations, we propose a hypothesis that the pigments serve as endogenous tissue protectors by multiple lines of mechanisms including antioxidant and anti-complement actions.


Subject(s)
Bile Pigments/physiology , Biliverdine/physiology , Inflammation/prevention & control , Anaphylaxis , Animals , Bile Pigments/administration & dosage , Biliverdine/administration & dosage , Cattle , Guinea Pigs
14.
Comp Biochem Physiol B ; 97(1): 37-45, 1990.
Article in English | MEDLINE | ID: mdl-2253479

ABSTRACT

1. The blue-green coloration of the blood plasma in some marine fishes, which is attributed to a protein bound tetrapyrrol (biliverdin), is an anomaly in vertebrates. 2. Recent studies have shown that biliverdin not only occurs in many fish, but is also present in the blood of tobacco hornworm, the wings of moth and butterfly, the shell of bird eggs, the serum and egg of frog, the placenta of dog and in the blood of humans suffering from hepatic diseases. 3. In this review, we begin with a historical account of the description of the presence of blue-green blood plasma in fish, and then consider the biochemistry, metabolism, physiology, and the ecological implications of biliverdin in fish. 4. A comparative description of the occurrence of biliverdin in fish and other animals is presented. 5. The mechanism of accumulation of biliverdin in fish blood and its evolutionary significance are also considered. It is suggested that this process may serve as a useful model for further research on bile pigment metabolism in other animals.


Subject(s)
Biliverdine/blood , Fishes/blood , Animals , Biliverdine/metabolism , Biliverdine/physiology , Blood Chemical Analysis , Pigmentation
15.
Biochem Biophys Res Commun ; 122(1): 40-6, 1984 Jul 18.
Article in English | MEDLINE | ID: mdl-6547608

ABSTRACT

Treatment of rats with biliverdin (48 h) resulted in an increase in microsomal heme oxygenase activity in the liver. This was accompanied by decreases in the microsomal heme and cytochrome P-450 contents. In these respects cellular responses elicited by biliverdin resembled those produced by hematin (48 h). When rats were treated with biliverdin for a short interval (3 h) an inhibition of the activities of heme oxygenase and biliverdin reductase, concomitant with an increase in microsomal heme and cytochrome P-450 contents, were observed. Hematin was ineffective in altering these parameters under similar conditions. Biliverdin, in a concentration-dependent manner, inhibited the activities of purified heme oxygenase and biliverdin reductase. The activity of purified rat liver heme oxygenase was refractory to bilirubin, whereas that of purified biliverdin reductase was severely inhibited. It is suggested that biliverdin regulates cellular heme degradation processes by occupying the heme binding site on heme oxygenase, thus hindering the access of the substrate to the catalytic site of the enzyme.


Subject(s)
Bilirubin/analogs & derivatives , Biliverdine/physiology , Heme Oxygenase (Decyclizing)/metabolism , Heme/metabolism , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Oxidoreductases Acting on CH-CH Group Donors , 5-Aminolevulinate Synthetase/metabolism , Animals , Cytochrome P-450 Enzyme Inhibitors , Hemin/physiology , In Vitro Techniques , Male , Oxidoreductases/metabolism , Rats , Rats, Inbred Strains
16.
Rev Esp Fisiol ; 39(2): 183-92, 1983 Jun.
Article in Spanish | MEDLINE | ID: mdl-6622809

ABSTRACT

Biliverdin has been proposed as the biological signal that triggers liver regeneration after partial hepatectomy, since a transitory increase of plasma levels is found shortly after partial hepatectomy. We have carried out a detailed study to establish the feasibility of such a hypothesis. When biliverdin is administered i.p., it binds to albumin. This became evident by electrophoresis and affinity chromatography. Biliverdin disappears from the peritoneal cavity following a 1st order kinetics (t 1/2 = 40 min, K = 0.0175 min -1). Biliverdin reductase activity in the peritoneal exudate was high (1.04 mumol Bv min -1/mg protein), and very small amounts of biliverdin in comparison to the administered dose were found in plasma. However, unconjugated bilirubin level was high in plasma and the conjugated bilirubin in bile flux reached its maximum 90 min after administration. On the basis of blood elimination of biliverdin when administered intravenously (t 1/2 = 11 min, K = 0.068 min -1), a series of experiments was designed to reproduce the levels of plasma biliverdin as found shortly after partial hepatectomy. Our results showed a total absence of liver regeneration. There is, however, increase of the mitotic index if biliverdin is administered intraperitoneally. This effect is not directly related to biliverdin but to unspecific (?) stimulation of peritoneal cavity since the same effect was obtained after administration of unrelated substances (diatomaceous earth, killed bacteria). The mitotic figures did not incorporate 3H-thymidine, and it is suggested that those cells came from a G2 blocked quiescent cell population in liver. We therefore conclude that biliverdin is not the physiological trigger of liver regeneration.


Subject(s)
Bilirubin/analogs & derivatives , Biliverdine/physiology , Liver Regeneration , Oxidoreductases Acting on CH-CH Group Donors , Albumins/metabolism , Animals , Ascitic Fluid/metabolism , Biliverdine/administration & dosage , Biliverdine/pharmacology , Hepatectomy , Injections, Intraperitoneal , Liver/drug effects , Mitotic Index/drug effects , Oxidoreductases/metabolism , Rats , Rats, Inbred Strains
17.
Arch Biochem Biophys ; 221(2): 590-2, 1983 Mar.
Article in English | MEDLINE | ID: mdl-6838212

ABSTRACT

Pterobilin 1 (biliverdin IX gamma), a butterfly bile pigment, is photocyclized into phorcabilin 2 and sarpedobilin 3 by irradiation in visible light. Selective irradiations have now been performed at the absorption maxima of pterobilin 1. The 650-nm radiations are responsible for the observed photocyclizations while the 375-nm radiations lead to decomposition products. These results are discussed in connection with a hypothesis concerning the biological role of pterobilin in butterfly larvae.


Subject(s)
Bile Pigments/radiation effects , Bilirubin/analogs & derivatives , Biliverdine/radiation effects , Animals , Bile Pigments/physiology , Biliverdine/physiology , Butterflies , Light
SELECTION OF CITATIONS
SEARCH DETAIL
...