1
|
Saito Y, Sugiura Y, Sakaguchi A, Sada T, Nishiyama C, Maeda R, Kaneko M, Kiyonari H, Kimura W. Redox-dependent purine degradation triggers postnatal loss of cardiac regeneration potential. Redox Biol 2025; 79:103442. [PMID: 39637598 PMCID: PMC11664147 DOI: 10.1016/j.redox.2024.103442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/23/2024] [Indexed: 12/07/2024] Open
Abstract
Postnatal cardiomyocyte cell cycle withdrawal is a critical step wherein the mammalian heart loses regenerative potential after birth. Here, we conducted interspecies multi-omic comparisons between the mouse heart and that of the opossum, which have different postnatal time-windows for cardiomyocyte cell cycle withdrawal. Xanthine metabolism was activated in both postnatal hearts in parallel with cardiomyocyte cell cycle arrest. The pentose phosphate pathway (PPP) which produces NADPH was found to decrease simultaneously. Postnatal myocardial tissues became oxidized accordingly, and administration of antioxidants to neonatal mice altered the PPP and suppressed the postnatal activation of cardiac xanthine metabolism. These results suggest a redox-driven postnatal switch from purine synthesis to degradation in the heart. Importantly, inhibition of xanthine metabolism in the postnatal heart extended postnatal duration of cardiomyocyte proliferation and maintained postnatal heart regeneration potential in mice. These findings highlight a novel role of xanthine metabolism as a redox-dependent metabolic regulator of cardiac regeneration potential.
Collapse
Affiliation(s)
- Yuichi Saito
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Yuki Sugiura
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan; Human Biology Microbiome Quantum Research Center (WPI-Bio2Q), Keio University School of Medicine, Tokyo, Japan
| | - Akane Sakaguchi
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Tai Sada
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Chihiro Nishiyama
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Rae Maeda
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Wataru Kimura
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| |
Collapse
|
2
|
Vora B, Brackman DJ, Zou L, Garcia-Cremades M, Sirota M, Savic RM, Giacomini KM. Oxypurinol pharmacokinetics and pharmacodynamics in healthy volunteers: Influence of BCRP Q141K polymorphism and patient characteristics. Clin Transl Sci 2021; 14:1431-1443. [PMID: 33931953 PMCID: PMC8301548 DOI: 10.1111/cts.12992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/23/2020] [Accepted: 01/14/2021] [Indexed: 12/30/2022] Open
Abstract
The missense variant, breast cancer resistance protein (BCRP) p.Q141K, which encodes a reduced function BCRP, has been linked to poor response to allopurinol. Using a multifaceted approach, we aimed to characterize the relationship(s) between BCRP p.Q141K, the pharmacokinetics (PK) and pharmacodynamics (PD) of oxypurinol (the active metabolite of allopurinol), and serum uric acid (SUA) levels. A prospective clinical study (NCT02956278) was conducted in which healthy volunteers were given a single oral dose of 300 mg allopurinol followed by intensive blood sampling. Data were analyzed using noncompartmental analysis and population PK/PD modeling. Additionally, electronic health records were analyzed to investigate whether clinical inhibitors of BCRP phenocopied the effects of the p.Q141K variant with respect to SUA. Subjects homozygous for p.Q141K had a longer half‐life (34.2 ± 12.2 h vs. 19.1 ± 1.42 h) of oxypurinol. The PK/PD model showed that women had a 24.8% lower volume of distribution. Baseline SUA was affected by p.Q141K genotype and renal function; that is, it changed by 48.8% for every 1 mg/dl difference in serum creatinine. Real‐world data analyses showed that patients prescribed clinical inhibitors of BCRP have higher SUA levels than those that have not been prescribed inhibitors of BCRP, consistent with the idea that BCRP inhibitors phenocopy the effects of p.Q141K on uric acid levels. This study identified important covariates of oxypurinol PK/PD that could affect its efficacy for the treatment of gout as well as a potential side effect of BCRP inhibitors on increasing uric acid levels, which has not been described previously.
Collapse
Affiliation(s)
- Bianca Vora
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Deanna J Brackman
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Ling Zou
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Maria Garcia-Cremades
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
| | - Radojka M Savic
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Al-Shehri SS, Duley JA, Bansal N. Xanthine oxidase-lactoperoxidase system and innate immunity: Biochemical actions and physiological roles. Redox Biol 2020; 34:101524. [PMID: 32334145 PMCID: PMC7183230 DOI: 10.1016/j.redox.2020.101524] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 02/01/2023] Open
Abstract
The innate immune system in mammals is the first-line defense that plays an important protective role against a wide spectrum of pathogens, especially during early life before the adaptive immune system develops. The enzymes xanthine oxidase (XO) and lactoperoxidase (LPO) are widely distributed in mammalian tissues and secretions, and have a variety of biological functions including in innate immunity, provoking much interest for both in vitro and in vivo applications. The enzymes are characterized by their generation of reactive oxygen and nitrogen species, including hydrogen peroxide, hypothiocyanite, nitric oxide, and peroxynitrite. XO is a major generator of hydrogen peroxide and superoxide that subsequently trigger a cascade of oxidative radical pathways, including those produced by LPO, which have bactericidal and bacteriostatic effects against pathogens including opportunistic bacteria. In addition to their role in host microbial defense, reactive oxygen and nitrogen species play important physiological roles as second messenger cell signaling molecules, including cellular proliferation, differentiation and gene expression. There are several indications that the reactive species generated by peroxide have positive effects on human health, particularly in neonates; however, some important in vivo aspects of this system remain obscure. The primary dependence of the system on hydrogen peroxide has led us to propose it is particularly relevant to neonate mammals during milk feeding.
Collapse
Affiliation(s)
- Saad S Al-Shehri
- College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia.
| | - John A Duley
- School of Pharmacy, The University of Queensland, St Lucia, 4102, Australia
| | - Nidhi Bansal
- School of Pharmacy, The University of Queensland, St Lucia, 4102, Australia; School of Agriculture and Food Science, The University of Queensland, St Lucia, 4102, Australia
| |
Collapse
|
4
|
Poles MZ, Bódi N, Bagyánszki M, Fekete É, Mészáros AT, Varga G, Szűcs S, Nászai A, Kiss L, Kozlov AV, Boros M, Kaszaki J. Reduction of nitrosative stress by methane: Neuroprotection through xanthine oxidoreductase inhibition in a rat model of mesenteric ischemia-reperfusion. Free Radic Biol Med 2018; 120:160-169. [PMID: 29550332 DOI: 10.1016/j.freeradbiomed.2018.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 01/01/2023]
Abstract
Our aim was to characterize the main components of the nitrosative response with quantitative changes of the nitrergic myenteric neurons in adjacent intestinal segments after transient superior mesenteric artery occlusion. We also tested the hypothesis that exogenous methane may modulate the evolution of nitroxidation by influencing xanthine oxidoreductase (XOR) activity. The microcirculatory consequences of a 50 min ischemia or ischemia-reperfusion were investigated in anesthetized rats (n = 124) inhaling normoxic air with or without 2.2% methane. XOR activities, nitrogen monoxide (NO), nitrite/nitrate (NOx), and nitrotyrosine levels were measured, together with relative nitrergic neuron ratios from duodenum, ileum and colon samples. The effects of methane on XOR were also examined in vitro. The intramural flow stopped only in the ileum during ischemia. The highest baseline XOR activity was found in the duodenum, which increased further during ischemia. NO and nitrotyrosine levels rose, and the nNOS-immunopositive neuron ratio and NOx level both dropped. Reperfusion uniformly elevated XOR activity and nitrotyrosine formation, with the highest level attained in the duodenum, where the nitrergic neuron ratio remained depressed. These alterations were eliminated in methane-treated animals, XOR activity and nitrotyrosine formation decreased in all sites, and the duodenal nitrergic neuron ratio was re-established. The inhibitory effect of methane on XOR-linked nitrate reductase activity was also demonstrated in vitro. With segment-specific microcirculatory alterations, the risk for nitrosative stress is highest in transiently hypoxic tissues with high endogenous XOR activities. The XOR-inhibitory effect of methane can reduce nitroxidation and protects the nitrergic neuron population in such conditions.
Collapse
Affiliation(s)
- Marietta Zita Poles
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52., H-6726, Szeged, Hungary.
| | - Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52., H-6726, Szeged, Hungary.
| | - Éva Fekete
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52., H-6726, Szeged, Hungary.
| | - András Tamás Mészáros
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Gabriella Varga
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Szilárd Szűcs
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Anna Nászai
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Liliána Kiss
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Andrey V Kozlov
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, Vienna 1200, Austria.
| | - Mihály Boros
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - József Kaszaki
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| |
Collapse
|
5
|
Badri N, Florea A, Mhamdi M, Matei H, Tekaya WH, Bâati R, Maghraoui S, Tekaya L. Toxicological effects and ultrastructural changes induced by lanthanum and cerium in ovary and uterus of Wistar rats. J Trace Elem Med Biol 2017; 44:349-355. [PMID: 28965598 DOI: 10.1016/j.jtemb.2017.09.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 11/30/2022]
Abstract
Rare earths have been widely used in a huge number of areas in industry and medicine. Therefore, they exist in the environment and possibly accumulated within the human body. However their effects in the living organism particularly in the female reproductive system are still unclear. In this work, the subcellular behavior of lanthanum and cerium was investigated through the Transmission Electron Microscopy (TEM), in different territories of the reproductive system of Wistar rats exposed intraperitoneally to soluble solution of these elements during 2 weeks. Ultrastructural investigations of ultrathin sections from uterus and ovary of treated rats revealed the existence of inclusions with high electron density and heterogeneous aspects in the lysosomes of uterus and ovary cells. Many disruptions of architecture were observed, accompanied with several changes like vacuolations, significant expansion of the endoplasmic reticulum, mitochondrial alterations and necrotic cells, demonstrating the toxicity of these elements with doses used. Phagolysosomes as well as eosinophils were also seen. Our experimental investigations revealed no intralysosomal inclusions in ultrathin sections of the uterus and ovary of pregnant control females. The original mechanism implicated in this insolubilization-concentration phenomenon of these elements, as non-soluble phosphate form, in the lysosomes is a biochemical one involving intralysosomal hydrolytic enzymes, the acid phosphatase.
Collapse
Affiliation(s)
- Nedra Badri
- Laboratry of Physiology, Faculty of Medecine of Tunis "Université de Tunis EL Manar", 15 Rue Djebel Lakhdhar, 1007, La rabta, Tunis, Tunisia; Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy 6 L. Pasteur St, 400349 Cluj Napoca, Romania.
| | - Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy 6 L. Pasteur St, 400349 Cluj Napoca, Romania.
| | - Maroua Mhamdi
- Laboratry of Physiology, Faculty of Medecine of Tunis "Université de Tunis EL Manar", 15 Rue Djebel Lakhdhar, 1007, La rabta, Tunis, Tunisia; Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy 6 L. Pasteur St, 400349 Cluj Napoca, Romania
| | - Horea Matei
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy 6 L. Pasteur St, 400349 Cluj Napoca, Romania
| | - Walid-Habib Tekaya
- Department of Stomatology, Faculty of Dentistry of Monastir (Université de Monastir-Monastir), Tunisia
| | - Rym Bâati
- Laboratry of Physiology, Faculty of Medecine of Tunis "Université de Tunis EL Manar", 15 Rue Djebel Lakhdhar, 1007, La rabta, Tunis, Tunisia
| | - Samira Maghraoui
- Laboratry of Physiology, Faculty of Medecine of Tunis "Université de Tunis EL Manar", 15 Rue Djebel Lakhdhar, 1007, La rabta, Tunis, Tunisia
| | - Leila Tekaya
- Laboratry of Physiology, Faculty of Medecine of Tunis "Université de Tunis EL Manar", 15 Rue Djebel Lakhdhar, 1007, La rabta, Tunis, Tunisia
| |
Collapse
|
6
|
Deoxynivalenol impairs hepatic and intestinal gene expression of selected oxidative stress, tight junction and inflammation proteins in broiler chickens, but addition of an adsorbing agent shifts the effects to the distal parts of the small intestine. PLoS One 2013; 8:e69014. [PMID: 23922676 PMCID: PMC3724867 DOI: 10.1371/journal.pone.0069014] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 06/10/2013] [Indexed: 01/10/2023] Open
Abstract
Broiler chickens are rather resistant to deoxynivalenol and thus, clinical signs are rarely seen. However, effects of subclinical concentrations of deoxynivalenol on both the intestine and the liver are less frequently studied at the molecular level. During our study, we investigated the effects of three weeks of feeding deoxynivalenol on the gut wall morphology, intestinal barrier function and inflammation in broiler chickens. In addition, oxidative stress was evaluated in both the liver and intestine. Besides, the effect of a clay-based mycotoxin adsorbing agent on these different aspects was also studied. Our results show that feeding deoxynivalenol affects the gut wall morphology both in duodenum and jejenum of broiler chickens. A qRT-PCR analysis revealed that deoxynivalenol acts in a very specific way on the intestinal barrier, since only an up-regulation in mRNA expression of claudin 5 in jejunum was observed, while no effects were seen on claudin 1, zona occludens 1 and 2. Addition of an adsorbing agent resulted in an up-regulation of all the investigated genes coding for the intestinal barrier in the ileum. Up-regulation of Toll-like receptor 4 and two markers of oxidative stress (heme-oxigenase or HMOX and xanthine oxidoreductase or XOR) were mainly seen in the jejunum and to a lesser extent in the ileum in response to deoxynivalenol, while in combination with an adsorbing agent main effect was seen in the ileum. These results suggest that an adsorbing agent may lead to higher concentrations of deoxynivalenol in the more distal parts of the small intestine. In the liver, XOR was up-regulated due to DON exposure. HMOX and HIF-1α (hypoxia-inducible factor 1α) were down-regulated due to feeding DON but also due to feeding the adsorbing agent alone or in combination with DON.
Collapse
|
7
|
Sasaki T, Shimizu T, Koyama T, Sakai M, Uchiyama S, Kawakami S, Noda Y, Shirasawa T, Kojima S. Superoxide dismutase deficiency enhances superoxide levels in brain tissues during oxygenation and hypoxia-reoxygenation. J Neurosci Res 2011; 89:601-10. [PMID: 21280062 DOI: 10.1002/jnr.22581] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 11/24/2010] [Accepted: 11/26/2010] [Indexed: 12/18/2022]
Abstract
To determine whether the mitochondria or cytoplasm produces superoxide during ischemia-reperfusion of the brain, we analyzed lucigenine-enhanced chemiluminescence emission in slices of brain tissue prepared from manganese-superoxide dismutase (Mn-SOD)-deficient (Sod2-deficient) and copper and zinc-superoxide dismutase (Cu,Zn-SOD)-deficient (Sod1-deficient) mice during oxygenation and hypoxia-reoxygenation. The steady-state level of chemiluminescence under oxygenated conditions was significantly enhanced by a lack of either Sod. We hypothesize that the enhanced chemiluminescence produced by Sod2 and Sod1 deficiency reflects in situ superoxide generation in the mitochondria and cytoplasm, respectively. Based on this hypothesis, the major site of intracellular superoxide generation was assumed to be the cytoplasm. However, mitochondria occupy less cellular space than the cytoplasm. In terms of volume, the superoxide concentration is assumed to be higher in mitochondria than in the cytoplasm. Mn-SOD activity was 18% of the Cu,Zn-SOD activity observed in the wild-type mouse brain. However, when mitochondrial SOD activity was expressed as per volume, it was assumed to be equal to that observed in the cytoplasm. This imbalance between superoxide and SOD activity is expected to cause mitochondrial oxidative damage. The chemiluminescence intensity increased significantly during reoxygenation and was enhanced by Sod2 deficiency but was not significantly affected by Sod1 deficiency. The superoxide concentration in the reoxygenated brain would be higher in the mitochondria than in the cytoplasm. The present study indicated that the major site of intracellular superoxide generation in the brain during oxygenation is the cytoplasm, whereas it is the mitochondria during reoxygenation.
Collapse
Affiliation(s)
- Toru Sasaki
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Ishiyama F, Iijima K, Asanuma K, Ara N, Yoshitake J, Abe Y, Koike T, Imatani A, Ohara S, Shimosegawa T. Exogenous luminal nitric oxide exacerbates esophagus tissue damage in a reflux esophagitis model of rats. Scand J Gastroenterol 2009; 44:527-37. [PMID: 19172433 DOI: 10.1080/00365520802699260] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Cytotoxic concentrations of nitric oxide are generated luminally at the gastroesophageal junction through the entero-salivary recirculation of dietary nitrate in humans. The site of luminal nitric oxide generation shifts to the lower esophagus when gastric acid is refluxed into the esophagus. The aim of this study was to investigate the influence of persistent administration of exogenous nitric oxide on esophageal damage. MATERIAL AND METHODS 0.1% sodium nitrite and/or 1% ascorbic acid was administered in an established rat acid-refluxed esophagitis model. Co-administration of both reactants in this model is thought to induce high concentrations of nitric oxide luminally in the esophagus by an acid-catalyzed chemical reaction when refluxed gastric acid is present. The tissue damage was evaluated by a macroscopic lesion index and myeloperoxidase activity. Nitrotyrosin was assessed immunohistochemically as a footprint of peroxynitrite formation. RESULTS Co-administration of sodium nitrite and ascorbic acid induced a 4- to 5-fold increase in the esophageal damage compared with baseline reflux esophagitis, while the damage was unchanged when either of the reagents alone was given. Nitrotyrosine was strongly stained in the tissue from the co-administration. Treatment of superoxide scavengers efficiently prevented the exacerbation of esophageal damage by exogenous nitric oxide exposure, suggesting an essential role of superoxide in esophageal damage. CONCLUSIONS Exogenous luminal nitric oxide greatly exacerbated the tissue damage of reflux esophagitis. Diffusion of the luminal nitric oxide into the adjacent superoxide-enriched inflamed tissue of the esophagus could lead to the production of the highly toxic agent peroxynitrite, thus causing exacerbation of the esophageal damage.
Collapse
Affiliation(s)
- Fumitake Ishiyama
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
|
10
|
|
11
|
Atmani D, Baghiani A, Harrison R, Benboubetra M. NADH oxidation and superoxide production by caprine milk xanthine oxidoreductase. Int Dairy J 2005. [DOI: 10.1016/j.idairyj.2004.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
12
|
Abstract
Xanthine oxidoreductase (XOR) is a major protein component of the milk fat globule membrane (MFGM) surrounding fat droplets in milk and its enzymology is well characterised. The enzyme is widely distributed in mammalian tissues and is generally accepted to be a key enzyme of purine catabolism. It catalyses the oxidation of a wide range of substrates and can pass electrons to molecular oxygen, generating reactive oxygen species (ROS); similar reduction of nitrite yields reactive nitrogen species (RNS). While XOR has been implicated in ischemia-reperfusion injury, its involvement in normal physiological processes has been less studied. It is argued here that XOR-derived ROS and RNS play a role in innate immunity, specifically in the inflammatory response and in anti-microbial defense of the gastrointestinal tract. XOR-derived species could also be involved in signalling. Additionally, XOR is likely to play a part in metabolism of xenobiotics and has recently been shown to mediate the secretion of milk fat globules. The human enzyme has only relatively recently been characterized. The enzyme purified from breast milk shows very low enzymatic activity, and it is suggested that human XOR has evolved so as to be regulated by an exceptional range of pre- and posttranslational mechanisms.
Collapse
Affiliation(s)
- Roger Harrison
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom.
| |
Collapse
|
13
|
Martin HM, Moore KP, Bosmans E, Davies S, Burroughs AK, Dhillon AP, Tosh D, Harrison R. Xanthine oxidoreductase is present in bile ducts of normal and cirrhotic liver. Free Radic Biol Med 2004; 37:1214-23. [PMID: 15451061 DOI: 10.1016/j.freeradbiomed.2004.06.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2004] [Revised: 06/22/2004] [Accepted: 06/24/2004] [Indexed: 11/21/2022]
Abstract
Xanthine oxidoreductase (XOR) is a widely distributed enzyme, involved in the metabolism of purines, which generates superoxide and is thought to be involved in free radical-generated tissue injury. It is present at high concentrations in the liver, from where it may be released during liver injury into the circulation, binding to vascular endothelium and causing vascular dysfunction. The cellular localization of the enzyme, essential to understanding its function, is, however, still debated. The present study has used a highly specific mouse monoclonal antibody to define the cellular distribution of XOR in normal and cirrhotic human liver. As shown previously, XOR is present in hepatocytes. However, the novel finding of this study is that XOR is present in bile duct epithelial cells, where it is concentrated toward the luminal surface. Moreover, in liver disease, proliferating bile ducts are also strongly positive for XOR. These findings suggest that the enzyme is secreted into bile, and this was confirmed by analysis of human and rat bile. Xanthine oxidase activity was 10 to 20-fold higher in liver tissue obtained from patients with liver disease, than in healthy liver. We conclude that XOR is expressed primarily in hepatocytes, but is also present in bile duct epithelial cells and is secreted into bile. Its role in bile is unknown but it may be involved in innate immunity of the bowel muscosa.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Bile/enzymology
- Bile Ducts/enzymology
- Blotting, Western
- Cell Polarity
- Cholangitis, Sclerosing/enzymology
- Electrophoresis, Polyacrylamide Gel
- Epithelial Cells/enzymology
- Hepatitis C/enzymology
- Hepatocytes/enzymology
- Humans
- Hyperoxaluria, Primary/enzymology
- Immunoenzyme Techniques
- Liver Cirrhosis/enzymology
- Liver Cirrhosis/etiology
- Liver Cirrhosis, Alcoholic/enzymology
- Liver Cirrhosis, Biliary/enzymology
- Liver Diseases/enzymology
- Liver Diseases, Alcoholic/enzymology
- Mice
- Mice, Inbred BALB C
- Microscopy, Confocal
- Rats
- Rats, Sprague-Dawley
- Xanthine Oxidase/analysis
- Xanthine Oxidase/immunology
- Xanthine Oxidase/metabolism
- Xanthine Oxidase/physiology
Collapse
Affiliation(s)
- Hannah M Martin
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Martin HM, Hancock JT, Salisbury V, Harrison R. Role of xanthine oxidoreductase as an antimicrobial agent. Infect Immun 2004; 72:4933-9. [PMID: 15321984 PMCID: PMC517476 DOI: 10.1128/iai.72.9.4933-4939.2004] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Hannah M Martin
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | | | | | | |
Collapse
|
15
|
Schellens JPM, Vreeling-Sindelárová H, Frederiks WM. Electron microscopical enzyme histochemistry on unfixed tissues and cells. Bridging the gap between LM and EM enzyme histochemistry. Acta Histochem 2003; 105:1-19. [PMID: 12666984 DOI: 10.1078/0065-1281-00697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In principle, enzyme histochemistry should be performed on unfixed tissues and cells to avoid inhibition of enzyme activity by chemical fixation. For EM enzyme histochemistry, unfixed tissue specimens include fresh tissue blocks, non-frozen tissue chopper sections, cryostat sections and cell preparations. Studies on localization of enzyme activity at the ultrastructural level in unfixed specimens, be it fresh or frozen, are reviewed here. Preservation of ultrastructural morphology is discussed with special attention to the effects of freezing. It is concluded that unfixed cryostat sections are the best alternative for EM histochemistry of tissues, when interposing a semipermeable membrane in between cryostat section and gelled incubation medium. It is an adequate method to preserve structural integrity of unfixed tissue on the one hand and to avoid inactivation of the enzyme by chemical fixation on the other. For EM cytochemistry on individual cells, a better preservation of ultrastructure may be obtained because freezing can be avoided, but mild pretreatment with a fixative or detergent may be necessary to permeabilize cellular membranes for demonstration of intracellular enzyme activity.
Collapse
Affiliation(s)
- Jacques P M Schellens
- Academic Medical Center, University of Amsterdam, Department of Cell Biology and Histology, Amsterdam, The Netherlands
| | | | | |
Collapse
|
16
|
Vorbach C, Harrison R, Capecchi MR. Xanthine oxidoreductase is central to the evolution and function of the innate immune system. Trends Immunol 2003; 24:512-7. [PMID: 12967676 DOI: 10.1016/s1471-4906(03)00237-0] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Claudia Vorbach
- Ontario Cancer Institute, Princess Margaret Hospital/University Health Network, Toronto, Ontario, M5G 2C1, Canada
| | | | | |
Collapse
|
17
|
Osman AM, van Noort PCM. Evidence for redox cycling of lawsone (2-hydroxy-1,4-naphthoquinone) in the presence of the hypoxanthine/xanthine oxidase system. J Appl Toxicol 2003; 23:209-12. [PMID: 12884402 DOI: 10.1002/jat.908] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This study reports that lawsone (2-hydroxy-1,4-naphthoquinone) undergoes redox cycling in the presence of the hypoxanthine/xanthine oxidase system. The rate of cytochrome c reduction obtained in the presence of 80 microM lawsone was almost three times the rate of cytochrome c reduction measured in its absence. This increase in the rate of cytochrome c reduction was partially inhibited by superoxide dismutase, suggesting the involvement of O(2)(.-) in this process. It is remarkable to note that, even though lawsone is considered to be a non-redox-cycling quinone in vitro, this quinone was shown to be more toxic in vivo in rats than menadione, causing haemolytic anemia of an oxidative nature and renal damage. The view that this quinone is a non-redox-cycling quinone was based on the inability of one-electron-transferring flavoenzymes such as NADPH-cytochrome c reductase to reduce this naphthoquinone. Our finding that lawsone, like menadione, undergoes redox cycling in the presence of the hypoxanthine/xanthine oxidase system could explain the observed oxidative damage of tissues inflicted by this quinone in rats in vivo. Such an observation therefore reconciles the in vivo toxicity results of this naphthoquinone with those of in vitro experiments.
Collapse
Affiliation(s)
- A M Osman
- Institute for Inland Water Management and Waste Water Treatment (RIZA), PO Box 17, 8200 AA Lelystad, The Netherlands.
| | | |
Collapse
|
18
|
Frederiks WM, Vreeling-Sindelárová H. Ultrastructural localization of xanthine oxidoreductase activity in isolated rat liver cells. Acta Histochem 2002; 104:29-37. [PMID: 11993848 DOI: 10.1078/0065-1281-00629] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Xanthine oxidoreductase (XOR) can exist in a dehydrogenase form (XD) and an oxidase form (XO). The D-form uses NAD as cofactor and the O-form uses oxygen as second substrate and produces oxygen radicals. Both enzymes have a high affinity for hypoxanthine and xanthine as substrate and produce uric acid, a potent antioxidant. In the present study, XOR activity was demonstrated with the ferricyanide method in permeabilized isolated rat liver cells at the electron microscopical level. Moreover, ultrastructural localization of XO activity in these cells was studied with the cerium salt method. Activity of both XOR and XO was found in matrix and core of peroxisomes of rat liver parenchymal cells. Only XOR activity was present as well in the cytoplasm of rat liver parenchymal cells. In Kupffer cells and sinusoidal endothelial cells, XOR activity was demonstrated in vesicles and occasionally on granular endoplasmic reticulum. XO activity was not found in Kupffer cells and sinusoidal endothelial cells. The presence of uric acid oxidase activity in matrix and core of peroxisomes as was found previously suggests further breakdown of purines to allantoin in peroxisomes. It is suggested that the major function of XOR activity in the cytoplasm of rat liver parenchymal cells and in sinusoidal cells is not the production of oxygen radicals, but rather the production of uric acid which can act as a potent antioxidant.
Collapse
Affiliation(s)
- Wilma M Frederiks
- Academic Medical Center, University of Amsterdam, Department of Cell Biology and Histology, The Netherlands.
| | | |
Collapse
|
19
|
Abstract
Xanthine oxidoreductase (XOR) is a complex molybdoflavoenzyme, present in milk and many other tissues, which has been studied for over 100 years. While it is generally recognized as a key enzyme in purine catabolism, its structural complexity and specialized tissue distribution suggest other functions that have never been fully identified. The publication, just over 20 years ago, of a hypothesis implicating XOR in ischemia-reperfusion injury focused research attention on the enzyme and its ability to generate reactive oxygen species (ROS). Since that time a great deal more information has been obtained concerning the tissue distribution, structure, and enzymology of XOR, particularly the human enzyme. XOR is subject to both pre- and post-translational control by a range of mechanisms in response to hormones, cytokines, and oxygen tension. Of special interest has been the finding that XOR can catalyze the reduction of nitrates and nitrites to nitric oxide (NO), acting as a source of both NO and peroxynitrite. The concept of a widely distributed and highly regulated enzyme capable of generating both ROS and NO is intriguing in both physiological and pathological contexts. The details of these recent findings, their pathophysiological implications, and the requirements for future research are addressed in this review.
Collapse
Affiliation(s)
- Roger Harrison
- Department of Biology and Biochemistry, University of Bath, Bath, UK.
| |
Collapse
|
20
|
Cancio I, Cajaraville MP. Cell biology of peroxisomes and their characteristics in aquatic organisms. INTERNATIONAL REVIEW OF CYTOLOGY 2000; 199:201-93. [PMID: 10874580 DOI: 10.1016/s0074-7696(00)99005-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
The general characteristics of peroxisomes in different organisms, including aquatic organisms such as fish, crustaceans, and mollusks, are reviewed, with special emphasis on different aspects of the organelle biogenesis and mechanistic aspects of peroxisome proliferation. Peroxisome proliferation and peroxisomal enzyme inductions elicited by xenobiotics or physiological conditions have become useful tools to study the mechanisms of peroxisome biogenesis. During peroxisome proliferation, the induction of peroxisomal proteins is heterogeneous, enzymes that show increased activity being involved in different aspects of lipid homeostasis. The process of peroxisome biogenesis is coordinately triggered by a whole array of structurally dissimilar compounds known as peroxisome proliferators, and investigating the effect of some of these compounds that commonly appear as pollutants in the environment on the peroxisomes of aquatic animals inhabiting marine and estuarine habitats seems interesting. It is also important to determine whether peroxisome proliferation in these animals is a phenomenon that might occur under normal physiological or season-related conditions and plays a metabolic or functional role. This would help set the basis for understanding the process of peroxisome biogenesis in aquatic animals.
Collapse
Affiliation(s)
- I Cancio
- Zoologia eta Animali Zelulen Dinamika Saila, Euskal Herriko Unibertsitatea, Bilbo/Basque Country, Spain
| | | |
Collapse
|
21
|
Godber BL, Doel JJ, Sapkota GP, Blake DR, Stevens CR, Eisenthal R, Harrison R. Reduction of nitrite to nitric oxide catalyzed by xanthine oxidoreductase. J Biol Chem 2000; 275:7757-63. [PMID: 10713088 DOI: 10.1074/jbc.275.11.7757] [Citation(s) in RCA: 289] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Xanthine oxidase (XO) was shown to catalyze the reduction of nitrite to nitric oxide (NO), under anaerobic conditions, in the presence of either NADH or xanthine as reducing substrate. NO production was directly demonstrated by ozone chemiluminescence and showed stoichiometry of approximately 2:1 versus NADH depletion. With xanthine as reducing substrate, the kinetics of NO production were complicated by enzyme inactivation, resulting from NO-induced conversion of XO to its relatively inactive desulfo-form. Steady-state kinetic parameters were determined spectrophotometrically for urate production and NADH oxidation catalyzed by XO and xanthine dehydrogenase in the presence of nitrite under anaerobic conditions. pH optima for anaerobic NO production catalyzed by XO in the presence of nitrite were 7.0 for NADH and </=6.0 for xanthine. Involvement of the molybdenum site of XO in nitrite reduction was shown by the fact that alloxanthine inhibits xanthine oxidation competitively with nitrite. Strong preference for Mo=S over Mo=O was shown by the relatively very low NADH-nitrite reductase activity shown by desulfo-enzyme. The FAD site of XO was shown not to influence nitrite reduction in the presence of xanthine, although it was clearly involved when NADH was the reducing substrate. Apparent production of NO decreased with increasing oxygen tensions, consistent with reaction of NO with XO-generated superoxide. It is proposed that XO-derived NO fulfills a bactericidal role in the digestive tract.
Collapse
Affiliation(s)
- B L Godber
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
22
|
Van den Munckhof RJ. In situ heterogeneity of peroxisomal oxidase activities: an update. THE HISTOCHEMICAL JOURNAL 1996; 28:401-29. [PMID: 8863047 DOI: 10.1007/bf02331433] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Oxidases are a widespread group of enzymes. They are present in numerous organisms and organs and in various tissues, cells, and subcellular compartments, such as mitochondria. An important source of oxidases, which is investigated and discussed in this study, are the (micro)peroxisomes. Oxidases share the ability to reduce molecular oxygen during oxidation of their substrate, yielding an oxidized product and hydrogen peroxide. Besides the hydrogen peroxide-catabolizing enzyme catalase, peroxisomes contain one or more hydrogen peroxide-generating oxidases, which participate in different metabolic pathways. During the last four decades, various methods have been developed and elaborated for the histochemical localization of the activities of these oxidases. These methods are based either on the reduction of soluble electron acceptors by oxidase activity or on the capture of hydrogen peroxide. Both methods yield a coloured and/or electron dense precipitate. The most reliable technique in peroxisomal oxidase histochemistry is the cerium salt capture method. This method is based on the direct capture of hydrogen peroxide by cerium ions to form a fine crystalline, insoluble, electron dense reaction product, cerium perhydroxide, which can be visualized for light microscopy with diaminobenzidine. With the use of this technique, it became clear that oxidase activities not only vary between different organisms, organs, and tissues, but that heterogeneity also exists between different cells and within cells, i.e. between individual peroxisomes. A literature review, and recent studies performed in our laboratory, show that peroxisomes are highly differentiated organelles with respect to the presence of active enzymes. This study gives an overview of the in situ distribution and heterogeneity of peroxisomal enzyme activities as detected by histochemical assays of the activities of catalase, and the peroxisomal oxidases D-amino acid oxidase, L-alpha-hydroxy acid oxidase, polyamine oxidase and uric acid oxidase.
Collapse
Affiliation(s)
- R J Van den Munckhof
- University of Amsterdam, Department of Cell Biology and Histology, The Netherlands
| |
Collapse
|