1
|
Seneff S, Kyriakopoulos AM. Taurine prevents mitochondrial dysfunction and protects mitochondria from reactive oxygen species and deuterium toxicity. Amino Acids 2025; 57:6. [PMID: 39789296 PMCID: PMC11717795 DOI: 10.1007/s00726-024-03440-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
Taurine, although not a coding amino acid, is the most common free amino acid in the body. Taurine has multiple and complex functions in protecting mitochondria against oxidative-nitrosative stress. In this comprehensive review paper, we introduce a novel potential role for taurine in protecting from deuterium (heavy hydrogen) toxicity. This can be of crucial impact to either normal or cancer cells that have highly different mitochondrial redox status. Deuterium is an isotope of hydrogen with a neutron as well as a proton, making it about twice as heavy as hydrogen. We first explain the important role that the gut microbiome and the gut sulfomucin barrier play in deuterium management. We describe the synergistic effects of taurine in the gut to protect against the deleterious accumulation of deuterium in the mitochondria, which disrupts ATP synthesis by ATPase pumps. Moreover, taurine's derivatives, N-chlorotaurine (NCT) and N-bromotaurine (NBrT), produced through spontaneous reaction of taurine with hypochlorite and hypobromite, have fascinating regulatory roles to protect from oxidative stress and beyond. We describe how taurine could potentially alleviate deuterium stress, primarily through metabolic collaboration among various gut microflora to produce deuterium depleted nutrients and deuterium depleted water, and in this way protect against leaky gut barrier, inflammatory bowel disease, and colon cancer.
Collapse
Affiliation(s)
- Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Anthony M Kyriakopoulos
- Laboratory of Molecular Biology and Immunology, Department of Pharmacy, University of Patras, 26500, Rio-Patras, Greece
| |
Collapse
|
2
|
Wilson VC, Kerr BJ. Feeding nursery pigs diets containing peroxidized soybean oil has minimal effects on oxidative status but dramatically reduces serum vitamin E concentrations. J Anim Sci 2025; 103:skaf016. [PMID: 39864064 PMCID: PMC11879175 DOI: 10.1093/jas/skaf016] [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: 11/21/2024] [Accepted: 01/23/2025] [Indexed: 01/28/2025] Open
Abstract
A subgroup of pigs from two experiments (EXP) was selected to evaluate the impact of pigs fed diets containing peroxidized soybean oil (SO) on plasma-based measures of oxidative stress and vitamin E. Pigs were fed diets containing SO that was either unprocessed (23 °C; peroxide value (PV) of 3 meq/kg and an anisidine value of 4) or thermally processed at 135 °C for 42 h (PV of 30 meq/kg and an anisidine value of 501). The corn-soybean meal-based diets contained either 10% SO (EXP 1) or 8% SO (EXP 2). Pigs were fed the experimental diets for 22 d (EXP 1, 13.5 to 24.0 kg, 2 pigs/pen) or 27 d (EXP 2, 21.3 to 37.5 kg, 1 pig/pen), each with 10 replications per dietary treatment. Pigs fed diets containing the peroxidized SO had reduced ADG, ADFI, and GF compared to pigs fed diets containing the unheated SO (P ≤ 0.01). Pigs fed diets containing peroxidized SO had increased plasma concentrations of F2-isoprostanes and reactive oxygen metabolites compared to pigs fed diets containing unheated SO (P ≤ 0.01). In contrast, plasma thiobarbituric acid reactive substances concentrations tended to decrease in pigs fed diets containing peroxidized SO compared to pigs fed diets containing unheated SO (P = 0.10). There was no apparent effect of pigs consuming diets containing peroxidized SO on plasma antioxidant adsorbent capacity or an oxidative stress index (P ≥ 0.19). Pigs fed diets containing peroxidized SO resulted in a reduction in plasma vitamin E compared to pigs fed diets containing unheated SO (P ≤ 0.01). Results indicate that adding SO that has been thermally processed thereby containing high concentrations of aldehydes resulted in inconsistent changes of markers of oxidative stress, but dramatically reduced plasma vitamin E concentrations.
Collapse
Affiliation(s)
- Victoria C Wilson
- Department of Animal Sciences, Iowa State University, Ames, IA 50011, USA
| | - Brian J Kerr
- USDA-ARS National Laboratory for Agriculture and the Environment, Ames, IA 50011, USA
| |
Collapse
|
3
|
Ramírez-Mejía MM, Castillo-Castañeda SM, Pal SC, Qi X, Méndez-Sánchez N. The Multifaceted Role of Bilirubin in Liver Disease: A Literature Review. J Clin Transl Hepatol 2024; 12:939-948. [PMID: 39544246 PMCID: PMC11557368 DOI: 10.14218/jcth.2024.00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Bilirubin, the primary breakdown product of hemoproteins, particularly hemoglobin, plays a key role in the diagnosis, prognosis, and monitoring of liver diseases. In acute liver diseases, such as acute liver failure, drug-induced liver injury, and viral hepatitis, bilirubin serves as a biomarker reflecting the extent of hepatocyte loss and liver damage. Chronic liver diseases, including alcohol-related liver disease, chronic hepatitis C virus infection, metabolic dysfunction-associated fatty liver disease, and autoimmune liver diseases, are marked by persistent liver injury and inflammation. Bilirubin levels in chronic liver diseases provide insight into liver function, disease severity, and prognosis. As a versatile biomarker, bilirubin offers valuable information on the pathophysiology of liver diseases and aids in guiding clinical decision-making regarding the treatment of liver diseases and their complications. This review aimed to explore the multifunctional role of bilirubin in liver diseases by analyzing its biological functions beyond its role as a biomarker of liver damage.
Collapse
Affiliation(s)
- Mariana M. Ramírez-Mejía
- Plan of Combined Studies in Medicine (PECEM-MD/PhD), Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Stephany M. Castillo-Castañeda
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Medical, Dental and Health Sciences Master and Doctorate Program, National Autonomous University of Mexico, Mexico City, Mexico
| | - Shreya C. Pal
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang, Liaoning, China
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
4
|
Dorresteijn MJ, Dekker D, Zwaag J, Heemskerk S, Roelofs HM, Smits P, van der Hoeven JG, Wagener FA, Pickkers P. Atazanavir-induced unconjugated hyperbilirubinemia prevents vascular hyporeactivity during experimental human endotoxemia. Front Immunol 2023; 14:1176775. [PMID: 37261364 PMCID: PMC10228648 DOI: 10.3389/fimmu.2023.1176775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/24/2023] [Indexed: 06/02/2023] Open
Abstract
Objective Inflammation-induced free radical release is important in the pathogenesis of several diseases, including atherosclerosis and sepsis. Heme oxygenase (HO) breaks down heme into carbon monoxide, iron, and biliverdin. Biliverdin IXα is directly converted to bilirubin by biliverdin reductase. Unconjugated bilirubin is a powerful antioxidant, and elevated levels have beneficial effects in preclinical models and human cardiovascular disease. However, its impact during acute inflammation in humans is unknown. In the present study, we investigated the impact of atazanavir-induced (unconjugated) hyperbilirubinemia on antioxidant capacity, inflammation, and vascular dysfunction in human experimental endotoxemia. Approach and results Following double-blinded four-day treatment with atazanavir 2dd300 mg (or placebo), twenty healthy male volunteers received 2 ng/kg Escherichia coli lipopolysaccharide intravenously. Blood was drawn to determine the bilirubin levels, antioxidant capacity, and cytokine response. It was demonstrated that following atazanavir treatment, total bilirubin concentrations increased to maximum values of 4.67 (95%CI 3.91-5.59) compared to 0.82 (95%CI 0.64-1.07) mg/dL in the control group (p<0.01). Furthermore, the anti-oxidant capacity, as measured by the ferric-reducing ability of plasma (FRAP), was significantly increased with 36% in hyperbilirubinemia subjects (p<0.0001), and FRAP concentrations correlated strongly to bilirubin concentrations (R2 = 0.77, p<0.001). Hyperbilirubinemia attenuated the release of interleukin-10 from 377 (95%CI 233-609) to 219 (95%CI 152-318) pg/mL (p=0.01), whereas the release of pro-inflammatory cytokines remained unaltered. In vitro, in the absence of hyperbilirubinemia, atazanavir did not influence lipopolysaccharide-induced cytokine release in a whole blood assay. Vascular function was assessed using forearm venous occlusion plethysmography after intra-arterial infusion of acetylcholine and nitroglycerin. Hyperbilirubinemia completely prevented the LPS-associated blunted vascular response to acetylcholine and nitroglycerin. Conclusions Atazanavir-induced hyperbilirubinemia increases antioxidant capacity, attenuates interleukin-10 release, and prevents vascular hyporesponsiveness during human systemic inflammation elicited by experimental endotoxemia. Clinical trial registration http://clinicaltrials.gov, identifier NCT00916448.
Collapse
Affiliation(s)
- Mirrin J. Dorresteijn
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pharmacology and Toxicology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Douwe Dekker
- Department of Pharmacology and Toxicology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jelle Zwaag
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Suzanne Heemskerk
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pharmacology and Toxicology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hennie M.J. Roelofs
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul Smits
- Department of Pharmacology and Toxicology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johannes G. van der Hoeven
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank A.D.T.G. Wagener
- Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
5
|
Mancuso C. The impact of heme oxygenase-2 on pharmacological research: A bibliometric analysis and beyond. Front Pharmacol 2023; 14:1156333. [PMID: 37153762 PMCID: PMC10154548 DOI: 10.3389/fphar.2023.1156333] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/06/2023] [Indexed: 05/10/2023] Open
Abstract
Heme oxygenase (HO-2) is an enzyme mainly involved in the physiologic turnover of heme and intracellular gas sensing, and it is very abundant in the brain, testes, kidneys and vessels. Since 1990, when HO-2 was discovered, the scientific community has underestimated the role of this protein in health and disease, as attested by the small amount of articles published and citations received. One of the reason that have contributed to the lack of interest in HO-2 was the difficulty in upregulating or inhibiting this enzyme. However, over the last 10 years, novel HO-2 agonists and antagonists have been synthesized, and the availability of these pharmacological tools should increase the appeal of HO-2 as drug target. In particular, these agonists and antagonists could help explain some controversial aspects, such as the neuroprotective versus neurotoxic roles of HO-2 in cerebrovascular diseases. Furthermore, the discovery of HO-2 genetic variants and their involvement in Parkinson's disease, in particular in males, opens new avenues for pharmacogenetic studies in gender medicine.
Collapse
Affiliation(s)
- Cesare Mancuso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica Del Sacro Cuore, Rome, Italy
- *Correspondence: Cesare Mancuso,
| |
Collapse
|
6
|
Mechanisms Underlying the Protective Effect of Maternal Zinc (ZnSO4 or Zn-Gly) against Heat Stress-Induced Oxidative Stress in Chicken Embryo. Antioxidants (Basel) 2022; 11:antiox11091699. [PMID: 36139773 PMCID: PMC9495990 DOI: 10.3390/antiox11091699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Environmental factors such as high temperature can cause oxidative stress and negatively affect the physiological status and meat quality of broiler chickens. The study was conducted to evaluate the effects of dietary maternal Zn-Gly or ZnSO4 supplementation on embryo mortality, hepatocellular mitochondrial morphology, liver antioxidant capacity and the expression of related genes involved in liver oxidative mechanisms in heat-stressed broilers. A total of 300 36-week-old Lingnan Yellow broiler breeders were randomly divided into three treatments: (1) control (basal diet, 24 mg zinc/kg); (2) inorganic ZnSO4 group (basal diet +80 mg ZnSO4/kg); (3) organic Zn-Gly group (basal diet +80 mg Zn-Gly/kg). The results show that maternal zinc alleviated heat stress-induced chicken embryo hepatocytes’ oxidative stress by decreasing the content of ROS, MDA, PC, 8-OHdG, and levels of HSP70, while enhancing T-SOD, T-AOC, CuZn-SOD, GSH-Px, CTA activities and the content of MT. Maternal zinc alleviated oxidative stress-induced mitochondrial damage in chick embryo hepatocytes by increasing mitochondrial membrane potential and UCP gene expression; and Caspase-3-mediated apoptosis was alleviated by increasing CuZn-SOD and MT gene expression and decreasing Bax gene expression and reducing the activity of caspase 3. Furthermore, maternal zinc treatment significantly increased Nrf2 gene expression. The results above suggest that maternal zinc can activate the Nrf2 signaling pathway in developing chick embryos, enhance its antioxidant function and reduce the apoptosis-effecting enzyme caspase-3 activities, thereby slowing oxidative stress injury and tissue cell apoptosis.
Collapse
|
7
|
Aggarwal A, Dinda AK, Mukhopadhyay CK. Effect of Cisplatin on Renal Iron Homeostasis Components: Implication in Nephropathy. ACS OMEGA 2022; 7:27804-27817. [PMID: 35990481 PMCID: PMC9386824 DOI: 10.1021/acsomega.1c06716] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cisplatin is an important chemotherapeutic drug for the treatment of solid tumors but often causes nephropathy as part of the off-target toxicity. Iron accumulation and related damage were implicated in cisplatin-induced kidney injury. However, the role of cisplatin in the renal iron sensing mechanism and its target genes responsible for iron uptake, storage, and release have not been investigated. Cellular iron homeostasis is controlled by the interaction of iron regulatory proteins (IRP1 and IRP2) and iron-responsive elements (IREs) present in the untranslated regions of iron transport and storage components. Here, we report that cisplatin does not influence the expressions of IRP targets such as transferrin receptor-1 (TfR1), divalent metal transporter-1 (DMT1), and ferroportin in renal cells despite the increased heme oxygenase-1 (HO-1) level. Ferritin subunits (Ft-H and Ft-L) are elevated in different magnitudes due to the increased mRNA expression. Intriguingly, a higher expression of Ft-L mRNA is detected than that of Ft-H mRNA. The inability of cisplatin in altering the IRE-IRP interaction is confirmed by examining IRE-containing luciferase activity, RNA electrophoretic mobility shift assay, and activation of IRPs. The labile iron pool is depleted but reversed by silencing of either Ft-H or Ft-L, suggesting increased iron storage by ferritin. Silencing of Ft-H or Ft-L promotes cell death, suggesting that ferritin acts to protect the renal cells from cisplatin-mediated toxicity. A differential increase of transcripts and equivalent increase of proteins of Ft-H and Ft-L and unaltered TfR1 and DMT1 transcripts are found in the kidneys of cisplatin-treated rats along with iron accumulation. Our results reveal that cisplatin does not influence the IRE-IRP interaction despite alteration of the cellular iron pool in renal cells. This insensitivity of the IRE-IRP system may be implicated in the accumulation of iron to contribute to cisplatin-induced nephropathy.
Collapse
Affiliation(s)
- Ayushi Aggarwal
- Department
of Pathology, All India Institute of Medical
Sciences, Ansari Nagar, New Delhi 110029, India
| | - Amit K. Dinda
- Department
of Pathology, All India Institute of Medical
Sciences, Ansari Nagar, New Delhi 110029, India
| | | |
Collapse
|
8
|
Aronson SJ, Junge N, Trabelsi M, Kelmemi W, Hubert A, Brigatti KW, Fox MD, de Knegt RJ, Escher JC, Ginocchio VM, Iorio R, Zhu Y, Özçay F, Rahim F, El-Shabrawi MHF, Shteyer E, Di Giorgio A, D'Antiga L, Mingozzi F, Brunetti-Pierri N, Strauss KA, Labrune P, Mrad R, Baumann U, Beuers U, Bosma PJ. Disease burden and management of Crigler-Najjar syndrome: Report of a world registry. Liver Int 2022; 42:1593-1604. [PMID: 35274801 DOI: 10.1111/liv.15239] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Sem J Aronson
- Department of Gastroenterology & Hepatology and Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology & Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Norman Junge
- Division for Pediatric Gastroenterology and Hepatology, Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Mediha Trabelsi
- Laboratoire de Génétique Humaine, Faculté de Médecine de Tunis (Laboratory of Human Genetics, Faculty of Medicine of Tunis, Université de Tunis El Manar (University of Tunis El Manar), Tunis, Tunisia
- Service des Maladies Congénitales et Héréditaires (Department of Hereditary and Congenital Disorders), Hôpital Charles Nicolle (Charles Nicolle Hospital), Tunis, Tunisia
| | - Wided Kelmemi
- Laboratoire de Génétique Humaine, Faculté de Médecine de Tunis (Laboratory of Human Genetics, Faculty of Medicine of Tunis, Université de Tunis El Manar (University of Tunis El Manar), Tunis, Tunisia
| | - Aurelie Hubert
- Department of Hereditary Diseases of Hepatic Metabolism, Hôpital Antoine Béclère, Clamart, France
| | | | - Michael D Fox
- Clinic for Special Children, Strasburg, Pennsylvania, USA
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert J de Knegt
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Johanna C Escher
- Department of Pediatric Gastroenterology, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Virginia M Ginocchio
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Raffaele Iorio
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Yan Zhu
- Third Military Medical University, Chongqing, China
| | - Figen Özçay
- Department of Pediatric Gastroenterology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Fakher Rahim
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Health research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mortada H F El-Shabrawi
- Department of Pediatrics and Pediatric Hepatology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eyal Shteyer
- Paediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Angelo Di Giorgio
- Department of Paediatric Gastroenterology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Lorenzo D'Antiga
- Department of Paediatric Gastroenterology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Kevin A Strauss
- Clinic for Special Children, Strasburg, Pennsylvania, USA
- Departments of Pediatrics and Molecular, Cell & Cancer Biology, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - Philippe Labrune
- Department of Hereditary Diseases of Hepatic Metabolism, Hôpital Antoine Béclère, Clamart, France
| | - Ridha Mrad
- Laboratoire de Génétique Humaine, Faculté de Médecine de Tunis (Laboratory of Human Genetics, Faculty of Medicine of Tunis, Université de Tunis El Manar (University of Tunis El Manar), Tunis, Tunisia
- Service des Maladies Congénitales et Héréditaires (Department of Hereditary and Congenital Disorders), Hôpital Charles Nicolle (Charles Nicolle Hospital), Tunis, Tunisia
| | - Ulrich Baumann
- Division for Pediatric Gastroenterology and Hepatology, Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Ulrich Beuers
- Department of Gastroenterology & Hepatology and Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology & Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Piter J Bosma
- Department of Gastroenterology & Hepatology and Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology & Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Growth promotion and antibiotic induced metabolic shifts in the chicken gut microbiome. Commun Biol 2022; 5:293. [PMID: 35365748 PMCID: PMC8975857 DOI: 10.1038/s42003-022-03239-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/08/2022] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial growth promoters (AGP) have played a decisive role in animal agriculture for over half a century. Despite mounting concerns about antimicrobial resistance and demand for antibiotic alternatives, a thorough understanding of how these compounds drive performance is missing. Here we investigate the functional footprint of microbial communities in the cecum of chickens fed four distinct AGP. We find relatively few taxa, metabolic or antimicrobial resistance genes similarly altered across treatments, with those changes often driven by the abundances of core microbiome members. Constraints-based modeling of 25 core bacterial genera associated increased performance with fewer metabolite demands for microbial growth, pointing to altered nitrogen utilization as a potential mechanism of narasin, the AGP with the largest performance increase in our study. Untargeted metabolomics of narasin treated birds aligned with model predictions, suggesting that the core cecum microbiome might be targeted for enhanced performance via its contribution to host-microbiota metabolic crosstalk. This study compares the functional profiles of the cecal microbiome among chickens fed four different antimicrobial growth promoters. Chickens receiving narasin exhibited the largest performance increase via apparent nitrogen recycling by the core cecal microbiome.
Collapse
|
10
|
Huang Z, Shi Y, Wang H, Chun C, Chen L, Wang K, Lu Z, Zhao Y, Li X. Protective Effects of Chitosan-Bilirubin Nanoparticles Against Ethanol-Induced Gastric Ulcers. Int J Nanomedicine 2022; 16:8235-8250. [PMID: 34992363 PMCID: PMC8709796 DOI: 10.2147/ijn.s344805] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022] Open
Abstract
Purpose Gastric ulcers (GU) are a disease of the gastrointestinal tract that can be caused by excessive alcohol consumption and heavy use of nonsteroidal anti-inflammatory drugs. GU manifests predominantly as pathological damage, such as extensive inflammatory erosion and superficial bleeding of the gastric mucosa. Oxidative stress damage and the inflammatory response are now considered important predisposing factors for GU, suggesting that antioxidant and anti-inflammatory drugs could be treatments for GU. Nanoparticle drug carriers offer many advantages over conventional drugs, such as improved drug efficiency, increased drug stability, and increased half-life. Methods We designed chitosan-bilirubin conjugate (CS-BR) nanoparticles and assessed the anti-inflammatory and antioxidant abilities of CS-BR in gastric epithelial cells. Then, we evaluated the intragastric retention time and the anti-ulcer effects of CS-BR in vivo. Results The in vitro data showed that CS-BR nanoparticles protect gastric epithelial cells against oxidative/inflammatory injury. The in vivo study demonstrated that CS-BR nanoparticles accumulate permanently in the stomach and exert powerful antioxidant and anti-inflammatory effects against GU. Conclusion This study applied bilirubin to the treatment of GU and confirmed that CS-BR nanoparticles are effective at alleviating acute GU in an experimental model. The findings provide innovative ideas for prophylaxis against or treatment of GU.
Collapse
Affiliation(s)
- Zhiwei Huang
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, People's Republic of China.,Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yannan Shi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, People's Republic of China
| | - Hengcai Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, People's Republic of China
| | - Changju Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Longwang Chen
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, People's Republic of China.,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, 325035, People's Republic of China
| | - Kang Wang
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, People's Republic of China.,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, 325035, People's Republic of China
| | - Zhongqiu Lu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, People's Republic of China.,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, 325035, People's Republic of China
| | - Yingzheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, People's Republic of China
| | - Xinze Li
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, People's Republic of China.,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, 325035, People's Republic of China
| |
Collapse
|
11
|
González-Silvera D, Cuesta A, Esteban MÁ. Immune defence mechanisms presented in liver homogenates and bile of gilthead seabream (Sparus aurata). JOURNAL OF FISH BIOLOGY 2021; 99:1958-1967. [PMID: 34486119 DOI: 10.1111/jfb.14901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 06/13/2023]
Abstract
Because the role of the liver of fishes in providing possible immunity remains largely unknown, the aim of this work was to identify and characterize different humoral defence mechanisms in the liver homogenates and bile of gilthead seabream (Sparus aurata) for the first time. Total protein levels and several immune parameters (complement activity, lysozyme and immunoglobulin M level) were studied. Furthermore, the activity of some lytic (proteases, antiproteases, esterase, alkaline phosphatase) and antioxidant (superoxide dismutase, catalase and peroxidase) enzymes was determined. Finally, bacteriostatic activity on three opportunist fish pathogens (Vibrio harveyi, Vibrio angillarum and Photobacterium damselae) was measured. Lysozyme and antiprotease activity were undetected in liver and bile, while natural haemolytic complement activity was only detected in bile, and immunoglobulin M was detected in both samples. The levels of proteases, esterase and antioxidant enzymes were greater in bile than in liver homogenates, while the level of alkaline phosphatase was very low in both samples. In addition, while no bacteriostatic activity was detected on liver homogenates, the bile revealed a very potent bacteriostatic activity against all the tested pathogenic bacteria. These results corroborate that fish liver - especially fish bile - contains many factors involved in innate immunity that could be useful for better understanding the role of the liver as an organ involved in fish immune functions as well as the possible contribution of bile to gut mucosal immunity.
Collapse
Affiliation(s)
- Daniel González-Silvera
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - Alberto Cuesta
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - Maria Ángeles Esteban
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, Murcia, Spain
| |
Collapse
|
12
|
Karg CA, Wang S, Al Danaf N, Pemberton RP, Bernard D, Kretschmer M, Schneider S, Zisis T, Vollmar AM, Lamb DC, Zahler S, Moser S. Tetrapyrrolische Pigmente aus dem Häm‐ und Chlorophyllabbau interagieren mit Aktin. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202107813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Cornelia A. Karg
- Pharmazeutische Biologie Department Pharmazie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Shuaijun Wang
- Pharmazeutische Biologie Department Pharmazie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Nader Al Danaf
- Center for Nanoscience (CeNS) und Nanosystems Initiative Munich (NIM) Department Chemie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Ryan P. Pemberton
- Atomwise Inc. 717 Market Street, Suite 800 San Francisco CA 94103 USA
| | - Denzil Bernard
- Atomwise Inc. 717 Market Street, Suite 800 San Francisco CA 94103 USA
| | - Maibritt Kretschmer
- Pharmazeutische Biologie Department Pharmazie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Sabine Schneider
- Department Chemie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Themistoklis Zisis
- Pharmazeutische Biologie Department Pharmazie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Angelika M. Vollmar
- Pharmazeutische Biologie Department Pharmazie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Don C. Lamb
- Center for Nanoscience (CeNS) und Nanosystems Initiative Munich (NIM) Department Chemie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Stefan Zahler
- Pharmazeutische Biologie Department Pharmazie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| | - Simone Moser
- Pharmazeutische Biologie Department Pharmazie Ludwig-Maximilians-Universität München Butenandtstraße 5–13 81377 München Deutschland
| |
Collapse
|
13
|
Karg CA, Wang S, Al Danaf N, Pemberton RP, Bernard D, Kretschmer M, Schneider S, Zisis T, Vollmar AM, Lamb DC, Zahler S, Moser S. Tetrapyrrolic Pigments from Heme- and Chlorophyll Breakdown are Actin-Targeting Compounds. Angew Chem Int Ed Engl 2021; 60:22578-22584. [PMID: 34310831 PMCID: PMC8519017 DOI: 10.1002/anie.202107813] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/22/2021] [Indexed: 11/11/2022]
Abstract
Chlorophyll and heme are among the "pigments of life", tetrapyrrolic structures, without which life on Earth would not be possible. Their catabolites, the phyllobilins and the bilins, respectively, share not only structural features, but also a similar story: Long considered waste products of detoxification processes, important bioactivities for both classes have now been demonstrated. For phyllobilins, however, research on physiological roles is sparse. Here, we introduce actin, the major component of the cytoskeleton, as the first discovered target of phyllobilins and as a novel target of bilins. We demonstrate the inhibition of actin dynamics in vitro and effects on actin and related processes in cancer cells. A direct interaction with G-actin is shown by in silico studies and confirmed by affinity chromatography. Our findings open a new chapter in bioactivities of tetrapyrroles-especially phyllobilins-for which they form the basis for broad implications in plant science, ecology, and physiology.
Collapse
Affiliation(s)
- Cornelia A. Karg
- Pharmaceutical BiologyDepartment of PharmacyLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| | - Shuaijun Wang
- Pharmaceutical BiologyDepartment of PharmacyLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| | - Nader Al Danaf
- Center for Nanoscience (CeNS) and Nanosystems Initiative Munich (NIM)Department of ChemistryLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| | | | - Denzil Bernard
- Atomwise Inc.717 Market Street, Suite 800San FranciscoCA94103USA
| | - Maibritt Kretschmer
- Pharmaceutical BiologyDepartment of PharmacyLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| | - Sabine Schneider
- Department of ChemistryLudwig-Maximilians University MunichButenandtstrasse 5–1381377MunichGermany
| | - Themistoklis Zisis
- Pharmaceutical BiologyDepartment of PharmacyLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| | - Angelika M. Vollmar
- Pharmaceutical BiologyDepartment of PharmacyLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| | - Don C. Lamb
- Center for Nanoscience (CeNS) and Nanosystems Initiative Munich (NIM)Department of ChemistryLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| | - Stefan Zahler
- Pharmaceutical BiologyDepartment of PharmacyLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| | - Simone Moser
- Pharmaceutical BiologyDepartment of PharmacyLudwig-Maximilians University of MunichButenandtstraße 5–1381377MunichGermany
| |
Collapse
|
14
|
Basiglio CL, Crocenzi FA, Sánchez Pozzi EJ, Roma MG. Oxidative Stress and Localization Status of Hepatocellular Transporters: Impact on Bile Secretion and Role of Signaling Pathways. Antioxid Redox Signal 2021; 35:808-831. [PMID: 34293961 DOI: 10.1089/ars.2021.0021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Significance: Most hepatopathies are primarily or secondarily cholestatic in nature. Oxidative stress (OS) is a frequent trait among them, and impairs the machinery to generate bile by triggering endocytic internalization of hepatocellular transporters, thus causing cholestasis. This is critical, since it leads to accelerated transporter degradation, which could explain the common post-transcriptional downregulation of transporter expression in human cholestatic diseases. Recent Advances: The mechanisms involved in OS-induced hepatocellular transporter internalization are being revealed. Filamentous actin (F-actin) cytoskeleton disorganization and/or detachment of crosslinking actin proteins that afford transporter stability have been characterized as causal factors. Activation of redox-sensitive signaling pathways leading to changes in phosphorylation status of these structures is involved, including Ca2+-mediated activation of "classical" and "novel" protein kinase C (PKC) isoforms or redox-signaling cascades downstream of NADPH oxidase. Critical Issues: Despite the well-known occurrence of hepatocellular transporter internalization in human hepatopathies, the cholestatic implications of this phenomenon have been overlooked. Accordingly, no specific treatment has been established in the clinical practice for its prevention/reversion. Future Directions: We need to improve our knowledge on the pro-oxidant triggering factors and the multiple signaling pathways that mediate this oxidative injury in each cholestatic hepatopathy, so as to envisage tailor-made therapeutic strategies for each case. Meanwhile, administration of antioxidants or heme oxygenase-1 induction to elevate the hepatocellular levels of the endogenous scavenger bilirubin are promising alternatives that need to be re-evaluated and implemented. They may complement current treatments in cholestasis aimed to enhance transcriptional carrier expression, by providing membrane stability to the newly synthesized carriers. Antioxid. Redox Signal. 35, 808-831.
Collapse
Affiliation(s)
- Cecilia L Basiglio
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Fernando A Crocenzi
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Enrique J Sánchez Pozzi
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Marcelo G Roma
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| |
Collapse
|
15
|
Abstract
Significance: As the central metabolic organ, the liver is exposed to a variety of potentially cytotoxic, proinflammatory, profibrotic, and carcinogenic stimuli. To protect the organism from these deleterious effects, the liver has evolved a number of defense systems, which include antioxidant substrates and enzymes, anti-inflammatory tools, enzymatic biotransformation systems, and metabolic pathways. Recent Advances: One of the pivotal systems that evolved during phylogenesis was the heme catabolic pathway. Comprising the important enzymes heme oxygenase and biliverdin reductase, this complex pathway has a number of key functions including enzymatic activities, but also cell signaling, and DNA transcription. It further generates two important bile pigments, biliverdin and bilirubin, as well as the gaseous molecule carbon monoxide. These heme degradation products have potent antioxidant, immunosuppressive, and cytoprotective effects. Recent data suggest that the pathway participates in the regulation of metabolic and hormonal processes implicated in the pathogenesis of hepatic and other diseases. Critical Issues: This review discusses the impact of the heme catabolic pathway on major liver diseases, with particular focus on the involvement of cellular targeting and signaling in the pathogenesis of these conditions. Future Directions: To utilize the biological consequences of the heme catabolic pathway, several unique therapeutic strategies have been developed. Research indicates that pharmaceutical, nutraceutical, and lifestyle modifications positively affect the pathway, delivering potentially long-term clinical benefits. However, further well-designed studies are needed to confirm the clinical benefits of these approaches. Antioxid. Redox Signal. 35, 734-752.
Collapse
Affiliation(s)
- Libor Vítek
- Fourth Department of Internal Medicine, and Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
16
|
Zöhrer PA, Hana CA, Seyed Khoei N, Mölzer C, Hörmann-Wallner M, Tosevska A, Doberer D, Marculescu R, Bulmer AC, Herbold CW, Berry D, Wagner KH. Gilbert's Syndrome and the Gut Microbiota - Insights From the Case-Control BILIHEALTH Study. Front Cell Infect Microbiol 2021; 11:701109. [PMID: 34604105 PMCID: PMC8481893 DOI: 10.3389/fcimb.2021.701109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
The heme catabolite bilirubin has anti-inflammatory, anti-oxidative and anti-mutagenic effects and its relation to colorectal cancer (CRC) risk is currently under evaluation. Although the main metabolic steps of bilirubin metabolism, including the formation of stercobilin and urobilin, take place in the human gastrointestinal tract, potential interactions with the human gut microbiota are unexplored. This study investigated, whether gut microbiota composition is altered in Gilbert's Syndrome (GS), a mild form of chronically elevated serum unconjugated bilirubin (UCB) compared to matched controls. Potential differences in the incidence of CRC-associated bacterial species in GS were also assessed. To this end, a secondary investigation of the BILIHEALTH study was performed, assessing 45 adults with elevated UCB levels (GS) against 45 age- and sex-matched controls (C). Fecal microbiota analysis was performed using 16S rRNA gene sequencing. No association between mildly increased UCB and the composition of the gut microbiota in this healthy cohort was found. The alpha and beta diversity did not differ between C and GS and both groups showed a typical representation of the known dominant phyla. Furthermore, no difference in abundance of Firmicutes and Proteobacteria, which have been associated with the mucosa of CRC patients were observed between the groups. A sequence related to the Christensenella minuta strain YIT 12065 was identified with a weak association value of 0.521 as an indicator species in the GS group. This strain has been previously associated with a lower body mass index, which is typical for the GS phenotype. Overall, sex was the only driver for an identifiable difference in the study groups, as demonstrated by a greater bacterial diversity in women. After adjusting for confounding factors and multiple testing, we can conclude that the GS phenotype does not affect the composition of the human gut microbiota in this generally healthy study group.
Collapse
Affiliation(s)
- Patrick A. Zöhrer
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
| | - Claudia A. Hana
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Nazlisadat Seyed Khoei
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
| | - Christine Mölzer
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Marlies Hörmann-Wallner
- Institute for Dietetics and Nutrition, University of Applied Sciences FH JOANNEUM, Graz, Austria
| | - Anela Tosevska
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Daniel Doberer
- Clinical Institute of Laboratory Medicine, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Department of Clinical Pharmacology, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Andrew C. Bulmer
- School of Pharmacy and Medical Sciences and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Craig W. Herbold
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - David Berry
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and University of Vienna, Vienna, Austria
| | - Karl-Heinz Wagner
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Reyes-Ramos CA, Gaxiola-Robles R, Vázquez-Medina JP, Ramírez-Jirano LJ, Bitzer-Quintero OK, Zenteno-Savín T. In silico Characterization of the Heme Oxygenase 1 From Bottlenose Dolphin ( Tursiops truncatus): Evidence of Changes in the Active Site and Purifying Selection. Front Physiol 2021; 12:711645. [PMID: 34456750 PMCID: PMC8388933 DOI: 10.3389/fphys.2021.711645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Cetacea is a clade well-adapted to the aquatic lifestyle, with diverse adaptations and physiological responses, as well as a robust antioxidant defense system. Serious injuries caused by boats and fishing nets are common in bottlenose dolphins (Tursiops truncatus); however, these animals do not show signs of serious infections. Evidence suggests an adaptive response to tissue damage and associated infections in cetaceans. Heme oxygenase (HO) is a cytoprotective protein that participates in the anti-inflammatory response. HO catalyzes the first step in the oxidative degradation of the heme group. Various stimuli, including inflammatory mediators, regulate the inducible HO-1 isoform. This study aims to characterize HO-1 of the bottlenose dolphin in silico and compare its structure to the terrestrial mammal protein. Upstream HO-1 sequence of the bottlenose dolphin was obtained from NCBI and Ensemble databases, and the gene structure was determined using bioinformatics tools. Five exons and four introns were identified, and proximal regulatory elements were detected in the upstream region. The presence of 10 α-helices, three 310 helices, the heme group lodged between the proximal and distal helices, and a histidine-25 in the proximal helix serving as a ligand to the heme group were inferred for T. truncatus. Amino acid sequence alignment suggests HO-1 is a conserved protein. The HO-1 "fingerprint" and histidine-25 appear to be fully conserved among all species analyzed. Evidence of positive selection within an α-helix configuration without changes in protein configuration and evidence of purifying selection were found, indicating evolutionary conservation of the coding sequence structure.
Collapse
Affiliation(s)
- Carlos A. Reyes-Ramos
- Centro de Investigaciones Biológicas del Noroeste, S.C. Planeación Ambiental y Conservación, La Paz, Mexico
| | - Ramón Gaxiola-Robles
- Centro de Investigaciones Biológicas del Noroeste, S.C. Planeación Ambiental y Conservación, La Paz, Mexico
- Hospital General de Zona No. 1, Instituto Mexicano del Seguro Social, La Paz, Mexico
| | | | - Luis Javier Ramírez-Jirano
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | - Oscar Kurt Bitzer-Quintero
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | - Tania Zenteno-Savín
- Centro de Investigaciones Biológicas del Noroeste, S.C. Planeación Ambiental y Conservación, La Paz, Mexico
| |
Collapse
|
18
|
Patients with Gilbert syndrome and type 2 diabetes have lower prevalence of microvascular complications. Metabol Open 2021; 11:100114. [PMID: 34386764 PMCID: PMC8346683 DOI: 10.1016/j.metop.2021.100114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Accumulating clinical evidence indicates an inverse relationship between oxidative stress and unconjugated hyperbilirubinemia. This study aimed to compare the prevalence of diabetes microvascular complications in patients with Gilbert syndrome and type 2 diabetes mellitus (T2D). Methods A total of 1200 electronic records with T2D were reviewed. From them, 50 patients with Gilbert syndrome (cases [indirect bilirubin ≥1.2 mg/dl without evidence of hemolysis or liver disease]) and 50 controls (T2D without hyperbilirubinemia) were included. Linear and logistic regression models were performed to evaluate the independent association between indirect hyperbilirubinemia with microvascular complications related with T2D. Results Both case and control group had the same proportion of gender (female = 20 [40 %]) and diabetes duration (14.0 ± 6.5 years) and similar mean of age (60 ± 9.6 and 60 ± 9.2 years, respectively, p = 0.91). The median of unconjugated bilirubin of case and control group was 1.4 (1.2–1.6) vs. 0.4 (0.2–0.6) mg/dl (p < 0.001), respectively. Patients with elevated unconjugated bilirubin had less urine albumin-creatinine ratio compared with control group (8.5 [4.3–23] vs. 80 [8–408] mg/g, p < 0.001), and lower rate of diabetes microvascular complications and metabolic syndrome. After adjustment for BMI, age, HbA1c, blood pressure, triglycerides, and the metabolic syndrome, the lineal regression analysis showed that unconjugated bilirubin protects against microalbuminuria in T2D patients (β = −414.11, 95 % CI [-747.9, −80.3], p = 0.006. Also, unconjugated hyperbilirubinemia was independently associated with a better glomerular filtration rate (GFR) (β = 9.87, 95 % CI [1.5, 18.3], P = 0.02). Conclusions Patients with Gilbert syndrome and T2D had a lower prevalence of diabetes microvascular complications.
Collapse
|
19
|
Nitti M, Ivaldo C, Traverso N, Furfaro AL. Clinical Significance of Heme Oxygenase 1 in Tumor Progression. Antioxidants (Basel) 2021; 10:antiox10050789. [PMID: 34067625 PMCID: PMC8155918 DOI: 10.3390/antiox10050789] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase 1 (HO-1) plays a key role in cell adaptation to stressors through the antioxidant, antiapoptotic, and anti-inflammatory properties of its metabolic products. For these reasons, in cancer cells, HO-1 can favor aggressiveness and resistance to therapies, leading to poor prognosis/outcome. Genetic polymorphisms of HO-1 promoter have been associated with an increased risk of cancer progression and a high degree of therapy failure. Moreover, evidence from cancer biopsies highlights the possible correlation between HO-1 expression, pathological features, and clinical outcome. Indeed, high levels of HO-1 in tumor specimens often correlate with reduced survival rates. Furthermore, HO-1 modulation has been proposed in order to improve the efficacy of antitumor therapies. However, contrasting evidence on the role of HO-1 in tumor biology has been reported. This review focuses on the role of HO-1 as a promising biomarker of cancer progression; understanding the correlation between HO-1 and clinical data might guide the therapeutic choice and improve the outcome of patients in terms of prognosis and life quality.
Collapse
|
20
|
Pathophysiological effects of Klebsiella pneumoniae infection on Galleria mellonella as an invertebrate model organism. Arch Microbiol 2021; 203:3509-3517. [PMID: 33909089 DOI: 10.1007/s00203-021-02346-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 12/27/2022]
Abstract
Klebsiella pneumoniae is an important human pathogen causing urinary tract infections and pneumonia. Due to the increase in resistant strains and being an opportunistic pathogen, it is very important to determine the virulence process, the cellular damage it causes in the host and the immunological response level of the host. In this study, invertebrate infection model Galleria mellonella larvae were used to investigate cellular damage, antioxidant response and changes in biochemical parameters due to K. pneumoniae infection. The activity of cell damage indicators alanine aminotransferase, aspartate aminotransferase and lactate dehydrogenase increased in hemolymph of G. mellonella larvae due to K. pneumoniae virulence. Creatine kinase, alkaline phosphatase, gamma glutamyl transferase and amylase activities were increased to regulate the disrupted energy metabolism due to infection. As a result of the damage caused by K. pneumoniae infection, changes occurred in the amount of non-enzymatic antioxidants, uric acid, bilirubin and albumin. Due to K. pneumoniae infection, the amount of calcium, potassium, magnesium and phosphorus altered. This study showed that G. mellonella larvae was important infection model in the investigation of infectious cell damage and physiological effects, given the opportunistic nature of the K. pneumoniae pathogen and the lack of adequate animal models.
Collapse
|
21
|
Cheung IM, Mcghee CN, Sherwin T. A new perspective on the pathobiology of keratoconus: interplay of stromal wound healing and reactive species‐associated processes. Clin Exp Optom 2021; 96:188-96. [DOI: 10.1111/cxo.12025] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 10/30/2012] [Indexed: 12/13/2022] Open
Affiliation(s)
- Isabella My Cheung
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand,
| | - Charles Nj Mcghee
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand,
| | - Trevor Sherwin
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand,
| |
Collapse
|
22
|
Zhao X, Wang Y, Liu C, Zhou P, Sheng Z, Li J, Zhou J, Chen R, Chen Y, Zhao H, Yan H. Prognostic Value of Total Bilirubin in Patients With ST-Segment Elevation Acute Myocardial Infarction Undergoing Primary Coronary Intervention. Front Cardiovasc Med 2021; 7:615254. [PMID: 33392275 PMCID: PMC7773653 DOI: 10.3389/fcvm.2020.615254] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/24/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Bilirubin, a natural product of heme catabolism, has antioxidant and anti-inflammatory activities and is inversely associated with stable coronary artery disease. However, the relationship between the bilirubin levels and long-term outcomes in patients with ST-segment elevation myocardial infarction (STEMI) who underwent primary percutaneous coronary intervention (PPCI) remains unknown. This study aimed to establish a score model based on bilirubin for predicting major adverse cardiovascular events (MACEs) and stratify patients to the level of care. Methods and Results: Data of 4,151 consecutive patients with STEMI who underwent PPCI were evaluated, and 3,708 cases were analyzed. The total bilirubin (TBil) levels were measured during admission, and the study population was divided into two groups. The high TBil group (n = 143) comprised patients who had a TBil level of ≥22 μmmol/L, and the low TBil group (n = 3,565) comprised patients who had a TBil level of <22 μmmol/L. The median follow-up period was 754 days (2.066 years). The MACE was significantly lower in the high TBil group than in the low TBil group (3.5% vs. 11.0%, p = 0.001). In the multivariate Cox regression analysis, a significant association was noted between the TBil levels and adjusted risk of MACE (hazard ratio, 0.279; 95% confidence interval, 0.088-0.877; p = 0.029). A prediction score model composed of TBil, age, hypertension history, and other eight variables was developed, with scores ranging from 0 to 500. The scores categorized patients into low-, medium-, and high-risk categories. The cumulative survival rate was significantly higher in the low-risk group than in the medium- and high-risk groups for MACE, all-cause death, cardiac death, recurrent myocardial infarction, and ischemic stroke (p < 0.001, p < 0.001, p < 0.001, p = 0.030, and p = 0.001, respectively). The area under the curve of the TBil score was 0.768; this was significantly greater in the pairwise comparison with the Global Registry of Acute Coronary Events score (p = 0.0012). Conclusion: The new prediction score model based on TBil could be used in clinical practice to support risk stratification as recommended in the clinical guidelines.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Ying Wang
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Chen Liu
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Peng Zhou
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Zhaoxue Sheng
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Jiannan Li
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Jinying Zhou
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Runzhen Chen
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Yi Chen
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Hanjun Zhao
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Hongbing Yan
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China.,Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
| |
Collapse
|
23
|
Fu J, Wang Q, Zhang L, Liu J, Wang G. Serum Bilirubin Level Is Increased in Metabolically Healthy Obesity. Front Endocrinol (Lausanne) 2021; 12:792795. [PMID: 35432184 PMCID: PMC9005889 DOI: 10.3389/fendo.2021.792795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Bilirubin is a biochemical substance with metabolic benefits. The objective of this research was to elucidate the association between serum bilirubin levels and metabolic alterations in different obesity phenotypes. METHODS In total, 1,042 drug-naive participants were included in the study. Of them, 541 were obese patients and 501 were age-matched and sex-matched healthy control subjects. The obese patients were divided into metabolically healthy obesity (MHO) group and metabolically unhealthy obesity (MUHO) group according to the levels of fasting plasma glucose (FBG), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C) and blood pressure (BP). Clinical and biochemical parameters including total bilirubin (TBil), indirect bilirubin (IBil) and direct bilirubin (DBil) were measured. ANOVA or Kruskal-Wallis H test was used to test differences among the three groups. Pearson and Spearman correlations were used to analyze the relationships between two parameters. The relationships between bilirubin and other variables were analyzed using Multivariate regression analysis. RESULTS MHO group had favorable blood pressure, glucose and lipids profiles, along with increased TBil and DBil, and decreased high-sensitivity C-reactive protein (hsCRP) and homeostasis model assessment of insulin resistance (HOMA-IR) levels when compared to MUHO group (P < 0.05 for all). TBil and DBil were negatively correlated with total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), fasting insulin (FINS), hsCRP and HOMA-IR, even after adjusted for age, gender and BMI (all P <0.01). Multivariate regression analysis demonstrated that HOMA-IR was independently correlated with TBil and DBIi levels (β = -0.400, P < 0.01). CONCLUSION MHO group harbors increased bilirubin level compared with MUHO group. HOMA-IR was independently correlated with TBil and DBIi levels.
Collapse
|
24
|
Puentes-Pardo JD, Moreno-SanJuan S, Carazo Á, León J. Heme Oxygenase-1 in Gastrointestinal Tract Health and Disease. Antioxidants (Basel) 2020; 9:antiox9121214. [PMID: 33276470 PMCID: PMC7760122 DOI: 10.3390/antiox9121214] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022] Open
Abstract
Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. HO-1, a stress inducible enzyme, is considered as an anti-oxidative and cytoprotective agent. As many studies suggest, HO-1 is highly expressed in the gastrointestinal tract where it is involved in the response to inflammatory processes, which may lead to several diseases such as pancreatitis, diabetes, fatty liver disease, inflammatory bowel disease, and cancer. In this review, we highlight the pivotal role of HO-1 and its downstream effectors in the development of disorders and their beneficial effects on the maintenance of the gastrointestinal tract health. We also examine clinical trials involving the therapeutic targets derived from HO-1 system for the most common diseases of the digestive system.
Collapse
Affiliation(s)
- Jose D. Puentes-Pardo
- Research Unit, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18011 Granada, Spain
- Correspondence: (J.D.P.-P.); (J.L.); Tel.: +34-958-023-706 (J.L.)
| | - Sara Moreno-SanJuan
- Cytometry and Microscopy Research Service, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain;
| | - Ángel Carazo
- Genomic Research Service, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain;
| | - Josefa León
- Research Unit, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain
- Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, 18016 Granada, Spain
- Correspondence: (J.D.P.-P.); (J.L.); Tel.: +34-958-023-706 (J.L.)
| |
Collapse
|
25
|
Jayanti S, Vítek L, Tiribelli C, Gazzin S. The Role of Bilirubin and the Other "Yellow Players" in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:900. [PMID: 32971784 PMCID: PMC7555389 DOI: 10.3390/antiox9090900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/16/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023] Open
Abstract
Bilirubin is a yellow endogenous derivate of the heme catabolism. Since the 1980s, it has been recognized as one of the most potent antioxidants in nature, able to counteract 10,000× higher intracellular concentrations of H2O2. In the recent years, not only bilirubin, but also its precursor biliverdin, and the enzymes involved in their productions (namely heme oxygenase and biliverdin reductase; altogether the "yellow players"-YPs) have been recognized playing a protective role in diseases characterized by a chronic prooxidant status. Based on that, there is an ongoing effort in inducing their activity as a therapeutic option. Nevertheless, the understanding of their specific contributions to pathological conditions of the central nervous system (CNS) and their role in these diseases are limited. In this review, we will focus on the most recent evidence linking the role of the YPs specifically to neurodegenerative and neurological conditions. Both the protective, as well as potentially worsening effects of the YP's activity will be discussed.
Collapse
Affiliation(s)
- Sri Jayanti
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
- Faculty of Medicine, Universitas Hasanuddin, Makassar 90245, Indonesia
- Molecular Biomedicine Ph.D. Program, University of Trieste, 34127 Trieste, Italy
| | - Libor Vítek
- Institute of Medical Biochemistry and Laboratory Diagnostics, and 4th Department of Internal Medicine, Faculty General Hospital and 1st Faculty of Medicine, Charles University, 12000 Prague, Czech Republic;
| | - Claudio Tiribelli
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
| | - Silvia Gazzin
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
| |
Collapse
|
26
|
Seyed Khoei N, Jenab M, Murphy N, Banbury BL, Carreras-Torres R, Viallon V, Kühn T, Bueno-de-Mesquita B, Aleksandrova K, Cross AJ, Weiderpass E, Stepien M, Bulmer A, Tjønneland A, Boutron-Ruault MC, Severi G, Carbonnel F, Katzke V, Boeing H, Bergmann MM, Trichopoulou A, Karakatsani A, Martimianaki G, Palli D, Tagliabue G, Panico S, Tumino R, Sacerdote C, Skeie G, Merino S, Bonet C, Rodríguez-Barranco M, Gil L, Chirlaque MD, Ardanaz E, Myte R, Hultdin J, Perez-Cornago A, Aune D, Tsilidis KK, Albanes D, Baron JA, Berndt SI, Bézieau S, Brenner H, Campbell PT, Casey G, Chan AT, Chang-Claude J, Chanock SJ, Cotterchio M, Gallinger S, Gruber SB, Haile RW, Hampe J, Hoffmeister M, Hopper JL, Hsu L, Huyghe JR, Jenkins MA, Joshi AD, Kampman E, Larsson SC, Le Marchand L, Li CI, Li L, Lindblom A, Lindor NM, Martín V, Moreno V, Newcomb PA, Offit K, Ogino S, Parfrey PS, Pharoah PDP, Rennert G, Sakoda LC, Schafmayer C, Schmit SL, Schoen RE, Slattery ML, Thibodeau SN, Ulrich CM, van Duijnhoven FJB, Weigl K, Weinstein SJ, White E, Wolk A, Woods MO, Wu AH, Zhang X, Ferrari P, Anton G, Peters A, Peters U, Gunter MJ, Wagner KH, Freisling H. Circulating bilirubin levels and risk of colorectal cancer: serological and Mendelian randomization analyses. BMC Med 2020; 18:229. [PMID: 32878631 PMCID: PMC7469292 DOI: 10.1186/s12916-020-01703-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/09/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Bilirubin, a byproduct of hemoglobin breakdown and purported anti-oxidant, is thought to be cancer preventive. We conducted complementary serological and Mendelian randomization (MR) analyses to investigate whether alterations in circulating levels of bilirubin are associated with risk of colorectal cancer (CRC). We decided a priori to perform analyses separately in men and women based on suggestive evidence that associations may differ by sex. METHODS In a case-control study nested in the European Prospective Investigation into Cancer and Nutrition (EPIC), pre-diagnostic unconjugated bilirubin (UCB, the main component of total bilirubin) concentrations were measured by high-performance liquid chromatography in plasma samples of 1386 CRC cases and their individually matched controls. Additionally, 115 single-nucleotide polymorphisms (SNPs) robustly associated (P < 5 × 10-8) with circulating total bilirubin were instrumented in a 2-sample MR to test for a potential causal effect of bilirubin on CRC risk in 52,775 CRC cases and 45,940 matched controls in the Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO), the Colon Cancer Family Registry (CCFR), and the Colorectal Transdisciplinary (CORECT) study. RESULTS The associations between circulating UCB levels and CRC risk differed by sex (Pheterogeneity = 0.008). Among men, higher levels of UCB were positively associated with CRC risk (odds ratio [OR] = 1.19, 95% confidence interval [CI] = 1.04-1.36; per 1-SD increment of log-UCB). In women, an inverse association was observed (OR = 0.86 (0.76-0.97)). In the MR analysis of the main UGT1A1 SNP (rs6431625), genetically predicted higher levels of total bilirubin were associated with a 7% increase in CRC risk in men (OR = 1.07 (1.02-1.12); P = 0.006; per 1-SD increment of total bilirubin), while there was no association in women (OR = 1.01 (0.96-1.06); P = 0.73). Raised bilirubin levels, predicted by instrumental variables excluding rs6431625, were suggestive of an inverse association with CRC in men, but not in women. These differences by sex did not reach formal statistical significance (Pheterogeneity ≥ 0.2). CONCLUSIONS Additional insight into the relationship between circulating bilirubin and CRC is needed in order to conclude on a potential causal role of bilirubin in CRC development.
Collapse
Affiliation(s)
- Nazlisadat Seyed Khoei
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Mazda Jenab
- Nutritional Epidemiology Group, Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Neil Murphy
- Nutritional Epidemiology Group, Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Barbara L Banbury
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Robert Carreras-Torres
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL). L'Hospitalet de Llobregat, Barcelona, Spain
| | - Vivian Viallon
- Nutritional Methodology and Biostatistics Group, Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), 150 cours Albert Thomas, 69372, Lyon CEDEX 08, France
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center, Utrecht, The Netherlands
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Krasimira Aleksandrova
- Group Nutrition, Immunity and Metabolism, Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | | | - Magdalena Stepien
- Nutritional Epidemiology Group, Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Andrew Bulmer
- School of Medicine, Griffith University, Brisbane, QLD, Australia
- Alliance for Vascular Access Teaching and Research (AVATAR), Menzies Health Institute Queensland, Griffith University, Brisbane, QLD, Australia
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Marie-Christine Boutron-Ruault
- CESP (Centre de Recherche en Epidémiologie et Santé des Populations), Fac. de médecine - Univ. Paris-Sud, Fac. de médecine - UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
| | - Gianluca Severi
- CESP (Centre de Recherche en Epidémiologie et Santé des Populations), Fac. de médecine - Univ. Paris-Sud, Fac. de médecine - UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
| | - Franck Carbonnel
- CESP (Centre de Recherche en Epidémiologie et Santé des Populations), Fac. de médecine - Univ. Paris-Sud, Fac. de médecine - UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
- Department of Gastroenterology, Bicêtre University Hospital, Public Assistance Hospitals of Paris, Le Kremlin Bicêtre, France
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Postdam-Rehbrücke, Nuthetal, Germany
| | - Manuela M Bergmann
- Department of Epidemiology, German Institute of Human Nutrition Postdam-Rehbrücke, Nuthetal, Germany
| | | | - Anna Karakatsani
- Hellenic Health Foundation, Athens, Greece
- 2nd Pulmonary Medicine Department, School of Medicine, National and Kapodistrian University of Athens, "ATTIKON" University Hospital, Haidari, Greece
| | | | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network - ISPRO, Florence, Italy
| | - Giovanna Tagliabue
- Lombardy Cancer Registry Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, "M.P. Arezzo" Hospital, ASP Ragusa, Ragusa, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Guri Skeie
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
- Nutritional Epidemiology Group, School of Food and Nutrition, University of Leeds, Leeds, UK
| | | | - Catalina Bonet
- Cancer Epidemiology Research Program, Unit of Nutrition and Cancer, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Miguel Rodríguez-Barranco
- Escuela Andaluza de Salud Pública. Instituto de Investigación Biosanitaria, ibs. GRANADA, Universidad de Granada, Granada, Spain
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Leire Gil
- Public Health Division of Gipuzkoa-BIODONOSTIA, Basque Regional Health Department, San Sebastian, Spain
| | - Maria-Dolores Chirlaque
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia University, Murcia, Spain
| | - Eva Ardanaz
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Robin Myte
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
| | - Johan Hultdin
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Nutrition, Bjørknes University College, Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John A Baron
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stéphane Bézieau
- Service de Génétique Médicale, Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter T Campbell
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, GA, USA
| | - Graham Casey
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Medical Centre Hamburg-Eppendorf, University Cancer Centre Hamburg (UCCH), Hamburg, Germany
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michelle Cotterchio
- Prevention and Cancer Control, Cancer Care Ontario, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Steven Gallinger
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Stephen B Gruber
- Department of Preventive Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert W Haile
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Jochen Hampe
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Epidemiology, School of Public Health and Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Jeroen R Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Amit D Joshi
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Ellen Kampman
- Division of Human Nutrition, Wageningen University and Research, Wageningen, The Netherlands
| | - Susanna C Larsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Christopher I Li
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Li Li
- Department of Family Medicine, University of Virginia, Charlottesville, VA, USA
| | - Annika Lindblom
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Noralane M Lindor
- Department of Health Science Research, Mayo Clinic, Scottsdale, AZ, USA
| | - Vicente Martín
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Biomedicine Institute (IBIOMED), University of León, León, Spain
| | - Victor Moreno
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL). L'Hospitalet de Llobregat, Barcelona, Spain
- Cancer Epidemiology Research Program, Unit of Nutrition and Cancer, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Kenneth Offit
- Clinical Genetics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, USA
| | - Shuji Ogino
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Patrick S Parfrey
- The Clinical Epidemiology Unit, Memorial University Medical School, Newfoundland, Canada
| | - Paul D P Pharoah
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Gad Rennert
- Department of Community Medicine and Epidemiology, Lady Davis Carmel Medical Center, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Clalit National Cancer Control Center, Haifa, Israel
| | - Lori C Sakoda
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Clemens Schafmayer
- Department of General, Visceral, Vascular, and Transplantation Surgery, University Hospital Rostock, Rostock, Germany
| | - Stephanie L Schmit
- Department of Preventive Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert E Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Stephen N Thibodeau
- Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Cornelia M Ulrich
- Huntsman Cancer Institute and Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | | | - Korbinian Weigl
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Michael O Woods
- Discipline of Genetics, Memorial University of Newfoundland, St. John's, Canada
| | - Anna H Wu
- University of Southern California, Preventative Medicine, Los Angeles, CA, USA
| | - Xuehong Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Pietro Ferrari
- Nutritional Methodology and Biostatistics Group, Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), 150 cours Albert Thomas, 69372, Lyon CEDEX 08, France
| | - Gabriele Anton
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Marc J Gunter
- Nutritional Epidemiology Group, Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Karl-Heinz Wagner
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Heinz Freisling
- Nutritional Methodology and Biostatistics Group, Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), 150 cours Albert Thomas, 69372, Lyon CEDEX 08, France.
| |
Collapse
|
27
|
Liu H, Perumal N, Manicam C, Mercieca K, Prokosch V. Proteomics Reveals the Potential Protective Mechanism of Hydrogen Sulfide on Retinal Ganglion Cells in an Ischemia/Reperfusion Injury Animal Model. Pharmaceuticals (Basel) 2020; 13:ph13090213. [PMID: 32867129 PMCID: PMC7557839 DOI: 10.3390/ph13090213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Glaucoma is the leading cause of irreversible blindness and is characterized by progressive retinal ganglion cell (RGC) degeneration. Hydrogen sulfide (H2S) is a potent neurotransmitter and has been proven to protect RGCs against glaucomatous injury in vitro and in vivo. This study is to provide an overall insight of H2S’s role in glaucoma pathophysiology. Ischemia-reperfusion injury (I/R) was induced in Sprague-Dawley rats (n = 12) by elevating intraocular pressure to 55 mmHg for 60 min. Six of the animals received intravitreal injection of H2S precursor prior to the procedure and the retina was harvested 24 h later. Contralateral eyes were assigned as control. RGCs were quantified and compared within the groups. Retinal proteins were analyzed via label-free mass spectrometry based quantitative proteomics approach. The pathways of the differentially expressed proteins were identified by ingenuity pathway analysis (IPA). H2S significantly improved RGC survival against I/R in vivo (p < 0.001). In total 1115 proteins were identified, 18 key proteins were significantly differentially expressed due to I/R and restored by H2S. Another 11 proteins were differentially expressed following H2S. IPA revealed a significant H2S-mediated activation of pathways related to mitochondrial function, iron homeostasis and vasodilation. This study provides first evidence of the complex role that H2S plays in protecting RGC against I/R.
Collapse
Affiliation(s)
- Hanhan Liu
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Natarajan Perumal
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Caroline Manicam
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Karl Mercieca
- Royal Eye Hospital, School of Medicine, University of Manchester, Manchester M13 9WH, UK;
| | - Verena Prokosch
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Correspondence: ; Tel.: +49-1703862250
| |
Collapse
|
28
|
Hansen TWR, Wong RJ, Stevenson DK. Molecular Physiology and Pathophysiology of Bilirubin Handling by the Blood, Liver, Intestine, and Brain in the Newborn. Physiol Rev 2020; 100:1291-1346. [PMID: 32401177 DOI: 10.1152/physrev.00004.2019] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Bilirubin is the end product of heme catabolism formed during a process that involves oxidation-reduction reactions and conserves iron body stores. Unconjugated hyperbilirubinemia is common in newborn infants, but rare later in life. The basic physiology of bilirubin metabolism, such as production, transport, and excretion, has been well described. However, in the neonate, numerous variables related to nutrition, ethnicity, and genetic variants at several metabolic steps may be superimposed on the normal physiological hyperbilirubinemia that occurs in the first week of life and results in bilirubin levels that may be toxic to the brain. Bilirubin exists in several isomeric forms that differ in their polarities and is considered a physiologically important antioxidant. Here we review the chemistry of the bilirubin molecule and its metabolism in the body with a particular focus on the processes that impact the newborn infant, and how differences relative to older children and adults contribute to the risk of developing both acute and long-term neurological sequelae in the newborn infant. The final section deals with the interplay between the brain and bilirubin and its entry, clearance, and accumulation. We conclude with a discussion of the current state of knowledge regarding the mechanism(s) of bilirubin neurotoxicity.
Collapse
Affiliation(s)
- Thor W R Hansen
- Division of Paediatric and Adolescent Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; and Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Ronald J Wong
- Division of Paediatric and Adolescent Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; and Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - David K Stevenson
- Division of Paediatric and Adolescent Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; and Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
29
|
Sugishima M, Wada K, Fukuyama K. Recent Advances in the Understanding of the Reaction Chemistries of the Heme Catabolizing Enzymes HO and BVR Based on High Resolution Protein Structures. Curr Med Chem 2020; 27:3499-3518. [PMID: 30556496 PMCID: PMC7509768 DOI: 10.2174/0929867326666181217142715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/21/2018] [Accepted: 12/11/2018] [Indexed: 01/15/2023]
Abstract
In mammals, catabolism of the heme group is indispensable for life. Heme is first cleaved by the enzyme Heme Oxygenase (HO) to the linear tetrapyrrole Biliverdin IXα (BV), and BV is then converted into bilirubin by Biliverdin Reductase (BVR). HO utilizes three Oxygen molecules (O2) and seven electrons supplied by NADPH-cytochrome P450 oxidoreductase (CPR) to open the heme ring and BVR reduces BV through the use of NAD(P)H. Structural studies of HOs, including substrate-bound, reaction intermediate-bound, and several specific inhibitor-bound forms, reveal details explaining substrate binding to HO and mechanisms underlying-specific HO reaction progression. Cryo-trapped structures and a time-resolved spectroscopic study examining photolysis of the bond between the distal ligand and heme iron demonstrate how CO, produced during the HO reaction, dissociates from the reaction site with a corresponding conformational change in HO. The complex structure containing HO and CPR provides details of how electrons are transferred to the heme-HO complex. Although the tertiary structure of BVR and its complex with NAD+ was determined more than 10 years ago, the catalytic residues and the reaction mechanism of BVR remain unknown. A recent crystallographic study examining cyanobacterial BVR in complex with NADP+ and substrate BV provided some clarification regarding these issues. Two BV molecules are bound to BVR in a stacked manner, and one BV may assist in the reductive catalysis of the other BV. In this review, recent advances illustrated by biochemical, spectroscopic, and crystallographic studies detailing the chemistry underlying the molecular mechanism of HO and BVR reactions are presented.
Collapse
Affiliation(s)
- Masakazu Sugishima
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, Japan
| | - Kei Wada
- Department of Medical Sciences, University of Miyazaki, Miyazaki, Japan
| | - Keiichi Fukuyama
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Japan.,Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Japan
| |
Collapse
|
30
|
Taboada C, Brunetti AE, Lyra ML, Fitak RR, Faigón Soverna A, Ron SR, Lagorio MG, Haddad CFB, Lopes NP, Johnsen S, Faivovich J, Chemes LB, Bari SE. Multiple origins of green coloration in frogs mediated by a novel biliverdin-binding serpin. Proc Natl Acad Sci U S A 2020; 117:18574-18581. [PMID: 32661155 PMCID: PMC7414155 DOI: 10.1073/pnas.2006771117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
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.
Collapse
Affiliation(s)
- Carlos Taboada
- Department of Biology, Duke University, Durham, NC 27708;
- División Herpetología, Museo Argentino de Ciencias Naturales "Bernardino Rivadavia," Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1405DJR, Argentina
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1428EHA, Argentina
- Núcleo de Pesquisa em Produtos Naturais e Sintéticos (NPPNS), Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14040903 Ribeirão Preto, São Paulo, Brazil
| | - Andrés E Brunetti
- Núcleo de Pesquisa em Produtos Naturais e Sintéticos (NPPNS), Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14040903 Ribeirão Preto, São Paulo, Brazil
- Laboratorio de Genética Evolutiva "Claudio Juan Bidau," Instituto de Biología Subtropical (CONICET-UNaM), Facultad de Ciencias Exactas, Universidad Nacional de Misiones, 3300 Posadas, Misiones, Argentina
| | - Mariana L Lyra
- Departamento de Biodiversidade e Centro de Aquicultura, Instituto de Biociências, Universidade Estadual Paulista, 13506-900 Rio Claro, São Paulo, Brazil
| | - Robert R Fitak
- Department of Biology, Duke University, Durham, NC 27708
- Department of Biology, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL 32816
| | - Ana Faigón Soverna
- División Herpetología, Museo Argentino de Ciencias Naturales "Bernardino Rivadavia," Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1405DJR, Argentina
| | - Santiago R Ron
- Museo de Zoología, Escuela de Biología, Pontificia Universidad Católica del Ecuador, Aptdo. 17-01-2184, Quito, Ecuador
| | - María G Lagorio
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1428EHA, Argentina
- Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1428EHA, Argentina
| | - Célio F B Haddad
- Departamento de Biodiversidade e Centro de Aquicultura, Instituto de Biociências, Universidade Estadual Paulista, 13506-900 Rio Claro, São Paulo, Brazil
| | - Norberto P Lopes
- Núcleo de Pesquisa em Produtos Naturais e Sintéticos (NPPNS), Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14040903 Ribeirão Preto, São Paulo, Brazil
| | - Sönke Johnsen
- Department of Biology, Duke University, Durham, NC 27708
| | - Julián Faivovich
- División Herpetología, Museo Argentino de Ciencias Naturales "Bernardino Rivadavia," Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1405DJR, Argentina
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1428EHA, Argentina
| | - Lucía B Chemes
- Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina;
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, CP1650 San Martín, Buenos Aires, Argentina
| | - Sara E Bari
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1428EHA, Argentina;
| |
Collapse
|
31
|
Corona D, Ekser B, Gioco R, Caruso M, Schipa C, Veroux P, Giaquinta A, Granata A, Veroux M. Heme-Oxygenase and Kidney Transplantation: A Potential for Target Therapy? Biomolecules 2020; 10:E840. [PMID: 32486245 PMCID: PMC7355572 DOI: 10.3390/biom10060840] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/24/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022] Open
Abstract
Kidney transplantation is a well-established therapy for patients with end-stage renal disease. While a significant improvement of short-term results has been achieved in the short-term, similar results were not reported in the long-term. Heme-oxygenase (HO) is the rate-limiting enzyme in heme catabolism, converting heme to iron, carbon monoxide, and biliverdin. Heme-oxygenase overexpression may be observed in all phases of transplant processes, including brain death, recipient management, and acute and chronic rejection. HO induction has been proved to provide a significant reduction of inflammatory response and a reduction of ischemia and reperfusion injury in organ transplantation, as well as providing a reduction of incidence of acute rejection. In this review, we will summarize data on HO and kidney transplantation, suggesting possible clinical applications in the near future to improve the long-term outcomes.
Collapse
Affiliation(s)
- Daniela Corona
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (D.C.); (M.C.)
- Organ Transplant Unit, University Hospital of Catania, 95123 Catania, Italy; (P.V.); (A.G.)
| | - Burcin Ekser
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46077, USA;
| | - Rossella Gioco
- General Surgery Unit, University Hospital of Catania, 95123 Catania, Italy; (R.G.); (C.S.)
| | - Massimo Caruso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (D.C.); (M.C.)
| | - Chiara Schipa
- General Surgery Unit, University Hospital of Catania, 95123 Catania, Italy; (R.G.); (C.S.)
| | - Pierfrancesco Veroux
- Organ Transplant Unit, University Hospital of Catania, 95123 Catania, Italy; (P.V.); (A.G.)
| | - Alessia Giaquinta
- Organ Transplant Unit, University Hospital of Catania, 95123 Catania, Italy; (P.V.); (A.G.)
| | | | - Massimiliano Veroux
- Organ Transplant Unit, University Hospital of Catania, 95123 Catania, Italy; (P.V.); (A.G.)
- General Surgery Unit, University Hospital of Catania, 95123 Catania, Italy; (R.G.); (C.S.)
| |
Collapse
|
32
|
Cannabidiol Protects Dopaminergic Neurons in Mesencephalic Cultures against the Complex I Inhibitor Rotenone Via Modulation of Heme Oxygenase Activity and Bilirubin. Antioxidants (Basel) 2020; 9:antiox9020135. [PMID: 32033040 PMCID: PMC7070382 DOI: 10.3390/antiox9020135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/25/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
Phytocannabinoids protect neurons against stressful conditions, possibly via the heme oxygenase (HO) system. In cultures of primary mesencephalic neurons and neuroblastoma cells, we determined the capability of cannabidiol (CBD) and tetrahydrocannabinol (THC) to counteract effects elicited by complex I-inhibitor rotenone by analyzing neuron viability, morphology, gene expression of IL6, CHOP, XBP1, HO-1 (stress response), and HO-2, and in vitro HO activity. Incubation with rotenone led to a moderate stress response but massive degeneration of dopaminergic neurons (DN) in primary mesencephalic cultures. Both phytocannabinoids inhibited in-vitro HO activity, with CBD being more potent. Inhibition of the enzyme reaction was not restricted to neuronal cells and occurred in a non-competitive manner. Although CBD itself decreased viability of the DNs (from 100% to 78%), in combination with rotenone, it moderately increased survival from 28.6% to 42.4%. When the heme degradation product bilirubin (BR) was added together with CBD, rotenone-mediated degeneration of DN was completely abolished, resulting in approximately the number of DN determined with CBD alone (77.5%). Using N18TG2 neuroblastoma cells, we explored the neuroprotective mechanism underlying the combined action of CBD and BR. CBD triggered the expression of HO-1 and other cell stress markers. Co-treatment with rotenone resulted in the super-induction of HO-1 and an increased in-vitro HO-activity. Co-application of BR completely mitigated the rotenone-induced stress response. Our findings indicate that CBD induces HO-1 and increases the cellular capacity to convert heme when stressful conditions are met. Our data further suggest that CBD via HO may confer full protection against (oxidative) stress when endogenous levels of BR are sufficiently high.
Collapse
|
33
|
Canesin G, Hejazi SM, Swanson KD, Wegiel B. Heme-Derived Metabolic Signals Dictate Immune Responses. Front Immunol 2020; 11:66. [PMID: 32082323 PMCID: PMC7005208 DOI: 10.3389/fimmu.2020.00066] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/10/2020] [Indexed: 12/21/2022] Open
Abstract
Heme is one of the most abundant molecules in the body acting as the functional core of hemoglobin/myoglobin involved in the O2/CO2 carrying in the blood and tissues, redox enzymes and cytochromes in mitochondria. However, free heme is toxic and therefore its removal is a significant priority for the host. Heme is a well-established danger-associated molecular pattern (DAMP), which binds to toll-like receptor 4 (TLR4) to induce immune responses. Heme-derived metabolites including the bile pigments, biliverdin (BV) and bilirubin (BR), were first identified as toxic drivers of neonatal jaundice in 1800 but have only recently been appreciated as endogenous drivers of multiple signaling pathways involved in protection from oxidative stress and regulators of immune responses. The tissue concentration of heme, BV and BR is tightly controlled. Heme oxygenase-1 (HO-1, encoded by HMOX1) produces BV by heme degradation, while biliverdin reductase-A (BLVR-A) generates BR by the subsequent conversion of BV. BLVR-A is a fascinating protein that possesses a classical protein kinase domain, which is activated in response to BV binding to its enzymatic site and initiates the downstream mitogen-activated protein kinases (MAPK) and phosphatidylinositol 3-kinase (PI3K) pathways. This links BLVR-A activity to cell growth and survival pathways. BLVR-A also contains a bZip DNA binding domain and a nuclear export sequence (NES) and acts as a transcription factor to regulate the expression of immune modulatory genes. Here we will discuss the role of heme-related immune response and the potential for targeting the heme system for therapies directed toward hepatitis and cancer.
Collapse
Affiliation(s)
- Giacomo Canesin
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Seyed M. Hejazi
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Kenneth D. Swanson
- Brain Tumor Center and Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Barbara Wegiel
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
34
|
Choi Y, Lee SJ, Spiller W, Jung KJ, Lee JY, Kimm H, Back JH, Lee S, Jee SH. Causal Associations Between Serum Bilirubin Levels and Decreased Stroke Risk: A Two-Sample Mendelian Randomization Study. Arterioscler Thromb Vasc Biol 2019; 40:437-445. [PMID: 31801373 PMCID: PMC6975519 DOI: 10.1161/atvbaha.119.313055] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE A number of epidemiological studies have reported that decreased serum bilirubin, an endogenous antioxidant, is associated with cardiovascular disease. However, previous Mendelian randomization analyses conducted using a single sample have shown no evidence of association. Approach and Results: A 2-sample summary Mendelian randomization study was performed by obtaining exposure and outcome data from separate nonoverlapping samples. We utilized data from the KoGES (Korean Genome and Epidemiology Study; n=25 406) and KCPS-II (Korean Cancer Prevention Study-II; n=14 541) biobank for serum bilirubin and stroke, respectively. Using KoGES, a total of 1784 single nucleotide polymorphisms associated with serum bilirubin levels were discovered using a genome-wide significance threshold (P<5×10-8), of which 10 single nucleotide polymorphisms were identified as independent (R2<0.005) and adopted as genetic instruments. From KCPS-II, total and ischemic stroke cases were identified (n=1489 and n=686), with 12 366 acting as controls. Various 2-sample summary Mendelian randomization methods were employed, with Mendelian randomization estimates showing an inverse causal association between serum bilirubin levels and total stroke risk (odds ratio, 0.481 [95% CI, 0.234-0.988]; P=0.046). This association increased in magnitude when restricting the analysis to ischemic stroke cases (odds ratio, 0.302 [95% CI, 0.105-0.868]; P=0.026). CONCLUSIONS Our findings provide evidence of significant causal relationship between high levels of bilirubin and decreased stroke risk in Korean population in agreement with observational approaches. This highlights the potential for bilirubin to serve as a therapeutic target for oxidative stress-related diseases such as stroke and suggests that previous findings were not a consequence of unmeasured confounding.
Collapse
Affiliation(s)
- Yoonjeong Choi
- From the Department of Public Health, Graduate School, Yonsei University, Seoul, Korea (Y.C., S.H.J.).,Department of Epidemiology, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea (Y.C., S.J.L., K.J.J., J.-Y.L., H.K., S.H.J.)
| | - Sun Ju Lee
- Department of Epidemiology, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea (Y.C., S.J.L., K.J.J., J.-Y.L., H.K., S.H.J.).,Health Insurance Policy Research Institute, National Health Insurance Service, Wonju, Korea (S.J.L., J.H.B., S.L.)
| | - Wes Spiller
- Population Health Science Institute, University of Bristol, Barley House, Oakfield Grove, United Kingdom (W.S.)
| | - Keum Ji Jung
- Department of Epidemiology, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea (Y.C., S.J.L., K.J.J., J.-Y.L., H.K., S.H.J.)
| | - Ji-Young Lee
- Department of Epidemiology, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea (Y.C., S.J.L., K.J.J., J.-Y.L., H.K., S.H.J.)
| | - Heejin Kimm
- Department of Epidemiology, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea (Y.C., S.J.L., K.J.J., J.-Y.L., H.K., S.H.J.)
| | - Joung Hwan Back
- Health Insurance Policy Research Institute, National Health Insurance Service, Wonju, Korea (S.J.L., J.H.B., S.L.)
| | - Sunmi Lee
- Health Insurance Policy Research Institute, National Health Insurance Service, Wonju, Korea (S.J.L., J.H.B., S.L.)
| | - Sun Ha Jee
- From the Department of Public Health, Graduate School, Yonsei University, Seoul, Korea (Y.C., S.H.J.).,Department of Epidemiology, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea (Y.C., S.J.L., K.J.J., J.-Y.L., H.K., S.H.J.)
| |
Collapse
|
35
|
Duvigneau JC, Esterbauer H, Kozlov AV. Role of Heme Oxygenase as a Modulator of Heme-Mediated Pathways. Antioxidants (Basel) 2019; 8:antiox8100475. [PMID: 31614577 PMCID: PMC6827082 DOI: 10.3390/antiox8100475] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/27/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023] Open
Abstract
The heme oxygenase (HO) system is essential for heme and iron homeostasis and necessary for adaptation to cell stress. HO degrades heme to biliverdin (BV), carbon monoxide (CO) and ferrous iron. Although mostly beneficial, the HO reaction can also produce deleterious effects, predominantly attributed to excessive product formation. Underrated so far is, however, that HO may exert effects additionally via modulation of the cellular heme levels. Heme, besides being an often-quoted generator of oxidative stress, plays also an important role as a signaling molecule. Heme controls the anti-oxidative defense, circadian rhythms, activity of ion channels, glucose utilization, erythropoiesis, and macrophage function. This broad spectrum of effects depends on its interaction with proteins ranging from transcription factors to enzymes. In degrading heme, HO has the potential to exert effects also via modulation of heme-mediated pathways. In this review, we will discuss the multitude of pathways regulated by heme to enlarge the view on HO and its role in cell physiology. We will further highlight the contribution of HO to pathophysiology, which results from a dysregulated balance between heme and the degradation products formed by HO.
Collapse
Affiliation(s)
- J Catharina Duvigneau
- Institute for Medical Biochemistry, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria.
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, 1210 Vienna, Austria.
| | - Andrey V Kozlov
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, 1200 Vienna, Austria.
- Laboratory of Navigational Redox Lipidomics, Department of Human Pathology, IM Sechenov Moscow State Medical University, 119992 Moscow, Russia.
| |
Collapse
|
36
|
Takei R, Inoue T, Sonoda N, Kohjima M, Okamoto M, Sakamoto R, Inoguchi T, Ogawa Y. Bilirubin reduces visceral obesity and insulin resistance by suppression of inflammatory cytokines. PLoS One 2019; 14:e0223302. [PMID: 31577826 PMCID: PMC6774504 DOI: 10.1371/journal.pone.0223302] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023] Open
Abstract
Objective Although previous studies have reported a negative relationship between serum bilirubin concentration and the development of diabetes mellitus (DM), the relationship between bilirubin and insulin resistance has not been thoroughly assessed. This study was designed to determine the relationships between bilirubin, body fat distribution, and adipose tissue inflammation in patients with type 2 DM and the effect of bilirubin in an obese animal model. Method Body fat distribution was measured using an abdominal dual bioelectrical impedance analyzer in patients with type 2 DM. We also measured glycemic control, lipid profile, serum bilirubin concentration and other clinical characteristics, and determined their relationships with body fat distribution. In the animal study, biliverdin (20 mg/kg daily) was orally administered to high-fat diet (HFD)-induced obese (DIO) mice for 2 weeks, after which intraperitoneal insulin tolerance testing was performed. Then, adipocyte area, adipocytokine expression, and macrophage polarization were evaluated in epididymal adipose tissues. Results In the clinical study, univariate analysis showed that a lower bilirubin concentration was significantly correlated with higher body mass index, waist circumference, triglyceride, uric acid, creatinine, visceral fat area and lower HDL-C. In multivariate analyses, bilirubin concentration significantly correlated with diastolic blood pressure, creatinine, and visceral fat area. However, there was no association between bilirubin concentration and subcutaneous fat area. In the animal study, DIO mice treated with biliverdin had smaller adipocytes than untreated DIO mice and biliverdin improved HFD-induced insulin resistance. Biliverdin treatment reversed the higher gene expression of Cd11c, encoding an M1 macrophage marker, and Tnfa, encoding the proinflammatory cytokine tumor necrosis factor-α, in the adipose tissues of DIO mice. These data suggest biliverdin administration alleviates insulin resistance by ameliorating inflammation and the dysregulation of adipocytokine expression in adipose tissues of DIO mice. Conclusions Bilirubin may protect against insulin resistance by ameliorating visceral obesity and adipose tissue inflammation.
Collapse
Affiliation(s)
- Ryoko Takei
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Fukuoka City Health Promotion Support Center, Fukuoka, Japan
| | - Tomoaki Inoue
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriyuki Sonoda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Motoyuki Kohjima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Misato Okamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuichi Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toyoshi Inoguchi
- Fukuoka City Health Promotion Support Center, Fukuoka, Japan
- * E-mail:
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
37
|
The complexity of porphyrin-like pigments in a marine annelid sheds new light on haem metabolism in aquatic invertebrates. Sci Rep 2019; 9:12930. [PMID: 31506557 PMCID: PMC6736840 DOI: 10.1038/s41598-019-49433-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/23/2019] [Indexed: 11/08/2022] Open
Abstract
True green pigments in the animal kingdom are scarce and are almost invariably porphyrinoids. Endogenous porphyrins resulting from the breakdown of haem are usually known as “bile pigments”. The pigmentation of intertidal Polychaeta has long gained attention due to its variety and vivid patterning that often seems incompatible with camouflage, as it occurs with Eulalia viridis, one of the few truly green Polychaeta. The present study combined UV and bright-field microscopy with HPLC to address the presence and distribution of pigments in several organs. The results showed two major types of porphyrin-like pigments, yellowish and greenish in colour, that are chiefly stored as intraplasmatic granules. Whereas the proboscis holds yellow pigments, the skin harbours both types in highly specialised cells. In their turn, oocytes and intestine have mostly green pigments. Despite some inter-individual variation, the pigments tend to be stable after prolonged storage at −20 °C, which has important implications for future studies. The results show that, in a foraging predator of the intertidal where melanins are circumscribed to lining the nervous system, porphyrinoid pigments have a key role in protection against UV light, in sensing and even as chemical defence against foulants and predators, which represents a remarkable adaptive feature.
Collapse
|
38
|
Heme, Heme Oxygenase, and Endoplasmic Reticulum Stress-A New Insight into the Pathophysiology of Vascular Diseases. Int J Mol Sci 2019; 20:ijms20153675. [PMID: 31357546 PMCID: PMC6695876 DOI: 10.3390/ijms20153675] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023] Open
Abstract
The prevalence of vascular disorders continues to rise worldwide. Parallel with that, new pathophysiological pathways have been discovered, providing possible remedies for prevention and therapy in vascular diseases. Growing evidence suggests that endoplasmic reticulum (ER) stress is involved in a number of vasculopathies, including atherosclerosis, vascular brain events, and diabetes. Heme, which is released from hemoglobin or other heme proteins, triggers various pathophysiological consequence, including heme stress as well as ER stress. The potentially toxic free heme is converted by heme oxygenases (HOs) into carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is reduced to bilirubin (BR). Redox-active iron is oxidized and stored by ferritin, an iron sequestering protein which exhibits ferroxidase activity. In recent years, CO, BV, and BR have been shown to control cellular processes such as inflammation, apoptosis, and antioxidant defense. This review covers our current knowledge about how heme induced endoplasmic reticulum stress (HIERS) participates in the pathogenesis of vascular disorders and highlights recent discoveries in the molecular mechanisms of HO-mediated cytoprotection in heme stress and ER stress, as well as crosstalk between ER stress and HO-1. Furthermore, we focus on the translational potential of HIERS and heme oxygenase-1 (HO-1) in atherosclerosis, diabetes mellitus, and brain hemorrhage.
Collapse
|
39
|
Jeon C, Lee JY, Lee SJ, Jung KJ, Kimm H, Jee SH. Bilirubin and risk of ischemic heart disease in Korea: a two-sample Mendelian randomization study. Epidemiol Health 2019; 41:e2019034. [PMID: 31319653 PMCID: PMC6759489 DOI: 10.4178/epih.e2019034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/12/2019] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES Bilirubin is an endogenous antioxidant that protects cells against oxidative stress. Increased plasma levels of bilirubin have been associated with a reduced risk of ischemic heart disease (IHD) in previous studies. Nonetheless, whether those associations reflect a true protective effect of bilirubin on IHD, rather than confounding or reverse causation, remains unknown. Therefore, we applied two-sample Mendelian randomization to evaluate the causal association between bilirubin levels and IHD risk in a Korean population. METHODS A total of 5 genetic variants-TRPM8 (rs10490012), USP40 (rs12993249), ATG16L1 (rs2119503), SLCO1B1 (rs4149014), and SLCO1B3 (rs73233620)-were selected as genetic instruments for serum bilirubin levels using a communitybased cohort, the Korean Genome and Epidemiology Study, comprising 33,598 subjects. We then evaluated their impact on IHD using the Korean Cancer Prevention Study-II cohort. RESULTS Among the 5 instrumental variables that showed significant associations with serum bilirubin levels, rs12993249 (USP40) showed the most significant association (p<2.36×10-105). However, we found no significant association between serum bilirubin levels and IHD. Sensitivity analyses demonstrated a consistent association, suggesting that our observations were robust. CONCLUSIONS Using two-sample Mendelian randomization, we found no association between serum bilirubin levels and IHD. Further studies that confirm the observed interactions among other ethnicities are warranted.
Collapse
Affiliation(s)
- Christina Jeon
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea.,Department of Public Health, Graduate School, Yonsei University, Seoul, Korea
| | - Ji-Young Lee
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Sun Ju Lee
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Keum Ji Jung
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Heejin Kimm
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| |
Collapse
|
40
|
Potential protective effects of bilirubin following the treatment of neonatal hypoxic-ischemic encephalopathy with hypothermia therapy. Biosci Rep 2019; 39:BSR20182332. [PMID: 31101685 PMCID: PMC6549084 DOI: 10.1042/bsr20182332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 04/21/2019] [Accepted: 05/07/2019] [Indexed: 12/02/2022] Open
Abstract
Background: Therapeutic hypothermia (TH) is the standard therapy for hypoxic-ischemic encephalopathy (HIE) and is associated with a wide range of physiological changes. Objective: We re-evaluated the effects of HIE and TH on bilirubin measurements following HIE in a center involved in the China cooling randomized controlled trial (RCT). Methods: Serial serum bilirubin concentrations measured during the first week of life were compared among the HIE + NT (normothermia) group, HIE + TH treatment group and control group (without HIE). Survivors of HIE were followed and assessed at approximately 2 years of age, and the results were correlated with peak bilirubin levels during the first week of life. Results: One hundred and thirty-eight infants were available for analysis. Significantly lower bilirubin levels were recorded in the HIE + NT group than in the controls (P<0.05). Significant differences were not observed among the patients in the HIE + NT group (mild to severe) or between the HIE + TH group and the HIE + NT group at any time point (P>0.05). The peak serum bilirubin concentrations recorded at 96 h of age showed a good correlation with the results of the Bayley Scales of Infant and Toddler Development, third edition (BSID-III) (P=0.02). Conclusion: Bilirubin potentially exerts a neuroprotective effect during the first week of life, and low temperature does not affect the possible antioxidant function of bilirubin during TH following HIE.
Collapse
|
41
|
LED-phototherapy does not induce oxidative DNA damage in hyperbilirubinemic Gunn rats. Pediatr Res 2019; 85:1041-1047. [PMID: 30851724 DOI: 10.1038/s41390-019-0367-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/18/2019] [Accepted: 02/26/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND Phototherapy (PT) is the standard treatment of neonatal unconjugated hyperbilirubinemia. Fluorescent tube (FT)-emitted PT light is known to induce oxidative DNA damage in neonates. Nowadays, however, FTs have largely been replaced by light-emitting diodes (LEDs) for delivering PT. Until now, it is unknown whether LED-PT causes oxidative DNA damage. We aim to determine whether LED-PT induces oxidative DNA damage in hyperbilirubinemic rats. METHODS Adult Gunn rats, with genetically unconjugated hyperbilirubinemia, received LED-PT in the clinically relevant doses of 10 or 30 µW/cm2/nm. Urine was collected at 0, 24, and 48 h of PT. A group of young Gunn rats received intensive LED-PT of 100 µW/cm2/nm for 24 h. Urine was collected every 8 h and analyzed for the levels of oxidative DNA damage marker 8-hydroxy-2'deoxyguanosine (8-OHdG) and creatinine. DNA damage was evaluated by immunohistochemistry (γH2AX) of skin and spleen samples. RESULTS LED-PT of 10 and 30 µW/cm2/nm did not affect urinary concentrations of 8-OHdG and creatinine or the 8-OHdG/creatinine ratio. Likewise, intensive LED-PT did not affect the 8-OHdG/creatinine ratio or the number of γH2AX-positive cells in the skin or spleen. CONCLUSIONS Our results show that LED-PT does not induce oxidative DNA damage in hyperbilirubinemic Gunn rats either at clinically relevant or intensive dosages.
Collapse
|
42
|
Pulaski L, Jatczak-Pawlik I, Sobalska-Kwapis M, Strapagiel D, Bartosz G, Sadowska-Bartosz I. 3-Bromopyruvate induces expression of antioxidant genes. Free Radic Res 2019; 53:170-178. [PMID: 30362385 DOI: 10.1080/10715762.2018.1541176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
An alkylating compound, 3-bromopyruvic acid (3-3-bromopyruvic acid (BP)) is a promising anti-cancer agent, potentially able to act on multidrug-resistant cells. Its action has been attributed mainly to inhibition of glycolysis. This compound induces also oxidative stress at a cellular level. The effects of 3-BP on gene expression have not been studied although they may determine the survival of cells exposed to 3-BP. The aim of this paper was to examine the effect 3-BP on gene expression pattern in breast MCF-7 cancer cells. Detection of the differences in gene expression was performed using microarrays and dysregulated genes were validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Exposure of cells to 100 µM 3-BP for 6, 12 and 24 increased expression and diminished expression of 39 and 6 genes, respectively. Among the induced genes, 22 belong to general cellular stress response genes, maintenance genes involved in redox homeostasis, responding to oxidative stress (among them metallothioneins, low-molecular-weight thiol homeostasis enzymes and genes coding for NAD(P)H-dependent oxidoreductases operating on complex organic substrates, including aldo-keto reductases). These results demonstrate that transient oxidative stress in cells exposed to 3-BP is followed by antioxidant response.
Collapse
Affiliation(s)
- Lukasz Pulaski
- a Laboratory of Transcriptional Regulation, Institute of Medical Biology , Polish Academy of Sciences , Lodz , Poland.,b Faculty of Biology and Environmental Protection, Department of Molecular Biophysics , University of Lodz , Lodz , Poland
| | - Izabela Jatczak-Pawlik
- b Faculty of Biology and Environmental Protection, Department of Molecular Biophysics , University of Lodz , Lodz , Poland
| | - Marta Sobalska-Kwapis
- c Biobank Lab, Faculty of Biology and Environmental Protection, Department of Molecular Biophysics , University of Lodz , Lodz , Poland
| | - Dominik Strapagiel
- c Biobank Lab, Faculty of Biology and Environmental Protection, Department of Molecular Biophysics , University of Lodz , Lodz , Poland
| | - Grzegorz Bartosz
- b Faculty of Biology and Environmental Protection, Department of Molecular Biophysics , University of Lodz , Lodz , Poland
| | - Izabela Sadowska-Bartosz
- d Department of Analytical Biochemistry Faculty of Biology and Agriculture , University of Rzeszow , Rzeszow , Poland
| |
Collapse
|
43
|
Affiliation(s)
- Jon Y. Takemoto
- Department of BiologyUtah State University, Logan Utah 84322-5305 U.S.A
| | - Cheng‐Wei T. Chang
- Department of Chemistry and BiochemistryUtah State University Logan, Utah 84322-0300 U.S.A
| | - Dong Chen
- Department of Biological EngineeringUtah State University Logan, Utah 843122 U.S.A
| | - Garrett Hinton
- Department of BiologyUtah State University Logan, Utah 84322-5305 U.S.A
| |
Collapse
|
44
|
Gundamaraju R, Vemuri R, Chong WC, Bulmer AC, Eri R. Bilirubin Attenuates ER Stress-Mediated Inflammation, Escalates Apoptosis and Reduces Proliferation in the LS174T Colonic Epithelial Cell Line. Int J Med Sci 2019; 16:135-144. [PMID: 30662337 PMCID: PMC6332492 DOI: 10.7150/ijms.29134] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Mildly elevated serum unconjugated bilirubin (UCB) concentrations are associated with protection against disease conditions underpinned by cellular and metabolic stress. To determine the potential therapeutic efficacy of UCB we tested it in an in vitro model of gut inflammation. Tunicamycin TUN (10 µg/mL) was used to induce endoplasmic reticular stress (ERS) affecting N-glycosylation in LS174T cells. Cultured cells were investigated with addition of UCB at doses 0.1, 1 and 10µM (resulting in bilirubin:albumin ratios of 0.325-0.003)against ER stress-mediated effects including inflammation, cell survival (determined by apoptosis) and proliferation. Gene expression of ER stress markers (Grp78, Perk, XBP1 and ATF6) were evaluated in addition to cytokine concentrations in media after six hours of treatment. We then verified the potential role of UCB in executing programmed cell death via PARP, Caspase3 and Annexin V assays and further explored cell proliferation using the Click-iT EdU assay. A dose of 10µM UCB most potently reduced tunicamycin-mediated effects on enhanced UPR markers, inflammatory cytokines and proliferation; however all the doses (i.e.0.1-10µM) reduced the expression of ER stress and inflammatory markers Grp78, NLRP3, IL1-b, XBP1, PERK and ATF6. Furthermore, media concentrations of pro-inflammatory cytokines IL-8, IL-4 and TNFα decreased and the anti-inflammatory cytokine IL-10 increased (P<0.05). A dose of 10µM UCB initiated intrinsic apoptosis via Caspase 3 and in addition reduced cellular proliferation. Collectively, these data indicate that co treatment with UCB resulted in reducing ER stress response to TUN in gastrointestinal epithelial cells, reduced the subsequent inflammatory response, induced cancer cell death and decreased cellular proliferation. These data suggest that mildly elevated circulating or enteric UCB might protect against gastrointestinal inflammatory disorders.
Collapse
Affiliation(s)
- Rohit Gundamaraju
- School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| | - Ravichandra Vemuri
- School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| | - Wai Chin Chong
- School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| | - Andrew Cameron Bulmer
- School of Medical Science and Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld, Australia
| | - Rajaraman Eri
- School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| |
Collapse
|
45
|
Keskin E, Karabakan M, Bozkurt A, Hirik E, Karabulut İ, Gunay M, Çakan M. Is there any relationship between serum levels of total bilirubin and the severity of erectile dysfunction? Urologia 2018; 85:106-110. [PMID: 29633653 DOI: 10.1177/0391560317749424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Recent studies have shown that atherosclerosis is associated with erectile dysfunction and the serum bilirubin level. In this study, the serum total bilirubin levels of patients with erectile dysfunction were measured to investigate the relationship between the levels of erectile dysfunction and total bilirubin. METHODS A total of 94 patients with erectile dysfunction were divided into three groups; severe erectile dysfunction (33 patients), moderate erectile dysfunction (31 patients), and mild erectile dysfunction (30 patients). In addition, a control group was formed with 31 healthy men. The International Index of Erectile Function-5 Questionnaire was used to measure the quality of erection in all the groups. The body mass index was calculated for all the participants. The serum glucose, low-density lipoprotein and high-density lipoprotein, cholesterol, triglyceride, total bilirubin, and total testosterone levels were also determined. RESULTS No statistically significant difference was observed between the groups in terms of the mean age, hypertension, smoking status, alcohol use, cardiovascular diseases, hepatobiliary disease, diabetes mellitus, and levels of total testosterone, low-density lipoprotein-cholesterol, and triglyceride. However, high-density lipoprotein, body mass index, and total bilirubin were significantly lower compared to the control group (p < 0.001). The serum total bilirubin level was found to be 0.41 ± 0.21 ng/dL in the severe erectile dysfunction, 0.43 ± 0.19 ng/dL in the moderate erectile dysfunction, and 0.48 ± 0.11 ng/dL in the mild erectile dysfunction groups (p < 0.001). CONCLUSION Considering the significant differences between the erectile dysfunction and control groups in terms of serum total bilirubin levels, a low level of bilirubin may have a role in the etiology of erectile dysfunction.
Collapse
Affiliation(s)
- Ercüment Keskin
- 1 Department of Urology, Erzincan University Mengucek Gazi Research and Training Hospital, Erzincan, Turkey
| | - Mehmet Karabakan
- 2 Department of Urology, Mersin Toros State Hospital, Mersin, Turkey
| | - Aliseydi Bozkurt
- 1 Department of Urology, Erzincan University Mengucek Gazi Research and Training Hospital, Erzincan, Turkey
| | - Erkan Hirik
- 1 Department of Urology, Erzincan University Mengucek Gazi Research and Training Hospital, Erzincan, Turkey
| | - İbrahim Karabulut
- 3 Department of Urology, Regional Research and Training Hospital, Erzurum, Turkey
| | - Murat Gunay
- 4 Department of Chemistry, Erzincan University Mengucek Gazi Research and Training Hospital, Erzincan, Turkey
| | - Murat Çakan
- 1 Department of Urology, Erzincan University Mengucek Gazi Research and Training Hospital, Erzincan, Turkey
| |
Collapse
|
46
|
Bulmer AC, Bakrania B, Du Toit EF, Boon AC, Clark PJ, Powell LW, Wagner KH, Headrick JP. Bilirubin acts as a multipotent guardian of cardiovascular integrity: more than just a radical idea. Am J Physiol Heart Circ Physiol 2018; 315:H429-H447. [PMID: 29600900 DOI: 10.1152/ajpheart.00417.2017] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bilirubin, a potentially toxic catabolite of heme and indicator of hepatobiliary insufficiency, exhibits potent cardiac and vascular protective properties. Individuals with Gilbert's syndrome (GS) may experience hyperbilirubinemia in response to stressors including reduced hepatic bilirubin excretion/increased red blood cell breakdown, with individuals usually informed by their clinician that their condition is of little consequence. However, GS appears to protect from all-cause mortality, with progressively elevated total bilirubin associated with protection from ischemic heart and chronic obstructive pulmonary diseases. Bilirubin may protect against these diseases and associated mortality by reducing circulating cholesterol, oxidative lipid/protein modifications, and blood pressure. In addition, bilirubin inhibits platelet activation and protects the heart from ischemia-reperfusion injury. These effects attenuate multiple stages of the atherosclerotic process in addition to protecting the heart during resultant ischemic stress, likely underpinning the profound reduction in cardiovascular mortality in hyperbilirubinemic GS. This review outlines our current knowledge of and uses for bilirubin in clinical medicine and summarizes recent progress in revealing the physiological importance of this poorly understood molecule. We believe that this review will be of significant interest to clinicians, medical researchers, and individuals who have GS.
Collapse
Affiliation(s)
- Andrew C Bulmer
- School of Medical Science and Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| | - Bhavisha Bakrania
- Department of Physiology and Biophysics, University of Mississippi Medical Centre , Jackson, Mississippi
| | - Eugene F Du Toit
- School of Medical Science and Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| | - Ai-Ching Boon
- School of Medical Science and Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| | - Paul J Clark
- QIMR-Berghofer Medical Research Institute, School of Medicine, University of Queensland and Princess Alexandra and Mater Hospitals , Brisbane, New South Wales , Australia
| | - Lawrie W Powell
- The Centre for the Advancement of Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland Centre for Clinical Research , Brisbane, Queensland , Australia
| | - Karl-Heinz Wagner
- Department of Nutritional Science, University of Vienna , Vienna , Austria
| | - John P Headrick
- School of Medical Science and Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| |
Collapse
|
47
|
Bakrania BA, Spradley FT, Satchell SC, Stec DE, Rimoldi JM, Gadepalli RSV, Granger JP. Heme oxygenase-1 is a potent inhibitor of placental ischemia-mediated endothelin-1 production in cultured human glomerular endothelial cells. Am J Physiol Regul Integr Comp Physiol 2018; 314:R427-R432. [PMID: 29212810 PMCID: PMC5899255 DOI: 10.1152/ajpregu.00370.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 01/07/2023]
Abstract
Preeclampsia is a pregnancy-specific disorder of maternal hypertension and reduced renal hemodynamics linked to reduced endothelial function. Placental ischemia is thought to be the culprit of this disease, as it causes the release of factors like tumor necrosis factor (TNF)-α that induce vascular endothelin-1 (ET-1) production. Interestingly, placental ischemia-induced hypertension in rats [reduced uterine perfusion pressure (RUPP) model] is abolished by ETA receptor blockade, suggesting a critical role for ET-1. Although it has been found that systemic induction of heme oxygenase (HO)-1 is associated with reduced ET-1 production and attenuated hypertension, it is unclear whether HO-1 directly modulates the increased ET-1 response to placental factors. We tested the hypothesis that HO-1 or its metabolites inhibit ET-1 production in human glomerular endothelial cells induced by serum of RUPP rats or TNF-α. Serum (5%) from RUPP hypertensive (mean arterial blood pressure 119 ± 9 mmHg) vs. normotensive pregnant (NP, 101 ± 6 mmHg, P < 0.001) rats increased ET-1 production (RUPP 168.8 ± 18.1 pg/ml, NP 80.3 ± 22.7 pg/ml, P < 0.001, n = 12/group). HO-1 induction [25 µM cobalt photoporphyrin (CoPP)] abolished RUPP serum-induced ET-1 production (1.6 ± 0.8 pg/ml, P < 0.001), whereas bilirubin (10 µM) significantly attenuated ET-1 release (125.3 ± 5.2 pg/ml, P = 0.005). Furthermore, TNF-α-induced ET-1 production (TNF-α 31.0 ± 8.4 vs. untreated 7.5 ± 0.4 pg/ml, P < 0.001) was reduced by CoPP (1.5 ± 0.8 pg/ml, P < 0.001) and bilirubin (10.5 ± 4.3 pg/ml, P < 0.001). These results suggest that circulating factors released during placental ischemia target the maternal glomerular endothelium to increase ET-1, and that pharmacological induction of HO-1 or bilirubin could be a treatment strategy to block this prohypertensive pathway in preeclampsia.
Collapse
Affiliation(s)
- Bhavisha A Bakrania
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Frank T Spradley
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Department of Surgery, University of Mississippi Medical Center , Jackson, Mississippi
| | - Simon C Satchell
- School of Clinical Sciences, University of Bristol , Bristol , United Kingdom
| | - David E Stec
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - John M Rimoldi
- Department of Biomolecular Sciences, University of Mississippi , Oxford, Mississippi
| | - Rama S V Gadepalli
- Department of Biomolecular Sciences, University of Mississippi , Oxford, Mississippi
| | - Joey P Granger
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW This review aims to highlight recent advances on the role of hyperbilirubinemia in hypertension and chronic kidney disease, with a focus on the pathophysiological mechanisms explaining the protective effects of bilirubin. An overview of pharmacologic induction of hyperbilirubinemia will also be discussed. RECENT FINDINGS The findings depict a protective role of bilirubin in the development of hypertension and cardiovascular diseases. Hyperbilirubinemia is also negatively correlated with the development and progression of chronic kidney disease. Commonly used drugs play a role in pharmacologic induction of hyperbilirubinemia. Bilirubin is therefore an exciting target for new therapeutic interventions for its antioxidant properties can be pivotal in the management of hypertension and in preventing and halting the progression of chronic kidney disease. Longitudinal studies are warranted to evaluate the prospective association between bilirubin levels and incident hypertension and chronic kidney disease in the general population. Interventions to induce hyperbilirubinemia need to be explored as a novel therapeutic approach in fighting disease burden.
Collapse
Affiliation(s)
- Ibrahim Mortada
- American University of Beirut Faculty of Medicine, Beirut, Lebanon.
| |
Collapse
|
49
|
Lai X, Fang Q, Yang L, Chen X, Wang H, Ma L, Guo W, Liu M, Yang H, Zhang C, Li X, Min X, Yuan J, He MA, Wu T, Zhang X. Direct, indirect and total bilirubin and risk of incident coronary heart disease in the Dongfeng-Tongji cohort. Ann Med 2018; 50:16-25. [PMID: 28879775 DOI: 10.1080/07853890.2017.1377846] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Total bilirubin (TBIL) is known to be inversely associated with coronary heart disease (CHD) risk, however, whether this association is dose-response remains inconsistent and it is unclear which subtype of bilirubin is responsible for the potential protective effect. METHODS We included 12,097 participants who were free of CHD, stroke, cancer and potential liver, biliary and renal diseases at baseline from September 2008 to June 2010 and were followed-up until October 2013. Cox proportional hazards models were used to assess the hazard ratios (HR) and 95% confidence interval (95% CI) of bilirubin with incident CHD risk. RESULTS The adjusted HRs for incident CHD increased with increasing direct bilirubin (DBIL) (p for trend = .013). Participants within the highest quintile of DBIL had 30% higher risk of incident CHD compared to those in the lowest quintile (95% CI: 1.07, 1.58). In contrast, compared with subjects in the lowest quintile of TBIL, those in the third quintile had the lowest of 24% risk for CHD incidence (95% CI: 0.63, 0.92), which showed a U-shaped association (p for quadratic trend = .040). CONCLUSIONS DBIL was associated with a dose-response increased risk for CHD incidence. However, a U-shaped association existed between TBIL, indirect bilirubin and incident CHD risk. Key messages Direct bilirubin is independently associated with incident coronary heart disease (CHD) in a dose-response manner. A similarly consistent U-shaped association was found between total bilirubin, indirect bilirubin and incident CHD. The potential protective effect of total bilirubin within the normal range on incident CHD should be mainly attributed to mild-to moderate elevated levels of indirect bilirubin.
Collapse
Affiliation(s)
- Xuefeng Lai
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Qin Fang
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Liangle Yang
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xuguang Chen
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Hao Wang
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Lin Ma
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Wenting Guo
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Miao Liu
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Handong Yang
- b Department of Cardiology and Epidemiology, Dongfeng Central Hospital, Dongfeng Motor Corporation and Hubei University of Medicine , Shiyan , China
| | - Ce Zhang
- b Department of Cardiology and Epidemiology, Dongfeng Central Hospital, Dongfeng Motor Corporation and Hubei University of Medicine , Shiyan , China
| | - Xiulou Li
- b Department of Cardiology and Epidemiology, Dongfeng Central Hospital, Dongfeng Motor Corporation and Hubei University of Medicine , Shiyan , China
| | - Xinwen Min
- b Department of Cardiology and Epidemiology, Dongfeng Central Hospital, Dongfeng Motor Corporation and Hubei University of Medicine , Shiyan , China
| | - Jing Yuan
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Mei-An He
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Tangchun Wu
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xiaomin Zhang
- a Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
50
|
Whiten SR, Eggleston H, Adelman ZN. Ironing out the Details: Exploring the Role of Iron and Heme in Blood-Sucking Arthropods. Front Physiol 2018; 8:1134. [PMID: 29387018 PMCID: PMC5776124 DOI: 10.3389/fphys.2017.01134] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/22/2017] [Indexed: 12/12/2022] Open
Abstract
Heme and iron are essential molecules for many physiological processes and yet have the ability to cause oxidative damage such as lipid peroxidation, protein degradation, and ultimately cell death if not controlled. Blood-sucking arthropods have evolved diverse methods to protect themselves against iron/heme-related damage, as the act of bloodfeeding itself is high risk, high reward process. Protective mechanisms in medically important arthropods include the midgut peritrophic matrix in mosquitoes, heme aggregation into the crystalline structure hemozoin in kissing bugs and hemosomes in ticks. Once heme and iron pass these protective mechanisms they are presumed to enter the midgut epithelial cells via membrane-bound transporters, though relatively few iron or heme transporters have been identified in bloodsucking arthropods. Upon iron entry into midgut epithelial cells, ferritin serves as the universal storage protein and transport for dietary iron in many organisms including arthropods. In addition to its role as a nutrient, heme is also an important signaling molecule in the midgut epithelial cells for many physiological processes including vitellogenesis. This review article will summarize recent advancements in heme/iron uptake, detoxification and exportation in bloodfeeding arthropods. While initial strides have been made at ironing out the role of dietary iron and heme in arthropods, much still remains to be discovered as these molecules may serve as novel targets for the control of many arthropod pests.
Collapse
Affiliation(s)
- Shavonn R Whiten
- Department of Entomology, Texas A&M University, College Station, TX, United States
| | - Heather Eggleston
- Genetics Graduate Program, Texas A&M University, College Station, TX, United States
| | - Zach N Adelman
- Department of Entomology, Texas A&M University, College Station, TX, United States
| |
Collapse
|