1
|
Dikalova A, Fehrenbach D, Mayorov V, Panov A, Ao M, Lantier L, Amarnath V, Lopez MG, Billings FT, Sack MN, Dikalov S. Mitochondrial CypD Acetylation Promotes Endothelial Dysfunction and Hypertension. Circ Res 2024; 134:1451-1464. [PMID: 38639088 PMCID: PMC11116043 DOI: 10.1161/circresaha.123.323596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/01/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Nearly half of adults have hypertension, a major risk factor for cardiovascular disease. Mitochondrial hyperacetylation is linked to hypertension, but the role of acetylation of specific proteins is not clear. We hypothesized that acetylation of mitochondrial CypD (cyclophilin D) at K166 contributes to endothelial dysfunction and hypertension. METHODS To test this hypothesis, we studied CypD acetylation in patients with essential hypertension, defined a pathogenic role of CypD acetylation in deacetylation mimetic CypD-K166R mutant mice and endothelial-specific GCN5L1 (general control of amino acid synthesis 5 like 1)-deficient mice using an Ang II (angiotensin II) model of hypertension. RESULTS Arterioles from hypertensive patients had 280% higher CypD acetylation coupled with reduced Sirt3 (sirtuin 3) and increased GCN5L1 levels. GCN5L1 regulates mitochondrial protein acetylation and promotes CypD acetylation, which is counteracted by mitochondrial deacetylase Sirt3. In human aortic endothelial cells, GCN5L1 depletion prevents superoxide overproduction. Deacetylation mimetic CypD-K166R mice were protected from vascular oxidative stress, endothelial dysfunction, and Ang II-induced hypertension. Ang II-induced hypertension increased mitochondrial GCN5L1 and reduced Sirt3 levels resulting in a 250% increase in GCN5L1/Sirt3 ratio promoting CypD acetylation. Treatment with mitochondria-targeted scavenger of cytotoxic isolevuglandins (mito2HOBA) normalized GCN5L1/Sirt3 ratio, reduced CypD acetylation, and attenuated hypertension. The role of mitochondrial acetyltransferase GCN5L1 in the endothelial function was tested in endothelial-specific GCN5L1 knockout mice. Depletion of endothelial GCN5L1 prevented Ang II-induced mitochondrial oxidative stress, reduced the maladaptive switch of vascular metabolism to glycolysis, prevented inactivation of endothelial nitric oxide, preserved endothelial-dependent relaxation, and attenuated hypertension. CONCLUSIONS These data support the pathogenic role of CypD acetylation in endothelial dysfunction and hypertension. We suggest that targeting cytotoxic mitochondrial isolevuglandins and GCN5L1 reduces CypD acetylation, which may be beneficial in cardiovascular disease.
Collapse
Affiliation(s)
- Anna Dikalova
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | - Mingfang Ao
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | | | | | - Sergey Dikalov
- Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
2
|
Caspa Gokulan R, Paulrasu K, Azfar J, El-Rifai W, Que J, Boutaud OG, Ban Y, Gao Z, Buitrago MG, Dikalov SI, Zaika AI. Protein adduction causes non-mutational inhibition of p53 tumor suppressor. Cell Rep 2023; 42:112024. [PMID: 36848235 PMCID: PMC9989503 DOI: 10.1016/j.celrep.2023.112024] [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: 04/05/2022] [Revised: 06/04/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
p53 is a key tumor suppressor that is frequently mutated in human tumors. In this study, we investigated how p53 is regulated in precancerous lesions prior to mutations in the p53 gene. Analyzing esophageal cells in conditions of genotoxic stress that promotes development of esophageal adenocarcinoma, we find that p53 protein is adducted with reactive isolevuglandins (isoLGs), products of lipid peroxidation. Modification of p53 protein with isoLGs diminishes its acetylation and binding to the promoters of p53 target genes causing modulation of p53-dependent transcription. It also leads to accumulation of adducted p53 protein in intracellular amyloid-like aggregates that can be inhibited by isoLG scavenger 2-HOBA in vitro and in vivo. Taken together, our studies reveal a posttranslational modification of p53 protein that causes molecular aggregation of p53 protein and its non-mutational inactivation in conditions of DNA damage that may play an important role in human tumorigenesis.
Collapse
Affiliation(s)
| | | | - Jamal Azfar
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Wael El-Rifai
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Olivier G Boutaud
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yuguang Ban
- Department of Public Health Sciences, University of Miami, Miami, FL, USA
| | - Zhen Gao
- Department of Public Health Sciences, University of Miami, Miami, FL, USA
| | | | - Sergey I Dikalov
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander I Zaika
- Department of Surgery, University of Miami, Miami, FL, USA; Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA.
| |
Collapse
|
3
|
Aiello G, Rescigno F, Meloni M, Zoanni B, Aldini G, Carini M, D’Amato A. The Effect of Carnosine on UVA-Induced Changes in Intracellular Signaling of Human Skin Fibroblast Spheroids. Antioxidants (Basel) 2023; 12:antiox12020300. [PMID: 36829859 PMCID: PMC9951876 DOI: 10.3390/antiox12020300] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
Dermis fibroblasts are very sensitive to penetrating UVA radiation and induce photo-damage. To protect skin cells against this environmental damage, there is an urgent need for effective compounds, specifically targeting UVA-induced mitochondrial injury. This study aimed to analyze the effect of carnosine on the proteome of UVA-irradiated human skin fibroblast, cultured in a three-dimensional (3D) biological system recapitulating dermal compartment as a test system to investigate the altered cellular pathways after 48 h and 7 days of culture with or without carnosine treatment. The obtained results indicate that UVA dysregulates Oxidative Phosphorylation, the Fibrosis Signaling Pathway, Glycolysis I and Nrf2-mediated Oxidative Stress Response. Carnosine exercises provide a protective function against the harmful effects of UVA radiation by activating the Nrf2 pathway with the upregulations of some ROS-detoxifying enzymes such as the glutathione S-transferase (GST) protein family. Additionally, carnosine regulates the activation of the Epithelial Adherens Junction and Wound Healing Signaling Pathway by mediating the activation of structural proteins such as vinculin and zyxin as well as fibronectin 1 and collagen type XVIII alpha 1 chain against UVA-induced changes.
Collapse
Affiliation(s)
- Gilda Aiello
- Department of Human Science and Quality of Life Promotion, Telematic University San Raffaele, 00166 Rome, Italy
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milan, Italy
| | | | - Marisa Meloni
- VitroScreen, In Vitro Innovation Center, 20149 Milan, Italy
| | - Beatrice Zoanni
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Marina Carini
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Alfonsina D’Amato
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milan, Italy
- Correspondence:
| |
Collapse
|
4
|
Lee SH, Tsutsui M, Matsunaga A, Oe T. Lipid hydroperoxide-derived insulin resistance and its inhibition by pyridoxamine in skeletal muscle cells. Toxicol Res 2023; 39:147-156. [PMID: 36726824 PMCID: PMC9839902 DOI: 10.1007/s43188-022-00155-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/28/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress is strongly associated with the onset and/or progression of diabetes. Under conditions of oxidative stress, lipid hydroperoxides are decomposed to reactive aldehydes that have been reported to induce insulin resistance by modifying proteins involved in insulin signaling. Pyridoxamine (PM) can inhibit the formation of advanced glycation/lipoxidation end products by scavenging reactive carbonyl species. Thus, PM has emerged as a promising drug candidate for various chronic conditions, including diabetic complications. In this study, L6 skeletal muscle cells were treated with 4-oxo-2(E)-nonenal (ONE), one of the most abundant and reactive lipid-derived aldehydes. Cellular insulin resistance was assessed by measuring insulin-stimulated glucose uptake using 2-deoxyglucose. ONE induced a time- and dose-dependent decrease in glucose uptake. Liquid chromatography/electrospray ionization-mass spectrometry analysis of the reaction between ONE and insulin receptor substrate 1 (IRS1) lysate identified multiple modifications that could disturb the interaction between IRS1 and activated IR, leading to insulin resistance. Pretreatment of the cells with PM restored the ONE-induced decrease in glucose uptake. Concomitantly, the formation of PM-ONE adducts in cell culture medium was increased in a PM-dose dependent manner. PM can therefore prevent lipid hydroperoxide-derived insulin resistance by quenching ONE. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-022-00155-z.
Collapse
Affiliation(s)
- Seon Hwa Lee
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578 Japan
| | - Mizuki Tsutsui
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578 Japan
| | - Atsushi Matsunaga
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578 Japan
| | - Tomoyuki Oe
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578 Japan
| |
Collapse
|
5
|
Huang J, Tao H, Yancey PG, Leuthner Z, May-Zhang LS, Jung JY, Zhang Y, Ding L, Amarnath V, Liu D, Collins S, Davies SS, Linton MF. Scavenging dicarbonyls with 5'-O-pentyl-pyridoxamine increases HDL net cholesterol efflux capacity and attenuates atherosclerosis and insulin resistance. Mol Metab 2022; 67:101651. [PMID: 36481344 PMCID: PMC9792904 DOI: 10.1016/j.molmet.2022.101651] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Oxidative stress contributes to the development of insulin resistance (IR) and atherosclerosis. Peroxidation of lipids produces reactive dicarbonyls such as Isolevuglandins (IsoLG) and malondialdehyde (MDA) that covalently bind plasma/cellular proteins, phospholipids, and DNA leading to altered function and toxicity. We examined whether scavenging reactive dicarbonyls with 5'-O-pentyl-pyridoxamine (PPM) protects against the development of IR and atherosclerosis in Ldlr-/- mice. METHODS Male or female Ldlr-/- mice were fed a western diet (WD) for 16 weeks and treated with PPM versus vehicle alone. Plaque extent, dicarbonyl-lysyl adducts, efferocytosis, apoptosis, macrophage inflammation, and necrotic area were measured. Plasma MDA-LDL adducts and the in vivo and in vitro effects of PPM on the ability of HDL to reduce macrophage cholesterol were measured. Blood Ly6Chi monocytes and ex vivo 5-ethynyl-2'-deoxyuridine (EdU) incorporation into bone marrow CD11b+ monocytes and CD34+ hematopoietic stem and progenitor cells (HSPC) were also examined. IR was examined by measuring fasting glucose/insulin levels and tolerance to insulin/glucose challenge. RESULTS PPM reduced the proximal aortic atherosclerosis by 48% and by 46% in female and male Ldlr-/- mice, respectively. PPM also decreased IR and hepatic fat and inflammation in male Ldlr-/- mice. Importantly, PPM decreased plasma MDA-LDL adducts and prevented the accumulation of plaque MDA- and IsoLG-lysyl adducts in Ldlr-/- mice. In addition, PPM increased the net cholesterol efflux capacity of HDL from Ldlr-/- mice and prevented both the in vitro impairment of HDL net cholesterol efflux capacity and apoAI crosslinking by MPO generated hypochlorous acid. Moreover, PPM decreased features of plaque instability including decreased proinflammatory M1-like macrophages, IL-1β expression, myeloperoxidase, apoptosis, and necrotic core. In contrast, PPM increased M2-like macrophages, Tregs, fibrous cap thickness, and efferocytosis. Furthermore, PPM reduced inflammatory monocytosis as evidenced by decreased blood Ly6Chi monocytes and proliferation of bone marrow monocytes and HSPC from Ldlr-/- mice. CONCLUSIONS PPM has pleotropic atheroprotective effects in a murine model of familial hypercholesterolemia, supporting the therapeutic potential of reactive dicarbonyl scavenging in the treatment of IR and atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Jiansheng Huang
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Huan Tao
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Patricia G. Yancey
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Zoe Leuthner
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Linda S. May-Zhang
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Ju-Yang Jung
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Youmin Zhang
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Lei Ding
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Venkataraman Amarnath
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Dianxin Liu
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sheila Collins
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Sean S. Davies
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - MacRae F. Linton
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States,Department of Pharmacology, Vanderbilt University, Nashville, TN, United States,Corresponding author. Department of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN, United States.
| |
Collapse
|
6
|
Fadaei R, Davies SS. Oxidative modification of HDL by lipid aldehydes impacts HDL function. Arch Biochem Biophys 2022; 730:109397. [PMID: 36116503 PMCID: PMC9670862 DOI: 10.1016/j.abb.2022.109397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/21/2022]
Abstract
Reduced levels of high-density lipoprotein (HDL) cholesterol correlate with increased risk for atherosclerotic cardiovascular diseases and HDL performs functions including reverse cholesterol transport, inhibition of lipid peroxidation, and suppression of inflammation, that would appear critical for cardioprotection. However, several large clinical trials utilizing pharmacologic interventions that elevated HDL cholesterol levels failed to provide cardioprotection to at-risk individuals. The reasons for these unexpected results have only recently begun to be elucidated. HDL cholesterol levels and HDL function can be significantly discordant, so that elevating HDL cholesterol levels may not necessarily lead to increased functional capacity, particularly under conditions that cause HDL to become oxidatively modified, resulting in HDL dysfunction. Here we review evidence that oxidative modifications of HDL, including by reactive lipid aldehydes generated by lipid peroxidation, reduce HDL functionality and that dicarbonyl scavengers that protect HDL against lipid aldehyde modification are beneficial in pre-clinical models of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sean S Davies
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
7
|
Guo J, Xu F, Ji H, Jing Y, Shen L, Weng X, Hu L. Isolevuglandins Scavenger Ameliorates Myocardial Ischemic Injury by Suppressing Oxidative Stress, Apoptosis, and Inflammation. Front Cell Dev Biol 2022; 10:836035. [PMID: 35356291 PMCID: PMC8959416 DOI: 10.3389/fcell.2022.836035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Augmented levels of reactive isolevuglandins (IsoLGs) are responsible for cardiovascular diseases. The role of IsoLGs in myocardial infarction (MI) remains elusive. Here we explored the effect of IsoLGs scavenger 2-hydroxybenzylamine (2-HOBA) in post-infarction cardiac repair. We observed that infarcted cardiac tissues expressed high IsoLGs in mice. Following MI injury, 2-HOBA treated mice displayed decreased infarction area and improved heart function compared with the saline-treated group. Moreover, 2-HOBA effectively attenuated MI-induced cardiac remodeling, oxidative stress, apoptosis, and inflammation. 4-hydroxybenzylamine (4-HOBA), a less reactive isomer of 2-HOBA, barely antagonized the MI-induced injury. These findings suggest that IsoLGs elimination may be helpful in MI therapy.
Collapse
Affiliation(s)
- Junjie Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, China
- *Correspondence: Junjie Guo, ; Xinyu Weng, ; Longgang Hu,
| | - Fengqiang Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, China
| | - Hongwei Ji
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, China
| | - Yajun Jing
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- *Correspondence: Junjie Guo, ; Xinyu Weng, ; Longgang Hu,
| | - Longgang Hu
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
- *Correspondence: Junjie Guo, ; Xinyu Weng, ; Longgang Hu,
| |
Collapse
|
8
|
Myeloperoxidase-induced modification of HDL by isolevuglandins inhibits paraoxonase-1 activity. J Biol Chem 2021; 297:101019. [PMID: 34331945 PMCID: PMC8390528 DOI: 10.1016/j.jbc.2021.101019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 07/09/2021] [Accepted: 07/27/2021] [Indexed: 11/21/2022] Open
Abstract
Reduced activity of paraoxonase 1 (PON1), a high-density lipoprotein (HDL)-associated enzyme, has been implicated in the development of atherosclerosis. Post-translational modifications of PON1 may represent important mechanisms leading to reduced PON1 activity. Under atherosclerotic conditions, myeloperoxidase (MPO) is known to associate with HDL. MPO generates the oxidants hypochlorous acid and nitrogen dioxide, which can lead to post-translational modification of PON1, including tyrosine modifications that inhibit PON1 activity. Nitrogen dioxide also drives lipid peroxidation, leading to the formation of reactive lipid dicarbonyls such as malondialdehyde and isolevuglandins, which modify HDL and could inhibit PON1 activity. Because isolevuglandins are more reactive than malondialdehyde, we used in vitro models containing HDL, PON1, and MPO to test the hypothesis that IsoLG formation by MPO and its subsequent modification of HDL contributes to MPO-mediated reductions in PON1 activity. Incubation of MPO with HDL led to modification of HDL proteins, including PON1, by IsoLG. Incubation of HDL with IsoLG reduced PON1 lactonase and antiperoxidation activities. IsoLG modification of recombinant PON1 markedly inhibited its activity, while irreversible IsoLG modification of HDL before adding recombinant PON1 only slightly inhibited the ability of HDL to enhance the catalytic activity of recombinant PON1. Together, these studies support the notion that association of MPO with HDL leads to lower PON1 activity in part via IsoLG-mediated modification of PON1, so that IsoLG modification of PON1 could contribute to increased risk for atherosclerosis, and blocking this modification might prove beneficial to reduce atherosclerosis.
Collapse
|
9
|
Aschner M, Nguyen TT, Sinitskii AI, Santamaría A, Bornhorst J, Ajsuvakova OP, da Rocha JBT, Skalny AV, Tinkov AA. Isolevuglandins (isoLGs) as toxic lipid peroxidation byproducts and their pathogenetic role in human diseases. Free Radic Biol Med 2021; 162:266-273. [PMID: 33099003 DOI: 10.1016/j.freeradbiomed.2020.10.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 12/14/2022]
Abstract
Lipid peroxidation results in generation of a variety of lipid hydroperoxides and other highly reactive species that covalently modify proteins, nucleic acids, and other lipids, thus resulting in lipotoxicity. Although biological relevance of 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA) is well studied, the existing data on the role of isolevuglandins (isoLGs) in pathology are insufficient. Therefore, the objective of the present study was to review the existing data on biological effects of isoLG and isoLG adducts and their role in multiple diseases. Sixty four highly reactive levuglandin-like γ-ketoaldehyde (γ-KA, or isoketals, IsoK, or isolevuglandins, IsoLG) regio- and stereo-isomers are formed as products of arachidonic acid oxidation. IsoLGs react covalently with lysyl residues of proteins to form a stable adduct and intramolecular aminal, bispyrrole, and trispyrrole cross-links. Phosphatidylethanolamine was also shown to be the target for isoLG binding as compared to proteins and DNA. Free IsoLGs are not detectable in vivo, although isolevuglandin adduction to amino acid residues of particular proteins may be evaluated with liquid chromatography-tandem mass spectrometry. Adducts formed were shown to play a significant role in the development and maintenance of oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction, and inflammation. These, and more specific molecular pathways, link isoLG and isoLG-adduct formation to develop a variety of pathologies, including cardiovascular diseases (atherosclerosis, hypertension, heart failure), obesity and diabetes, cancer, neurodegeneration, eye diseases (retinal degeneration and glaucoma), as well as ageing. Hypothetically, isoLGs and isoLG adduct formation may be considered as the potential target for treatment of oxidative stress-related diseases.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; IM Sechenov First Moscow State Medical University, Moscow, Russia.
| | - Thuy T Nguyen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Julia Bornhorst
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Olga P Ajsuvakova
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
| | | | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia
| |
Collapse
|
10
|
Cheng YS, Linetsky M, Li H, Ayyash N, Gardella A, Salomon RG. 4-Hydroxy-7-oxo-5-heptenoic acid lactone can induce mitochondrial dysfunction in retinal pigmented epithelial cells. Free Radic Biol Med 2020; 160:719-733. [PMID: 32920040 PMCID: PMC7704664 DOI: 10.1016/j.freeradbiomed.2020.09.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/25/2020] [Accepted: 09/04/2020] [Indexed: 11/30/2022]
Abstract
Oxidation of docosahexaenoate (DHA)-containing phospholipids in the cell plasma membrane leads to release of the α,β-unsaturated aldehyde 4-hydroxy-7-oxo-5-heptenoic acid (HOHA) lactone which is capable of inducing retinal pigmented epithelial (RPE) cell dysfunction. Previously, HOHA lactone was shown to induce apoptosis and angiogenesis, and to activate the alternative complement pathway. RPE cells metabolize HOHA lactone through enzymatic conjugation with glutathione (GSH). Competing with this process is the adduction of HOHA lactone to protein lysyl residues generating 2-(ω-carboxyethyl)pyrrole (CEP) derivatives that have pathological relevance to age-related macular degeneration (AMD). We now find that HOHA lactone induces mitochondrial dysfunction. It decreases ATP levels, mitochondrial membrane potentials, enzymatic activities of mitochondrial complexes, depletes GSH and induces oxidative stress in RPE cells. The present study confirmed that pyridoxamine and other primary amines, which have been shown to scavenge γ-ketoaldehydes formed by carbohydrate or lipid peroxidation, are ineffective for scavenging the α,β-unsaturated aldehydes. Histidyl hydrazide (HH), that has both hydrazide and imidazole nucleophile functionalities, is an effective scavenger of HOHA lactone and it protects ARPE-19 cells against HOHA lactone-induced cytotoxicity. The HH α-amino group is not essential for this electrophile trapping activity. The Nα-acyl L-histidyl hydrazide derivatives with 2- to 7-carbon acyl groups with increasing lipophilicities are capable of maintaining the effectiveness of HH in protecting ARPE-19 cells against HOHA lactone toxicity, which potentially has therapeutic utility for treatment of age related eye diseases.
Collapse
Affiliation(s)
- Yu-Shiuan Cheng
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Mikhail Linetsky
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA; Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Haoting Li
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Naji Ayyash
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Anthony Gardella
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Robert G Salomon
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA; Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
11
|
Dikalova A, Mayorov V, Xiao L, Panov A, Amarnath V, Zagol-Ikapitte I, Vergeade A, Ao M, Yermalitsky V, Nazarewicz RR, Boutaud O, Lopez MG, Billings FT, Davies S, Roberts LJ, Harrison DG, Dikalov S. Mitochondrial Isolevuglandins Contribute to Vascular Oxidative Stress and Mitochondria-Targeted Scavenger of Isolevuglandins Reduces Mitochondrial Dysfunction and Hypertension. Hypertension 2020; 76:1980-1991. [PMID: 33012204 DOI: 10.1161/hypertensionaha.120.15236] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hypertension remains a major health problem in Western Societies, and blood pressure is poorly controlled in a third of patients despite use of multiple drugs. Mitochondrial dysfunction contributes to hypertension, and mitochondria-targeted agents can potentially improve treatment of hypertension. We have proposed that mitochondrial oxidative stress produces reactive dicarbonyl lipid peroxidation products, isolevuglandins, and that scavenging of mitochondrial isolevuglandins improves vascular function and reduces hypertension. To test this hypothesis, we have studied the accumulation of mitochondrial isolevuglandins-protein adducts in patients with essential hypertension and Ang II (angiotensin II) model of hypertension using mass spectrometry and Western blot analysis. The therapeutic potential of targeting mitochondrial isolevuglandins was tested by the novel mitochondria-targeted isolevuglandin scavenger, mito2HOBA. Mitochondrial isolevuglandins in arterioles from hypertensive patients were 250% greater than in arterioles from normotensive subjects, and ex vivo mito2HOBA treatment of arterioles from hypertensive subjects increased deacetylation of a key mitochondrial antioxidant, SOD2 (superoxide dismutase 2). In human aortic endothelial cells stimulated with Ang II plus TNF (tumor necrosis factor)-α, mito2HOBA reduced mitochondrial superoxide and cardiolipin oxidation, a specific marker of mitochondrial oxidative stress. In Ang II-infused mice, mito2HOBA diminished mitochondrial isolevuglandins-protein adducts, raised Sirt3 (sirtuin 3) mitochondrial deacetylase activity, reduced vascular superoxide, increased endothelial nitric oxide, improved endothelium-dependent relaxation, and attenuated hypertension. Mito2HOBA preserved mitochondrial respiration, protected ATP production, and reduced mitochondrial permeability pore opening in Ang II-infused mice. These data support the role of mitochondrial isolevuglandins in endothelial dysfunction and hypertension. We conclude that scavenging of mitochondrial isolevuglandins may have therapeutic potential in treatment of vascular dysfunction and hypertension.
Collapse
Affiliation(s)
- Anna Dikalova
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | | | - Liang Xiao
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Alexander Panov
- Scientific Centre for Family Health and Human Reproduction Problems, Irkutsk, Russian Federation (A.P.)
| | - Venkataraman Amarnath
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Irene Zagol-Ikapitte
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Aurelia Vergeade
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Mingfang Ao
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Valery Yermalitsky
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Rafal R Nazarewicz
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Olivier Boutaud
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Marcos G Lopez
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Frederic T Billings
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Sean Davies
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - L Jackson Roberts
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - David G Harrison
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| | - Sergey Dikalov
- From the Vanderbilt University Medical Center, Nashville, TN (A.D., L.X., V.A., I.Z.-I., A.V., M.A., V.Y., R.R.N., O.B., M.G.L., F.T.B., S. Davies, L.J.R., D.G.H., S. Dikalov)
| |
Collapse
|
12
|
May-Zhang LS, Kirabo A, Huang J, Linton MF, Davies SS, Murray KT. Scavenging Reactive Lipids to Prevent Oxidative Injury. Annu Rev Pharmacol Toxicol 2020; 61:291-308. [PMID: 32997599 DOI: 10.1146/annurev-pharmtox-031620-035348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Oxidative injury due to elevated levels of reactive oxygen species is implicated in cardiovascular diseases, Alzheimer's disease, lung and liver diseases, and many cancers. Antioxidant therapies have generally been ineffective at treating these diseases, potentially due to ineffective doses but also due to interference with critical host defense and signaling processes. Therefore, alternative strategies to prevent oxidative injury are needed. Elevated levels of reactive oxygen species induce lipid peroxidation, generating reactive lipid dicarbonyls. These lipid oxidation products may be the most salient mediators of oxidative injury, as they cause cellular and organ dysfunction by adducting to proteins, lipids, and DNA. Small-molecule compounds have been developed in the past decade to selectively and effectively scavenge these reactive lipid dicarbonyls. This review outlines evidence supporting the role of lipid dicarbonyls in disease pathogenesis, as well as preclinical data supporting the efficacy of novel dicarbonyl scavengers in treating or preventing disease.
Collapse
Affiliation(s)
- Linda S May-Zhang
- Division of Clinical Pharmacology, Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6602, USA;
| | - Annet Kirabo
- Division of Clinical Pharmacology, Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6602, USA;
| | - Jiansheng Huang
- Division of Clinical Pharmacology, Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6602, USA;
| | - MacRae F Linton
- Division of Clinical Pharmacology, Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6602, USA;
| | - Sean S Davies
- Division of Clinical Pharmacology, Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6602, USA;
| | - Katherine T Murray
- Division of Clinical Pharmacology, Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6602, USA;
| |
Collapse
|
13
|
Scavenging of reactive dicarbonyls with 2-hydroxybenzylamine reduces atherosclerosis in hypercholesterolemic Ldlr -/- mice. Nat Commun 2020; 11:4084. [PMID: 32796843 PMCID: PMC7429830 DOI: 10.1038/s41467-020-17915-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 07/27/2020] [Indexed: 12/21/2022] Open
Abstract
Lipid peroxidation generates reactive dicarbonyls including isolevuglandins (IsoLGs) and malondialdehyde (MDA) that covalently modify proteins. Humans with familial hypercholesterolemia (FH) have increased lipoprotein dicarbonyl adducts and dysfunctional HDL. We investigate the impact of the dicarbonyl scavenger, 2-hydroxybenzylamine (2-HOBA) on HDL function and atherosclerosis in Ldlr−/− mice, a model of FH. Compared to hypercholesterolemic Ldlr−/− mice treated with vehicle or 4-HOBA, a nonreactive analogue, 2-HOBA decreases atherosclerosis by 60% in en face aortas, without changing plasma cholesterol. Ldlr−/− mice treated with 2-HOBA have reduced MDA-LDL and MDA-HDL levels, and their HDL display increased capacity to reduce macrophage cholesterol. Importantly, 2-HOBA reduces the MDA- and IsoLG-lysyl content in atherosclerotic aortas versus 4-HOBA. Furthermore, 2-HOBA reduces inflammation and plaque apoptotic cells and promotes efferocytosis and features of stable plaques. Dicarbonyl scavenging with 2-HOBA has multiple atheroprotective effects in a murine FH model, supporting its potential as a therapeutic approach for atherosclerotic cardiovascular disease. Hypercholesterolemia is associated with lipid peroxidation induced reactive dicarbonyl adducts. Here the authors show that the dicarbonyl scavenger, 2-hydroxybenzylamine(2-HOBA), decreases reactive dicarbonyl modifications of LDL and HDL, improves HDL function, reduces atherosclerosis and promotes features of stable plaques in a mouse model of hypercholestrolemia.
Collapse
|
14
|
Prinsen JK, Kannankeril PJ, Sidorova TN, Yermalitskaya LV, Boutaud O, Zagol-Ikapitte I, Barnett JV, Murphy MB, Subati T, Stark JM, Christopher IL, Jafarian-Kerman SR, Saleh MA, Norlander AE, Loperena R, Atkinson JB, Fogo AB, Luther JM, Amarnath V, Davies SS, Kirabo A, Madhur MS, Harrison DG, Murray KT. Highly Reactive Isolevuglandins Promote Atrial Fibrillation Caused by Hypertension. JACC Basic Transl Sci 2020; 5:602-615. [PMID: 32613146 PMCID: PMC7315188 DOI: 10.1016/j.jacbts.2020.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 01/11/2023]
Abstract
Oxidative damage is implicated in atrial fibrillation (AF), but antioxidants are ineffective therapeutically. The authors tested the hypothesis that highly reactive lipid dicarbonyl metabolites, or isolevuglandins (IsoLGs), are principal drivers of AF during hypertension. In a hypertensive murine model and stretched atriomyocytes, the dicarbonyl scavenger 2-hydroxybenzylamine (2-HOBA) prevented IsoLG adducts and preamyloid oligomers (PAOs), and AF susceptibility, whereas the ineffective analog 4-hydroxybenzylamine (4-HOBA) had minimal effect. Natriuretic peptides generated cytotoxic oligomers, a process accelerated by IsoLGs, contributing to atrial PAO formation. These findings support the concept of pre-emptively scavenging reactive downstream oxidative stress mediators as a potential therapeutic approach to prevent AF.
Collapse
Key Words
- 2-HOBA, 2-hydroxylbenzylamine
- 4-HOBA, 4-hydroxylbenzylamine
- AF, atrial fibrillation
- ANP, atrial natriuretic peptide
- B-type natriuretic peptide
- BNP, B-type natriuretic peptide
- BP, blood pressure
- ECG, electrocardiogram
- G/R, green/red ratio
- IsoLG, isolevuglandin
- PAO, preamyloid oligomer
- PBS, phosphate-buffered saline
- ROS, reactive oxygen species
- ang II, angiotensin II
- atrial fibrillation
- atrial natriuretic peptide
- hypertension
- isolevuglandins
- oxidative stress
- preamyloid oligomers
Collapse
Affiliation(s)
- Joseph K. Prinsen
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Prince J. Kannankeril
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Tatiana N. Sidorova
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Liudmila V. Yermalitskaya
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Olivier Boutaud
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Irene Zagol-Ikapitte
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Joey V. Barnett
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Matthew B. Murphy
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Tuerdi Subati
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Joshua M. Stark
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Isis L. Christopher
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Scott R. Jafarian-Kerman
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mohamed A. Saleh
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Allison E. Norlander
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Roxana Loperena
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James B. Atkinson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Agnes B. Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James M. Luther
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Venkataraman Amarnath
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Sean S. Davies
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Meena S. Madhur
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - David G. Harrison
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Katherine T. Murray
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
15
|
Arriagada-Petersen C, Fernandez P, Gomez M, Ravello N, Palomo I, Fuentes E, Ávila F. Effect of advanced glycation end products on platelet activation and aggregation: a comparative study of the role of glyoxal and methylglyoxal. Platelets 2020; 32:507-515. [PMID: 32449466 DOI: 10.1080/09537104.2020.1767770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Advanced glycation end products (AGEs) arising from dietary intake have been associated with numerous chronic diseases including cardiovascular diseases. The interaction between platelets and AGEs has been proposed to play a role in the etiology of cardiovascular diseases. However, the effects of the interaction between platelets and Maillard reaction products generated from glyoxal (Gly) or methylglyoxal (MG) are poorly understood. In this work, the effects of AGEs generated by the reaction between Gly or MG with Lys or bovine serum albumin (BSA) on platelet activation and aggregation were assessed. AGEs were generated incubating Gly or MG with Lys or BSA during 5 hours or 14 days, respectively. AGEs generation were characterized by kinetic studies and by amino acid analysis. Human platelet-rich plasma (PRP) was incubated with different concentrations of AGEs from Lys-MG or Lys-Gly and BSA-MG or BSA-Gly. Platelet activation was determined quantifying the expression of CD62 (P-selectin) in PRP exposed to different AGEs concentrations. It was found that Lys-MG and Lys-Gly induced an increase in P-selectin expression (p < .05), being 33.9% higher for Lys-MG when compared to Lys-Gly. Platelets incubated in the presence of BSA-MG and BSA-Gly did not show an increase in the P-selectin expression. Platelet aggregation was significantly higher for the mixture Lys-MG (in all the range of concentrations evaluated), whereas for Lys-Gly it was only significant the highest concentration (Lys 168 µM/Gly 168 µM). It was observed a significant increase in platelet aggregation induced by ADP for samples BSA-Gly. AGEs formed with MG-Lys induce a higher activation and aggregation of platelets when compared to those formed from Gly-Lys.
Collapse
Affiliation(s)
| | - Paula Fernandez
- Escuela De Nutrición Y Dietética, Facultad De Ciencias De La Salud, Universidad De Talca, Talca, Chile
| | - Maira Gomez
- Escuela De Nutrición Y Dietética, Facultad De Ciencias De La Salud, Universidad De Talca, Talca, Chile
| | - Natalia Ravello
- Escuela De Nutrición Y Dietética, Facultad De Ciencias De La Salud, Universidad De Talca, Talca, Chile
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad De Talca, Talca, Chile
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad De Talca, Talca, Chile
| | - Felipe Ávila
- Escuela De Nutrición Y Dietética, Facultad De Ciencias De La Salud, Universidad De Talca, Talca, Chile
| |
Collapse
|
16
|
Davies SS, May-Zhang LS, Boutaud O, Amarnath V, Kirabo A, Harrison DG. Isolevuglandins as mediators of disease and the development of dicarbonyl scavengers as pharmaceutical interventions. Pharmacol Ther 2019; 205:107418. [PMID: 31629006 DOI: 10.1016/j.pharmthera.2019.107418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
Products of lipid peroxidation include a number of reactive lipid aldehydes such as malondialdehyde, 4-hydroxy-nonenal, 4-oxo-nonenal, and isolevuglandins (IsoLGs). Although these all contribute to disease processes, the most reactive are the IsoLGs, which rapidly adduct to lysine and other cellular primary amines, leading to changes in protein function, cross-linking and immunogenicity. Their rapid reactivity means that only IsoLG adducts, and not the unreacted aldehyde, can be readily measured. This high reactivity also makes it challenging for standard cellular defense mechanisms such as aldehyde reductases and oxidases to dispose of them before they react with proteins and other cellular amines. This led us to seek small molecule primary amines that might trap and inactivate IsoLGs before they could modify cellular proteins or other endogenous cellular amines such as phosphatidylethanolamines to cause disease. Our studies identified 2-aminomethylphenols including 2-hydroxybenzylamine as IsoLG scavengers. Subsequent studies showed that they also trap other lipid dicarbonyls that react with primary amines such as 4-oxo-nonenal and malondialdehyde, but not hydroxyalkenals like 4-hydroxy-nonenal that preferentially react with soft nucleophiles. This review describes the use of these 2-aminomethylphenols as dicarbonyl scavengers to assess the contribution of IsoLGs and other amine-reactive lipid dicarbonyls to disease and as therapeutic agents.
Collapse
Affiliation(s)
- Sean S Davies
- Division of Clinical Pharmacology and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN, United States.
| | - Linda S May-Zhang
- Division of Clinical Pharmacology and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN, United States
| | - Olivier Boutaud
- Division of Clinical Pharmacology and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN, United States
| | - Venkataraman Amarnath
- Division of Clinical Pharmacology and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN, United States
| | - David G Harrison
- Division of Clinical Pharmacology and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
17
|
Beavers WN, Monteith AJ, Amarnath V, Mernaugh RL, Roberts LJ, Chazin WJ, Davies SS, Skaar EP. Arachidonic Acid Kills Staphylococcus aureus through a Lipid Peroxidation Mechanism. mBio 2019; 10:e01333-19. [PMID: 31575763 PMCID: PMC6775451 DOI: 10.1128/mbio.01333-19] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/21/2019] [Indexed: 01/14/2023] Open
Abstract
Staphylococcus aureus infects every niche of the human host. In response to microbial infection, vertebrates have an arsenal of antimicrobial compounds that inhibit bacterial growth or kill bacterial cells. One class of antimicrobial compounds consists of polyunsaturated fatty acids, which are highly abundant in eukaryotes and encountered by S. aureus at the host-pathogen interface. Arachidonic acid (AA) is one of the most abundant polyunsaturated fatty acids in vertebrates and is released in large amounts during the oxidative burst. Most of the released AA is converted to bioactive signaling molecules, but, independently of its role in inflammatory signaling, AA is toxic to S. aureus Here, we report that AA kills S. aureus through a lipid peroxidation mechanism whereby AA is oxidized to reactive electrophiles that modify S. aureus macromolecules, eliciting toxicity. This process is rescued by cotreatment with antioxidants as well as in a S. aureus strain genetically inactivated for lcpA (USA300 ΔlcpA mutant) that produces lower levels of reactive oxygen species. However, resistance to AA stress in the USA300 ΔlcpA mutant comes at a cost, making the mutant more susceptible to β-lactam antibiotics and attenuated for pathogenesis in a murine infection model compared to the parental methicillin-resistant S. aureus (MRSA) strain, indicating that resistance to AA toxicity increases susceptibility to other stressors encountered during infection. This report defines the mechanism by which AA is toxic to S. aureus and identifies lipid peroxidation as a pathway that can be modulated for the development of future therapeutics to treat S. aureus infections.IMPORTANCE Despite the ability of the human immune system to generate a plethora of molecules to control Staphylococcus aureus infections, S. aureus is among the pathogens with the greatest impact on human health. One class of host molecules toxic to S. aureus consists of polyunsaturated fatty acids. Here, we investigated the antibacterial properties of arachidonic acid, one of the most abundant polyunsaturated fatty acids in humans, and discovered that the mechanism of toxicity against S. aureus proceeds through lipid peroxidation. A better understanding of the molecular mechanisms by which the immune system kills S. aureus, and by which S. aureus avoids host killing, will enable the optimal design of therapeutics that complement the ability of the vertebrate immune response to eliminate S. aureus infections.
Collapse
Affiliation(s)
- William N Beavers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew J Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Venkataraman Amarnath
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Raymond L Mernaugh
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - L Jackson Roberts
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Walter J Chazin
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Sean S Davies
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
18
|
Shang L, Weng X, Wang D, Yue W, Mernaugh R, Amarnath V, Weir EK, Dudley SC, Xu Y, Hou M, Chen Y. Isolevuglandin scavenger attenuates pressure overload-induced cardiac oxidative stress, cardiac hypertrophy, heart failure and lung remodeling. Free Radic Biol Med 2019; 141:291-298. [PMID: 31254620 DOI: 10.1016/j.freeradbiomed.2019.06.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 12/20/2022]
Abstract
Increased levels of reactive isolevuglandins (IsoLGs) are associated with vascular inflammation and hypertension, two important factors affect heart failure (HF) development. The role of IsoLGs in HF development is unknown. Here we studied the role of IsoLG scavenger 2-hydroxybenzylamine (2-HOBA) in transverse aortic constriction (TAC) induced heart failure. We observed that TAC caused a significant increase of IsoLG protein adducts in cardiac and lung tissues in mice. Both IsoLG scavenger 2-hydroxybenzylamine (2-HOBA) and its less reactive isomer 4-hydroxybenzylamine (4-HOBA) significantly attenuated the left ventricular (LV) and lung IsoLGs in mice after TAC. 2-HOBA and 4-HOBA attenuated TAC-induced LV hypertrophy, heart failure, and the increase of lung weight in mice, and also improved TAC-induced LV dysfunction. Moreover, both 2-HOBA and 4-HOBA effectively attenuated LV cardiomyocyte hypertrophy, lung inflammation, lung fibrosis. These findings suggest that methods to reduce IsoLGs may be useful for HF therapy.
Collapse
Affiliation(s)
- Linlin Shang
- Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyu Weng
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongzhi Wang
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenhui Yue
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ray Mernaugh
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| | | | - E Kenneth Weir
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Samuel C Dudley
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Mingxiao Hou
- Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Yingjie Chen
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
19
|
Caspa Gokulan R, Adcock JM, Zagol-Ikapitte I, Mernaugh R, Williams P, Washington KM, Boutaud O, Oates JA, Dikalov SI, Zaika AI. Gastroesophageal Reflux Induces Protein Adducts in the Esophagus. Cell Mol Gastroenterol Hepatol 2018; 7:480-482.e7. [PMID: 30827415 PMCID: PMC6410348 DOI: 10.1016/j.jcmgh.2018.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 02/08/2023]
Affiliation(s)
| | - Jamie M. Adcock
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Irene Zagol-Ikapitte
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Raymond Mernaugh
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Phillip Williams
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kay M. Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Olivier Boutaud
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - John A. Oates
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Sergey I. Dikalov
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alexander I. Zaika
- Department of Surgery, University of Miami, Miami, Florida,Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Veterans Affairs, Miami VA Healthcare System, Miami, Florida,Corresponding author:
| |
Collapse
|
20
|
Xiao L, Patrick DM, Aden LA, Kirabo A. Mechanisms of isolevuglandin-protein adduct formation in inflammation and hypertension. Prostaglandins Other Lipid Mediat 2018; 139:48-53. [PMID: 30278231 PMCID: PMC6299826 DOI: 10.1016/j.prostaglandins.2018.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/15/2018] [Accepted: 09/20/2018] [Indexed: 12/15/2022]
Abstract
Inflammation has been implicated in the pathogenesis of hypertension and recent evidence suggests that isolevuglandin (IsoLG)-protein adducts play a role. Several hypertensive stimuli contribute to formation of IsoLG-protein adducts including excess dietary salt and catecholamines. The precise intracellular mechanisms by which these hypertensive stimuli lead to IsoLG-protein adduct formation are still not well understood; however, there is now evidence implicating NADPH-oxidase derived reactive oxygen species (ROS) in this process. ROS oxidize arachidonic acid leading to formation of IsoLGs, which non-covalently adduct to lysine residues and alter protein structure and function. Recent studies suggest that these altered proteins act as neo-antigens leading to an autoimmune state that results in hypertension. The goal of this mini-review is to highlight some of the hypertensive stimuli and the mechanisms contributing to IsoLG-protein adduct formation leading to inflammation and hypertension.
Collapse
Affiliation(s)
- Liang Xiao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - David M Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Luul A Aden
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States; Vanderbilt Institute for Infection, Immunology & Inflammation (VI4), Nashville, TN, United States.
| |
Collapse
|
21
|
Fuller JC, Pitchford LM, Abumrad NN, Rathmacher JA. Subchronic (90-day) repeated dose oral toxicity study of 2-hydroxybenzylamine acetate in rabbit. Regul Toxicol Pharmacol 2018; 100:52-58. [PMID: 30359704 DOI: 10.1016/j.yrtph.2018.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/05/2018] [Accepted: 10/20/2018] [Indexed: 11/25/2022]
Abstract
2-hydroxybenzylamine (2-HOBA), a naturally occurring compound found in buckwheat, has potential for use as a nutrition supplement due to its ability to protect against the damaging effects of oxidative stress. In a series of rodent toxicity studies, 2-HOBA acetate was well-tolerated and did not produce any toxic effects over 28 or 90 days of repeated oral administration. However, it remained necessary to test the potential toxicity of 2-HOBA acetate in a non-rodent species. In this investigation, 2-HOBA acetate was orally administered to male and female New Zealand White Rabbits for 90 days at doses of 100, 500, and 1000 mg·kg BW-1·day-1 (n = 5 per sex/group). As previously observed in rodents, 2-HOBA acetate administration was well tolerated. No toxic effects of 2-HOBA acetate were detected in body weight, feed consumption, hematology, blood chemistry, urine chemistry, organ weights, gross pathology or histopathology. Based on these findings, the no-observed-adverse-effect-level of 2-HOBA acetate in rabbits was determined to be 1000 mg·kg BW-1·day-1, which was the highest dose tested. These results provide further support for the safety of 2-HOBA acetate administration.
Collapse
Affiliation(s)
| | | | - Naji N Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - John A Rathmacher
- Metabolic Technologies, Inc., Ames, IA, 50010, USA; Department of Animal Science, Iowa State University, Ames, IA, 50010, USA.
| |
Collapse
|
22
|
Davies SS, May-Zhang LS. Isolevuglandins and cardiovascular disease. Prostaglandins Other Lipid Mediat 2018; 139:29-35. [PMID: 30296489 DOI: 10.1016/j.prostaglandins.2018.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/25/2018] [Accepted: 10/03/2018] [Indexed: 11/30/2022]
Abstract
Isolevuglandins are 4-ketoaldehydes formed by peroxidation of arachidonic acid. Isolevuglandins react rapidly with primary amines including the lysyl residues of proteins to form irreversible covalent modifications. This review highlights evidence for the potential role of isolevuglandin modification in the disease processes, especially atherosclerosis, and some of the tools including small molecule dicarbonyl scavengers utilized to assess their contributions to disease.
Collapse
Affiliation(s)
- Sean S Davies
- Department of Pharmacology, Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, United States.
| | - Linda S May-Zhang
- Department of Pharmacology, Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
23
|
Fuller JC, Pitchford LM, Abumrad NN, Rathmacher JA. Subchronic (90-day) repeated dose toxicity study of 2-hydroxybenzylamine acetate in rats. Regul Toxicol Pharmacol 2018; 99:225-232. [PMID: 30266239 DOI: 10.1016/j.yrtph.2018.09.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/20/2018] [Accepted: 09/23/2018] [Indexed: 11/16/2022]
Abstract
2-Hydroxybenzylamine (2-HOBA), a naturally occurring compound found in buckwheat, can protect cells and tissues from oxidative stress. In this study, 2-HOBA acetate was orally administered to male and female rats for 90 consecutive days at doses of 100, 500, and 1000 mg·kg BW-1·d-1 (n = 20 per sex/group). Subchronic administration of 2-HOBA was well tolerated at all dose levels. 2-HOBA-treated male rats were slightly heavier in the last weeks of the study, but this difference was very small (<5%), did not show a dose-response relationship, and was not observed in female rats. Similarly, some statistically significant changes in serum biochemistry and hematology parameters were noted, but these were not considered to be of biological or toxicological significance. Sporadic differences in organ weights were observed between groups, but all were small (<10%) and unlikely to indicate toxicity. The incidence of histopathological lesions was similar between treated and control groups across all organs. Based upon these findings, the no-observed-adverse-effect level was determined to be ≥ 1000 mg·kg BW-1·d-1, which was the highest dose tested. These results further support no toxicity associated with oral consumption of 2-HOBA acetate in rats and the continued development of 2-HOBA as a dietary supplement or functional food.
Collapse
Affiliation(s)
| | | | - Naji N Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - John A Rathmacher
- Metabolic Technologies, Inc, Ames, IA 50010, USA; Department of Animal Science, Iowa State University, Ames, IA, 50010, USA.
| |
Collapse
|
24
|
In vitro safety pharmacology evaluation of 2-hydroxybenzylamine acetate. Food Chem Toxicol 2018; 121:541-548. [PMID: 30253245 DOI: 10.1016/j.fct.2018.09.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/19/2018] [Accepted: 09/21/2018] [Indexed: 12/14/2022]
Abstract
2-hydroxybenzylamine (2-HOBA), a compound found in buckwheat, is a potent scavenger of reactive γ-ketoaldehydes, which are increased in diseases associated with inflammation and oxidative stress. While the potential of 2-HOBA is promising, studies were needed to characterize the safety of the compound before clinical trials. In a series of experiments, the risks of 2-HOBA-mediated mutagenicity and cardio-toxicity were assessed in vitro. The effects of 2-HOBA on the mRNA expression of select cytochrome P450 (CYP) enzymes were also assessed in cryopreserved human hepatocytes. Further, the distribution and metabolism of 2-HOBA in blood were determined. Our results indicate that 2-HOBA is not cytotoxic or mutagenic in vitro and does not induce the expression of CYP1A2, CYP2B6, or CYP3A4 in human hepatocytes. The results of the hERG testing showed a low risk of cardiac QT wave prolongation. Plasma protein binding and red blood cell distribution characteristics indicate low protein binding and no preferential distribution into erythrocytes. The major metabolites identified were salicylic acid and the glycoside conjugate of 2-HOBA. Together, these findings support development of 2-HOBA as a nutritional supplement and provide important information for the design of further preclinical safety studies in animals as well as for human clinical trials with 2-HOBA.
Collapse
|
25
|
Pitchford LM, Smith JD, Abumrad NN, Rathmacher JA, Fuller JC. Acute and 28-day repeated dose toxicity evaluations of 2-hydroxybenzylamine acetate in mice and rats. Regul Toxicol Pharmacol 2018; 98:190-198. [PMID: 30075181 DOI: 10.1016/j.yrtph.2018.07.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/27/2018] [Accepted: 07/28/2018] [Indexed: 11/30/2022]
Abstract
2-hydroxybenzylamine (2-HOBA), a compound naturally found in buckwheat, has been shown to protect cells and tissues from the damaging effects of oxidative stress. The purpose of this report was to evaluate 2-HOBA in preclinical oral rodent toxicity studies. This report includes the results from three oral toxicity studies in rodents: a preliminary 28-day feeding study in mice, a 14-day acute oral toxicity study in rats, and a 28-day repeated dose oral toxicity study in rats. The preliminary mouse feeding study showed no adverse effects of 2-HOBA at concentrations up to 0.456% by weight in feed, but decreased food intake and weight loss were observed at 1.56% 2-HOBA in the diet, likely due to poor palatability. In the acute dosing study, 2000 mg/kg BW 2-HOBA resulted in mortality in one of the six tested female rats, indicating a median lethal dose of 2500 mg/kg BW. In the 28-day repeated oral dose study, small differences were observed between 2-HOBA treated and control group rats, but none of these differences were determined to be of toxicological significance. Together, these studies support the lack of toxicity of oral administration of 2-HOBA acetate at doses up to 1000 mg/kg BW d-1 in rodents.
Collapse
Affiliation(s)
| | - Jodi D Smith
- Department of Veterinary Pathology, Iowa State University College of Veterinary Medicine, Ames, IA 50010, USA.
| | - Naji N Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - John A Rathmacher
- Metabolic Technologies, Inc, Ames, IA 50010, USA; Department of Animal Science, Iowa State University, Ames, IA 50010, USA.
| | | |
Collapse
|
26
|
May-Zhang LS, Yermalitsky V, Huang J, Pleasent T, Borja MS, Oda MN, Jerome WG, Yancey PG, Linton MF, Davies SS. Modification by isolevuglandins, highly reactive γ-ketoaldehydes, deleteriously alters high-density lipoprotein structure and function. J Biol Chem 2018; 293:9176-9187. [PMID: 29712723 DOI: 10.1074/jbc.ra117.001099] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/30/2018] [Indexed: 01/17/2023] Open
Abstract
Cardiovascular disease risk depends on high-density lipoprotein (HDL) function, not HDL-cholesterol. Isolevuglandins (IsoLGs) are lipid dicarbonyls that react with lysine residues of proteins and phosphatidylethanolamine. IsoLG adducts are elevated in atherosclerosis. The consequences of IsoLG modification of HDL have not been studied. We hypothesized that IsoLG modification of apoA-I deleteriously alters HDL function. We determined the effect of IsoLG on HDL structure-function and whether pentylpyridoxamine (PPM), a dicarbonyl scavenger, can preserve HDL function. IsoLG adducts in HDL derived from patients with familial hypercholesterolemia (n = 10, 233.4 ± 158.3 ng/mg) were found to be significantly higher than in healthy controls (n = 7, 90.1 ± 33.4 pg/mg protein). Further, HDL exposed to myeloperoxidase had elevated IsoLG-lysine adducts (5.7 ng/mg protein) compared with unexposed HDL (0.5 ng/mg protein). Preincubation with PPM reduced IsoLG-lysine adducts by 67%, whereas its inactive analogue pentylpyridoxine did not. The addition of IsoLG produced apoA-I and apoA-II cross-links beginning at 0.3 molar eq of IsoLG/mol of apoA-I (0.3 eq), whereas succinylaldehyde and 4-hydroxynonenal required 10 and 30 eq. IsoLG increased HDL size, generating a subpopulation of 16-23 nm. 1 eq of IsoLG decreased HDL-mediated [3H]cholesterol efflux from macrophages via ABCA1, which corresponded to a decrease in HDL-apoA-I exchange from 47.4% to only 24.8%. This suggests that IsoLG inhibits apoA-I from disassociating from HDL to interact with ABCA1. The addition of 0.3 eq of IsoLG ablated HDL's ability to inhibit LPS-stimulated cytokine expression by macrophages and increased IL-1β expression by 3.5-fold. The structural-functional effects were partially rescued with PPM scavenging.
Collapse
Affiliation(s)
- Linda S May-Zhang
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6602
| | - Valery Yermalitsky
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6602
| | - Jiansheng Huang
- the Division of Cardiovascular Medicine, Department of Medicine, and
| | | | - Mark S Borja
- the Department of Chemistry and Biochemistry, California State University East Bay, Hayward, California 94542, and
| | - Michael N Oda
- the Children's Hospital Oakland Research Institute, Oakland, California 94609
| | - W Gray Jerome
- the Department of Pathology, Vanderbilt Medical Center, Nashville, Tennessee 37232
| | - Patricia G Yancey
- the Division of Cardiovascular Medicine, Department of Medicine, and
| | - MacRae F Linton
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6602.,the Division of Cardiovascular Medicine, Department of Medicine, and
| | - Sean S Davies
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6602,
| |
Collapse
|
27
|
Lee SH, Matsunaga A, Oe T. Inhibition effect of pyridoxamine on lipid hydroperoxide-derived modifications to human serum albumin. PLoS One 2018; 13:e0196050. [PMID: 29672562 PMCID: PMC5908094 DOI: 10.1371/journal.pone.0196050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/01/2018] [Indexed: 02/02/2023] Open
Abstract
Pyridoxamine (PM) is a promising drug candidate for treating various chronic conditions/diseases in which oxidative stress and carbonyl compounds are important factors affecting pathogenicity. These abilities of PM are mainly attributed to its inhibition of advanced glycation and lipoxidation end product formation, by scavenging reactive carbonyl species. PM might therefore prevent protein damage from lipid hydroperoxide-derived aldehydes such as 4-oxo-2(E)-nonenal (ONE) and 4-hydroxy-2(E)-nonenal (HNE) by trapping them. It was previously reported that PM reacts with ONE to produce pyrrolo-1,3-oxazine (PO8) through the formation of pyrido-1,3-oxazine (PO1/PO2). In this study, we found that ONE and HNE yield an identical product containing a pyrrole ring (PO7, PH2) upon reaction with PM. The structure of PO7/PH2 was shown by LC-MS and NMR analyses to be 1-(2-hydroxy-6-hydroxymethyl-3-methylpyridin-4-ylmethyl)-2-pentylpyrrole. PO1, PO7/PH2, and PO8 were the main stable PM-ONE/HNE adducts. In the incubation of human serum albumin (HSA) with ONE or HNE, Lys residues provided the most favorable modification sites for both aldehydes, and the number of HNE-modified sites was higher than that of ONE-modified sites. When HSA was allowed to react with a linoleic acid hydroperoxide in the presence of ascorbic acid, ONE modified more residues (10 Lys, 3 His, 2 Arg) than did HNE (8 His, 2 Lys), indicating the relative reactivity of aldehydes towards amino acid residues. Upon treatment with increasing concentrations of PM, the concentrations of ONE-modified HSA peptides, but not of HNE-modified peptides, were reduced significantly and dose-dependently. Concomitantly, the formation of PM-ONE adducts increased in a dose-dependent manner. The inhibition effect of PM was also confirmed in the cell system subjected to oxidative stress. Our results demonstrate that PM can inhibit lipid hydroperoxide-derived damage to proteins by trapping ONE preferentially, and the resulting PM-ONE adducts can be used as a dosimeter for ONE production to determine the levels of lipid peroxidation.
Collapse
Affiliation(s)
- Seon Hwa Lee
- Department of Bio-analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- * E-mail: (SHL); (TO)
| | - Atsushi Matsunaga
- Department of Bio-analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tomoyuki Oe
- Department of Bio-analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- * E-mail: (SHL); (TO)
| |
Collapse
|
28
|
Nguyen TT, Caito SW, Zackert WE, West JD, Zhu S, Aschner M, Fessel JP, Roberts LJ. Scavengers of reactive γ-ketoaldehydes extend Caenorhabditis elegans lifespan and healthspan through protein-level interactions with SIR-2.1 and ETS-7. Aging (Albany NY) 2017; 8:1759-80. [PMID: 27514077 PMCID: PMC5032694 DOI: 10.18632/aging.101011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/20/2016] [Indexed: 11/25/2022]
Abstract
Isoketals (IsoKs) are highly reactive γ-ketoaldehyde products of lipid peroxidation that covalently adduct lysine side chains in proteins, impairing their function. Using C. elegans as a model organism, we sought to test the hypothesis that IsoKs contribute to molecular aging through adduction and inactivation of specific protein targets, and that this process can be abrogated using salicylamine (SA), a selective IsoK scavenger. Treatment with SA extends adult nematode longevity by nearly 56% and prevents multiple deleterious age-related biochemical and functional changes. Testing of a variety of molecular targets for SA's action revealed the sirtuin SIR-2.1 as the leading candidate. When SA was administered to a SIR-2.1 knockout strain, the effects on lifespan and healthspan extension were abolished. The SIR-2.1-dependent effects of SA were not mediated by large changes in gene expression programs or by significant changes in mitochondrial function. However, expression array analysis did show SA-dependent regulation of the transcription factor ets-7 and associated genes. In ets-7 knockout worms, SA's longevity effects were abolished, similar to sir-2.1 knockouts. However, SA dose-dependently increases ets-7 mRNA levels in non-functional SIR-2.1 mutant, suggesting that both are necessary for SA's complete lifespan and healthspan extension.
Collapse
Affiliation(s)
- Thuy T Nguyen
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Samuel W Caito
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Environmental Medicine, University of Rochester, Rochester, NY 14642, USA
| | - William E Zackert
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - James D West
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Shijun Zhu
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael Aschner
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joshua P Fessel
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - L Jackson Roberts
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
29
|
Davies SS, Zhang LS. Reactive Carbonyl Species Scavengers-Novel Therapeutic Approaches for Chronic Diseases. ACTA ACUST UNITED AC 2017; 3:51-67. [PMID: 28993795 DOI: 10.1007/s40495-017-0081-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF THE REVIEW To summarize recent evidence supporting the use of reactive carbonyl species scavengers in the prevention and treatment of disease. RECENT FINDINGS The newly developed 2-aminomethylphenol class of scavengers shows great promise in preclinical trials for a number of diverse conditions including neurodegenerative diseases and cardiovascular disease. In addition, new studies with the thiol-based and imidazole-based scavengers have found new applications outside of adjunctive therapy for chemotherapeutics. SUMMARY Reactive oxygen species (ROS) generated by cells and tissues act as signaling molecules and as cytotoxic agents to defend against pathogens, but ROS also cause collateral damage to vital cellular components. The polyunsaturated fatty acyl chains of phospholipids in the cell membranes are particularly vulnerable to damaging peroxidation by ROS. Evidence suggests that the breakdown of these peroxidized lipids to reactive carbonyls species plays a critical role in many chronic diseases. Antioxidants that abrogate ROS-induced formation of reactive carbonyl species also abrogate normal ROS signaling and thus exert both beneficial and adverse functional effects. The use of scavengers of reactive dicarbonyl species represent an alternative therapeutic strategy to potentially mitigate the adverse effects of ROS without abrogating normal signaling by ROS. In this review, we focus on three classes of reactive carbonyl species scavengers: thiol-based scavengers (2-mercaptoethanesulfonate and amifostine), imidazole-based scavengers (carnosine and its analogs), and 2-aminomethylphenols-based scavengers (pyridoxamine, 2-hydroxybenzylamine, and 5'-O-pentyl-pyridoxamine) that are either undergoing pre-clinical studies, advancing to clinical trials, or are already in clinical use.
Collapse
Affiliation(s)
- Sean S Davies
- Department of Pharmacology and Division of Clinical Pharmacology, Vanderbilt University, 556 Robinson Research Building, 2220 Pierce Avenue, Nashville, TN 37232-6602
| | - Linda S Zhang
- Department of Pharmacology and Division of Clinical Pharmacology, Vanderbilt University, 556 Robinson Research Building, 2220 Pierce Avenue, Nashville, TN 37232-6602
| |
Collapse
|
30
|
Longato L, Andreola F, Davies SS, Roberts JL, Fusai G, Pinzani M, Moore K, Rombouts K. Reactive gamma-ketoaldehydes as novel activators of hepatic stellate cells in vitro. Free Radic Biol Med 2017; 102:162-173. [PMID: 27890721 DOI: 10.1016/j.freeradbiomed.2016.11.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 11/28/2022]
Abstract
AIMS Products of lipid oxidation, such as 4-hydroxynonenal (4-HNE), are key activators of hepatic stellate cells (HSC) to a pro-fibrogenic phenotype. Isolevuglandins (IsoLG) are a family of acyclic γ-ketoaldehydes formed through oxidation of arachidonic acid or as by-products of the cyclooxygenase pathway. IsoLGs are highly reactive aldehydes which are efficient at forming protein adducts and cross-links at concentrations 100-fold lower than 4-hydroxynonenal. Since the contribution of IsoLGs to liver injury has not been studied, we synthesized 15-E2-IsoLG and used it to investigate whether IsoLG could induce activation of HSC. RESULTS Primary human HSC were exposed to 15-E2-IsoLG for up to 48h. Exposure to 5μM 15-E2-IsoLG in HSCs promoted cytotoxicity and apoptosis. At non-cytotoxic doses (50 pM-500nM) 15-E2-IsoLG promoted HSC activation, indicated by increased expression of α-SMA, sustained activation of ERK and JNK signaling pathways, and increased mRNA and/or protein expression of cytokines and chemokines, which was blocked by inhibitors of JNK and NF-kB. In addition, IsoLG promoted formation of reactive oxygen species, and induced an early activation of ER stress, followed by autophagy. Inhibition of autophagy partially reduced the pro-inflammatory effects of IsoLG, suggesting that it might serve as a cytoprotective response. INNOVATION This study is the first to describe the biological effects of IsoLG in primary HSC, the main drivers of hepatic fibrosis. CONCLUSIONS IsoLGs represent a newly identified class of activators of HSC in vitro, which are biologically active at concentrations as low as 500 pM, and are particularly effective at promoting a pro-inflammatory response and autophagy.
Collapse
Affiliation(s)
- Lisa Longato
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Fausto Andreola
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK
| | - Sean S Davies
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jackson L Roberts
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Giuseppe Fusai
- Division of Surgery, University College London, Royal Free, London, UK
| | - Massimo Pinzani
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Kevin Moore
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Krista Rombouts
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK.
| |
Collapse
|
31
|
Pearson JN, Warren E, Liang LP, Roberts LJ, Patel M. Scavenging of highly reactive gamma-ketoaldehydes attenuates cognitive dysfunction associated with epileptogenesis. Neurobiol Dis 2016; 98:88-99. [PMID: 27932305 DOI: 10.1016/j.nbd.2016.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/21/2016] [Accepted: 11/25/2016] [Indexed: 02/02/2023] Open
Abstract
Cognitive dysfunction is a major comorbidity of the epilepsies; however, treatments targeting seizure-associated cognitive dysfunction, particularly deficits in learning and memory are not available. Isoketals and neuroketals, collectively known as gamma-ketoaldehydes are formed via the non-enzymatic, free radical catalyzed oxidation of arachidonic acid and docosahexaenoic acid, respectively. They are attractive candidates for oxidative protein damage and resultant cognitive dysfunction due to their formation within the plasma membrane and their high proclivity to form cytotoxic adducts on protein lysine residues. We tested the hypothesis that gamma-ketoaldehydes mechanistically contribute to seizure-associated memory impairment using a specific gamma-ketoaldehyde scavenger, salicylamine in the kainic acid and pilocarpine rat models of temporal lobe epilepsy. We show that gamma-ketoaldehydes are increased following epileptogenic injury in hippocampus and perirhinal cortex, two brain regions imperative for learning and memory. Treatment with an orally bioavailable, brain permeable scavenger, salicylamine attenuated 1) spatial memory deficits 2) reference memory deficits and 3) neuronal loss and astrogliosis in two mechanistically distinct models of epilepsy without affecting the epileptogenic injury or the development of chronic epilepsy. We have previously demonstrated that reactive oxygen species and the lipid peroxidation biomarkers, F2-isoprostanes are produced following status epilepticus. However, which reactive species specifically mediate oxidative damage to cellular macromolecules remains at large. We provide novel data suggesting that memory impairment occurs via gamma-ketoaldehyde production in two models of epilepsy and that treatment with a gamma-ketoaldehyde scavenger can protect vulnerable neurons. This work suggests a novel target and therapy to treat seizure-induced memory deficits in epilepsy.
Collapse
Affiliation(s)
- Jennifer N Pearson
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 80045, United States
| | - Eric Warren
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 80045, United States
| | - Li-Ping Liang
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 80045, United States
| | - L Jackson Roberts
- Division of Clinical Pharmacology, Vanderbilt University, 37235, United States; Department of Pharmacology, Vanderbilt University, 37235, United States
| | - Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 80045, United States.
| |
Collapse
|
32
|
Reddy IA, Pino JA, Weikop P, Osses N, Sørensen G, Bering T, Valle C, Bluett RJ, Erreger K, Wortwein G, Reyes JG, Graham D, Stanwood GD, Hackett TA, Patel S, Fink-Jensen A, Torres GE, Galli A. Glucagon-like peptide 1 receptor activation regulates cocaine actions and dopamine homeostasis in the lateral septum by decreasing arachidonic acid levels. Transl Psychiatry 2016; 6:e809. [PMID: 27187231 PMCID: PMC5070047 DOI: 10.1038/tp.2016.86] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/19/2016] [Accepted: 03/30/2016] [Indexed: 12/12/2022] Open
Abstract
Agonism of the glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) has been effective at treating aspects of addictive behavior for a number of abused substances, including cocaine. However, the molecular mechanisms and brain circuits underlying the therapeutic effects of GLP-1R signaling on cocaine actions remain elusive. Recent evidence has revealed that endogenous signaling at the GLP-1R within the forebrain lateral septum (LS) acts to reduce cocaine-induced locomotion and cocaine conditioned place preference, both considered dopamine (DA)-associated behaviors. DA terminals project from the ventral tegmental area to the LS and express the DA transporter (DAT). Cocaine acts by altering DA bioavailability by targeting the DAT. Therefore, GLP-1R signaling might exert effects on DAT to account for its regulation of cocaine-induced behaviors. We show that the GLP-1R is highly expressed within the LS. GLP-1, in LS slices, significantly enhances DAT surface expression and DAT function. Exenatide (Ex-4), a long-lasting synthetic analog of GLP-1 abolished cocaine-induced elevation of DA. Interestingly, acute administration of Ex-4 reduces septal expression of the retrograde messenger 2-arachidonylglycerol (2-AG), as well as a product of its presynaptic degradation, arachidonic acid (AA). Notably, AA reduces septal DAT function pointing to AA as a novel regulator of central DA homeostasis. We further show that AA oxidation product γ-ketoaldehyde (γ-KA) forms adducts with the DAT and reduces DAT plasma membrane expression and function. These results support a mechanism in which postsynaptic septal GLP-1R activation regulates 2-AG levels to alter presynaptic DA homeostasis and cocaine actions through AA.
Collapse
Affiliation(s)
- I A Reddy
- Neuroscience Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - J A Pino
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - P Weikop
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Psychiatric Centre Copenhagen, University Hospital Copenhagen, Copenhagen, Denmark
| | - N Osses
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - G Sørensen
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Psychiatric Centre Copenhagen, University Hospital Copenhagen, Copenhagen, Denmark
| | - T Bering
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - C Valle
- Departamento de Ciencias Básicas, Universidad de Viña del Mar, Viña del Mar, Chile
| | - R J Bluett
- Neuroscience Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - K Erreger
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - G Wortwein
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - J G Reyes
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - D Graham
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University, Tallahassee, FL, USA
| | - G D Stanwood
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University, Tallahassee, FL, USA
| | - T A Hackett
- Neuroscience Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - S Patel
- Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - A Fink-Jensen
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Psychiatric Centre Copenhagen, University Hospital Copenhagen, Copenhagen, Denmark
| | - G E Torres
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - A Galli
- Neuroscience Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
33
|
Roscales S, Csákÿ AG. Transition-metal free reactions of boronic acids: cascade addition – ring-opening of furans towards functionalized γ-ketoaldehydes. Chem Commun (Camb) 2016; 52:3018-21. [DOI: 10.1039/c5cc08809g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We describe the first ring-opening of furfuryl alcohols with boronic acids to afford functionalized γ-ketoaldehydes.
Collapse
Affiliation(s)
- S. Roscales
- Instituto Pluridisciplinar
- Universidad Complutense
- Campus de Excelencia Internacional Moncloa
- 28040-Madrid
- Spain
| | - A. G. Csákÿ
- Instituto Pluridisciplinar
- Universidad Complutense
- Campus de Excelencia Internacional Moncloa
- 28040-Madrid
- Spain
| |
Collapse
|
34
|
Abstract
4-Oxo-2-nonenal (ONE), a product of cellular lipid oxidation, reacts nonspecifically with the lysine residues of proteins and is generated in increased amounts during degenerative diseases and cancer. We show that pyridoxamine, salicylamine, and related 2-aminomethylphenols react with ONE, to form pyrrolo[2,1-b][1,3]oxazines with the participation of both the amino and the phenolic groups. 2-Aminomethylphenols react with ONE as well as with the Michael adducts of ONE much more rapidly than lysine, suggesting their use for therapeutically scavenging ONE.
Collapse
Affiliation(s)
- Venkataraman Amarnath
- Department of Pathology, Microbiology and Immunology, and ‡Division of Clinical Pharmacology Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Kalyani Amarnath
- Department of Pathology, Microbiology and Immunology, and ‡Division of Clinical Pharmacology Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| |
Collapse
|
35
|
Abstract
SIGNIFICANCE A diverse family of lipid-derived levulinaldehydes, isolevuglandins (isoLGs), is produced by rearrangement of endoperoxide intermediates generated through both cyclooxygenase (COX) and free radical-induced cyclooxygenation of polyunsaturated fatty acids and their phospholipid esters. The formation and reactions of isoLGs with other biomolecules has been linked to alcoholic liver disease, Alzheimer's disease, age-related macular degeneration, atherosclerosis, cardiac arythmias, cancer, end-stage renal disease, glaucoma, inflammation of allergies and infection, mitochondrial dysfunction, multiple sclerosis, and thrombosis. This review chronicles progress in understanding the chemistry of isoLGs, detecting their production in vivo and understanding their biological consequences. CRITICAL ISSUES IsoLGs have never been isolated from biological sources, because they form adducts with primary amino groups of other biomolecules within seconds. Chemical synthesis enabled investigation of isoLG chemistry and detection of isoLG adducts present in vivo. RECENT ADVANCES The first peptide mapping and sequencing of an isoLG-modified protein present in human retina identified the modification of a specific lysyl residue of the sterol C27-hydroxylase Cyp27A1. This residue is preferentially modified by iso[4]LGE2 in vitro, causing loss of function. Adduction of less than one equivalent of isoLG can induce COX-associated oligomerization of the amyloid peptide Aβ1-42. Adduction of isoLGE2 to phosphatidylethanolamines causes gain of function, converting them into proinflammatory isoLGE2-PE agonists that foster monocyte adhesion to endothelial cells. FUTURE DIRECTIONS Among the remaining questions on the biochemistry of isoLGs are the dependence of biological activity on isoLG isomer structure, the structures and mechanism of isoLG-derived protein-protein and DNA-protein cross-link formation, and its biological consequences.
Collapse
Affiliation(s)
- Robert G Salomon
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio
| | - Wenzhao Bi
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
36
|
Sidorova TN, Yermalitskaya LV, Mace LC, Wells KS, Boutaud O, Prinsen JK, Davies SS, Roberts LJ, Dikalov SI, Glabe CG, Amarnath V, Barnett JV, Murray KT. Reactive γ-ketoaldehydes promote protein misfolding and preamyloid oligomer formation in rapidly-activated atrial cells. J Mol Cell Cardiol 2015; 79:295-302. [PMID: 25463275 PMCID: PMC4302000 DOI: 10.1016/j.yjmcc.2014.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/24/2014] [Accepted: 11/12/2014] [Indexed: 01/15/2023]
Abstract
Rapid activation causes remodeling of atrial myocytes resembling that which occurs in experimental and human atrial fibrillation (AF). Using this cellular model, we previously observed transcriptional upregulation of proteins implicated in protein misfolding and amyloidosis. For organ-specific amyloidoses such as Alzheimer's disease, preamyloid oligomers (PAOs) are now recognized to be the primary cytotoxic species. In the setting of oxidative stress, highly-reactive lipid-derived mediators known as γ-ketoaldehydes (γ-KAs) have been identified that rapidly adduct proteins and cause PAO formation for amyloid β1-42 implicated in Alzheimer's. We hypothesized that rapid activation of atrial cells triggers oxidative stress with lipid peroxidation and formation of γ-KAs, which then rapidly crosslink proteins to generate PAOs. To investigate this hypothesis, rapidly-paced and control, spontaneously-beating atrial HL-1 cells were probed with a conformation-specific antibody recognizing PAOs. Rapid stimulation of atrial cells caused the generation of cytosolic PAOs along with a myocyte stress response (e.g., transcriptional upregulation of Nppa and Hspa1a), both of which were absent in control, unpaced cells. Rapid activation also caused the formation of superoxide and γ-KA adducts in atriomyocytes, while direct exposure of cells to γ-KAs resulted in PAO production. Increased cytosolic atrial natriuretic peptide (ANP), and the generation of ANP oligomers with exposure to γ-KAs and rapid atrial HL-1 cell stimulation, strongly suggest a role for ANP in PAO formation. Salicylamine (SA) is a small molecule scavenger of γ-KAs that can protect proteins from modification by these reactive compounds. PAO formation and transcriptional remodeling were inhibited when cells were stimulated in the presence of SA, but not with the antioxidant curcumin, which is incapable of scavenging γ-KAs. These results demonstrate that γ-KAs promote protein misfolding and PAO formation as a component of the atrial cell stress response to rapid activation, and they provide a potential mechanistic link between oxidative stress and atrial cell injury.
Collapse
Affiliation(s)
- Tatiana N Sidorova
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Liudmila V Yermalitskaya
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lisa C Mace
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - K Sam Wells
- Departments of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Olivier Boutaud
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joseph K Prinsen
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sean S Davies
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - L Jackson Roberts
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sergey I Dikalov
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Venkataraman Amarnath
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joey V Barnett
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Katherine T Murray
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
37
|
Dwyer JP, Greco BA, Umanath K, Packham D, Fox JW, Peterson R, Broome BR, Greene LE, Sika M, Lewis JB. Pyridoxamine dihydrochloride in diabetic nephropathy (PIONEER-CSG-17): lessons learned from a pilot study. Nephron Clin Pract 2014; 129:22-8. [PMID: 25532068 DOI: 10.1159/000369310] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 10/02/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Pyridoxamine dihydrochloride (Pyridorin™) blocks pathogenic oxidative pathways in the progression of diabetic nephropathy. The pyridoxamine pilot study was designed to test entry criteria and outcomes. Subjects had SCr 1.3-3.5 mg/dl, protein-to-creatinine ≥1,200 mg/g and used a surrogate outcome of ΔSCr over 52 weeks. Subjects had to be on a maximally tolerated dose of ACE/ARB for 3 months; stable other antihypertensive doses for 2 months; stable diuretic dose for 2 weeks, and BP ≤160/90 mm Hg; or enter a Pharmaco-Stabilization Phase (PSP). This pilot failed to detect an effect on ΔSCr in intent-to-treat analysis. METHODS We queried the locked clinical trial database for subgroups in which there was a treatment effect. RESULTS Subjects not requiring PSP and those with entry SCr <2.0 mg/dl had a treatment effect. Subjects entering PSP required more changes in antihypertensive medications and experienced larger ΔSCr over 52 weeks. PSP subjects with BP >140/90 mm Hg had no treatment effect, but those ≤140/90 mm Hg did. CONCLUSION Time required for acute effects of ACE/ARB to stabilize is unknown, but these data suggest >3 months. Thus, subjects in the pivotal trial must be on ACE/ARB for 6 months. Frequent antihypertensive adjustment could engender SCr changes unrelated to CKD progression. Thus, we will require subjects to have BP ≤150/90 mm Hg and on stable antihypertensives for 26 weeks, or ≤140/90 mm Hg and on stable antihypertensives for 13 weeks. Since ΔSCr over 52 weeks is limited as a surrogate outcome, the pivotal trial uses a time-to-event analysis of baseline SCr to at least a 50% increase in SCr or ESRD as the primary outcome. This substantial ΔSCr is protected from noise and is clinically relevant. The pyridoxamine pilot provided critical information to inform the design of PIONEER-CSG-17, which we conducted under the SPA agreement with FDA.
Collapse
|
38
|
Kirabo A, Fontana V, de Faria APC, Loperena R, Galindo CL, Wu J, Bikineyeva AT, Dikalov S, Xiao L, Chen W, Saleh MA, Trott DW, Itani HA, Vinh A, Amarnath V, Amarnath K, Guzik TJ, Bernstein KE, Shen XZ, Shyr Y, Chen SC, Mernaugh RL, Laffer CL, Elijovich F, Davies SS, Moreno H, Madhur MS, Roberts J, Harrison DG. DC isoketal-modified proteins activate T cells and promote hypertension. J Clin Invest 2014; 124:4642-56. [PMID: 25244096 DOI: 10.1172/jci74084] [Citation(s) in RCA: 390] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 08/04/2014] [Indexed: 12/21/2022] Open
Abstract
Oxidative damage and inflammation are both implicated in the genesis of hypertension; however, the mechanisms by which these stimuli promote hypertension are not fully understood. Here, we have described a pathway in which hypertensive stimuli promote dendritic cell (DC) activation of T cells, ultimately leading to hypertension. Using multiple murine models of hypertension, we determined that proteins oxidatively modified by highly reactive γ-ketoaldehydes (isoketals) are formed in hypertension and accumulate in DCs. Isoketal accumulation was associated with DC production of IL-6, IL-1β, and IL-23 and an increase in costimulatory proteins CD80 and CD86. These activated DCs promoted T cell, particularly CD8+ T cell, proliferation; production of IFN-γ and IL-17A; and hypertension. Moreover, isoketal scavengers prevented these hypertension-associated events. Plasma F2-isoprostanes, which are formed in concert with isoketals, were found to be elevated in humans with treated hypertension and were markedly elevated in patients with resistant hypertension. Isoketal-modified proteins were also markedly elevated in circulating monocytes and DCs from humans with hypertension. Our data reveal that hypertension activates DCs, in large part by promoting the formation of isoketals, and suggest that reducing isoketals has potential as a treatment strategy for this disease.
Collapse
|
39
|
Davies SS, Guo L. Lipid peroxidation generates biologically active phospholipids including oxidatively N-modified phospholipids. Chem Phys Lipids 2014; 181:1-33. [PMID: 24704586 DOI: 10.1016/j.chemphyslip.2014.03.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/28/2014] [Accepted: 03/18/2014] [Indexed: 12/25/2022]
Abstract
Peroxidation of membranes and lipoproteins converts "inert" phospholipids into a plethora of oxidatively modified phospholipids (oxPL) that can act as signaling molecules. In this review, we will discuss four major classes of oxPL: mildly oxygenated phospholipids, phospholipids with oxidatively truncated acyl chains, phospholipids with cyclized acyl chains, and phospholipids that have been oxidatively N-modified on their headgroups by reactive lipid species. For each class of oxPL we will review the chemical mechanisms of their formation, the evidence for their formation in biological samples, the biological activities and signaling pathways associated with them, and the catabolic pathways for their elimination. We will end by briefly highlighting some of the critical questions that remain about the role of oxPL in physiology and disease.
Collapse
Affiliation(s)
- Sean S Davies
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University, United States.
| | - Lilu Guo
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University, United States
| |
Collapse
|
40
|
Charvet CD, Saadane A, Wang M, Salomon RG, Brunengraber H, Turko IV, Pikuleva IA. Pretreatment with pyridoxamine mitigates isolevuglandin-associated retinal effects in mice exposed to bright light. J Biol Chem 2013; 288:29267-80. [PMID: 23970548 DOI: 10.1074/jbc.m113.498832] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The benefits of antioxidant therapy for treating age-related macular degeneration, a devastating retinal disease, are limited. Perhaps species other than reactive oxygen intermediates should be considered as therapeutic targets. These could be lipid peroxidation products, including isolevuglandins (isoLGs), prototypical and extraordinarily reactive γ-ketoaldehydes that avidly bind to proteins, phospholipids, and DNA and modulate the properties of these biomolecules. We found isoLG adducts in aged human retina but not in the retina of mice kept under dim lighting. Hence, to test whether scavenging of isoLGs could complement or supplant antioxidant therapy, we exposed mice to bright light and found that this insult leads to retinal isoLG-adduct formation. We then pretreated mice with pyridoxamine, a B6 vitamer and efficient scavenger of γ-ketoaldehydes, and found that the levels of retinal isoLG adducts are decreased, and morphological changes in photoreceptor mitochondria are not as pronounced as in untreated animals. Our study demonstrates that preventing the damage to biomolecules by lipid peroxidation products, a novel concept in vision research, is a viable strategy to combat oxidative stress in the retina.
Collapse
|
41
|
Charvet CD, Laird J, Xu Y, Salomon RG, Pikuleva IA. Posttranslational modification by an isolevuglandin diminishes activity of the mitochondrial cytochrome P450 27A1. J Lipid Res 2013; 54:1421-9. [PMID: 23479405 DOI: 10.1194/jlr.m035790] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Posttranslational modification by isolevuglandins (isoLGs), arachidonate oxidation products, is an important yet understudied process associated with altered protein properties. This type of modification is detected in cytochrome P450 27A1 (CYP27A1), a multifunction enzyme expressed in almost every cell and involved in the metabolism of cholesterol and other sterols. Previously, the CYP27A1 Lys(358)-isoLG adduct was found in human retina afflicted with age-related macular degeneration. Yet, the effect of Lys(358) modification on enzyme activity was not investigated. Herein, we characterized catalytic properties of Lys(358) as well as Lys(476) CYP27A1 mutants before and after isoLG treatment and quantified the extent of modification by multiple reaction monitoring. The K358R mutant was less susceptible to isoLG-induced loss of catalytic activity than the wild type (WT), whereas the K476R mutant was nearly as vulnerable as the WT. Both mutants showed less isoLG modification than WT. Thus, modification of Lys(358), a residue involved in redox partner interactions, is the major contributor to isoLG-associated loss of CYP27A1 activity. Our data show the specificity of isoLG modification, provide direct evidence that isoLG adduction impairs enzyme activity, and support our hypothesis that isoLG modification in the retina is detrimental to CYP27A1 enzyme activity, potentially disrupting cholesterol homeostasis.
Collapse
Affiliation(s)
- Casey D Charvet
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|
42
|
Guo L, Davies SS. Bioactive aldehyde-modified phosphatidylethanolamines. Biochimie 2012; 95:74-8. [PMID: 22819995 DOI: 10.1016/j.biochi.2012.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 07/10/2012] [Indexed: 12/21/2022]
Abstract
Lipid peroxidation generates a variety of lipid aldehydes, which have been recognized to modify protein and DNA, causing inflammation and cancer. However, recent studies demonstrate that phosphatidylethanolamine (PE) is a major target for these aldehydes, forming aldehyde-modified PEs (al-PEs) as a novel family of mediators for inflammation. This review summarizes our current understanding of these al-PEs, including formation, detection, structural characterization, physiological relevance and mechanism of action.
Collapse
Affiliation(s)
- Lilu Guo
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232-6602, USA.
| | | |
Collapse
|
43
|
Davies SS, Bodine C, Matafonova E, Pantazides BG, Bernoud-Hubac N, Harrison FE, Olson SJ, Montine TJ, Amarnath V, Roberts LJ. Treatment with a γ-ketoaldehyde scavenger prevents working memory deficits in hApoE4 mice. J Alzheimers Dis 2012; 27:49-59. [PMID: 21709376 DOI: 10.3233/jad-2011-102118] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Both inflammation and oxidative injury are features of Alzheimer's disease (AD), but the contribution of these intertwined phenomena to the loss of working memory in this disease is unclear. We tested the hypothesis that highly reactive γ-ketoaldehydes that are formed both by non-enzymatic free radical catalyzed lipid peroxidation and by cyclooxygenases may be causally linked to the development of memory impairment in AD. We found that levels of γ-ketoaldehyde protein adducts were increased in the hippocampus of brains obtained postmortem from patients with AD compared to age-matched controls, but that levels of γ-ketoaldehyde protein adducts in the cerebellum were not different in the two groups. Moreover, immunohistochemistry revealed that adducts localized to hippocampal pyramidal neurons. We tested the effect of an orally available γ-ketoaldehyde scavenger, salicylamine, on the development of spatial working memory deficits in hApoE4 targeted replacement mice, a mouse model of dementia. Long-term salicylamine supplementation did not significantly alter body weight or survival, but protected against the development of age-related deficits in spatial working memory in 12-14 month old ApoE4 mice. These findings suggest that γ-ketoaldehyde adduct formation is associated with damage to hippocampal neurons in patients with AD and can contribute to the pathogenesis of spatial working memory deficits in hApoE4 mice. These data provide a rational basis for future studies exploring whether γ-ketoaldehyde scavengers may mitigate the development of cognitive dysfunction in patients with AD.
Collapse
Affiliation(s)
- Sean S Davies
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232-6602, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Ginger L Milne
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6602, USA.
| | | | | | | | | |
Collapse
|
45
|
Caldés C, Vilanova B, Adrover M, Muñoz F, Donoso J. Phenol Group in Pyridoxamine Acts as a Stabilizing Element for Its Carbinolamines and Schiff Bases. Chem Biodivers 2011; 8:1318-32. [DOI: 10.1002/cbdv.201000296] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
46
|
Picklo MJ, Azenkeng A, Hoffmann MR. Trans-4-oxo-2-nonenal potently alters mitochondrial function. Free Radic Biol Med 2011; 50:400-7. [PMID: 21092757 DOI: 10.1016/j.freeradbiomed.2010.11.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 11/08/2010] [Accepted: 11/09/2010] [Indexed: 11/27/2022]
Abstract
Alzheimer disease elevates lipid peroxidation in the brain and data indicate that the resulting lipid-aldehydes are pathological effectors of lipid peroxidation. The disposition of 4-substituted nonenals derived from arachidonate (20:4, n-6) and linoleate (18:2, n-6) oxidation is modulated by their protein adduction targets, their metabolism, and the nature of the 4-substitutent. Trans-4-oxo-2-nonenal (4-ONE) has a higher toxicity in some systems than the more commonly studied trans-4-hydroxy-2-nonenal (HNE). In this work, we performed a structure-function analysis of 4-hydroxy/oxoalkenal upon mitochondrial endpoints. We tested the hypotheses that 4-ONE, owing to a highly reactive nature, is more toxic than HNE and that HNE toxicity is enantioselective. We chose to study freshly isolated brain mitochondria because of the role of mitochondrial dysfunction in neurodegenerative disorders. Whereas there was little effect related to HNE chirality, our data indicate that in the mitochondrial environment, the order of toxic potency under most conditions was 4-ONE>HNE. 4-ONE uncoupled mitochondrial respiration at a concentration of 5μM and inhibited aldehyde dehydrogenase 2 (ALDH2) activity with an IC(50) of approximately 0.5μM. The efficacy of altering mitochondrial endpoints was ALDH2 inhibition>respiration=mitochondrial swelling=ALDH5A inhibition>GSH depletion. Thiol-based alkenal scavengers, but not amine-based scavengers, were effective in blocking the effects of 4-ONE upon respiration. Quantum mechanical calculations provided insights into the basis for the elevated reactivity of 4-ONE>HNE. Our data demonstrate that 4-ONE is a potent effector of lipid peroxidation in the mitochondrial environment.
Collapse
Affiliation(s)
- Matthew J Picklo
- Agricultural Research Center, Grand Forks Human Nutrition Research Center, U.S. Department of Agriculture, Grand Forks, ND 58203-9034, USA.
| | | | | |
Collapse
|
47
|
Zagol-Ikapitte I, Amarnath V, Bala M, Roberts LJ, Oates JA, Boutaud O. Characterization of scavengers of gamma-ketoaldehydes that do not inhibit prostaglandin biosynthesis. Chem Res Toxicol 2010; 23:240-50. [PMID: 20041722 DOI: 10.1021/tx900407a] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Expression of cyclooxygenase-2 (COX-2) is associated with the development of many pathologic conditions. The product of COX-2, prostaglandin H(2) (PGH(2)), can spontaneously rearrange to form reactive gamma-ketoaldehydes called levuglandins (LGs). This gamma-ketoaldehyde structure confers a high degree of reactivity on the LGs, which rapidly form covalent adducts with primary amines of protein residues. Formation of LG adducts of proteins has been demonstrated in pathologic conditions (e.g., increased levels in the hippocampus in Alzheimer's disease) and during physiologic function (platelet activation). On the basis of knowledge that lipid modification of proteins is known to cause their translocation and to alter their function, we hypothesize that modification of proteins by LG could have functional consequences. Testing this hypothesis requires an experimental approach that discriminates between the effects of protein modification by LG and the effects of cyclooxygenase-derived prostanoids acting through their G-protein coupled receptors. To achieve this goal, we have synthesized and evaluated a series of scavengers that react with LG with a potency more than 2 orders of magnitude greater than that with the epsilon-amine of lysine. A subset of these scavengers are shown to block the formation of LG adducts of proteins in cells without inhibiting the catalytic activity of the cyclooxygenases. Ten of these selective scavengers did not produce cytotoxicity. These results demonstrate that small molecules can scavenge LGs in cells without interfering with the formation of prostaglandins. They also provide a working hypothesis for the development of pharmacologic agents that could be used in experimental animals in vivo to assess the pathophysiological contribution of levuglandins in diseases associated with cyclooxygenase up-regulation.
Collapse
Affiliation(s)
- Irene Zagol-Ikapitte
- Departments of Pharmacology, Pathology, and Medicine, Vanderbilt University, Nashville, Tennessee 37232-6602, USA
| | | | | | | | | | | |
Collapse
|
48
|
Determination of the Pharmacokinetics and Oral Bioavailability of Salicylamine, a Potent γ-Ketoaldehyde Scavenger, by LC/MS/MS. Pharmaceutics 2010; 2:18-29. [PMID: 21822464 PMCID: PMC3150493 DOI: 10.3390/pharmaceutics2010018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Levels of reactive γ-ketoaldehydes derived from arachidonate increase in diseases associated with inflammation and oxidative injury. To assess the biological importance of these γ-ketoaldehydes, we previously identified salicylamine as an effective γ-ketoaldehyde scavenger in vitro and in cells. To determine if salicylamine could be administered in vivo, we developed an LC/MS/MS assay to measure salicylamine in plasma and tissues. In mice, half-life (t1/2) was 62 minutes. Drinking water supplementation (1-10 g/L) generated tissue concentrations (10-500 μM) within the range previously shown to inhibit γ-ketoaldehydes in cells. Therefore, oral administration of salicylamine can be used to assess the contribution of γ-ketoaldehydes in animal models of disease.
Collapse
|
49
|
Selective gamma-ketoaldehyde scavengers protect Nav1.5 from oxidant-induced inactivation. J Mol Cell Cardiol 2009; 48:352-9. [PMID: 19962379 DOI: 10.1016/j.yjmcc.2009.11.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 11/10/2009] [Accepted: 11/24/2009] [Indexed: 11/23/2022]
Abstract
The cardiac sodium channel (SCN5A, Na(V)1.5) is a key determinant of electrical impulse conduction in cardiac tissue. Acute myocardial infarction leads to diminished sodium channel availability, both because of decreased channel expression and because of greater inactivation of channels already present. Myocardial infarction leads to significant increases in reactive oxygen species and their downstream effectors including lipoxidation products. The effects of reactive oxygen species on Na(V)1.5 function in whole hearts can be modeled in cultured myocytes, where oxidants shift the availability curve of I(Na) to hyperpolarized potentials, decreasing cardiac sodium current at the normal activation threshold. We recently examined potential mediators of the oxidant-induced inactivation and found that one specific lipoxidation product, the isoketals, recapitulated the effects of oxidant on sodium currents. Isoketals are highly reactive gamma-ketoaldehydes formed by the peroxidation of arachidonic acid that covalently modify the lysine residues of proteins. We now confirm that exposure to oxidants induces lipoxidative modification of Na(V)1.5 and that the selective isoketal scavengers block voltage-dependent changes in sodium current by the oxidant tert-butylhydroperoxide, both in cells heterologously expressing Na(V)1.5 and in a mouse cardiac myocyte cell line (HL-1). Thus, inhibition of this lipoxidative modification pathway is sufficient to protect the sodium channel from oxidant induced inactivation and suggests the potential use of isoketal scavengers as novel therapeutics to prevent arrhythmogenesis during myocardial infarction.
Collapse
|
50
|
Sullivan CB, Matafonova E, Roberts LJ, Amarnath V, Davies SS. Isoketals form cytotoxic phosphatidylethanolamine adducts in cells. J Lipid Res 2009; 51:999-1009. [PMID: 19965577 DOI: 10.1194/jlr.m001040] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Levuglandins and their stereo- and regio-isomers (termed isolevuglandins or isoketals) are gamma-ketoaldehydes (IsoK) that rapidly react with lysines to form stable protein adducts. IsoK protein adduct levels increase in several pathological conditions including cardiovascular disease. IsoKs can induce ion channel dysfunction and cell death, potentially by adducting to cellular proteins. However, IsoKs also adduct to phosphatidylethanolamine (PE) in vitro, and whether PE adducts form in cells or contribute to the effects of IsoKs is unknown. When radiolabeled IsoK was added to HEK293 cells, 40% of the radiolabel extracted into the chloroform lower phase suggesting the possible formation of PE adducts. We therefore developed methods to measure IsoK-PE adducts in cells. IsoK-PE was quantified by LC/MS/MS after hydrolysis to IsoK-ethanolamine by Streptomyces chromofuscus phospholipase D. In HEK293 and human umbilical vein endothelial cells (HUVEC), IsoK dose-dependently increased PE adduct concentrations to a greater extent than protein adduct. To test the biological significance of IsoK-PE formation, we treated HUVEC with IsoK-PE. IsoK-PE dose dependently induced cytotoxicity (LC(50) 2.2 muM). These results indicate that cellular PE is a significant target of IsoKs, and that formation of PE adducts may mediate some of the biological effects of IsoKs relevant to disease.
Collapse
Affiliation(s)
- C Blake Sullivan
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | | | | | | | | |
Collapse
|